1
|
Wilkins HN, Knerler SA, Warshanna A, Ortiz RC, Haas K, Orsburn BC, Williams DW. Drug Metabolism and Transport Capacity of Endothelial Cells, Pericytes, and Astrocytes: Implications for CNS Drug Disposition. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.01.606165. [PMID: 39149336 PMCID: PMC11326144 DOI: 10.1101/2024.08.01.606165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Therapeutically targeting the brain requires interactions with endothelial cells, pericytes, and astrocytes at the blood brain barrier (BBB). We evaluated regional and cell-type specific drug metabolism and transport mechanisms using rhesus macaques and in vitro treatment of primary human cells. Here, we report heterogenous distribution of representative drugs, tenofovir (TFV), emtricitabine (FTC), and their active metabolites, which cerebrospinal fluid measures could not reflect. We found that all BBB cell types possessed functional drug metabolizing enzymes and transporters that promoted TFV and FTC uptake and pharmacologic activation. Pericytes and astrocytes emerged as pharmacologically dynamic cells that rivaled hepatocytes and were uniquely susceptible to modulation by disease and treatment. Together, our findings demonstrate the importance of considering the BBB as a unique pharmacologic entity, rather than viewing it as an extension of the liver, as each cell type possesses distinct drug metabolism and transport capacities that contribute to differential brain drug disposition.
Collapse
Affiliation(s)
- Hannah N. Wilkins
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Stephen A. Knerler
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, USA
| | - Ahmed Warshanna
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Rodnie Colón Ortiz
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kate Haas
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Benjamin C. Orsburn
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Dionna W. Williams
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
2
|
Chawki S, Goldwirt L, Mouhebb ME, Gabassi A, Taouk M, Bichard I, Loze B, Amara A, Brand R, Siegel A, McGowan I, Costagliola D, Assoumou L, Molina JM, Delaugerre C. Ex-vivo rectal tissue infection with HIV-1 to assess time to protection following oral preexposure prophylaxis with tenofovir disoproxil/emtricitabine. AIDS 2024; 38:455-464. [PMID: 37976073 PMCID: PMC10906210 DOI: 10.1097/qad.0000000000003789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 09/21/2023] [Accepted: 10/13/2023] [Indexed: 11/19/2023]
Abstract
OBJECTIVES We wished to assess time to protection from HIV-1 infection following oral tenofovir disoproxil and emtricitabine (TDF/FTC) as preexposure prophylaxis (PrEP), using ex-vivo rectal tissue infections and drug concentration measures in blood and rectal tissue. DESIGN/METHODS Participants from the ANRS PREVENIR study (NCT03113123) were offered this sub-study after a 14-day wash-out. We used an ex-vivo model to evaluate rectal tissue HIV-1 susceptibility before and after PrEP, 2 h after two pills or 7 days of a daily pill of TDF/FTC. PrEP efficacy was expressed by the difference (after-before) of 14-day cumulative p24 antigen levels. TFV-DP and FTC-TP levels were measured in rectal tissue and PBMCs and correlated with HIV-1 infection. RESULTS Twelve and 11 men were analyzed in the 2 h-double dose and 7 days-single dose groups, respectively. Cumulative p24 differences after-before PrEP were -144 pg/ml/mg (IQR[-259;-108]) for the 2 h-double dose group ( P = 0.0005) and -179 pg/ml/mg (IQR [-253;-86]) for the 7 days-single dose group ( P = 0.001), with no differences between groups ( P = 0.93). Rectal TFV-DP was below quantification after a double dose, but FTC-TP levels were similar to levels at 7 days. There was a significant correlation between rectal FTC-TP levels and p24 changes after a double dose ( R = -0.84; P = 0.0001). CONCLUSION Oral TDF/FTC provided similar protection against HIV-1 infection of rectal tissue 2 h after a double dose or 7 days of a daily dose. At 2 h, this protection seems driven by high FTC-TP concentrations in rectal tissue. This confirms the importance of combining TDF and FTC to achieve early protection.
