Pharmacokinetics and pharmacodynamics of the reverse transcriptase inhibitor tenofovir and prophylactic efficacy against HIV-1 infection

Sulav Duwal, Christof Schütte, Max von Kleist, Sulav Duwal, Christof Schütte, Max von Kleist

Abstract

Antiviral pre-exposure prophylaxis (PrEP) through daily drug administration can protect healthy individuals from HIV-1 infection. While PrEP was recently approved by the FDA, the potential long-term consequences of PrEP implementation remain entirely unclear. The aim of this study is to predict the efficacy of different prophylactic strategies with the pro-drug tenofovir-disoproxil-fumarate (TDF) and to assess the sensitivity towards timing- and mode of TDF administration (daily- vs. single dose), adherence and the number of transmitted viruses. We developed a pharmacokinetic model for TDF and its active anabolite tenofovir-diphosphate (TFV-DP) and validated it with data from 4 different trials, including 4 distinct dosing regimes. Pharmacokinetics were coupled to an HIV model and viral decay following TDF mono-therapy was predicted, consistent with available data. Subsequently, a stochastic approach was used to estimate the % infections prevented by (i) daily TDF-based PrEP, (ii) one week TDF started either shortly before, or -after viral exposure and (iii) a single dose oral TDF before viral challenge (sd-PrEP). Analytical solutions were derived to assess the relation between intracellular TFV-DP concentrations and prophylactic efficacy. The predicted efficacy of TDF was limited by a slow accumulation of active compound (TFV-DP) and variable TFV-DP half-life and decreased with increasing numbers of transmitted viruses. Once daily TDF-based PrEP yielded [Formula: see text]80% protection, if at least 40% of pills were taken. Sd-PrEP with 300 mg or 600 mg TDF could prevent [Formula: see text]50% infections, when given at least before virus exposure. The efficacy dropped to [Formula: see text]10%, when given 1 h before 24 h exposure. Efficacy could not be increased with increasing dosage or prolonged administration. Post-exposure prophylaxis poorly prevented infection. The use of drugs that accumulate more rapidly, or local application of tenofovir gel may overcome the need for drug administration long before virus exposure.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. Pharmacokinetic model of TFV and…
Figure 1. Pharmacokinetic model of TFV and intracellular TFV-DP and model of viral kinetics.
A: Pharmacokinetic model. Parameters and are the absorption and elimination rate constants of the central compartment C1 (which resembles plasma pharmacokinetics of TFV) respectively. The parameters and denote the influx and outflux rate constant to-/from the peripheral compartment C2 respectively. Both compartments (central-/peripheral-) have the same volume of distribution . The dotted line from the central compartment to the intracellular compartment C3 represents subsumed processes, namely the cellular uptake of TFV and subsequent phosphorylation to TFV-DP, which were related to the plasma concentration of TFV (C1) by Michaelis-Menten kinetics, with parameters and individual parameter . The parameter is the individual, cellular elimination rate constant of TFV-DP. B: Virus dynamics model. T-cell and macrophage target cells (, ) can become successfully infected by infective virus with lumped infection rate constants and , respectively, creating early infected cells and . Infection can also be unsuccessful after the irreversible step of fusion (rate constant and , dashed lines), eliminating the virus and rendering the cell uninfected. Early infected cells and can destroy essential viral proteins or DNA prior to integration with rate constants and (dashed lines) returning the cell to an uninfected stage. The genomic viral DNA can become integrated with rate constants and creating late infected cells and , which can release new infectious- and non infectious virus and with rate constants and , respectively. All cellular compartments can get destroyed by the immune system with respective rate constants and the free virus gets cleared with rate constant (thin dashed lines). The pharmacologically active form of tenofovir (tenofovir-diphosphate, TFV-DP, green box) inhibits successful cell-infection (parameter ) and increases the rate of unsuccessful infection (parameter ).
Figure 2. Pharmacokinetics of TFV for different…
Figure 2. Pharmacokinetics of TFV for different doses of oral TDF at plateau and intracellular TFV-DP concentrations after treatment cessation.
