Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
2.
EBioMedicine ; 58: 102894, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32707451

ABSTRACT

BACKGROUND: Daily oral pre- or post-exposure prophylaxis (PrEP or PEP) is highly effective in preventing HIV infection. However, many people find it challenging to adhere to a daily oral regimen. Chemoprophylaxis with single oral doses of antiretroviral drugs taken before or after sex may better adapt to changing or unanticipated sexual practices and be a desirable alternative to daily PrEP or PEP. We investigated willingness to use a single oral pill before or after sex among men who have sex with men (MSM) and assessed the biological efficacy of a potent antiretroviral combination containing elvitegravir (EVG), emtricitabine (FTC), and tenofovir alafenamide (TAF). METHODS: Data on willingness to use single-dose PrEP or PEP were obtained from the 2017 cycle of the American Men's Internet Survey (AMIS), an annual online behavioral surveillance survey of MSM in the United States. Antiretroviral drug levels were measured in humans and macaques to define drug distribution in rectal tissue and identify clinically relevant doses for macaque modeling studies. The biological efficacy of a single dose of FTC/TAF/EVG as PrEP or PEP was investigated using a repeat-challenge macaque model of rectal HIV infection. FINDINGS: Through pharmacokinetic assessment in humans and macaques we found that EVG penetrates and concentrates in rectal tissues supporting its addition to FTC/TAF to boost and extend chemoprophylactic activity. Efficacy estimates for a single oral dose given to macaques 4h before or 2h after SHIV exposure was 91•7%[35•7%-98•9%] and 100%, respectively, compared to 80•1%[13•9%-95•4%] and 64•6%[-19•4%-89•5%] when single doses were given 6 and 24h post challenge, respectively. A two-dose regimen at 24h and 48h after exposure was also protective [77•1%[1•7%-94•7%]. INTERPRETATION: Informed by user willingness, human and macaque pharmacokinetic data, and preclinical efficacy we show that single-dose prophylaxis before or after sex is a promising HIV prevention strategy. Carefully designed clinical trials are needed to determine if any of these strategies will be effective in humans. FUNDING: Funded by CDC intramural funds, CDC contract HCVJCG2-2016-03948 (to CFK), and a grant from the MAC AIDS Fund and by the National Institutes of Health [P30AI050409] - the Emory Center for AIDS Research (to MZ and TS).


Subject(s)
Adenine/analogs & derivatives , Emtricitabine/administration & dosage , HIV Infections/prevention & control , Homosexuality, Male/psychology , Patient Compliance/statistics & numerical data , Quinolones/administration & dosage , Tenofovir/administration & dosage , Adenine/administration & dosage , Adenine/pharmacokinetics , Administration, Oral , Animals , Cross-Sectional Studies , Drug Combinations , Emtricitabine/pharmacokinetics , Humans , Macaca , Male , Patient Compliance/psychology , Pre-Exposure Prophylaxis , Quinolones/pharmacokinetics , Rectum/chemistry , Surveys and Questionnaires , Tenofovir/pharmacokinetics
3.
J Infect Dis ; 220(11): 1826-1833, 2019 10 22.
Article in English | MEDLINE | ID: mdl-31362305

ABSTRACT

BACKGROUND: Tenofovir alafenamide (TAF)-based regimens are being evaluated for pre-exposure prophylaxis (PrEP). We used a macaque model of repeated exposures to simian human immunodeficiency virus (SHIV) to investigate whether TAF alone or the combination of TAF and emtricitabine (FTC) can prevent vaginal infection. METHODS: Pigtail macaques were exposed vaginally to SHIV162p3 once a week for up to 15 weeks. Animals received clinical doses of FTC/TAF (n = 6) or TAF (n = 9) orally 24 hours before and 2 hours after each weekly virus exposure. Infection was compared with 21 untreated controls. RESULTS: Five of the 6 animals in the FTC/TAF and 4 of the 9 animals in the TAF alone group were protected against infection (P = .001 and P = .049, respectively). The calculated efficacy of FTC/TAF and TAF was 91% (95% confidence interval [CI], 34.9%-98.8%) and 57.8% (95% CI, -8.7% to 83.6%), respectively. Infection in FTC/TAF but not TAF-treated macaques was delayed relative to controls (P = .005 and P = .114). Median tenofovir diphosphate (TFV-DP) levels in peripheral blood mononuclear cells (PBMCs) were similar among infected and uninfected macaques receiving TAF PrEP (351 and 143 fmols/106 cells, respectively; P = .921). CONCLUSIONS: Emtricitabine/TAF provided a level of protection against vaginal challenge similar to FTC/TFV disoproxil fumarate combination in the macaque model. Our results support the clinical evaluation of FTC/TAF for PrEP in women.