Collapse
Affiliation(s)
- Sylvain Chawki
- Université de Paris Cité, INSERM U-944, Institut Recherche Saint Louis
- Assistance Publique - Hôpitaux de Paris (APHP), Hôpital Saint Louis, Service de Maladies Infectieuses
| | - Lauriane Goldwirt
- Assistance Publique - Hôpitaux de Paris (APHP), Hôpital Saint Louis, Laboratoire de Pharmacologie Biologique
| | - Mayssam El Mouhebb
- Sorbonne Université, INSERM, Institut Pierre Louis d’Epidémiologie et de Santé Publique
| | - Audrey Gabassi
- Université de Paris Cité, INSERM U-944, Institut Recherche Saint Louis
- Assistance Publique - Hôpitaux de Paris (APHP), Hôpital Saint Louis, Service de Virologie
| | - Milad Taouk
- Assistance Publique - Hôpitaux de Paris (APHP), Hôpital Saint Louis, Service de Gastro-entérologie, Paris, France
| | - Iris Bichard
- Assistance Publique - Hôpitaux de Paris (APHP), Hôpital Saint Louis, Service de Maladies Infectieuses
| | - Bénédicte Loze
- Assistance Publique - Hôpitaux de Paris (APHP), Hôpital Saint Louis, Service de Maladies Infectieuses
| | - Ali Amara
- Université de Paris Cité, INSERM U-944, Institut Recherche Saint Louis
| | - Rhonda Brand
- University of Pittsburgh, Magee-Women's Research Institute and Foundation, Pittsburgh, Pennsylvania, USA
| | - Aaron Siegel
- University of Pittsburgh, Magee-Women's Research Institute and Foundation, Pittsburgh, Pennsylvania, USA
| | - Ian McGowan
- University of Pittsburgh, Magee-Women's Research Institute and Foundation, Pittsburgh, Pennsylvania, USA
- Orion Biotechnology, Ottawa, Ontario, Canada
| | - Dominique Costagliola
- Sorbonne Université, INSERM, Institut Pierre Louis d’Epidémiologie et de Santé Publique
| | - Lambert Assoumou
- Sorbonne Université, INSERM, Institut Pierre Louis d’Epidémiologie et de Santé Publique
| | - Jean-Michel Molina
- Université de Paris Cité, INSERM U-944, Institut Recherche Saint Louis
- Assistance Publique - Hôpitaux de Paris (APHP), Hôpital Saint Louis, Service de Maladies Infectieuses
| | - Constance Delaugerre
- Université de Paris Cité, INSERM U-944, Institut Recherche Saint Louis
- Assistance Publique - Hôpitaux de Paris (APHP), Hôpital Saint Louis, Service de Virologie
| |
Collapse
|
3
|
Seneviratne HK. Nucleoside Triphosphate Diphosphohydrolase 1 Exhibits Enzymatic Activity toward Tenofovir Diphosphate. Drug Metab Dispos 2023; 51:385-391. [PMID: 36396461 DOI: 10.1124/dmd.122.000855] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 10/24/2022] [Accepted: 10/31/2022] [Indexed: 11/19/2022] Open
Abstract
Tenofovir (TFV; prescribed as TFV disoproxil fumarate and TFV alafenamide prodrugs) is currently used for HIV prevention and treatment. TFV must be phosphorylated twice into TFV-diphosphate (TFV-DP) to become pharmacologically active. Previously, we reported heterogeneity in TFV-DP distribution in colorectal tissue (a putative site of HIV infection) sections collected from research participants receiving a TFV-containing enema. This observed heterogeneity is likely multifactorial. Of note, TFV-DP is structurally similar to ATP. It is known that nucleotidases such as nucleoside triphosphate diphosphohydrolases (NTPDases) dephosphorylate ATP. Thus, it was hypothesized that NTPDase-mediated dephosphorylation plays a role in regulating TFV-DP levels in colorectal tissue. To test this hypothesis, recombinant NTPDase proteins (NTPDase 1, 3, 4, 5, 6, and 8) were incubated, individually, with TFV-DP to determine their abilities to dephosphorylate TFV-DP in vitro. Following incubations, TFV-DP dephosphorylation was determined using both malachite green phosphate assays and ultrahigh-performance liquid chromatography tandem mass spectrometry. From these, NTPDase 1 exhibited the highest activity toward TFV-DP. Further, enzyme kinetic analysis revealed Michaelis-Menten kinetics for NTPDase 1-mediated TFV-DP dephosphorylation. Next, immunoblot analyses were conducted to confirm the expression of NTPDase 1 protein in human colorectal tissue. Liquid chromatography coupled to mass spectrometry proteomics analysis was used to measure the relative abundance of NTPDases in human colorectal tissue among healthy adult individuals (n = 4). These analyses confirmed the high abundance of NTPDase 1 in human colorectal tissue. Taken together, results suggest that NTPDase 1 may contribute to the regulation of TFV-DP levels. The above data provide important insights into the dephosphorylation of TFV-DP. SIGNIFICANCE STATEMENT: Nucleoside triphosphate diphosphohydrolases (NTPDases) that are involved in enzymatic ATP dephosphorylation may contribute to tenofovir-diphosphate (TFV-DP) dephosphorylation, leading to its inactivation. In this study, the NTPDases responsible for TFV-DP dephosphorylation in vitro and their expression in human colorectal tissue were investigated. Through this work, it was demonstrated that NTPDase 1 has the highest activity toward TFV-DP dephosphorylation, and it was abundant in human colorectal tissue. Importantly, these studies will increase our understanding of TFV-DP disposition.