A: Predicted pharmacokinetics of TFV after once daily 75-, 150-, 300- and 600 mg oral TDF (lines) together with data from , , (markers). B: Goodness-of-fit plot for the plasma pharmacokinetics of TFV with data from 3 clinical studies and 4 different dosing schemes , , . The dashed red line indicates the line of unity, whereas the green squares, -diamonds, triangles and filled dots represent the observed TFV concentrations in following 75-, 150-, 300- or 600 mg once daily administration of TDF. The blue left-pointing triangles and the magenta right-pointing triangles represent observed TFV concentrations after 300 mg once daily oral administration from and respectively. C: Predicted pharmacokinetics of intracellular TFV-DP after stopping of 300 mg once daily oral TDF dosing (lines) together with data from (markers). D: Goodness-of-fit plot for intracellular TFV-DP. The up- and downward pointing filled and open triangles, open- and filled circles, filled squares and filled diamonds indicate intracellular TFV-DP pharmacokinetics after stopping 300 mg once daily oral TDF dosing in 8 different individuals from .
Figure 3. Predicted TFV-DP intracellular pharmacokinetics following…
Figure 3. Predicted TFV-DP intracellular pharmacokinetics following a single dose oral 300 mg TDF and accumulation of TFV-DP after daily 300 mg oral TDF.
A: Predicted intracellular pharmacokinetics of TFV-DP in PBMCs after a single 300 mg oral TDF dose. Solid black circle and horizontal error bar indicate the value and its range. B: Trough levels of TFV-DP in PBMCs following 300 mg oral TDF every 24hours, indicating the accumulation of active compound. The solid black circle and the horizontal error bar indicate the time until plateau concentrations are reached and the range for this parameter. Blue cirles, black squares, green diamonds, red downward pointing triangles, magenta upward-pointing triangles, cyan right-ward pointing triangles, black left-pointing triangles and blue asterisks indicate individual predictions for 8 patients.
Figure 4. Viral load log 10 kinetics…
Figure 4. Viral load log10 kinetics during- and after 28 days of TDF mono-therapy.
Black dashed vertical lines indicate the withdrawal of TDF dosing. Solid lines represent predicted median viral kinetics using the coupled PK-PD model, whereas dashed lines represent the observed viral kinetics . Once daily 75mg TDF dosing. B: Once daily 150 mg TDF dosing. C: Once daily 300 mg TDF dosing. D: Once daily 600 mg TDF dosing.
Figure 5. Predicted % infections prevented by…
Figure 5. Predicted % infections prevented by distinct TDF-based prophylactic strategies for various parameter sets.
A: Predicted % infections prevented by once daily 300 mg TDF taken at different levels of adherence and with distinct virus inoculum sizes. prophylactic efficacy depends on adherence at the p 0.05 or p 0.01 level respectively. B: Predicted % infections prevented by a one week 300 mg TDF (1w-PrEP/PEP) when started at distinct times before/after exposure with distinct numbers of viruses. prophylactic efficacy depends on the timing of start of TDF administration at the p 0.05 or p 0.01 level respectively. C: Predicted % infections prevented by a single dose 300 mg TDF (sd-PrEP) when taken at distinct times before exposure with distinct virus inoculum sizes. prophylactic efficacy depends on the timing of TDF single dose administration at the p 0.05 or p 0.01 level respectively. D: Predicted % infections prevented by a single dose 600 mg TDF (sd-PrEP) when taken at distinct times before exposure with distinct virus inoculum sizes. Error bars represent confidence bounds calculated using Greenwood’s formula. prophylactic efficacy depends on the inoculum size. The predicted probability of infection in the absence of drugs was , , and when  = 1, 5, 20 or 100, respectively, viruses were inoculated.
Figure 6. Predicted % infections prevented vs.…
Figure 6. Predicted % infections prevented vs. intracellular TFV-DP concentrations for distinct virus inoculum sizes.
The solid blue-, dash-dotted green, dashed red and dotted black lines show the concentration-response profile for virus inoculum size 1, 5, 20 and 100 respectively. The thick dashed horizontal black line indicates the TFV-DP concentration, which prevents 50% of infections (). The dark grey area indicates the TFV-DP concentration range achieved during once daily 300 mg oral TDF dosing with 100% adherence, whereas the light grey extension to the left indicates the range of concentrations resulting from imperfect adherence. Predictions are based on the approximate analytic solution derived in Text S2.

References

    1. Chapman T, McGavin J, Noble S. Tenofovir disoproxil fumarate. Drugs. 2003;63:1597–1608.
    1. National Institute of Health (NIH). Accessed: 27 Feb. 2012. Current HIV treatment guidelines: Available: .
    1. Anderson PL, Kiser JJ, Gardner EM, Rower JE, Meditz A, et al. Pharmacological considerations for tenofovir and emtricitabine to prevent HIV infection. J Antimicrob Chemother. 2011;66:240–250.
