Indian Journal of Human Genetics
Home Current Issue Archives Guidelines Subscriptions e-Alerts Login 
Users online: 36
Print this page  Email this page Small font sizeDefault font sizeIncrease font size


 
            Table of Contents  
REVIEW ARTICLE
Year : 2011  |  Volume : 17  |  Issue : 4  |  Page : 22-26
 

Human immunodeficiency virus therapeutics and pharmacogenomics


National Institute of Immunohaematology, 13th floor, KEM Hospital, Parel, Mumbai, Maharashtra, India

Date of Web Publication3-May-2011

Correspondence Address:
U Shankarkumar
National Institute of Immunohaematology, 13th Floor, KEM Hospital, Parel, Mumbai - 400012, Maharashtra
India
Login to access the Email id

Source of Support: None, Conflict of Interest: None


DOI: 10.4103/0971-6866.80354

Get Permissions

 

   Abstract 

Pharmacogenomics and pharmacogenetics are promising in development of a personalized treatment approach They are of paramount importance for basic immunology, for peptide based vaccine design (vaccinomics) drug monitoring in clinical setting and molecular pathophysiology of multifactorial diseases like cancer, tuberculosis, cardiac disorders, diabetes, asthma, HIV, etc



How to cite this article:
Shankarkumar U, Shankarkumar A, Ghosh K. Human immunodeficiency virus therapeutics and pharmacogenomics. Indian J Hum Genet 2011;17, Suppl S1:22-6

How to cite this URL:
Shankarkumar U, Shankarkumar A, Ghosh K. Human immunodeficiency virus therapeutics and pharmacogenomics. Indian J Hum Genet [serial online] 2011 [cited 2016 May 13];17, Suppl S1:22-6. Available from: http://www.ijhg.com/text.asp?2011/17/4/22/80354


Pharmacogenomics is the branch of Pharmacology which deals with the influence of genetic variation on drug response in diseased person studied by correlating gene expression and/or single-nucleotide polymorphism (SNPs), with drug toxicity and efficacy. Phamacogenomics uses whole genome wide sequencing or target oriented sequencing to identify single gene interaction with drugs. It has been extensively studied in patients of cancer, tuberculosis, cardiac disorders, diabetes, asthma and HIV.

AIDS offers the greatest threat to humans of all infectious diseases in history. To its challenge, more than 25 FDA approved antiretroviral drugs are available for clinical use which target HIV reverse transcriptase, protease, or viral entry receptors. The lifelong administration of multiple drugs necessiates constant monitoring of drug efficacy. Though these medications significantly reduce AIDS-related mortality [1] but their efficacy is not only compromised by their toxicity, viral resistance, and nonadherence to treatment, but also by comorbidities lik hepatitis, diabetes, and cardiovascula disease.

The field of pharmacogenomics strive to understand relationship between human genetic variations and response to treatment. [2-6] 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 to explore new targets for therapeutic intervention.

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. [7],[8],[9] 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. [10],[11],[12] This experiment of nature suggested that CCR5 inhibitors could be effectiv antiretroviral agents, and several CCR5 inhibitors are now under clinical trials. Other cellular factors which restrict HIV replication are: mRNA-editing enzyme 'apolipoprotein B', a catalytic polypeptide '3G (APOBEC3G) [13] and tripartite motif '5a (TRIM5a)'. [14] The naturally occurring variants in these and associated genes which affect progression of HIV disease are potential intervention targets.

Progress in pharmacogenomics require access to DNA specimens from large, well-characterized patient population by genetic investigators. 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 136,000 individuals into diverse prospective trials with 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 prospective study on stored DNA wher informed consent was obtained for other AACTG trials. [15] One challenge for the identification of genetic associations in cohort studies is to define control group with all relevant factors except the phenotype. [16]

Antiretroviral treatment is characterized by differential rates of adverse events and responses in seropositive individuals. Genetic variations between human beings are the major cause for this variablity. A number of associations of genetic variants with predisposition to drug adversities are well characterized, like hypersensitivity to abacavir. Although the drug is generally well tolerated, 5%-9% of Caucasians who receive abacavir experience hypersensitivity reactions proove life threatening unless intervened. Two research groups independently reported an association between major histocompatibility complex alleles and hypersensitivity to abacavir. [17],[18] In patients exposed to abacavir in Perth, Australia, the presence of HLA-B*5701, HLA-DR7, and HLADQ3 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 [19] HLA Class II allele DRB1*0101 has been associated with Nevirepine-associted hypersentivity. [20] HLA B*3505 allele has been a strong predictor for neverpine-induced skin adverse drug reactions in Thai HIV patients. [21] In nevirapine induced rash HIV-1 positive infected individuals from Mumbai, India a highly significant association with HLA B35 an protection with HLA B7 is found. [22]