Collapse
Affiliation(s)
- Herana Kamal Seneviratne
- Department of Chemistry and Biochemistry, University of Maryland, Baltimore County and Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
4
|
The predictive value of macaque models of preexposure prophylaxis for HIV prevention. Curr Opin HIV AIDS 2022; 17:179-185. [PMID: 35762371 DOI: 10.1097/coh.0000000000000738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW We review macaque models for preexposure prophylaxis (PrEP) for HIV prevention and highlight their role in advancing currently approved and novel PrEP agents. RECENT FINDINGS The development of the repeat low dose simian HIV (SHIV) challenge models represented a significant advancement in preclinical PrEP modeling that has allowed the investigation of PrEP under conditions that better mimic HIV exposures in humans. These models incorporate relevant drug pharmacology to inform drug correlates of PrEP protection. Models of rectal, vaginal, and penile infection are now available and have been found to predict clinical efficacy of all the currently approved PrEP strategies including daily oral PrEP with the combination of emtricitabine and tenofovir disoproxil fumarate or tenofovir alafenamide, and a long-acting formulation of the integrase inhibitor cabotegravir. These models are being used to test new PrEP modalities including the nucleoside reverse transcriptase-translocation inhibitor islatravir and long-acting capsid inhibitors. The SHIV models have also been supplemented by sexually transmitted infection co-infections with Chlamydia trachomatis, Treponema pallidum or Trichomonas vaginalis to assess the impact of inflammation on PrEP efficacy. SUMMARY Clinical efficacy validated current PrEP macaque models supporting their continued use to advance novel PrEP agents to improve global PrEP coverage.
Collapse
|
5
|
Islatravir for the treatment and prevention of infection with the human immunodeficiency virus type 1. Curr Opin HIV AIDS 2021; 15:27-32. [PMID: 31658118 DOI: 10.1097/coh.0000000000000599] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW To discuss the potential role of islatravir (ISL), a novel reverse transcriptase translocation inhibitor, in the treatment and prevention of human immunodeficiency virus type 1 (HIV-1) infection. RECENT FINDINGS Islatravir (4'-ethynyl-2-fluoro-2'-deoxyadenosine, MK-8591) is a long-acting first-in-class nucleoside reverse transcriptase translocation inhibitor with the potential for versatile dosing routes and dosing intervals. It demonstrated robust antiviral activity when dosed once daily and once weekly in HIV-1-infected individuals and SIV-infected rhesus macaques. In clinical trials of ISL in combination with doravirine and lamivudine, daily oral administration resulted in high levels of virologic suppression in HIV-infected individuals. In preclinical studies, ISL dosed orally once-weekly as preexposure prophylaxis (PrEP), protected rhesus macaques against SHIV infection via the mucosal route in the low-dose rectal challenge model. Most recently, data in healthy HIV-1-uninfected individuals demonstrated the feasibility of formulating of ISL as an implant. In these studies, levels of intracellular ISL-triphosphate were consistent with the potential for a once-yearly implantable administration of ISL as PrEP. SUMMARY Islatravir is a promising new agent for both the treatment and prevention of HIV-1 infection.
Collapse
|
6
|
Markowitz M, Gettie A, St Bernard L, Andrews CD, Mohri H, Horowitz A, Grasperge BF, Blanchard JL, Niu T, Sun L, Fillgrove K, Hazuda DJ, Grobler JA. Once-Weekly Oral Dosing of MK-8591 Protects Male Rhesus Macaques From Intrarectal Challenge With SHIV109CP3. J Infect Dis 2021; 221:1398-1406. [PMID: 31175822 DOI: 10.1093/infdis/jiz271] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 05/20/2019] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND MK-8591 (4'-ethynyl-2-fluoro-2'-deoxyadenosine [EFdA]) is a novel reverse transcriptase-translocation inhibitor. METHODS We assessed MK-8591 as preexposure prophylaxis in the rhesus macaque model of intrarectal challenge with simian/human immunodeficiency virus (SHIV). In study 1, 8 rhesus macaques received 3.9 mg/kg of MK-8591 orally on day 0 and once weekly for the next 14 weeks. Eight controls were treated with vehicle. All rhesus macaques were challenged with SHIV109CP3 on day 6 and weekly for up to 12 challenges or until infection was confirmed. The dose of MK-8591 was reduced to 1.3 and 0.43 mg/kg/week in study 2 and further to 0.1 and 0.025 mg/kg/week in study 3. In studies 2 and 3, each dose was given up to 6 times once weekly, and animals were challenged 4 times once weekly with SHIV109CP3. RESULTS Control macaques were infected after a median of 1 challenge (range, 1-4 challenges). All treated animals in studies 1 and 2 were protected, consistent with a 41.5-fold lower risk of infection (P < .0001, by the log-rank test). In study 3, at a 0.1-mg/kg dose, 2 rhesus macaques became infected, consistent with a 7.2-fold lower risk of infection (P = .0003, by the log-rank test). The 0.025-mg/kg dose offered no protection. CONCLUSIONS These data support MK-8591's potential as a preexposure prophylaxis agent.