    1. Naesens L, Balzarini J, Bischofberger N, Clercq ED. Antiretroviral activity and pharmacokinetics in mice of oral bis(pivaloyloxymethyl)-9-(2- phosphonylmethoxyethyl)adenine, the bis(pivaloyloxymethyl) ester prodrug of 9-(2- phosphonylmethoxyethyl)adenine. Antimicrob Agents Chemother. 1996;40:22–28.
    1. Kearney BP, Flaherty JF, Shah J. Tenofovir disoproxil fumarate: clinical pharmacology and pharmacokinetics. Clin Pharmacokinet. 2004;43:595–612.
    1. von Kleist M, Metzner P, Marquet R, Scutte C. Polymerase inhibition by nucleoside analogs: Cellular- and kinetic parameters of efficacy, susceptibility and resistance selection. Plos Comput Biol. 2012;8:e1002359.
    1. Droste JAH, van Wissen CPWGMV, Kearney BP, Buffels R, Vanhorssen PJ, et al. Pharmacokinetic study of tenofovir disoproxil fumarate combined with rifampin in healthy volunteers. Antimicrob Agents Chemother. 2005;49:680–684.
    1. Blum MR, Chittick GE, Begley JA, Zong J. Steady-state pharmacokinetics of emtricitabine and tenofovir disoproxil fumarate administered alone and in combination in healthy volunteers. J Clin Pharmacol. 2007;47:751–759.
    1. Chittick GE, Zong J, Blum MR, Sorbel JJ, Begley JA, et al. Pharmacokinetics of tenofovir disoproxil fumarate and ritonavir-boosted saquinavir mesylate administered alone or in combination at steady state. Antimicrob Agents Chemother. 2006;50:1304–1310.
    1. Jullien V, Téluyer JM, Rey E, Jaffray P, Krivine A, et al. Population pharmacokinetics of tenofovir in human immunodeficiency virus-infected patients taking highly active antiretroviral therapy. Antimicrob Agents Chemother. 2005;49:3361–3366.
    1. Hawkins T, Veikley W, Claire RLS, Guyer B, Clark N, et al. Intracellular pharmacokinetics of tenofovir diphosphate, carbovir triphosphate, and lamivudine triphosphate in patients receiving triple-nucleoside regimens. J Acquir Immune Defic Syndr. 2005;39:406–411.
    1. Barditch-Crovo P, Deeks SG, Collier A, Safrin S, Coakley DF, et al. Phase I/II trial of the pharmacokinetics, safety, and antiretroviral activity of tenofovir disoproxil fumarate in human immunodeficiency virus-infected adults. Antimicrob Agents Chemother. 2001;45:2733–2739.
    1. Hirt D, Ekouvi DK, Pruvost A, Urien S, Arriv E, et al. Plasma and intracellular tenofovir pharmacokinetics in the neonate (anrs 12109 trial, step 2). Antimicrob Agents Chemother. 2011;55:2961–2967.
    1. Baheti G, Kiser JJ, Havens PL, Fletcher CV. Plasma and intracellular population pharmacokinetic analysis of tenofovir in HIV-1 infected patients. Antimicrob Agents Chemother. 2011.
    1. von Kleist M, Huisinga W. Pharmacokinetic-pharmacodynamic relationship of NRTIs and its connection to viral escape: an example based on zidovudine. Eur J Pharm Sci. 2009;36:532–543.
    1. Sharma PL, Nurpeisov V, Hernandez-Santiago B, Beltran T, Schinazi RF. Nucleoside inhibitors of human immunodeficiency virus type 1 reverse transcriptase. Curr Top Med Chem. 2004;4:895–919.
    1. Hurwitz SJ, Asif G, Schinazi RF. Development of a population simulation model for HIV monotherapy virological outcomes using lamivudine. Antivir Chem Chemother. 2007;18:329–341.
    1. Garca-Lerma JG, Paxton L, Kilmarx PH, Heneine W. Oral pre-exposure prophylaxis for HIV prevention. Trends Pharmacol Sci. 2010;31:74–81.
    1. [No authors listed] Early end for FEM-PrEP HIV prevention trial. AIDS Patient Care STDS. 2011;25:383.