Subject(s)
Adenine/analogs & derivatives , Anti-HIV Agents/administration & dosage , Disease Transmission, Infectious/prevention & control , Emtricitabine/administration & dosage , HIV Infections/prevention & control , Pre-Exposure Prophylaxis/methods , Vagina/virology , Adenine/administration & dosage , Alanine , Animals , Chemoprevention/methods , Disease Models, Animal , Female , HIV/genetics , HIV/isolation & purification , Macaca , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/isolation & purification , Tenofovir/analogs & derivatives , Treatment Outcome
4.
Nat Commun ; 10(1): 2005, 2019 05 01.
Article in English | MEDLINE | ID: mdl-31043606

ABSTRACT

A long-acting injectable formulation of the HIV integrase inhibitor cabotegravir (CAB-LA) is currently in clinical development for PrEP. Although the long plasma half-life of CAB-LA is an important attribute for PrEP, it also raises concerns about drug resistance emergence if someone becomes infected with HIV, or if PrEP is initiated during undiagnosed acute infection. Here we use a macaque model of SHIV infection to model risks of drug resistance to CAB-LA PrEP. Six macaques infected with SHIV received CAB-LA before seroconversion. We show integrase mutations G118R, E92G/Q, or G140R in plasma from 3/6 macaques as early as day 57, and identify G118R and E92Q in viruses from vaginal and rectal fluids. G118R and G140R confer > 800-fold resistance to CAB and cross-resistance to all licensed integrase inhibitors. Our results emphasize the need for appropriate HIV testing strategies before and possibly shortly after initiating CAB LA PrEP to exclude acute infection.


Subject(s)
Drug Resistance, Viral/genetics , HIV Infections/prevention & control , HIV Integrase Inhibitors/pharmacology , Pre-Exposure Prophylaxis/methods , Pyridones/pharmacology , Simian Acquired Immunodeficiency Syndrome/prevention & control , Acute Disease , Animals , Disease Models, Animal , Drug Resistance, Viral/drug effects , Female , HEK293 Cells , HIV Infections/blood , HIV Infections/immunology , HIV Infections/virology , HIV Integrase/genetics , HIV Integrase Inhibitors/blood , HIV Integrase Inhibitors/therapeutic use , HIV-1/drug effects , HIV-1/genetics , Half-Life , Humans , Macaca , Male , Pyridones/blood , Pyridones/therapeutic use , Seroconversion , Simian Acquired Immunodeficiency Syndrome/blood , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/drug effects , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/immunology , Time Factors
5.
J Virol ; 91(18)2017 09 15.
Article in English | MEDLINE | ID: mdl-28679753

ABSTRACT

Progressive T cell depletion during chronic human immunodeficiency virus type 1 (HIV) infection is a key mechanism that leads to the development of AIDS. Recent studies have suggested that most T cells in the tissue die through pyroptosis triggered by abortive infection, i.e., infection of resting T cells in which HIV failed to complete reverse transcription. However, the contribution of abortive infection to T cell loss and how quickly abortively infected cells die in vivo, key parameters for a quantitative understanding of T cell population dynamics, are not clear. Here, we infected rhesus macaques with simian-human immunodeficiency viruses (SHIV) and followed the dynamics of both plasma SHIV RNA and total cell-associated SHIV DNA. Fitting mathematical models to the data, we estimate that upon infection a majority of CD4+ T cells (approximately 65%, on average) become abortively infected and die at a relatively high rate of 0.27 day-1 (half-life, 2.6 days). This confirms the importance of abortive infection in driving T cell depletion. Further, we find evidence suggesting that an immune response may be restricting viral infection 1 to 3 weeks after infection. Our study serves as a step forward toward a quantitative understanding of the mechanisms driving T cell depletion during HIV infection.IMPORTANCE In HIV-infected patients, progressive CD4+ T cell loss ultimately leads to the development of AIDS. The mechanisms underlying this T cell loss are not clear. Recent experimental data suggest that the majority of CD4+ T cells in tissue die through abortive infection, where the accumulation of incomplete HIV transcripts triggers cell death. To investigate the role of abortive infection in driving CD4+ T cell loss in vivo, we infected macaques with simian-human immunodeficiency viruses (SHIV) and followed the viral kinetics of both plasma RNA and cell-associated DNA during infection. Fitting mathematical models, we estimated that a large fraction of infected cells dies through abortive infection and has a half-life of approximately 2.6 days. Our results provide the first in vivo quantitative estimates of parameters characterizing abortive infection and support the notion that abortive infection represents an important mechanism underlying progressive CD4+ T cell depletion in vivo.


Subject(s)
Cell Death , HIV/growth & development , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/growth & development , T-Lymphocytes/virology , Animals , DNA, Viral/analysis , Macaca mulatta , Models, Theoretical , RNA, Viral/blood , Viral Load
6.
PLoS One ; 11(11): e0164821, 2016.
Article in English | MEDLINE | ID: mdl-27806064