Collapse
Affiliation(s)
- Martin Markowitz
- Aaron Diamond AIDS Research Center, an affiliate of the Rockefeller University, New York, New York
| | - Agegnehu Gettie
- Aaron Diamond AIDS Research Center, an affiliate of the Rockefeller University, New York, New York
| | - Leslie St Bernard
- Aaron Diamond AIDS Research Center, an affiliate of the Rockefeller University, New York, New York
| | - Chasity D Andrews
- Aaron Diamond AIDS Research Center, an affiliate of the Rockefeller University, New York, New York
| | - Hiroshi Mohri
- Aaron Diamond AIDS Research Center, an affiliate of the Rockefeller University, New York, New York
| | - Amir Horowitz
- Aaron Diamond AIDS Research Center, an affiliate of the Rockefeller University, New York, New York
| | | | | | - Tao Niu
- Merck Research Laboratories, West Point, Pennsylvania
| | - Li Sun
- Merck Research Laboratories, West Point, Pennsylvania
| | | | | | | |
Collapse
|
7
|
Herrera C. The Pre-clinical Toolbox of Pharmacokinetics and Pharmacodynamics: in vitro and ex vivo Models. Front Pharmacol 2019; 10:578. [PMID: 31178736 PMCID: PMC6543330 DOI: 10.3389/fphar.2019.00578] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 05/06/2019] [Indexed: 01/05/2023] Open
Abstract
Prevention strategies against sexual transmission of human immunodeficiency virus (HIV) are essential to curb the rate of new infections. In the absence of a correlate of protection against HIV infection, pre-clinical evaluation is fundamental to facilitate and accelerate prioritization of prevention candidates and their formulations in a rapidly evolving clinical landscape. Characterization of pharmacokinetic (PK) and pharmacodynamic (PD) properties for candidate inhibitors is the main objective of pre-clinical evaluation. in vitro and ex vivo systems for pharmacological assessment allow experimental flexibility and adaptability at a relatively low cost without raising as significant ethical concerns as in vivo models. Applications and limitations of pre-clinical PK/PD models and future alternatives are reviewed in the context of HIV prevention.
Collapse
Affiliation(s)
- Carolina Herrera
- Section of Virology, Division of Infectious Diseases, Department of Medicine, Faculty of Medicine, Imperial College London, London, United Kingdom
| |
Collapse
|
8
|
Mandal S, Kang G, Prathipati PK, Zhou Y, Fan W, Li Q, Destache CJ. Nanoencapsulation introduces long-acting phenomenon to tenofovir alafenamide and emtricitabine drug combination: A comparative pre-exposure prophylaxis efficacy study against HIV-1 vaginal transmission. J Control Release 2018; 294:216-225. [PMID: 30576746 DOI: 10.1016/j.jconrel.2018.12.027] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 11/20/2018] [Accepted: 12/15/2018] [Indexed: 02/08/2023]
Abstract
Daily oral antiretroviral (ARV) drugs for pre-exposure prophylaxis (PrEP) has proven efficacy for diverse groups of high-risk individuals. However, daily dosing regimen has augmented non-adherence. These experiments comparatively investigated the long-acting (LA) PrEP potency of subcutaneous (SubQ) administrated tenofovir alafenamide (TAF) and emtricitabine (FTC) loaded nanoparticles (NPs) to solution in humanized (hu) mice. TAF + FTC NPs and TAF + FTC solution (each drug at 200 mg/kg) were administered to hu-CD34-NSG mice (n = 3/time point) for plasma and tissue pharmacokinetic parameter estimation using LC-MS/MS. NP enhanced tissue ARV assimilation compared to plasma. The same dose was administered for PrEP efficacy in HIV-1 challenged hu-BLT mice (n = 5/group). The hu-BLT mice were vaginally challenged with a transmission-founder (T/F) virus at 5 × 105 TCID50 inoculation, on day 4, 7 and 14 post-SubQ treatments (PT) and were compared to infected-untreated-control hu-BLT mice. By 21 days PT, 100% TAF + FTC solution-treated and control-untreated mice were infected. However, TAF + FTC NPs resulted in significant (p = .0002) protection from HIV-1 (day 4: 80%, day 7 and 14: 60%, respectively) compared to control mice. This proof-of-concept study demonstrated detectable TAF/FTC vaginal levels among TAF + FTC NP-treated hu-BLT mice correlating with prolonged PrEP efficacy, thus establishing long-acting TAF + FTC NPs as a potential PrEP modality.
