Prevention of rectal SHIV transmission in macaques by daily or intermittent prophylaxis with emtricitabine and tenofovir

J Gerardo García-Lerma, Ron A Otten, Shoukat H Qari, Eddie Jackson, Mian-Er Cong, Silvina Masciotra, Wei Luo, Caryn Kim, Debra R Adams, Michael Monsour, Jonathan Lipscomb, Jeffrey A Johnson, David Delinsky, Raymond F Schinazi, Robert Janssen, Thomas M Folks, Walid Heneine, J Gerardo García-Lerma, Ron A Otten, Shoukat H Qari, Eddie Jackson, Mian-Er Cong, Silvina Masciotra, Wei Luo, Caryn Kim, Debra R Adams, Michael Monsour, Jonathan Lipscomb, Jeffrey A Johnson, David Delinsky, Raymond F Schinazi, Robert Janssen, Thomas M Folks, Walid Heneine

Abstract

Background: In the absence of an effective vaccine, HIV continues to spread globally, emphasizing the need for novel strategies to limit its transmission. Pre-exposure prophylaxis (PrEP) with antiretroviral drugs could prove to be an effective intervention strategy if highly efficacious and cost-effective PrEP modalities are identified. We evaluated daily and intermittent PrEP regimens of increasing antiviral activity in a macaque model that closely resembles human transmission.

Methods and findings: We used a repeat-exposure macaque model with 14 weekly rectal virus challenges. Three drug treatments were given once daily, each to a different group of six rhesus macaques. Group 1 was treated subcutaneously with a human-equivalent dose of emtricitabine (FTC), group 2 received orally the human-equivalent dosing of both FTC and tenofovir-disoproxil fumarate (TDF), and group 3 received subcutaneously a similar dosing of FTC and a higher dose of tenofovir. A fourth group of six rhesus macaques (group 4) received intermittently a PrEP regimen similar to group 3 only 2 h before and 24 h after each weekly virus challenge. Results were compared to 18 control macaques that did not receive any drug treatment. The risk of infection in macaques treated in groups 1 and 2 was 3.8- and 7.8-fold lower than in untreated macaques (p = 0.02 and p = 0.008, respectively). All six macaques in group 3 were protected. Breakthrough infections had blunted acute viremias; drug resistance was seen in two of six animals. All six animals in group 4 that received intermittent PrEP were protected.

Conclusions: This model suggests that single drugs for daily PrEP can be protective but a combination of antiretroviral drugs may be required to increase the level of protection. Short but potent intermittent PrEP can provide protection comparable to that of daily PrEP in this SHIV/macaque model. These findings support PrEP trials for HIV prevention in humans and identify promising PrEP modalities.

Conflict of interest statement

Competing Interests: Authors JGGL, RAO, RJ, TMF, and WH are named in a US Government patent application related to methods for HIV prophylaxis.

Figures

Figure 1. Study Design and Interventions
Figure 1. Study Design and Interventions
(A) Daily PrEP. Macaques were drug-treated 7–9 d before the first virus inoculation. Treated animals that remained negative during the 14 challenges received drug for an additional 28 d. Treated animals that became infected continued treatment to monitor plasma viremia and drug resistance emergence. (B) Intermittent PrEP. Macaques received FTC and tenofovir only 2 h before and 24 h after each weekly rectal challenge. (C) Untreated control macaques. Infection of macaques was monitored weekly by PCR and serologic testing. The treatment groups were staggered; four of 33 macaques were used in two separate arms (see Methods, Text S1, and Table S1).
Figure 2. Protection against Repeated Rectal Virus…
Figure 2. Protection against Repeated Rectal Virus Exposures by Daily or Intermittent PrEP
Each survival curve represents the cumulative proportion of uninfected macaques as a function of the number of weekly rectal exposures. Protected animals in groups 1–4 remained negative after a mean washout out of 27 wk (range, 17–60 wk).
Figure 3. Breakthrough Infections during PrEP Show…
Figure 3. Breakthrough Infections during PrEP Show Blunted Acute Viremias
Each time point represents the mean virus load observed in treated (n = 6) or untreated control (n = 10) macaques. Time 0 indicates the peak plasma virus load. Bars denote the standard error of the mean.
Figure 4. Dynamics of Infection in Animals…
Figure 4. Dynamics of Infection in Animals Infected during PrEP with FTC (AG-80, AG-46, AH-04, and AG-07) or FTC/TDF (AG-81 and AI-54)
Red circles indicate detectable M184V/I mutation; wild-type sequences are shown in blue. Open circles indicate time points that were not genotyped. The first detectable antibody response is shown as red arrows. The broken line denotes the limit of detection of the virus load assay (50 copies/ml).

