Literature
Home医源资料库在线期刊传染病学杂志2005年第191卷第9期

Pharmacogenomics and HIV Therapeutics

来源:传染病学杂志
摘要:Inresponsetothischallenge,atleast19distinctantiretroviralagentsthattargetHIVreversetranscriptase,protease,orviralentryhavebeenapprovedforclinicaluse。Suchabroadarmamentariumiscritical,giventhenatureofantiretroviraltherapy,whichrequiresthelifelongadministr......

点击显示 收起

    Departments of Medicine, Microbiology, and Immunology, and Center for Human Genetics Research, Vanderbilt University School of Medicine, Nashville, Tennessee

    AIDS offers perhaps the greatest threat to humans by any infectious disease in history. In response to this challenge, at least 19 distinct antiretroviral agents that target HIV reverse transcriptase, protease, or viral entry have been approved for clinical use. Such a broad armamentarium is critical, given the nature of antiretroviral therapy, which requires the lifelong administration of multiple drugs. Use of these medications greatly reduces AIDS-related mortality [1], but their efficacy is often compromised by their toxicity, viral resistance, and incomplete adherence to treatment, as well as by comorbidities, such as viral hepatitis, diabetes, and cardiovascular disease.

    The field of pharmacogenomics strives to understand relationships between human genetic variation and responses to treatment [26]. The relevance of pharmacogenomics to HIV therapeutics spans basic science, patient care, and public health disciplines. Laboratory-based investigators use genomic techniques to study viral pathogenesis in the hope of identifying new cellular targets for therapeutic intervention. Standard HIV clinical practice may soon include human genetic testing to help individualize antiretroviral therapy. From a public health perspective, as antiretroviral medications become increasingly available to racially and ethnically diverse populations worldwide, understanding the genetic structures of each population may allow us to anticipate the impact of adverse responses, even in groups that were not represented in drug registration trials.

    The potential for human genetic research to identify novel therapeutic targets is highlighted by previous studies of CCR5. This cellular chemokine receptor is required for infectivity of many HIV strains [79]. Soon after its role in HIV replication was elucidated, individuals were identified who were highly resistant to HIV infection and lacked functional CCR5 as the result of a 32-bp deletion in the CCR5 gene but were otherwise healthy [1012]. This experiment of nature suggested that CCR5 inhibitors could be particularly attractive antiretroviral agents, and several CCR5 inhibitors are now being studied in clinical trials. As other cellular factors that restrict HIV replication, such as apolipoprotein B mRNA-editing enzyme, catalytic polypeptidelike 3G (APOBEC3G) [13] and tripartite motif 5 (TRIM5) [14], are discovered, the identification of naturally occurring variants in these and associated genes that influence the progression of HIV disease may suggest additional new targets for drug development.

    Progress in pharmacogenomics requires that accomplished genomic investigators have access to DNA specimens from large, well-characterized patient populations. DNA banks that are associated with clinical trials and cohorts must therefore be established. The Adult AIDS Clinical Trials Group (AACTG), funded by the National Institutes of Health, has created an important repository. Since 1986, the AACTG has enrolled >36,000 individuals into diverse prospective trials that have well-defined entry criteria and on-study evaluations. To establish a usable DNA bank, a group of clinical researchers, genetic investigators, ethicists, statisticians, data managers, regulatory specialists, and community representatives worked in collaboration to develop AACTG Protocol A5128, which allows the use of stored DNA for studies that were not planned when informed consent was provided for other AACTG trials [15].

    Since 2001, 7500 different participants in these clinical trials19% female, 51% white, 28% black, and 18% Hispanichave contributed specimens to the AACTG Human DNA Repository under A5128, and accrual is ongoing. Because its specimens have an average yield of 400 g of extracted DNA, this repository can support an almost unlimited number of projects. One challenge for the identification of genetic associations in cohort studies is to define control groups that share all relevant factors except phenotype [16]. Because most AACTG studies are randomized, approximately equal numbers of subjects with a given genotype are likely to be assigned to different treatments. The AACTG encourages proposals to utilize this resource for studies relevant to HIV infection and its complications, and a well-established procedure for the consideration of such proposals is in place.