ABSTRACT

BACKGROUND: Pre-exposure prophylaxis (PrEP) with emtricitabine and tenofovir disoproxil fumarate (FTC/TDF) is a novel HIV prevention strategy. Suboptimal PrEP adherence and HIV infection creates an opportunity for continued antiretroviral drug activity during undiagnosed infection. We previously showed that macaques infected with SHIV during PrEP with FTC/TDF display reduced acute plasma viremias and limited virus diversity. We investigated the effect of PrEP on acute SHIV DNA dynamics and on the size of the persistent virus reservoir in lymphoid tissues. DESIGN: Cell-associated SHIV DNA levels in PBMCs were measured in 8 macaques infected during PrEP with FTC/TDF or single-agent TAF and was compared to those seen in untreated infections (n = 10). PrEP breakthrough infections continued treatment with 1-2 weekly drug doses to model suboptimal drug exposure during undiagnosed HIV infection in humans. SHIV DNA was also measured in lymphoid tissues collected from FTC/TDF PrEP breakthroughs after 1 year of infection. RESULTS: Compared to untreated controls, PrEP infections had reduced plasma RNA viremias both at peak and throughout weeks 1-12 (p<0.005). SHIV DNA levels were also reduced at peak and during the first 12 weeks of infection (p<0.043) but not throughout weeks 12-20. At 1 year, SHIV DNA reservoirs in lymphoid tissues were similar in size among macaques that received PrEP or placebo. CONCLUSIONS: Antiviral drug activity due to PrEP limits acute SHIV replication but has only a transient effect on cell-associated SHIV DNA levels. Our model suggests that suboptimal drug exposure in persons that are taking PrEP and become infected with HIV may not be sufficient to reduce the pool of HIV-infected cells, and that treatment intensification may be needed to sustain potential virological benefits from the PrEP regimen.


Subject(s)
Anti-HIV Agents/pharmacology , Pre-Exposure Prophylaxis , Proviruses/drug effects , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/drug effects , Animals , Anti-HIV Agents/administration & dosage , Case-Control Studies , Disease Models, Animal , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/virology , Lymphoid Tissue/drug effects , Lymphoid Tissue/virology , Macaca mulatta , Proviruses/genetics , Simian Acquired Immunodeficiency Syndrome/transmission , Time Factors , Viral Load
7.
Retrovirology ; 12: 69, 2015 Aug 08.
Article in English | MEDLINE | ID: mdl-26253002

ABSTRACT

BACKGROUND: Topically delivered tenofovir (TFV) from intravaginal rings, tablets, or gels is being evaluated for HIV prevention. We previously demonstrated that TFV delivered vaginally by gel protected macaques from vaginal infection with SHIV. Here we investigated efficacy of the TFV gel against vaginal transmission of a TFV-resistant SHIV containing the K65R mutation (SHIV162P3K65R) and its relationship to drug levels in vaginal tissues. RESULTS: SHIV162P3K65R shows approximately a 5-fold reduction in susceptibility to TFV compared to wild-type SHIV. Efficacy was evaluated in pig-tailed macaques exposed vaginally twice-weekly (up to 10 weeks) to SHIV162P3K65R 30 min after receiving placebo (n = 6) or 1% TFV (n = 6) gel. Four of the six controls were infected after a median of 5 exposures. In contrast, five of six macaques that received TFV gel remained uninfected after 20 vaginal SHIV162P3K65R exposures, resulting in an estimated efficacy of 75%. The mean intracellular TFV-diphosphate (TFV-DP) concentrations in vaginal lymphocytes 4 h after a single gel dose were found to be high (1,631 fmol/10(6) cells, range 492-3,847) and within the in vitro IC75 range (1,206 fmol/10(6) cells) for SHIV162P3K65R. CONCLUSION: Both the modest resistance conferred by K65R and the high TFV-DP exposure in vaginal lymphocytes, likely explain the observed protection. The findings in this model do not predict complete loss of protection by topical TFV against vaginal exposure to HIV-1K65R viruses and provide a tissue drug target for high efficacy. These data will facilitate the development of TFV delivery platforms that have high activity on both wild-type and TFV-resistant viruses.


Subject(s)
Administration, Intravaginal , HIV/drug effects , Reverse Transcriptase Inhibitors/administration & dosage , Simian Acquired Immunodeficiency Syndrome/transmission , Simian Immunodeficiency Virus/drug effects , Tenofovir/administration & dosage , Vagina/drug effects , Animals , Disease Models, Animal , Drug Resistance, Viral , Female , Gels , HIV Infections/virology , Humans , Macaca radiata , Pre-Exposure Prophylaxis , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/genetics , Vagina/virology
8.
J Infect Dis ; 208(3): 463-7, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23633402

ABSTRACT

Daily preexposure prophylaxis (PrEP) with emtricitabine/tenofovir disoproxil fumarate (FTC/TDF) is a novel strategy for preventing human immunodeficiency virus infection. We investigated in macaques whether FTC/TDF prevents transmission of a tenofovir-resistant simian/human immunodeficiency virus (SHIV) containing the K65R mutation. Six macaques received weekly a dose of FTC/TDF 3 days before rectal SHIV exposures and a second dose 2 hours after. Six untreated animals were controls. Animals were exposed rectally to escalating virus doses weekly for up to 28 weeks. PrEP significantly delayed infection with SHIVK65R (P = .028), although 4 of 6 FTC/TDF-treated macaques were infected at the end of the challenges. These findings highlight the need to closely monitor PrEP efficacy in areas with prevalent K65R.