Collapse
Affiliation(s)
- Subhra Mandal
- Creighton University School of Pharmacy & Health Professions, Omaha, NE, United States.
| | - Guobin Kang
- Nebraska Center for Virology, School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE, United States
| | | | - You Zhou
- Nebraska Center for Virology and Center for Biotechnology, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Wenjin Fan
- Nebraska Center for Virology, School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Qingsheng Li
- Nebraska Center for Virology, School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE, United States
| | | |
Collapse
|
9
|
Transcutaneously refillable nanofluidic implant achieves sustained level of tenofovir diphosphate for HIV pre-exposure prophylaxis. J Control Release 2018; 286:315-325. [PMID: 30092254 DOI: 10.1016/j.jconrel.2018.08.010] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 06/20/2018] [Accepted: 08/03/2018] [Indexed: 02/07/2023]
Abstract
Pre-exposure prophylaxis (PrEP) with antiretroviral (ARV) drugs are effective at preventing human immunodeficiency virus (HIV) transmission. However, implementation of PrEP presents significant challenges due to poor user adherence, low accessibility to ARVs and multiple routes of HIV exposure. To address these challenges, we developed the nanochannel delivery implant (NDI), a subcutaneously implantable device for sustained and constant delivery of tenofovir alafenamide (TAF) and emtricitabine (FTC) for HIV PrEP. Unlike existing drug delivery platforms with finite depots, the NDI incorporates ports allowing for transcutaneous refilling upon drug exhaustion. NDI-mediated drug delivery in rhesus macaques resulted in sustained release of both TAF and FTC for 83 days, as indicated by concentrations of TAF, FTC and their respectively metabolites in plasma, PBMCs, rectal mononuclear cells and tissues associated with HIV transmission. Notably, clinically relevant preventative levels of tenofovir diphosphate were achieved as early as 3 days after NDI implantation. We also demonstrated the feasibility of transcutaneous drug refilling to extend the duration of PrEP drug delivery in NHPs. Overall, the NDI represents an innovative strategy for long-term HIV PrEP administration in both developed and developing countries.
Collapse
|
10
|
Dunn DT, Glidden DV, Stirrup OT, McCormack S. The averted infections ratio: a novel measure of effectiveness of experimental HIV pre-exposure prophylaxis agents. Lancet HIV 2018; 5:e329-e334. [PMID: 29893246 PMCID: PMC6035730 DOI: 10.1016/s2352-3018(18)30045-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 03/02/2018] [Accepted: 03/16/2018] [Indexed: 06/08/2023]
Abstract
Tenofovir disoproxil fumarate combined with emtricitabine is a highly effective oral pre-exposure prophylaxis (PrEP) agent for preventing the acquisition of HIV. This effectiveness has consequences for the design and analysis of trials assessing experimental PrEP regimens, which now generally include an active-control tenofovir disoproxil fumarate plus emtricitabine group, rather than a placebo group, as a comparator. Herein, we describe major problems in the interpretation of the primary measure of effectiveness proposed for these trials, namely the ratio of HIV incidence in the experimental agent group to that in the active-control group. We argue that valid interpretation requires an assumption about one of two parameters: either the incidence among trial participants had they not received PrEP or the effectiveness of tenofovir disoproxil fumarate plus emtricitabine within the trial. However, neither parameter is directly observed because of the absence of a no-treatment group, thus requiring the use of external evidence or subjective judgment. We propose an alternative measure of effectiveness based on the concept of averted infections, which incorporates one of these parameters. The measure is simple to interpret, has clinical and public health relevance, and is a natural preservation-of-effect criterion for assessing statistical non-inferiority. Its adoption could also allow the use of smaller sample sizes, currently a major barrier to the assessment of experimental PrEP regimens.