    In this issue of the Journal of Infectious Diseases, Tarr et al. describe an association between variant alleles of APOE and APOC3 and protease inhibitorassociated hyperlipidemia in participants in the Swiss HIV Cohort Study [17]. Their study expands the number of putative associations between human genetic variants and responses to antiretroviral therapy.

    To date, the most impressive such association regards hypersensitivity to abacavir. Although the drug is generally well tolerated, 5%9% of whites who receive abacavir experience hypersensitivity reactions that can be life threatening. Two research groups independently reported an association between major histocompatibility complex alleles and hypersensitivity to abacavir [18, 19]. In patients exposed to abacavir in Perth, Australia, the presence of HLA-B*5701, HLA-DR7, and HLA-DQ3 had a positive predictive value of 100% and a negative predictive value of 97% for hypersensitivity [18]. An association between hypersensitivity to abacavir and HLA-B*5701 and HLA-DR7 was confirmed in patients in North America [19]. More-recent analyses have extended this association to include a polymorphism in Hsp70-Hom, a member of the heat shock protein family of chaperonins [20]. In western Australia, the routine screening of patients' genetic makeup before they are prescribed abacavir has markedly reduced the incidence of hypersensitivity reactions. Such screening is rarely performed in the United States.

    The nonnucleoside reverse-transcriptase inhibitor efavirenz is one of the most widely prescribed antiretroviral medications [21, 22], but many recipients of efavirenz experience central nervous system side effects during the initial weeks of therapy [2224]. Efavirenz is metabolized primarily by hepatic cytochrome P450 (CYP) 2B6 [25], and a large amount of interindividual variability in the amount of CYP2B6 in the liver has been reported [2629], as have functional differences between genetic variants [28, 3032]. Specimens from the AACTG Human DNA Repository and associated data from clinical trials were used to show that a CYP2B6 exon 4 polymorphism that occurs more frequently in blacks than in whites is associated with 3-fold higher plasma concentrations of efavirenz (P < .0001) and with increased central nervous system side effects (P = .036) [33]. Differences in the frequency of this polymorphism in different populations may explain the lower clearance of efavirenz noted in blacks [3436]. Additional studies are needed to assess the implications for long-term responses to efavirenz, as well as to nevirapine, which is also metabolized by CYP2B6 [37, 38].

    Peripheral fat wasting and central fat deposition often complicate antiretroviral therapy. Independent risk factors for antiretroviral-associated lipoatrophy include white race and prolonged exposure to nucleoside analogues, particularly stavudine [39]. Tumor necrosis factor (TNF) has been implicated in the pathogenesis of lipodystrophy [40, 41], and TNF- expression varies according to race and ethnicity [42]. At least 2 research groups have reported relationships between antiretroviral-associated lipodystrophy and a TNF- promoter polymorphism that may affect gene expression. In 96 white patients in England, a TNF- position -238 polymorphism was present only in subjects with lipodystrophy (P = .01) [43]. Similarly, in 191 white patients in Australia, all of whom had lipoatrophy, this polymorphism was associated with a more-rapid onset of fat wasting (P = .014) [44]. Although these findings support a role for TNF- in the pathogenesis of lipoatrophy, this variant allele may simply be a marker for other genes with which it is linked, such as members of the major histocompatibility complex [42].

    Bilirubin is the primary product of heme metabolism. Its efficient elimination requires conjugation with glucuronic acid in a reaction catalyzed by hepatic UDP-glucuronosyltransferase (UGT) 1A1. Approximately 5%10% of individuals have decreased bilirubin-conjugating activity that is caused by a TA insertion into the UGT1A1 promoter (Gilbert syndrome) [45, 46]. The HIV protease inhibitors indinavir and atazanavir commonly cause unconjugated hyperbilirubinemia by competing with bilirubin for binding to UGT1A1. Although the condition is usually asymptomatic, some patients discontinue treatment with these drugs because of jaundice. In a study of 15 HIV-positive men receiving indinavir, mean increases in serum bilirubin levels were significantly greater in patients with at least 1 Gilbert polymorphism (P = .012) [47]. Similarly, of 138 healthy volunteers who participated in phase 1 studies of atazanavir, those homozygous for the Gilbert genotype had significantly higher median total bilirubin levels than did heterozygous or wild-type subjects (P = .0001) [48]. Fortunately, not all patients with Gilbert syndrome who receive atazanavir or indinavir experience marked elevations in bilirubin levels.