    1. Grant RM, Lama JR, Anderson PL, McMahan V, Liu AY, et al. Pre-exposure chemoprophylaxis for HIV prevention in men who have sex with men. N Engl J Med. 2010;363:2587–2599.
    1. University of Washington. Pivotal study finds that HIV medications are highly effective as prophylaxis against HIV infection in men and women in africa. 2011.
    1. Centers for Disease Control and Prevention (CDC) CDC trial and another major study find PrEP can reduce risk of HIV infection among heterosexuals. 2011.
    1. Hayden EC. HIV drug-prevention strategy carries risks. Nature. 2011;476:260–261.
    1. Van Damme L, Corneli A, Ahmed K, Agot K, Lombaard J, et al. The FEMPrEP trial of emtricitabine/tenofovir disoproxil fumarate (truvada) among African women. In: The 19th Conference on Retroviruses and Oportunistic Infections, Abstract 32LB, March 5–8. 2012.
    1. Chapman EH, Kurec AS, Davey FR. Cell volumes of normal and malignant mononuclear cells. J Clin Pathol. 1981;34:1083–1090.
    1. Bisset LR, Lung TL, Kaelin M, Ludwig E, Dubs RW. Reference values for peripheral blood lymphocyte phenotypes applicable to the healthy adult population in switzerland. Eur J Haematol. 2004;72:203–212.
    1. Bonate PL. Pharmacokinetic-Pharmacodynamic Modeling and Simulation. Springer. 2006.
    1. Gagnieu MC, Barkil ME, Livrozet JM, Cotte L, Miailhes P, et al. Population pharmacokinetics of tenofovir in AIDS patients. J Clin Pharmacol. 2008;48:1282–1288.
    1. von Kleist M, Menz S, Huisinga W. Drug-class specific impact of antivirals on the reproductive capacity of HIV. PLoS Comput Biol. 2010;6:e1000720.
    1. von Kleist M, Menz S, Stocker H, Arasteh K, Schutte C, et al. HIV quasispecies dynamics during pro-active treatment switching: impact on multi-drug resistance and resistance archiving in latent reservoirs. PLoS One. 2011;6:e18204.
    1. Zhou Y, Zhang H, Siliciano JD, Siliciano RF. Kinetics of human immunodeficiency virus type 1 decay following entry into resting CD4+ T cells. J Virol. 2005;79:2199–2210.
    1. Shen L, Peterson S, Sedaghat AR, McMahon MA, Callender M, et al. Doseresponse curve slope sets class-specific limits on inhibitory potential of anti-HIV drugs. Nat Med. 2008;14:762–766.
    1. Baggaley RF, White RG, Boily MC. Infectiousness of HIV-infected homosexual men in the era of highly active antiretroviral therapy. AIDS. 2010;24:2418–2420.
    1. Royce RA, Sea A, Cates W, Cohen MS. Sexual transmission of HIV. N Engl J Med. 1997;336:1072–1078.
    1. Keele BF, Giorgi EE, Salazar-Gonzalez JF, Decker JM, Pham KT, et al. Identification and characterization of transmitted and early founder virus envelopes in primary HIV-1 infection. Proc Natl Acad Sci U S A. 2008;105:7552–7557.
    1. Salazar-Gonzalez JF, Bailes E, Pham KT, Salazar MG, Guffey MB, et al. Deciphering human immunodeficiency virus type 1 transmission and early envelope diversification by single-genome amplification and sequencing. J Virol. 2008;82:3952–3970.
    1. Frank M, von Kleist M, Kunz A, Harms G, Schutte C, et al. Quantifying the impact of nevirapine-based prophylaxis strategies to prevent mother-to-child transmission of HIV-1: a combined pharmacokinetic, pharmacodynamic, and viral dynamic analysis to predict clinical outcomes. Antimicrob Agents Chemother. 2011;55:5529–5540.
    1. Mayer KH, Mimiaga MJ, Gelman M, Grasso C. Raltegravir, tenofovir df, and emtricitabine for post-exposure prophylaxis to prevent the sexual transmission of HIV: Safety, tolerability and adherence. J Acquir Immune Defic Syndr. 2012.
    1. Tosini W, Muller P, Prazuck T, Benabdelmoumen G, Peyrouse E, et al. Tolerability of HIV postexposure prophylaxis with tenofovir/emtricitabine and lopinavir/ritonavir tablet formulation. AIDS. 2010;24:2375–2380.