Collapse
Affiliation(s)
- David T Dunn
- Medical Research Council Clinical Trials Unit, University College London, London, UK; Institute for Global Health, University College London, London, UK.
| | - David V Glidden
- Division of Biostatistics, University of California, San Francisco, CA, USA
| | - Oliver T Stirrup
- Institute for Global Health, University College London, London, UK
| | - Sheena McCormack
- Medical Research Council Clinical Trials Unit, University College London, London, UK
| |
Collapse
|
11
|
Collins JW, Heyward Hull J, Dumond JB. Comparison of tenofovir plasma and tissue exposure using a population pharmacokinetic model and bootstrap: a simulation study from observed data. J Pharmacokinet Pharmacodyn 2017; 44:631-640. [PMID: 29119381 DOI: 10.1007/s10928-017-9554-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 11/03/2017] [Indexed: 11/29/2022]
Abstract
Sparse tissue sampling with intensive plasma sampling creates a unique data analysis problem in determining drug exposure in clinically relevant tissues. Tissue exposure may govern drug efficacy, as many drugs exert their actions in tissues. We compared tissue area-under-the-curve (AUC) generated from bootstrapped noncompartmental analysis (NCA) methods and compartmental nonlinear mixed effect (NLME) modeling. A model of observed data after single-dose tenofovir disoproxil fumarate was used to simulate plasma and tissue concentrations for two destructive tissue sampling schemes. Two groups of 100 data sets with densely-sampled plasma and one tissue sample per individual were created. The bootstrapped NCA (SAS 9.3) used a trapezoidal method to calculate geometric mean tissue AUC per dataset. For NLME, individual post hoc estimates of tissue AUC were determined, and the geometric mean from each dataset calculated. Median normalized prediction error (NPE) and absolute normalized prediction error (ANPE) were calculated for each method from the true values of the modeled concentrations. Both methods produced similar tissue AUC estimates close to true values. Although the NLME-generated AUC estimates had larger NPEs, it had smaller ANPEs. Overall, NLME NPEs showed AUC under-prediction but improved precision and fewer outliers. The bootstrapped NCA method produced more accurate estimates but with some NPEs > 100%. In general, NLME is preferred, as it accommodates less intensive tissue sampling with reasonable results, and provides simulation capabilities for optimizing tissue distribution. However, if the main goal is an accurate AUC for the studied scenario, and relatively intense tissue sampling is feasible, the NCA bootstrap method is a reasonable, and potentially less time-intensive solution.
Collapse
Affiliation(s)
- Jon W Collins
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, 1093 Genetic Medicine Building, 120 Mason Farm Rd, CB 7361, Chapel Hill, NC, 27599-7361, USA
| | - J Heyward Hull
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, 1093 Genetic Medicine Building, 120 Mason Farm Rd, CB 7361, Chapel Hill, NC, 27599-7361, USA
| | - Julie B Dumond
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, 1093 Genetic Medicine Building, 120 Mason Farm Rd, CB 7361, Chapel Hill, NC, 27599-7361, USA.
| |
Collapse
|
12
|
Cong ME, Pau CP, Heneine W, García-Lerma JG. Antiretroviral Drug Activity in Macaques Infected during Pre-Exposure Prophylaxis Has a Transient Effect on Cell-Associated SHIV DNA Reservoirs. PLoS One 2016; 11:e0164821. [PMID: 27806064 PMCID: PMC5091888 DOI: 10.1371/journal.pone.0164821] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 09/30/2016] [Indexed: 01/12/2023] Open
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.
Collapse
Affiliation(s)
- Mian-er Cong
- Laboratory Branch, Division of HIV/AIDS Prevention, National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Chou-Pong Pau
- Laboratory Branch, Division of HIV/AIDS Prevention, National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Walid Heneine
- Laboratory Branch, Division of HIV/AIDS Prevention, National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - J. Gerardo García-Lerma
- Laboratory Branch, Division of HIV/AIDS Prevention, National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
13
|
Pre-exposure prophylaxis: a useful tool to prevent human immunodeficiency virus infection? Clin Microbiol Infect 2016; 22:757-767. [PMID: 27615724 DOI: 10.1016/j.cmi.2016.08.029] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2016] [Revised: 08/07/2016] [Accepted: 08/31/2016] [Indexed: 01/14/2023]
Abstract
The aim of preventive measures against human immunodeficiency virus (HIV) is to reduce the incidence of HIV infection in the general population and in high-risk groups, such as men having sex with men (MSM), and to reduce the risk that a given individual will contract or spread the virus. Male and female condoms, post-exposure prophylaxis and circumcision are preventive methods currently recognized or promoted worldwide. Although modest success has been reported in a phase-III vaccine trial, other methods are being evaluated, such as vaginal and rectal microbicides, and pre-exposure prophylaxis (PrEP). Herein, we discuss results from prevention trials, especially those focusing on PrEP and particularly on recent results from 'on-demand' PrEP regimens. The efficacy of PrEP (rates of 0%-86%) is strongly correlated with adherence and plasma concentrations of antiretrovirals. Adverse events are rare. Selection of emtricitabine-resistant strains is mainly reported in individuals with an undiagnosed HIV infection using PrEP. PrEP is now strongly recommended in WHO prevention programmes for individuals at substantial risk for HIV with a view to controlling this epidemic by 2030.