    The HIV protease inhibitors are substrates for P-glycoprotein, the multidrug efflux pump encoded by MDR1 [4954], and a frequent MDR1 exon 26 polymorphism has been associated with altered P-glycoprotein expression [55]. P-glycoprotein in the intestine, liver, and kidney is predicted to decrease oral bioavailability of these drugs and enhance their elimination. P-glycoprotein is also present in CD4 T cells [56], and its expression in the brain limits entry of protease inhibitors [51]. Importantly, a provocative report noted an association between the MDR1 exon 26 polymorphism, increases in CD4 T cells in response to antiretroviral therapy, and plasma concentrations of efavirenz and nelfinavir [57]. Subsequent studies, however, have not confirmed these findings [33, 58, 59]. Genetic associations suggested by initial studies are often not confirmed by subsequent analyses [60], and the relevance of MDR1 polymorphisms for HIV therapeutics remains uncertain.

    As pharmacogenomics moves from bench to bedside, most genotype-phenotype relationships will reflect the combined influences of multiple genes and polymorphisms. The growing number of identified genetic associations will increase the impetus to make human genetic testing a routine part of HIV clinical care. Prospective clinical trials will ultimately be needed to determine whether the use of human genetic testing to guide the administration of antiretroviral therapy results in an improved response to treatment. Because genetic variants are stable throughout one's lifetime, genetic testing performed on a single occasion could potentially inform every subsequent treatment decision for a patient, and this makes such an approach to HIV clinical care even more attractive.

    References

    1.  Palella FJJ, Delaney KM, Moorman AC, et al. Declining morbidity and mortality among patients with advanced human immunodeficiency virus infection. HIV Outpatient Study Investigators. N Engl J Med 1998; 338:85360. First citation in article

    2.  Evans WE, Relling MV. Pharmacogenomics: translating functional genomics into rational therapeutics. Science 1999; 286:48791. First citation in article

    3.  Sachidanandam R, Weissman D, Schmidt SC, et al. A map of human genome sequence variation containing 1.42 million single nucleotide polymorphisms. Nature 2001; 409:92833. First citation in article

    4.  Venter JC, Adams MD, Myers EW, et al. The sequence of the human genome. Science 2001; 291:130451. First citation in article

    5.  Syvanen AC. Accessing genetic variation: genotyping single nucleotide polymorphisms. Nat Rev Genet 2001; 2:93042. First citation in article

    6.  Phillips KA, Veenstra DL, Oren E, Lee JK, Sadee W. Potential role of pharmacogenomics in reducing adverse drug reactions: a systematic review. JAMA 2001; 286:22709. First citation in article

    7.  Deng H, Liu R, Ellmeier W, et al. Identification of a major co-receptor for primary isolates of HIV-1. Nature 1996; 381:6616. First citation in article

    8.  Dragic T, Litwin V, Allaway GP, et al. HIV-1 entry into CD4+ cells is mediated by the chemokine receptor CC-CKR-5. Nature 1996; 381:66773. First citation in article

    9.  Choe H, Farzan M, Sun Y, et al. The -chemokine receptors CCR3 and CCR5 facilitate infection by primary HIV-1 isolates. Cell 1996; 85:113548. First citation in article

    10.  Liu R, Paxton WA, Choe S, et al. Homozygous defect in HIV-1 coreceptor accounts for resistance of some multiply-exposed individuals to HIV-1 infection. Cell 1996; 86:36777. First citation in article

    11.  Samson M, Libert F, Doranz BJ, et al. Resistance to HIV-1 infection in caucasian individuals bearing mutant alleles of the CCR-5 chemokine receptor gene. Nature 1996; 382:7225. First citation in article