    1. Winston A, McAllister J, Amin J, Cooper DA, Carr A. The use of a triple nucleoside-nucleotide regimen for nonoccupational HIV post-exposure prophylaxis. HIV Med. 2005;6:191–197.
    1. Otten RA, Smith DK, Adams DR, Pullium JK, Jackson E, et al. Efficacy of postexposure prophylaxis after intravaginal exposure of pig-tailed macaques to a human-derived retrovirus (human immunodeficiency virus type 2). J Virol. 2000;74:9771–9775.
    1. Tsai CC, Emau P, Follis KE, Beck TW, Benveniste RE, et al. Effectiveness of postinoculation (r)-9-(2-phosphonylmethoxypropyl) adenine treatment for prevention of persistent simian immunodeficiency virus sivmne infection depends critically on timing of initiation and duration of treatment. J Virol. 1998;72:4265–4273.
    1. Lifson JD, Piatak M, Cline AN, Rossio JL, Purcell J, et al. Transient early post-inoculation anti-retroviral treatment facilitates controlled infection with sparing of CD4+ T cells in gut-associated lymphoid tissues in SIVmac239-infected rhesus macaques, but not resistance to rechallenge. J Med Primatol. 2003;32:201–210.
    1. Benaboud S, Hirt D, Launay O, Pannier E, Firtion G, et al. Pregnancy-related effects on tenofovir pharmacokinetics: a population study with 186 women. Antimicrob Agents Chemother. 2012;56:857–862.
    1. Bouazza N, Urien S, Hirt D, Frange P, Rey E, et al. Population pharmacokinetics of tenofovir in HIV-1-infected pediatric patients. J Acquir Immune Defic Syndr. 2011;58:283–288.
    1. Hurwitz SJ, Asif G, Kivel NM, Schinazi RF. Development of an optimized dose for coformulation of zidovudine with drugs that select for the K65R mutation using a population pharmacokinetic and enzyme kinetic simulation model. Antimicrob Agents Chemother. 2008;52:4241–4250.
    1. Piliero PJ. Pharmacokinetic properties of nucleoside/nucleotide reverse transcriptase inhibitors. J Acquir Immune Defic Syndr. 2004;37:S2–S12.
    1. Peter K, Gambertoglio JG. Intracellular phosphorylation of zidovudine (ZDV) and other nucleoside reverse transcriptase inhibitors (RTI) used for human immunodeficiency virus (HIV) infection. Pharm Res. 1998;15:819–825.
    1. Pruvost A, Negredo E, Benech H, Theodoro F, Puig J, et al. Measurement of intracellular didanosine and tenofovir phosphorylated metabolites and possible interaction of the two drugs in human immunodeficiency virus-infected patients. Antimicrob Agents Chemother. 2005;49:1907–1914.
    1. Pruvost A, Negredo E, Théodoro F, Puig J, Levi M, et al. Pilot pharmacokinetic study of human immunodeficiency virus-infected patients receiving tenofovir disoproxil fumarate (TDF): investigation of systemic and intracellular interactions between TDF and abacavir, lamivudine, or lopinavir-ritonavir. Antimicrob Agents Chemother. 2009;53:1937–1943.
    1. Patterson KB, Prince HA, Kraft E, Jenkins AJ, Shaheen NJ, et al. Penetration of tenofovir and emtricitabine in mucosal tissues: implications for prevention of hiv-1 transmission. Sci Transl Med. 2011;3:112re4.
    1. Donnell D, Baeten J, Hendrix C, Bumpus N, Bangsberg D, et al. Tenofovir disoproxil fumarate drug levels indicate PrEP use is strongly correlated with HIV-1 protective effects: Kenya and uganda. In: The 19th Conference on Retroviruses and Oportunistic Infections, Abstract 30, March 5–8. 2012.
    1. Cohen J. Aids research. FDA panel recommends anti-hiv drug for prevention. Science. 2012;336:792.
    1. Emau P, Jiang Y, Agy MB, Tian B, Bekele G, et al. Post-exposure prophylaxis for SIV revisited: animal model for HIV prevention. AIDS Res Ther. 2006;3:29.
    1. Haase AT. Targeting early infection to prevent HIV-1 mucosal transmission. Nature. 2010;464:217–223.
    1. Pudney J, Quayle AJ, Anderson DJ. Immunological microenvironments in the human vagina and cervix: mediators of cellular immunity are concentrated in the cervical transformation zone. Biol Reprod. 2005;73:1253–1263.