Collapse
|
14
|
Massud I, Mitchell J, Babusis D, Deyounks F, Ray AS, Rooney JF, Heneine W, Miller MD, García-Lerma JG. Chemoprophylaxis With Oral Emtricitabine and Tenofovir Alafenamide Combination Protects Macaques From Rectal Simian/Human Immunodeficiency Virus Infection. J Infect Dis 2016; 214:1058-62. [DOI: 10.1093/infdis/jiw312] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 07/18/2016] [Indexed: 11/13/2022] Open
|
15
|
Srinivasan P, Moss JA, Gunawardana M, Churchman SA, Yang F, Dinh CT, Mitchell JM, Zhang J, Fanter R, Miller CS, Butkyavichene I, McNicholl JM, Smith TJ, Baum MM, Smith JM. Topical Delivery of Tenofovir Disoproxil Fumarate and Emtricitabine from Pod-Intravaginal Rings Protects Macaques from Multiple SHIV Exposures. PLoS One 2016; 11:e0157061. [PMID: 27275923 PMCID: PMC4898685 DOI: 10.1371/journal.pone.0157061] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 05/23/2016] [Indexed: 11/19/2022] Open
Abstract
Topical preexposure prophylaxis (PrEP) against HIV has been marginally successful in recent clinical trials with low adherence rates being a primary factor for failure. Controlled, sustained release of antiretroviral (ARV) drugs may help overcome these low adherence rates if the product is protective for extended periods of time. The oral combination of tenofovir disoproxil fumarate (TDF) and emtricitabine (FTC) is currently the only FDA-approved ARV drug for HIV PrEP. A novel pod-intravaginal ring (IVR) delivering TDF and FTC at independently controlled rates was evaluated for efficacy at preventing SHIV162p3 infection in a rigorous, repeat low-dose vaginal exposure model using normally cycling female pigtailed macaques. Six macaques received pod-IVRs containing TDF (65 mg) and FTC (68 mg) every two weeks, and weekly vaginal exposures to 50 TCID50 of SHIV162p3 began one week after the first pod-IVR insertion. All pod-IVR-treated macaques were fully protected throughout the study (P = 0.0002, Log-rank test), whereas all control animals became infected with a median of 4 exposures to infection. The topical, sustained release of TDF and FTC from the pod-IVR maintained protective drug levels in macaques over four months of virus exposures. This novel and versatile delivery system has the capacity to deliver and maintain protective levels of multiple drugs and the protection observed here warrants clinical evaluation of this pod-IVR design.
Collapse
Affiliation(s)
- Priya Srinivasan
- Laboratory Branch, Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - John A. Moss
- Department of Chemistry, Oak Crest Institute of Science, Monrovia, California, United States of America
| | - Manjula Gunawardana
- Department of Chemistry, Oak Crest Institute of Science, Monrovia, California, United States of America
| | - Scott A. Churchman
- Auritec Pharmaceuticals, Inc., Pasadena, California, United States of America
| | - Flora Yang
- Department of Chemistry, Oak Crest Institute of Science, Monrovia, California, United States of America
| | - Chuong T. Dinh
- Total Solutions, Inc., Atlanta, Georgia, United States of America
| | - James M. Mitchell
- Laboratory Branch, Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Jining Zhang
- Total Solutions, Inc., Atlanta, Georgia, United States of America
| | - Rob Fanter
- Department of Chemistry, Oak Crest Institute of Science, Monrovia, California, United States of America
| | - Christine S. Miller
- Department of Chemistry, Oak Crest Institute of Science, Monrovia, California, United States of America
| | - Irina Butkyavichene
- Auritec Pharmaceuticals, Inc., Pasadena, California, United States of America
| | - Janet M. McNicholl
- Laboratory Branch, Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Thomas J. Smith
- Department of Chemistry, Oak Crest Institute of Science, Monrovia, California, United States of America
- Auritec Pharmaceuticals, Inc., Pasadena, California, United States of America
| | - Marc M. Baum
- Department of Chemistry, Oak Crest Institute of Science, Monrovia, California, United States of America
| | - James M. Smith
- Laboratory Branch, Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
16
|
Ariën KK, Venkatraj M, Michiels J, Joossens J, Vereecken K, Van der Veken P, Heeres J, De Winter H, Heyndrickx L, Augustyns K, Vanham G. Resistance and cross-resistance profile of the diaryltriazine NNRTI and candidate microbicide UAMC01398. J Antimicrob Chemother 2016; 71:1159-68. [PMID: 26850721 DOI: 10.1093/jac/dkv501] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 12/26/2015] [Indexed: 02/03/2023] Open