    12.  Dean M, Carrington M, Winkler C, et al. Genetic restriction of HIV-1 infection and progression to AIDS by a deletion allele of the CKR5 structural gene. Hemophilia Growth and Development Study, Multicenter AIDS Cohort Study, Multicenter Hemophilia Cohort Study, San Francisco City Cohort, ALIVE Study. Science 1996; 273:185662. First citation in article

    13.  Mangeat B, Turelli P, Caron G, Friedli M, Perrin L, Trono D. Broad antiretroviral defence by human APOBEC3G through lethal editing of nascent reverse transcripts. Nature 2003; 424:99103. First citation in article

    14.  Stremlau M, Owens CM, Perron MJ, Kiessling M, Autissier P, Sodroski J. The cytoplasmic body component TRIM5 restricts HIV-1 infection in Old World monkeys. Nature 2004; 427:84853. First citation in article

    15.  Haas DW, Wilkinson GR, Kuritzkes DR, et al. A multi-investigator/institutional DNA bank for AIDS-related human genetic studies: AACTG protocol A5128. HIV Clin Trials 2003; 4:287300. First citation in article

    16.  Merikangas KR, Swendsen JD. Genetic epidemiology of psychiatric disorders. Epidemiol Rev 1997; 19:14455. First citation in article

    17.  Tarr PE, Taffé P, Bleiber G, et al. Modeling the influence of APOC3, APOE, and TNF polymorphisms on the risk of antiretroviral therapyassociated lipid disorders. J Infect Dis 2005; 191:14207 (in this issue). First citation in article

    18.  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; 359:72732. First citation in article

    19.  Hetherington S, Hughes AR, Mosteller M, et al. Genetic variations in HLA-B region and hypersensitivity reactions to abacavir. Lancet 2002; 359:11212. First citation in article

    20.  Martin AM, Nolan D, Gaudieri S, et al. Predisposition to abacavir hypersensitivity conferred by HLA-B*5701 and a haplotypic Hsp70-Hom variant. Proc Natl Acad Sci USA 2004; 101:41805. First citation in article

    21.  The Panel on Clinical Practices for Treatment of HIV Infection. Guidelines for the use of antiretroviral agents in HIV-1infected adults and adolescents. Available at: http://www.aidsinfo.nih.gov/guidelines. Accessed 14 March 2005. First citation in article

    22.  Staszewski S, Morales-Ramirez J, Tashima KT, et al. Efavirenz plus zidovudine and lamivudine, efavirenz plus indinavir, and indinavir plus zidovudine and lamivudine in the treatment of HIV-1 infection in adults. Study 006 Team. N Engl J Med 1999; 341:186573. First citation in article

    23.  Marzolini C, Telenti A, Decosterd LA, Greub G, Biollaz J, Buclin T. Efavirenz plasma levels can predict treatment failure and central nervous system side effects in HIV-1infected patients. AIDS 2001; 15:715. First citation in article

    24.  Efavirenz prescribing information. Physician's desk reference. 57th ed. Montvale, NJ: Thompson's PDR, 2004:108894. First citation in article

    25.  Ward BA, Gorski JC, Jones DR, Hall SD, Flockhart DA, Desta Z. 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; 306:287300. First citation in article

    26.  Shimada T, Yamazaki H, Mimura M, Inui Y, Guengerich FP. Interindividual variations in human liver cytochrome P-450 enzymes involved in the oxidation of drugs, carcinogens and toxic chemicals: studies with liver microsomes of 30 Japanese and 30 Caucasians. J Pharmacol Exp Ther 1994; 270:41423. First citation in article

    27.  Code EL, Crespi CL, Penman BW, Gonzalez FJ, Chang TK, Waxman DJ. Human cytochrome P4502B6: interindividual hepatic expression, substrate specificity, and role in procarcinogen activation. Drug Metab Dispos 1997; 25:98593. First citation in article

    28.  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; 11:399415. First citation in article