Subject(s)
Adenine/analogs & derivatives , Anti-HIV Agents/administration & dosage , Deoxycytidine/analogs & derivatives , Drug Resistance, Viral , HIV/drug effects , Organophosphonates/administration & dosage , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Immunodeficiency Virus/drug effects , Adenine/administration & dosage , Administration, Oral , Animals , Deoxycytidine/administration & dosage , Disease Transmission, Infectious/prevention & control , Emtricitabine , HIV/genetics , Macaca , Mutation, Missense , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/genetics , Tenofovir , Treatment Outcome
9.
J Infect Dis ; 206(5): 770-9, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22740713

ABSTRACT

BACKGROUND: The impact of pre-exposure prophylaxis (PrEP) with antiretrovirals on breakthrough HIV or SHIV infection is not fully documented. We addressed the hypothesis that SHIV(SF162P3) infection despite active PrEP results in altered early immune parameters, compared with untreated infection. METHODS: Eleven rhesus macaques were infected during repeated, rectal, low-dose SHIV(SF162P3) exposures while receiving concurrent oral PrEP (Truvada [n = 2] or GS7340 [n = 4]) or as untreated controls (n = 5). We measured SHIV RNA, inflammatory cytokines, CD4 cells, and SHIV-specific and memory T cells until 20 weeks after peak viremia. RESULTS: SHIV infection during PrEP resulted in 100-fold lower peak viremia and lower IL-15, IL-18, and IL-1Ra levels, compared with controls (P < .05; Wilcoxon rank-sum test). Unlike controls, PrEP-treated macaques showed no significant CD4 cell count reduction during acute infection and developed more SHIV-specific central memory T cells, relative to controls. After in vivo CD8 cell depletion, viral load increased to similar levels, indicating that CD8 cells were critical for viral control in both groups. CONCLUSIONS: PrEP with antiretrovirals has beneficial effects on early SHIV infection even when infection is not prevented. Although long-term immune control could not be examined in this SHIV infection model, our results suggest that PrEP results in improved early disease parameters in breakthrough infections.


Subject(s)
Adenine/analogs & derivatives , Anti-HIV Agents/pharmacology , CD4-Positive T-Lymphocytes/immunology , Deoxycytidine/analogs & derivatives , Organophosphorus Compounds/pharmacology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Immunodeficiency Virus/immunology , Adenine/pharmacology , Alanine , Animals , CD4 Lymphocyte Count , CD4-Positive T-Lymphocytes/virology , Cytokines/blood , Cytokines/immunology , Deoxycytidine/pharmacology , Drug Combinations , Emtricitabine, Tenofovir Disoproxil Fumarate Drug Combination , Longitudinal Studies , Macaca mulatta , Male , RNA, Viral/chemistry , RNA, Viral/genetics , Reverse Transcriptase Polymerase Chain Reaction , Simian Acquired Immunodeficiency Syndrome/virology , Statistics, Nonparametric , Tenofovir/analogs & derivatives , Viremia/immunology , Viremia/virology
10.
J Virol ; 85(15): 7933-6, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21632769

ABSTRACT

Daily preexposure prophylaxis (PrEP) with Truvada (emtricitabine [FTC] and tenofovir disoproxil fumarate [TDF]) is a novel HIV prevention strategy recently found to reduce HIV incidence among men who have sex with men. We used a macaque model of HIV transmission to investigate if Truvada maintains prophylactic efficacy against an FTC-resistant isolate containing the M184V mutation. Five macaques received a dose of Truvada 3 days before exposing them rectally to the simian/human immunodeficiency virus mutant SHIV162p3(M184V), followed by a second dose 2 h after exposure. Five untreated animals were used as controls. Virus exposures were done weekly for up to 14 weeks. Despite the high (>100-fold) level of FTC resistance conferred by M184V, all five treated animals were protected from infection, while the five untreated macaques were infected (P = 0.0008). Our results show that Truvada maintains high prophylactic efficacy against an FTC-resistant isolate. Increased susceptibility to tenofovir due to M184V and other factors, including residual antiviral activity by FTC and/or reduced virus fitness due to M184V, may all have contributed to the observed protection.


Subject(s)
Antiviral Agents/pharmacology , Deoxycytidine/analogs & derivatives , HIV/drug effects , Mutation , Organophosphorus Compounds/pharmacology , Simian Acquired Immunodeficiency Syndrome/transmission , Simian Immunodeficiency Virus/drug effects , Animals , Cell Line , Deoxycytidine/pharmacology , Drug Combinations , Drug Resistance, Viral , Emtricitabine , Emtricitabine, Tenofovir Disoproxil Fumarate Drug Combination , HIV/genetics , Humans , Macaca mulatta , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Immunodeficiency Virus/genetics
11.
J Acquir Immune Defic Syndr ; 57(5): 355-62, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21637113