References

    1. Joint United Nations Programme on HIV/AIDS. 2007 AIDS Epidemic update. 2007. Available: .
    1. Cohen MS, Gay C, Kashuba AD, Blower S, Paxton L. Narrative review: Antiretroviral therapy to prevent the sexual transmission of HIV-1. Ann Intern Med. 2007;146:591–601.
    1. Derdelinckx I, Wainberg MA, Lange JM, Hill A, Halima Y, et al. Criteria for drugs used in pre-exposure prophylaxis trials against HIV infection. PLoS Med. 2006;3:e454.
    1. Liu AY, Grant RM, Buchbinder SP. Preexposure prophylaxis for HIV. JAMA. 2006;296:863–865.
    1. Grant RM, Buchbinder S, Cates W, Jr, Clarke E, Coates T, et al. Promote HIV chemoprophylaxis research, don't prevent it. Science. 2005;309:2170–2171.
    1. Abbas U, Anderson R, Mellors J. Potential impact of antiretroviral chemoprophylaxis on HIV-1 transmission in resource-limited settings. PLoS ONE. 2007;2:e875.
    1. Haase AT. Perils at the mucosal front lines for HIV and SIV and their hosts. Nature Rev Immunol. 2005;5:783–792.
    1. Miller CJ, Li Q, Abel K, Kim EY, Ma ZM, et al. Propagation and dissemination of infection after vaginal transmission of simian immunodeficiency virus. J Virol. 2005;79:9217–9227.
    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, Follis KE, Sabo A, Beck TW, Grant RF, et al. Prevention of SIV infection in macaques by (R)-9-(2-phosphonylmethoxypropyl)adenine. Science. 1995;270:1197–1199.
    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. Subbarao S, Otten RA, Ramos A, Kim C, Jackson E, et al. Chemoprophylaxis with tenofovir disoproxil fumarate provided partial protection against infection with simian human immunodeficiency virus in macaques given multiple virus challenges. J Infect Dis. 2006;194:904–911.
    1. van Rompay KK, Kearney BP, Sexton JJ, Colon R, Lawson JR, et al. Evaluation of oral tenofovir disoproxil fumarate and topical tenofovir GS-7340 to protect infant macaques against repeated oral challenges with virulent simian immunodeficiency virus. J Acquir Immune Defic Syndr. 2006;43:6–14.
    1. Peterson L, Taylor D, Roddy R, Belai G, Phillips P, et al. Tenofovir disoproxil fumarate for prevention of HIV infection in women: A phase 2, double-blind, randomized, placebo-controlled trial. PLoS Clin Trials. 2007;2:e27.
    1. Borroto-Esoda K, Vela JE, Myrick F, Ray AS, Miller MD. In vitro evaluation of the anti-HIV activity and metabolic interactions of tenofovir and emtricitabine. Antivir Ther. 2006;11:377–384.
    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. Wang LH, Begley J, St Claire RL, 3rd, Harris J, Wakeford C, et al. Pharmacokinetic and pharmacodynamic characteristics of emtricitabine support its once daily dosing for the treatment of HIV infection. AIDS Res Hum Retroviruses. 2004;20:1173–1182.
    1. Hawkins T, Veikley W, St Claire RL, 3rd, 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. Otten RA, Adams DR, Kim CN, Jackson E, Pullium JK, et al. Multiple vaginal exposures to low doses of R5 simian-human immunodeficiency virus: Strategy to study HIV preclinical interventions in nonhuman primates. J Infect Dis. 2005;19:164–173.
    1. Pilcher CD, Tien HC, Eron JJ, Jr, Vernazza PL, Leu SY, et al. Brief but efficient: Acute HIV infection and the sexual transmission of HIV. J Infect Dis. 2004;189:1785–1792.
    1. van Rompay KK, Matthews TB, Higgins J, Canfield DR, Tarara RP, et al. Virulence and reduced fitness of SIV with the M184V mutation in reverse transcriptase. J Virol. 2002;76:6083–6092.
    1. van Rompay KK, Singh RP, Heneine W, Johnson JA, Montefiori DC, et al. Structured treatment interruptions with tenofovir monotherapy for simian immunodeficiency virus-infected newborn macaques. J Virol. 2006;80:6399–6410.
    1. Harouse JM, Gettie A, Tan RC, Blanchard J, Cheng-Mayer C. Distinct pathogenic sequela in rhesus macaques infected with CCR5 or CXCR4 utilizing SHIVs. Science. 1999;284:816–819.
    1. Luciw PA, Pratt-Lowe E, Shaw KE, Levy JA, Cheng-Mayer C. Persistent infection of rhesus macaques with T-cell-line-tropic and macrophage-tropic clones of simian/human immunodeficiency viruses (SHIV) Proc Natl Acad Sci U S A. 1995;92:7490–7494.
    1. Pilcher CD, Joaki G, Hoffman IF, Martinson FE, Mapanje C, et al. Amplified transmission of HIV-1: Comparison of HIV-1 concentrations in semen and blood during acute and chronic infection. AIDS. 2007;21:1723–1730.
    1. Mordenti J. Man versus beast: pharmacokinetic scaling in mammals. J Pharm Sci. 1986;75:1028–1040.