    1. Bobardt MD, Chatterji U, Selvarajah S, der Schueren BV, David G, et al. Cell-free human immunodeficiency virus type 1 transcytosis through primary genital epithelial cells. J Virol. 2007;81:395–405.
    1. Wu L. Biology of HIV mucosal transmission. Curr Opin HIV AIDS. 2008;3:534–540.
    1. Morrow G, Vachot L, Vagenas P, Robbiani M. Current concepts of HIV transmission. Curr Infect Dis Rep. 2008;10:133–139.
    1. Galvin SR, Cohen MS. The role of sexually transmitted diseases in HIV transmission. Nat Rev Microbiol. 2004;2:33–42.
    1. Hallett TB, Baeten JM, Heffron R, Barnabas R, de Bruyn G, et al. Optimal uses of antiretrovirals for prevention in HIV-1 serodiscordant heterosexual couples in south africa: a modelling study. PLoS Med. 2011;8:e1001123.
    1. Cohen MS, Chen YQ, McCauley M, Gamble T, Hosseinipour MC, et al. Prevention of HIV-1 infection with early antiretroviral therapy. N Engl J Med. 2011;365:493–505.
    1. Garca-Lerma JG, Aung W, er Cong M, Zheng Q, Youngpairoj AS, et al. Natural substrate concentrations can modulate the prophylactic efficacy of nucleotide HIV reverse transcriptase inhibitors. J Virol. 2011;85:6610–6617.
    1. Karim QA, Karim SSA, Frohlich JA, Grobler AC, Baxter C, et al. Effectiveness and safety of tenofovir gel, an antiretroviral microbicide, for the prevention of HIV infection in women. Science. 2010;329:1168–1174.
    1. Dobard C, Sharma S, Martin A, Pau CP, Holder A, et al. Durable protection from vaginal simian-human immunodeficiency virus infection in macaques by tenofovir gel and its relationship to drug levels in tissue. J Virol. 2012;86:718–725.
    1. Schwartz JL, Rountree W, Kashuba ADM, Brache V, Creinin MD, et al. A multi-compartment, single and multiple dose pharmacokinetic study of the vaginal candidate microbicide 1% tenofovir gel. PLoS One. 2011;6:e25974.
    1. Katz DF, Gao Y, Kang M. Using modeling to help understand vaginal microbicide functionality and create better products. Drug Deliv Transl Res. 2011;1:256–276.
    1. Durand-Gasselin L, Silva DD, Benech H, Pruvost A, Grassi J. Evidence and possible consequences of the phosphorylation of nucleoside reverse transcriptase inhibitors in human red blood cells. Antimicrob Agents Chemother. 2007;51:2105–2111.
    1. Markowitz M, Louie M, Hurley A, Sun E, Mascio MD, et al. A novel antiviral intervention results in more accurate assessment of human immunodeficiency virus type 1 replication dynamics and T-cell decay in vivo. J Virol. 2003;77:5037–5038.
    1. Ramratnam B, Bonhoeffer S, Binley J, Hurley A, Zhang L, et al. Rapid production and clearance of HIV-1 and hepatitis C virus assessed by large volume plasma apheresis. Lancet. 1999;354:1782–1785.
    1. Tan WY, Wu H. Stochastic modeling of the dynamics of CD4+ T-cell infection by HIV and some monte carlo studies. Math Biosci. 1998;147:173–205.
    1. Tuckwell HC, Shipman PD, Perelson AS. The probability of HIV infection in a new host and its reduction with microbicides. Math Biosci. 2008;214:81–86.
    1. Wei X, Ghosh SK, Taylor ME, Johnson VA, Emini EA, et al. Viral dynamics in human immunodeficiency virus type 1 infection. Nature. 1995;373:117–122.
    1. Sedaghat AR, Siliciano RF, Wilke CO. Constraints on the dominant mechanism for HIV viral dynamics in patients on raltegravir. Antivir Ther. 2009;14:263–271.
    1. Koelsch KK, Liu L, Haubrich R, May S, Havlir D, et al. Dynamics of total, linear nonintegrated, and integrated HIV-1 DNA in vivo and in vitro. J Infect Dis. 2008;197:411–419.
    1. Sedaghat AR, Dinoso JB, Shen L, Wilke CO, Siliciano RF. Decay dynamics of HIV-1 depend on the inhibited stages of the viral life cycle. Proc Natl Acad Sci U S A. 2008;105:4832–4837.

Source: PubMed

3
Se inscrever