ABSTRACT

BACKGROUND: Pre-exposure prophylaxis (PrEP) is a novel intervention strategy for the prevention HIV transmission. Because several clinical trials are at various stages of completion, it is important to understand the impact of PrEP treatment on the development of the immune response to HIV, particularly in individuals who exhibit breakthrough infections despite PrEP. METHODS: A model of HIV infection, using rhesus macaques and the simian/human immunodeficiency virus (SHIV), was used to evaluate the effects of PrEP on the evolution of the humoral immune response. Time to seroconversion, neutralizing and binding antibody levels, and antibody avidity were measured in 12 rhesus macaques infected during daily or intermittent PrEP with FTC (emtricitabine) or Truvada (FTC/tenofovir combination) and compared with 11 untreated, simian HIV-infected controls. RESULTS: Macaques that became infected while receiving PrEP exhibited significantly lower peak virus loads during acute infection as compared with untreated animals. Although the timing of seroconversion and SHIV binding and neutralizing antibody levels were not impacted by treatment, lower maturation rates of antibody avidity for anti-p27, gp120, gp160, and gp41 were observed. CONCLUSIONS: This study suggests that reduced virus loads associated with PrEP treatment have little impact on timing of seroconversion and neutralizing/binding antibody levels; however, maturation of antibody avidity was suppressed.


Subject(s)
Anti-HIV Agents/therapeutic use , Antibodies, Viral/physiology , Antibody Affinity , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/immunology , Adenine/administration & dosage , Adenine/analogs & derivatives , Adenine/therapeutic use , Administration, Oral , Animals , Anti-HIV Agents/administration & dosage , Antibodies, Viral/blood , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Deoxycytidine/therapeutic use , Drug Combinations , Emtricitabine , HIV , HIV Antibodies/physiology , Human Immunodeficiency Virus Proteins/immunology , Macaca mulatta , Organophosphonates/administration & dosage , Organophosphonates/therapeutic use , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/drug effects , Tenofovir , Viral Load
12.
PLoS One ; 6(4): e19295, 2011 Apr 26.
Article in English | MEDLINE | ID: mdl-21541293

ABSTRACT

Pre-exposure prophylaxis (PrEP) with anti-viral drugs is currently in clinical trials for the prevention of HIV infection. Induction of adaptive immune responses to virus exposures during anti-viral drug administration, i.e., a "chemo-vaccination" effect, could contribute to PrEP efficacy. To study possible chemo-vaccination, we monitored humoral and cellular immune responses in nine rhesus macaques undergoing up to 14 weekly, low-dose SHIV(SF162P3) rectal exposures. Six macaques concurrently received PrEP with intermittent, oral Truvada; three were no-PrEP controls. PrEP protected 4 macaques from infection. Two of the four showed evidence of chemo-vaccination, because they developed anti-SHIV CD4(+) and CD8(+) T cells; SHIV-specific antibodies were not detected. Control macaques showed no anti-SHIV immune responses before infection. Chemo-vaccination-induced T cell responses were robust (up to 3,940 SFU/10(6) PBMCs), predominantly central memory cells, short-lived (≤22 weeks), and appeared intermittently and with changing specificities. The two chemo-vaccinated macaques were virus-challenged again after 28 weeks of rest, after T cell responses had waned. One macaque was not protected from infection. The other macaque concurrently received additional PrEP. It remained uninfected and T cell responses were boosted during the additional virus exposures. In summary, we document and characterize PrEP-induced T cell chemo-vaccination. Although not protective after subsiding in one macaque, chemo-vaccination-induced T cells warrant more comprehensive analysis during peak responses for their ability to prevent or to control infections after additional exposures. Our findings highlight the importance of monitoring these responses in clinical PrEP trials and suggest that a combination of vaccines and PrEP potentially might enhance efficacy.


Subject(s)
Deoxycytidine/analogs & derivatives , Mucous Membrane/virology , Organophosphorus Compounds/therapeutic use , Simian Acquired Immunodeficiency Syndrome/drug therapy , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Immunodeficiency Virus/immunology , Vaccination , Administration, Oral , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , CD4-Positive T-Lymphocytes/virology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/virology , Cell Differentiation/drug effects , Cytokines/biosynthesis , Deoxycytidine/administration & dosage , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Drug Combinations , Emtricitabine, Tenofovir Disoproxil Fumarate Drug Combination , Epitopes/immunology , Macaca mulatta/immunology , Macaca mulatta/virology , Male , Mucous Membrane/drug effects , Mucous Membrane/immunology , Organophosphorus Compounds/administration & dosage , Organophosphorus Compounds/pharmacology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/drug effects
13.
J Virol ; 85(13): 6610-7, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21525346