The nonnucleoside reverse-transcriptase inhibitor efavirenz is one of the most widely prescribed antiretroviral medications [23],[24] but many recipients of efavirenz experience central nervous system side effects during the initial weeks of therapy. [24] 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 [26],[27],[28],[29] as have functional differences between genetic variants. [28],[30],[31],[32] 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.000)) and with increased central nervous system side effects (P p.036). [33] Differences in the frequency of this polymorphism in different populations may explain the lower clearance of efavirenz noted in blacks. [34],[35],[36] Recently the importance of human CYP3A pharmacogenetics with the discovery of the Null allelle CYP3A4*20 have contributed in predicting efficacy and/or toxicity in HIV patients. [37] The CYP3A4*1B polymorphism influences the pharmacokinetics of indinavir and to some extent its biochemical safety. [38]

Tumor necrosis factor (TNF)-a has been implicated in the pathogenesis of lipodystroph [39],[40],[41] and TNF-a expression varies according to race and ethnicity. [42] Two research groups have reported relationships between antiretroviral-associated lipodystrophy and a TNF-a promoter polymorphism that may affect gene expression. In 96 white patients in England, a TNF-α position 238 polymorphism was present only in subjects with lipodystrophy (P p .0)). [43],[44] These findings support a role for TNF-a 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 UDPglucuronosyltransferase (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. The HIV protease inhibitors are substrates for P-glycoprotein, the multidrug efflux pump encoded by MDR1 [47] and a frequent MDR1 exon 26 polymorphism has been associated with altered P-glycoprotein expression. [48] 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, [49] and its expression in the brain limits entry of protease inhibitors. 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. [50]

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. Such approaches promise advent of "personalized medicine" in which drugs and drug combinations are optimized for each patients individual genetic makeup.

 
   References Top

1.Palella FJ Jr, Delaney KM, Moorman AC, Loveless MO, Fuhrer J, Satten GAJ et al. Declining morbidity and mortality among patients with advanced human immunodeficiency virus infection. HIV Outpatient Study Investigators. N Engl J Med 1998;338:853-60.   Back to cited text no. 1
    
2.Evans WE, Relling MV. Pharmacogenomics: Translating functional genomics into rational therapeutics. Science 1999;286:487-91.   Back to cited text no. 2
    
3.Sachidanandam R, Weissman D, Schmidt SC, Kakol JM, Stein LD, Marth G, et al. A map of human genome sequence variation containing 1.42 million single nucleotide polymorphisms. Nature 2001;409:928-33.   Back to cited text no. 3
    
4.Venter JC, Adams MD, Myers EW, Li PW, Mural RJ, Sutton GG, et al. The sequence of the human genome. Science 2001;291:1304-51.   Back to cited text no. 4
    
5.Syvanen AC. Accessing genetic variation: Genotyping single nucleotide polymorphisms. Nat Rev Genet 2001;2:930-42.   Back to cited text no. 5
    
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;286:2270-9.   Back to cited text no. 6
    
7.Deng H, Liu R, Ellmeier W, Choe S, Unutmaz D, Burkhart M, et al. Identification of a major co-receptor for primary isolates of HIV-1. Nature 1996;381:661-6.   Back to cited text no. 7
    
8.Dragic T, Litwin V, Allaway GP, Martin SR, Huang Y, Nagashima KA, et al. HIV-1 entry into CD4+ cells is mediated by the chemokine receptor CC-CKR-5. Nature 1996;381:667-73.   Back to cited text no. 8
    
9.Choe H, Farzan M, Sun Y, Sullivan N, Rollins B, Ponath PD, et al. The b-chemokine receptors CCR3 and CCR5 facilitate infection by primary HIV-1 isolates. Cell 1996;85:1135-48.   Back to cited text no. 9
    
10.Liu R, Paxton WA, Choe S, Ceradini D, Martin SR, Horuk R, 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.   Back to cited text no. 10
    
11.Samson M, Libert F, Doranz BJ, Rucker J, Liesnard C, Farber CM, 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.   Back to cited text no. 11
    