    29.  Lamba V, Lamba J, Yasuda K, et al. Hepatic CYP2B6 expression: gender and ethnic differences and relationship to CYP2B6 genotype and CAR expression. J Pharmacol Exp Ther 2003; 307:90622. First citation in article

    30.  Ariyoshi N, Miyazaki M, Toide K, Sawamura Y, Kamataki T. A single nucleotide polymorphism of CYP2B6 found in Japanese enhances catalytic activity by autoactivation. Biochem Biophys Res Commun 2001; 281:125660. First citation in article

    31.  Jinno H, Tanaka-Kagawa T, Ohno A, et al. Functional characterization of cytochrome P450 2B6 allelic variants. Drug Metab Dispos 2003; 31:398403. First citation in article

    32.  Kirchheiner J, Klein C, Meineke I, et al. Bupropion and 4-OH-bupropion pharmacokinetics in relation to genetic polymorphisms in CYP2B6. Pharmacogenetics 2003; 13:61926. First citation in article

    33.  Haas DW, Ribaudo HJ, Kim RB, et al. Pharmacogenetics of efavirenz and central nervous system side effects: an Adult AIDS Clinical Trials Group study. AIDS 2004; 18:2391400. First citation in article

    34.  Barrett JS, Joshi AS, Chai M, Ludden TM, Fiske WD, Pieniaszek HJ Jr. Population pharmacokinetic meta-analysis with efavirenz. Int J Clin Pharmacol Ther 2002; 40:50719. First citation in article

    35.  Pfister M, Labbe L, Hammer SM, et al. Population pharmacokinetics and pharmacodynamics of efavirenz, nelfinavir, and indinavir: Adult AIDS Clinical Trial Group Study 398. Antimicrob Agents Chemother 2003; 47:1307. First citation in article

    36.  Ribaudo H, Clifford D, Gulick R, et al. Relationships between efavirenz pharmacokinetics, side effects, drug discontinuation, virologic response, and race: results from ACTG A5095/A5097s [abstract 132]. In: Program and abstracts of the 11th Conference on Retroviruses and Opportunistic Infections. Alexandria, VA: Foundation for Retrovirology and Human Health, 2004. First citation in article

    37.  Riska P, Lamson M, MacGregor T, et al. Disposition and biotransformation of the antiretroviral drug nevirapine in humans. Drug Metab Dispos 1999; 27:895901. First citation in article

    38.  Erickson DA, Mather G, Trager WF, Levy RH, Keirns JJ. Characterization of the in vitro biotransformation of the HIV-1 reverse transcriptase inhibitor nevirapine by human hepatic cytochromes P-450. Drug Metab Dispos 1999; 27:148895. First citation in article

    39.  Mallal SA, John M, Moore CB, James IR, McKinnon EJ. Contribution of nucleoside analogue reverse transcriptase inhibitors to subcutaneous fat wasting in patients with HIV infection. AIDS 2000; 14:130916. First citation in article

    40.  Bastard JP, Caron M, Vidal H, et al. Association between altered expression of adipogenic factor SREBP1 in lipoatrophic adipose tissue from HIV-1infected patients and abnormal adipocyte differentiation and insulin resistance. Lancet 2002; 359:102631. First citation in article

    41.  Sethi JK, Hotamisligil GS. The role of TNF in adipocyte metabolism. Semin Cell Dev Biol 1999; 10:1929. First citation in article

    42.  Hajeer AH, Hutchinson IV. Influence of TNF gene polymorphisms on TNF production and disease. Hum Immunol 2001; 62:11919. First citation in article

    43.  Maher B, Alfirevic A, Vilar FJ, Wilkins EG, Park BK, Pirmohamed M. TNF- promoter region gene polymorphisms in HIV-positive patients with lipodystrophy. AIDS 2002; 16:20138. First citation in article

    44.  Nolan D, Moore C, Castley A, et al. Tumour necrosis factor- gene -238G/A promoter polymorphism associated with a more rapid onset of lipodystrophy. AIDS 2003; 17:1213. First citation in article