ABSTRACT

Preexposure prophylaxis (PrEP) with antiretroviral drugs is a novel human immunodeficiency virus (HIV) prevention strategy. It is generally thought that high systemic and mucosal drug levels are sufficient for protection. We investigated whether GS7340, a next-generation tenofovir (TFV) prodrug that effectively delivers tenofovir diphosphate (TFV-DP) to lymphoid cells and tissues, could protect macaques against repeated weekly rectal simian-human immunodeficiency virus (SHIV) exposures. Macaques received prophylactic GS7340 treatment 3 days prior to each virus exposure. At 3 days postdosing, TFV-DP concentrations in peripheral blood mononuclear cells (PBMCs) were about 50-fold higher than those seen with TFV disoproxil fumarate (TDF), and they remained above 1,000 fmol/10(6) cells for as long as 7 days. TFV-DP accumulated in lymphoid and rectal tissues, with concentrations at 3 days exceeding 500 fmol/10(6) mononuclear cells. Despite high mucosal and systemic TFV levels, GS7340 was not protective. Since TFV-DP blocks reverse transcription by competing with the natural dATP substrate, we measured dATP contents in peripheral lymphocytes, lymphoid tissue, and rectal mononuclear cells. Compared to those in circulating lymphocytes and lymphoid tissue, rectal lymphocytes had 100-fold higher dATP concentrations and dATP/TFV-DP ratios, likely reflecting the activated status of the cells and suggesting that TFV-DP may be less active at the rectal mucosa. Our results identify dATP/TFV-DP ratios as a possible correlate of protection by TFV and suggest that natural substrate concentrations at the mucosa will likely modulate the prophylactic efficacy of nucleotide reverse transcriptase inhibitors.


Subject(s)
Adenine/analogs & derivatives , Anti-HIV Agents/therapeutic use , Deoxyadenine Nucleotides/metabolism , HIV Infections/prevention & control , Organophosphonates/therapeutic use , Prodrugs/therapeutic use , Reverse Transcriptase Inhibitors/therapeutic use , Simian Acquired Immunodeficiency Syndrome/prevention & control , Adenine/administration & dosage , Adenine/pharmacokinetics , Adenine/therapeutic use , Animals , Anti-HIV Agents/administration & dosage , Anti-HIV Agents/pharmacokinetics , Chemoprevention , HIV Infections/virology , HIV Reverse Transcriptase/antagonists & inhibitors , HIV-1/drug effects , Humans , Lymphocytes/metabolism , Lymphoid Tissue/metabolism , Macaca , Organophosphonates/administration & dosage , Organophosphonates/pharmacokinetics , Prodrugs/administration & dosage , Prodrugs/pharmacokinetics , Rectum/virology , Reverse Transcriptase Inhibitors/administration & dosage , Reverse Transcriptase Inhibitors/pharmacokinetics , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/drug effects , Substrate Specificity , Tenofovir
14.
Virology ; 412(2): 435-40, 2011 Apr 10.
Article in English | MEDLINE | ID: mdl-21334708

ABSTRACT

Transmission of drug-resistant HIV has been widely documented. We generated tenofovir (TFV)- and emtricitabine (FTC)-resistant SHIV162P3 mutants that can be used to investigate the transmission efficiency of drug-resistant viruses and their impact on the efficacy of pre-exposure prophylaxis. Both SHIV162p3(M184V) and SHIV162p3(K65R) replicated in vitro at high titers. Drug resistance profiles were similar to those seen in HIV. Virus infectivity to virion particle ratios were 4- and 10-fold lower in SHIV162p3(M184V) and SHIV162p3(K65R), compared to a concurrently generated WT SHIV162p3, respectively. Mucosal transmissibility studies using a repeat low-dose macaque model of rectal and vaginal transmission showed that both mutants were able to efficiently infect macaques only after the dose was increased to adjust for fitness reductions due to K65R and M184V. Our results in limited number of macaques suggest that the reduction in fitness due to M184V and K65R decreases virus transmissibility, and identify in vitro infectivity parameters that associate with mucosal transmissibility.


Subject(s)
Adenine/analogs & derivatives , Anti-HIV Agents/pharmacology , Deoxycytidine/analogs & derivatives , Drug Resistance, Viral , HIV/drug effects , Organophosphonates/pharmacology , Simian Acquired Immunodeficiency Syndrome/transmission , Simian Immunodeficiency Virus/drug effects , Adenine/pharmacology , Animals , Deoxycytidine/pharmacology , Disease Models, Animal , Emtricitabine , HIV/genetics , HIV/growth & development , HIV/pathogenicity , Humans , Macaca , Mutation , Recombination, Genetic , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/growth & development , Simian Immunodeficiency Virus/pathogenicity , Tenofovir , Virus Cultivation
15.
Sci Transl Med ; 2(14): 14ra4, 2010 Jan 13.
Article in English | MEDLINE | ID: mdl-20371467

ABSTRACT

HIV continues to spread globally, mainly through sexual contact. Despite advances in treatment and care, preventing transmission with vaccines or microbicides has proven difficult. A promising strategy to avoid transmission is prophylactic treatment with antiretroviral drugs before exposure to HIV. Clinical trials evaluating the efficacy of daily treatment with the reverse transcriptase inhibitors tenofovir disoproxil fumarate (TDF) or Truvada (TDF plus emtricitabine) are under way. We hypothesized that intermittent prophylactic treatment with long-acting antiviral drugs would be as effective as daily dosing in blocking the earliest stages of viral replication and preventing mucosal transmission. We tested this hypothesis by intermittently giving prophylactic Truvada to macaque monkeys and then exposing them rectally to simian-human immunodeficiency virus (SHIV) once a week for 14 weeks. A simple regimen with an oral dose of Truvada given 1, 3, or 7 days before exposure followed by a second dose 2 hours after exposure was as protective as daily drug administration, possibly because of the long intracellular persistence of the drugs. In addition, a two-dose regimen initiated 2 hours before or after virus exposure was effective, and full protection was obtained by doubling the Truvada concentration in both doses. We saw no protection if the first dose was delayed until 24 hours after exposure, underscoring the importance of blocking initial replication in the mucosa. Our results show that intermittent prophylactic treatment with an antiviral drug can be highly effective in preventing SHIV infection, with a wide window of protection. They strengthen the possibility of developing feasible, cost-effective strategies to prevent HIV transmission in humans.