12.Dean M, Carrington M, Winkler C, Huttley GA, Smith MW, Allikmets R, 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;273:1856-62.   Back to cited text no. 12
    
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;424:99-103.   Back to cited text no. 13
    
14.Stremlau M, Owens CM, Perron MJ, Kiessling M, Autissier P, Sodroski J. The cytoplasmic body component TRIM5a restricts HIV-1 infection in Old World monkeys. Nature 2004;427:848-53.   Back to cited text no. 14
    
15.Haas DW, Wilkinson GR, Kuritzkes DR, Richman DD, Nicotera J, Mahon LFR, et al. A multi-investigator/institutional DNA bank for AIDS-related human genetic studies: AACTG protocol A5128. HIV Clin Trials 2003;4:287-300.   Back to cited text no. 15
    
16.Merikangas KR, Swendsen JD. Genetic epidemiology of psychiatric disorders. Epidemiol Rev 1997;19:144-55.   Back to cited text no. 16
    
17.Mallal S, Nolan D, Witt C, Masel G, Martin AM, Moore C, et al. Association between presence of HLA-B5701, HLA-DR7, and HLA-DQ3 and hypersensitivity to HIV-1 reverse-transcriptase inhibitor abacavir. Lancet 2002;359:727-32.   Back to cited text no. 17
    
18.Hetherington S, Hughes AR, Mosteller M, Shortino D, Baker KL, Spreen W, et al. Genetic variations in HLA-B region and hypersensitivity reactions to abacavir. Lancet 2002;359:1121-2.   Back to cited text no. 18
    
19.Martin AM, Nolan D, Gaudieri S, Almeida CA, Nolan R, James I, et al. Predisposition to abacavir hypersensitivity conferred by HLA-B5701 and a haplotypic Hsp70Hom variant. Proc Natl Acad Sci U S A 2004;101:4180-5.   Back to cited text no. 19
    
20.Martin et al 2005 Martin AM, Nolan D, James I, Cameron P, Keller J, Moore C,Set al. Predisposition to nevirapine hypersensitivity associated with HLA -DRB10101 and abrogated by CD4 T cell counts. AIDS 2005;1:979-9.  Back to cited text no. 20
    
21.Chantarangsu S, Mushiroda T, Mahasirimongkol S, Kiertiburanakul S, Sungkanuparph, Manosuthi W,Yet al. HLA B3505 allele is a strong predictor for nevirapine-induced skin adverse reactions in HIV-infected Thai patients. Pharmacogene. Genomics 2009;19:139-46.  Back to cited text no. 21
    
22.Shankarkumar U,.Pawar A, Bajpai S, Pazare AR, Ghosh K. HLA involvement in nevirapine-induced skin adverse drug reaction in ARV treated HIV-1 patients from Mumbai, Maharashtra, Western India. J Pharmacol Pharmacother 2011 in press.  Back to cited text no. 22
    
23.Staszewski S, Morales-Ramirez J, Tashima KT, Rachlis A, Skiest D, Stanford J, 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;341:1865-73.   Back to cited text no. 23
    
24.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-1-infected patients. AIDS 2001;15:71-5.   Back to cited text no. 24
    
25.Efavirenz prescribing information. Physician's desk reference. 57 th ed. Montvale, NJ: Thompson's PD ; 200:. p. 1088-94.   Back to cited text no. 25
    
26.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;306:287-300.   Back to cited text no. 26
    
27.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;270:414-23.   Back to cited text no. 27
    
28.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;25:985-93.   Back to cited text no. 28
    
29.Lang T, Klein K, Fischer J, Nüssler AK, Neuhaus P, Hofmann U, et al. Extensive genetic polymorphism in the human CYP2B6 gene with impact on expression and function in human liver. Pharmacogenetics 2001;11:399-415.   Back to cited text no. 29
    
30.Lamba V, Lamba J, Yasuda K, Strom S, Davila J, Hancock ML, et al. Hepatic CYP2B6 expression: Gender and ethnic differences and relationship to CYP2B6 genotype and CAR expression. J Pharmacol Exp Ther 2003;307:906-22.   Back to cited text no. 30
    
31.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;281:1256-60.   Back to cited text no. 31
    
32.Jinno H, Tanaka-Kagawa T, Ohno A, Makino Y, Matsushima E, Hanioka NA, et al. Functional characterization of cytochrome P450 2B6 allelic variants. Drug Metab Dispos 2003;31:398-403.   Back to cited text no. 32
    