    1. van Rompay KK, Brignolo LL, Meyer DJ, Jerome C, Tarara R, et al. Biological effects of short-term or prolonged administration of 9-[2-(phosphonomethoxy) propyl] adenine (tenofovir) to newborn and infant rhesus macaques. Antimicrob Agents Chemother. 2004;48:1469–1487.
    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. Johnson JA, Rompay KK, Delwart E, Heneine W. A rapid and sensitive real-time PCR assay for the K65R drug resistance mutation in SIV reverse transcriptase. AIDS Res Hum Retroviruses. 2006;22:912–916.
    1. Johnson JA, Li JF, Wei X, Lipscomb J, Bennett D, et al. Simple PCR assays improve the sensitivity of HIV-1 subtype B drug resistance testing and allow linking of resistance mutations. PLoS ONE. 2007;2:e638.
    1. Veazey RS, Klasse PJ, Schader SM, Hu Q, Ketas TJ, et al. Protection of macaques from vaginal SHIV challenge by vaginally delivered inhibitors of virus-cell fusion. Nature. 2005;438:99–102.
    1. Grant RM, Wainberg MA. Chemoprophylaxis of HIV infection: Moving forward with caution. J Infect Dis. 2006;194:874–876.
    1. van Rompay KK, Berardi CJ, Aguirre NL, Bischofberger N, Lietman PS, et al. Two doses of PMPA protect newborn macaques against oral simian immunodeficiency virus infection. AIDS. 1998;12:F79–F83.
    1. van Rompay KK, McChesney MB, Aguirre NL, Schmidt KA, Bischofberger N, et al. Two low doses of tenofovir protect newborn macaques against oral simian immunodeficiency virus infection. J Infect Dis. 2001;184:429–438.
    1. Couedel-Courteille A, Pretet JL, Barget N, Jacques S, Petitprez K, et al. Delayed viral replication and CD4(+) T cell depletion in the rectosigmoid mucosa of macaques during primary rectal SIV infection. Virology. 2003;316:290–301.
    1. Regoes RR, Longini IM, Feinberg MB, Staprans SI. Preclinical assessment of HIV vaccines and microbicides by repeated low-dose virus challenges. PLoS Med. 2005;2:e249.
    1. Kim CN, Adams DR, Bashirian S, Butera S, Folks TM, et al. Repetitive exposures with simian/human immunodeficiency viruses: Strategy to study HIV pre-clinical interventions in non-human primates. J Med Primatol. 2006;35:210–216.
    1. Johnson MA, Gathe JC, Jr, Podzamczer D, Molina JM, Naylor CT, et al. A once-daily lopinavir/ritonavir-based regimen provides noninferior antiviral activity compared with a twice-daily regimen. J Acquir Immune Defic Syndr. 2006;43:153–160.
    1. Veazey RS, Springer MS, Marx PA, Dufour J, Klasse PJ, et al. Protection of macaques from vaginal SHIV challenge by an orally delivered CCR5 inhibitor. Nature Med. 2005;11:1293–1294.
    1. Magierowska M, Bernardin F, Garg S, Staprans S, Miller MD, et al. Highly uneven distribution of tenofovir-selected simian immunodeficiency virus in different anatomical sites of rhesus macaques. J Virol. 2004;78:2434–2444.
    1. van Rompay KK, Cherrington JM, Marthas ML, Berardi CJ, Mulato AS, et al. 9-[2-(Phosphonomethoxy)propyl]adenine therapy of established simian immunodeficiency virus infection in infant rhesus macaques. Antimicrob Agents Chemother. 1996;40:2586–2591.
    1. Mattapallil JJ, Douek DC, Hill B, Nishimura Y, Martin M, et al. Massive infection and loss of memory CD4+ T cells in multiple tissues during acute SIV infection. Nature. 2005;434:1093–1097.
    1. Mehandru S, Poles MA, Tenner-Racz K, Horowitz A, Hurley A, et al. Primary HIV-1 infection is associated with preferential depletion of CD4+ T lymphocytes from effector sites in the gastrointestinal tract. J Exp Med. 2004;200:761–770.
    1. Veazey RS, DeMaria M, Chalifoux LV, Shvetz DE, Pauley DR, et al. Gastrointestinal tract as a major site of CD4+ T cell depletion and viral replication in SIV infection. Science. 1998;280:427–431.
    1. Gupta SB, Jacobson LP, Margolick JB, Rinaldo CR, Phair JP, et al. Estimating the benefit of an HIV-1 vaccine that reduces viral load set point. J Infect Dis. 2007;195:546–550.
    1. Dyer JR, Kazembe P, Vernazza PL, Gilliam BL, Maida M, et al. High levels of human immunodeficiency virus type 1 in blood and semen of seropositive men in sub-Saharan Africa. J Infect Dis. 1998;177:1742–1746.
    1. Wawer MJ, Gray RH, Sewankambo NK, Serwadda D, Li X, et al. Rates of HIV-1 transmission per coital act, by stage of HIV-1 infection, in Rakai, Uganda. J Infect Dis. 2005;191:1403–1409.
    1. Operskalski EA, Stram DO, Busch MP, Huang W, Harris M, et al. Role of viral load in heterosexual transmission of human immunodeficiency virus type 1 by blood transfusion recipients. Am J Epidemiol. 1997;146:655–661.

Source: PubMed

3
Suscribir