Subject(s)
Adenine/analogs & derivatives , Deoxycytidine/analogs & derivatives , Macaca/virology , Organophosphonates/pharmacokinetics , Organophosphorus Compounds/pharmacokinetics , Rectum/virology , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Immunodeficiency Virus/metabolism , Adenine/pharmacokinetics , Administration, Oral , Animals , Deoxycytidine/pharmacokinetics , Drug Combinations , Emtricitabine, Tenofovir Disoproxil Fumarate Drug Combination , Kinetics , Leukocytes, Mononuclear/virology , Proportional Hazards Models , Research Design , Tenofovir , Time Factors , Treatment Outcome , Viral Load
16.
J Chromatogr B Analyt Technol Biomed Life Sci ; 877(29): 3659-66, 2009 Nov 01.
Article in English | MEDLINE | ID: mdl-19783232

ABSTRACT

We developed an automated on-line weak anion exchange (WAX) solid-phase extraction (SPE) method coupled with ion-pair (IP) chromatography-tandem mass spectrometry (MS/MS) detection for quantitatively measuring triphosphorylated metabolites of three reverse transcriptase inhibitors (RTI). The administered pro-drugs were Tenofovir disoproxil fumarate (TDF), Emtricitabine (FTC) and Lamivudine (3TC). Their intracellular metabolites Tenofovir-diphosphate (TFV-DP), Emtricitabine-triphosphate (FTC-TP), and Lamivudine-triphosphate (3TC-TP) were measured in peripheral blood mononuclear cells (PBMC). We coupled the WAX and IP chromatography systems using a combination of 6-port and 10-port switching valves, and we mixed the WAX elute with 1,5-dimethyl-hexyl-amine before IP chromatography separation. Multiple waste outlets allowed for eliminating potential matrix components interfering with MS/MS detection. Limits of detection were 9, 200 and 75 pg per sample for TFV-DP (448/176 m/z), FTC-TP (488/130 m/z) and 3TC-TP (468/119 m/z), respectively.


Subject(s)
Chromatography, Ion Exchange/methods , Mass Spectrometry/methods , Polyphosphates/chemistry , Polyphosphates/metabolism , Reverse Transcriptase Inhibitors/chemistry , Reverse Transcriptase Inhibitors/metabolism , Adenine/analogs & derivatives , Adenine/chemistry , Adenine/metabolism , Deoxycytidine/analogs & derivatives , Deoxycytidine/chemistry , Deoxycytidine/metabolism , Emtricitabine , Humans , Lamivudine/chemistry , Lamivudine/metabolism , Leukocytes, Mononuclear/metabolism , Organophosphonates/chemistry , Organophosphonates/metabolism , Tenofovir
17.
J Virol ; 83(20): 10358-65, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19656878

ABSTRACT

New-generation gels that deliver potent antiretroviral drugs against human immunodeficiency virus type 1 have renewed hopes for topical prophylaxis as a prevention strategy. Previous preclinical research with monkey models suggested that high concentrations and drug combinations are needed for high efficacy. We evaluated two long-acting reverse transcriptase inhibitors, tenofovir (TFV) and emtricitabine (FTC), by using a twice-weekly repeat challenge macaque model and showed that a preexposure vaginal application of gel with 1% TFV alone or in combination with 5% FTC fully protected macaques from a total of 20 exposures to simian-human immunodeficiency virus SF162p3. FTC and TFV were detected in plasma 30 min after vaginal application, suggesting rapid absorption. FTC was detected more frequently than TFV and showed higher levels, reflecting the fivefold-higher concentration of this drug than of TFV. Two of 12 repeatedly exposed but protected macaques showed limited T-cell priming, which did not induce resistance to infection when macaques were rechallenged. Thus, single drugs with durable antiviral activity can provide highly effective topical prophylaxis and overcome the need for noncoital use or for drug combinations which are more complex and costly to formulate and approve.