33.Kirchheiner J, Klein C, Meineke I, Sasse J, Zanger UM, Mürdter TE, et al. Bupropion and 4-OH-bupropion pharmacokinetics in relation to genetic polymorphisms in CYP2B6. Pharmacogenetics 2003;13:619-26.   Back to cited text no. 33
    
34.Haas DW, Ribaudo HJ, Kim RB, Tierney C, Wilkinson GR, Gulick RM, et al. Pharmacogenetics of efavirenz and central nervous system side effects: An Adult AIDS Clinical Trials Group study. AIDS 2004;18:2391-400.   Back to cited text no. 34
    
35.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;40:507-19.   Back to cited text no. 35
    
36.Pfister M, Labbé L, Hammer SM, Mellors J, Bennett KK, Rosenkranz S, et al. Population pharmacokinetics and pharmacodynamics of efavirenz, nelfinavir, and indinavir: Adult AIDS Clinical Trial Group Study 398. Antimicrob Agents Chemother 2003;47:130-7.   Back to cited text no. 36
    
37.Lakhman SS, Maa Q, Morse GD. Pharmacogenomics of CYP3A: Considerations for HIV treatment. Pharmacogenomics 2009;10:1323-39.  Back to cited text no. 37
    
38.Bertrand J, Trehuyer JM, Panhard X, Tran A, Auleley S, Rey E, Fet al,; COPHAR2-ANRS 111 Study group. Influence of pharmacogenetics on indinavir disposition and short-term response in HIV patients initiating HAART. Eur J Cli Pharmaco 2009;65:667-78.  Back to cited text no. 38
    
39.Bastard JP, Caron M, Vidal H, Jan V, Auclair M, Vigouroux C, et al. Association between altered expression of adipogenic factor SREBP1 in lipoatrophic adipose tissue from HIV-1-infected patients and abnormal adipocyte differentiation and insulin resistance. Lancet 2002;359:1026-31.   Back to cited text no. 39
    
40.Sethi JK, Hotamisligil GS. The role of TNF-a in adipocyte metabolism. Semin Cell Dev Biol 1999;10:19-29.   Back to cited text no. 40
    
41.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;14:1309-16.   Back to cited text no. 41
    
42.Hajeer AH, Hutchinson IV. Influence of TNF-a gene polymorphisms on TNF-a production and disease. Hum Immunol 2001;62:1191-9.   Back to cited text no. 42
    
43.Maher B, Alfirevic A, Vilar FJ, Wilkins EG, Park BK, Pirmohamed M. TNF-a promoter region gene polymorphisms in HIV-positive patients with lipodystrophy. AIDS 2002;16:2013-8.   Back to cited text no. 43
    
44.Nolan D, Moore C, Castley A, Sayer D, Mamotte C, John MA, et al. Tumour necrosis factor-a gene 238G/A promoter polymorphism associated with a more rapid onset of lipodystrophy. AIDS 2003;17:121-3.   Back to cited text no. 44
    
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 U S A 1998;95:8170-4.   Back to cited text no. 45
    
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;33:348-51.   Back to cited text no. 46
    
47.Lee CG, Gottesman MM. HIV-1 protease inhibitors and the MDR1 multidrug transporter. J Clin Invest 1998;101:287-8.   Back to cited text no. 47
    
48.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;42:3157-62.   Back to cited text no. 48
    
49.Polli JW, Jarrett JL, Studenberg SD, Humphreys JE, Dennis SW, Brouwer KR, et al. Role of P-glycoprotein on the CNS disposition of amprenavir (141W94), an HIV protease inhibitor. Pharm Res 1999;16:1206-12.   Back to cited text no. 49
    
50.Nasi M, Borghi V, Pinti M, Bellodi C, Lugli E, Maffei S, et al. MDR1 C3435T genetic polymorphism does not influence the response to antiretroviral therapy in drug-naive HIV-positive patients. AIDS 2003;17:1696-8.  Back to cited text no. 50
    




 

Top
Print this article  Email this article
           

    

 
   Search
 
  
    Similar in PUBMED
   Search Pubmed for
   Search in Google Scholar for
    Article in PDF (221 KB)
    Citation Manager
    Access Statistics
    Reader Comments
    Email Alert *
    Add to My List *
* Registration required (free)  


    Abstract
    References

 Article Access Statistics
    Viewed1778    
    Printed142    
    Emailed0    
    PDF Downloaded78    
    Comments [Add]    

Recommend this journal