    45.  Beutler E, Gelbart T, Demina A. Racial variability in the UDP-glucuronosyltransferase 1 (UGT1A1) promoter: a balanced polymorphism for regulation of bilirubin metabolism Proc Natl Acad Sci USA 1998; 95:81704. First citation in article

    46.  Raijmakers MT, Jansen PL, Steegers EA, Peters WH. Association of human liver bilirubin UDP-glucuronyltransferase activity with a polymorphism in the promoter region of the UGT1A1 gene. J Hepatol 2000; 33:34851. First citation in article

    47.  Zucker SD, Qin X, Rouster SD, et al. Mechanism of indinavir-induced hyperbilirubinemia. Proc Natl Acad Sci USA 2001; 98:126716. First citation in article

    48.  O'Mara E, Randall D, Passarell J, et al. Population pharmacodynamic assessment of atazanavir exposure, uridine diphosphate-glucuronosyl transferase (UGT) 1A1 genotype and safety in healthy subjects [abstract A-1253]. 42nd Interscience Conference on Antimicrobial Agents and Chemotherapy. San Diego, 2002. First citation in article

    49.  Lee CG, Gottesman MM. HIV-1 protease inhibitors and the MDR1 multidrug transporter. J Clin Invest 1998; 101:2878. First citation in article

    50.  Srinivas RV, Middlemas D, Flynn P, Fridland A. Human immunodeficiency virus protease inhibitors serve as substrates for multidrug transporter proteins MDR1 and MRP1 but retain antiviral efficacy in cell lines expressing these transporters. Antimicrob Agents Chemother 1998; 42:315762. First citation in article

    51.  Kim RB, Fromm MF, Wandel C, et al. The drug transporter P-glycoprotein limits oral absorption and brain entry of HIV-1 protease inhibitors. J Clin Invest 1998; 101:28994. First citation in article

    52.  Kim AE, Dintaman JM, Waddell DS, Silverman JA. Saquinavir, an HIV protease inhibitor, is transported by P-glycoprotein. J Pharmacol Exp Ther 1998; 286:143945. First citation in article

    53.  Polli JW, Jarrett JL, Studenberg SD, et al. Role of P-glycoprotein on the CNS disposition of amprenavir (141W94), an HIV protease inhibitor. Pharm Res 1999; 16:120612. First citation in article

    54.  Washington CB, Wiltshire HR, Man M, et al. The disposition of saquinavir in normal and P-glycoprotein deficient mice, rats, and in cultured cells. Drug Metab Dispos 2000; 28:105862. First citation in article

    55.  Marzolini C, Paus E, Buclin T, Kim RB. Polymorphisms in human MDR1 (P-glycoprotein): recent advances and clinical relevance. Clin Pharmacol Ther 2004; 75:1333. First citation in article

    56.  Chaudhary PM, Roninson IB. Expression and activity of P-glycoprotein, a multidrug efflux pump, in human hematopoietic stem cells. Cell 1991; 66:8594. First citation in article

    57.  Fellay J, Marzolini C, Meaden ER, et al. Response to antiretroviral treatment in HIV-1infected individuals with allelic variants of the multidrug resistance transporter 1: a pharmacogenetics study. Lancet 2002; 359:306. First citation in article

    58.  Nasi M, Borghi V, Pinti M, et al. MDR1 C3435T genetic polymorphism does not influence the response to antiretroviral therapy in drug-naive HIV-positive patients. AIDS 2003; 17:16968. First citation in article

    59.  Haas DW, Wu H, Li H, et al. MDR1 gene polymorphisms and phase 1 viral decay during HIV-1 infection: an adult AIDS Clinical Trials Group study. J Acquir Immune Defic Syndr 2003; 34:2958. First citation in article

    60.  Ioannidis JP, Ntzani EE, Trikalinos TA, Contopoulos-Ioannidis DG. Replication validity of genetic association studies. Nature Genetics 2001; 29:3069. First citation in article

作者: David W. Haas 2007-5-15
医学百科App—中西医基础知识学习工具
  • 相关内容
  • 近期更新
  • 热文榜
  • 医学百科App—健康测试工具