Subject(s)
Adenine/analogs & derivatives , Anti-HIV Agents , Deoxycytidine/analogs & derivatives , Gels , Organophosphonates , Reverse Transcriptase Inhibitors , Simian Acquired Immunodeficiency Syndrome/drug therapy , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Immunodeficiency Virus/drug effects , Vagina/virology , Adenine/administration & dosage , Adenine/pharmacology , Adenine/therapeutic use , Administration, Intravaginal , Animals , Anti-HIV Agents/administration & dosage , Anti-HIV Agents/therapeutic use , Deoxycytidine/administration & dosage , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Drug Therapy, Combination , Emtricitabine , Female , Gels/administration & dosage , Gels/pharmacology , Gels/therapeutic use , Humans , Macaca nemestrina , Organophosphonates/administration & dosage , Organophosphonates/pharmacology , Organophosphonates/therapeutic use , Reverse Transcriptase Inhibitors/administration & dosage , Simian Acquired Immunodeficiency Syndrome/transmission , Simian Acquired Immunodeficiency Syndrome/virology , Tenofovir , Treatment Outcome
18.
PLoS Med ; 5(2): e28, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18254653

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.


Subject(s)
Adenine/analogs & derivatives , Deoxycytidine/analogs & derivatives , Organophosphonates/administration & dosage , Rectum/drug effects , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Acquired Immunodeficiency Syndrome/transmission , Simian Immunodeficiency Virus/drug effects , Adenine/administration & dosage , Adenine/blood , Animals , Deoxycytidine/administration & dosage , Deoxycytidine/blood , Drug Administration Schedule , Emtricitabine , Macaca , Macaca mulatta , Organophosphonates/blood , Rectum/metabolism , Rectum/pathology , Simian Acquired Immunodeficiency Syndrome/blood , Simian Immunodeficiency Virus/metabolism , Tenofovir
19.
PLoS One ; 2(7): e638, 2007 Jul 25.
Article in English | MEDLINE | ID: mdl-17653265

ABSTRACT

BACKGROUND: The success of antiretroviral therapy is known to be compromised by drug-resistant HIV-1 at frequencies detectable by conventional bulk sequencing. Currently, there is a need to assess the clinical consequences of low-frequency drug resistant variants occurring below the detection limit of conventional genotyping. Sensitive detection of drug-resistant subpopulations, however, requires simple and practical methods for routine testing. METHODOLOGY: We developed highly-sensitive and simple real-time PCR assays for nine key drug resistance mutations and show that these tests overcome substantial sequence heterogeneity in HIV-1 clinical specimens. We specifically used early wildtype virus samples from the pre-antiretroviral drug era to measure background reactivity and were able to define highly-specific screening cut-offs that are up to 67-fold more sensitive than conventional genotyping. We also demonstrate that sequencing the mutation-specific PCR products provided a direct and novel strategy to further detect and link associated resistance mutations, allowing easy identification of multi-drug-resistant variants. Resistance mutation associations revealed in mutation-specific amplicon sequences were verified by clonal sequencing. SIGNIFICANCE: Combined, sensitive real-time PCR testing and mutation-specific amplicon sequencing provides a powerful and simple approach that allows for improved detection and evaluation of HIV-1 drug resistance mutations.


Subject(s)
Drug Resistance, Viral/genetics , HIV-1/genetics , Polymerase Chain Reaction/methods , Anti-HIV Agents/pharmacology , Base Sequence , Cloning, Molecular , DNA Primers , Gene Amplification , HIV-1/classification , HIV-1/drug effects , Mutation , Oligodeoxyribonucleotides/chemistry , Plasmids/genetics , Polymorphism, Genetic , Reverse Transcriptase Polymerase Chain Reaction , Sensitivity and Specificity
20.
AIDS Res Hum Retroviruses ; 23(11): 1330-7, 2007 Nov.
Article in English | MEDLINE | ID: mdl-18184074

ABSTRACT

Persons occupationally exposed to nonhuman primates (NHPs) can be persistently infected with simian foamy virus (SFV). The clinical significance and person-to-person transmissibility of zoonotic SFV infection is unclear. Seven SFV-infected men responded to annual structured interviews and provided whole blood, oral, and urogenital specimens for study. Wives were tested for SFV infection. Proviral DNA was consistently detected by PCR in PBMCs of infected men and inconsistently in oral or urogenital samples. SFV was infrequently cultured from their PBMCs and throat swabs. Despite this and a long period of intimate exposure (median 20 years), wives were SFV negative. Most participants reported nonspecific symptoms and diseases common to aging. However, one of two persons with mild thrombocytopenia had clinically asymptomatic nonprogressive, monoclonal natural killer cell lymphocytosis of unclear relationship to SFV. All participants worked with NHPs before 1988 using mucocutaneous protection inconsistently; 57% described percutaneous injuries involving the infecting NHP species. SFV likely transmits to humans through both percutaneous and mucocutaneous exposures to NHP body fluids. Limited follow-up has not identified SFV-associated illness and secondary transmission among humans.


Subject(s)
Retroviridae Infections/physiopathology , Retroviridae Infections/virology , Simian foamy virus/genetics , Zoonoses/virology , Adult , Animals , Blood/virology , DNA, Viral/genetics , Family Health , Female , Humans , Interviews as Topic , Male , Middle Aged , Primates , Proviruses/isolation & purification , Retroviridae Infections/pathology , Saliva/virology , Semen/virology , Simian foamy virus/isolation & purification , Urine/virology
SELECTION OF CITATIONS
SEARCH DETAIL