Abstract
OBJECTIVES The resistance development, cross-resistance to other NNRTIs and the impact of resistance on viral replicative fitness were studied for the new and potent NNRTI UAMC01398. METHODS Resistance was selected by dose escalation and by single high-dose selection against a comprehensive panel of NNRTIs used as therapeutics and NNRTIs under investigation for pre-exposure prophylaxis of sexual HIV transmission. A panel of 27 site-directed mutants with single mutations or combinations of mutations involved in reverse transcriptase (RT) inhibitor-mediated resistance was developed and used to confirm resistance to UAMC01398. Cross-resistance to other NNRTIs was assessed, as well as susceptibility of UAMC01398-resistant HIV to diarylpyrimidine-resistant viruses. Finally, the impact of UAMC01398 resistance on HIV replicative fitness was studied. RESULTS We showed that UAMC01398 has potent activity against dapivirine-resistant HIV, that at least four mutations in the RT are required in concert for resistance and that the resistance profile is similar to rilpivirine, both genotypically and phenotypically. Resistance development to UAMC01398 is associated with a severe fitness cost. CONCLUSIONS These data, together with the enhanced safety profile and good solubility in aqueous gels, make UAMC01398 an excellent candidate for HIV topical prevention.
Collapse
Affiliation(s)
- Kevin K Ariën
- Department of Biomedical Sciences, Institute of Tropical Medicine, B-2000 Antwerp, Belgium
| | - Muthusamy Venkatraj
- Laboratory of Medicinal Chemistry, University of Antwerp, B-2000 Antwerp, Belgium
| | - Johan Michiels
- Department of Biomedical Sciences, Institute of Tropical Medicine, B-2000 Antwerp, Belgium
| | - Jurgen Joossens
- Laboratory of Medicinal Chemistry, University of Antwerp, B-2000 Antwerp, Belgium
| | - Katleen Vereecken
- Department of Biomedical Sciences, Institute of Tropical Medicine, B-2000 Antwerp, Belgium
| | - Pieter Van der Veken
- Laboratory of Medicinal Chemistry, University of Antwerp, B-2000 Antwerp, Belgium
| | - Jan Heeres
- Laboratory of Medicinal Chemistry, University of Antwerp, B-2000 Antwerp, Belgium
| | - Hans De Winter
- Laboratory of Medicinal Chemistry, University of Antwerp, B-2000 Antwerp, Belgium
| | - Leo Heyndrickx
- Department of Biomedical Sciences, Institute of Tropical Medicine, B-2000 Antwerp, Belgium
| | - Koen Augustyns
- Laboratory of Medicinal Chemistry, University of Antwerp, B-2000 Antwerp, Belgium
| | - Guido Vanham
- Department of Biomedical Sciences, Institute of Tropical Medicine, B-2000 Antwerp, Belgium Department of Biomedical Sciences, University of Antwerp, B-2000 Antwerp, Belgium
| |
Collapse
|
17
|
Hayes J, Powell N, Lathrop G, Heneine W, Dobard CW. Assessment of penile erection methods in rhesus macaques to model pharmacokinetics of antiretroviral drugs and penile infection with simian immunodeficiency virus. J Med Primatol 2016; 45:34-41. [PMID: 26778321 DOI: 10.1111/jmp.12207] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/24/2015] [Indexed: 11/29/2022]
Abstract
BACKGROUND An established macaque model to assess HIV interventions against penile transmission is currently not available. Physiological changes during penile erections may affect susceptibility to infection and drug pharmacokinetics (PK). Here, we identify methods to establish erections in macaques to evaluate penile transmission, PK, and efficacy under physiologic conditions. METHODS Penile rigidity and length were evaluated in eight rhesus macaques following rectal electrostimulation (RES), vibratory stimulation (VS), or pharmacological treatment with Sildenafil Citrate (Viagra) or Alprostadil. RESULTS Rectal electrostimulation treatment increased penile rigidity (>82%) and length (2.5 ± 0.58 cm), albeit the response was transient. In contrast, VS alone or coupled with Viagra or Alprostadil failed to elicit an erection response. CONCLUSION Rectal electrostimulation treatment elicits transient but consistent penile erections in macaques. High rigidity following RES treatment demonstrates increased blood flow and may provide a functional model for penile PK evaluations and possibly simian immunodeficiency virus (SIV) transmission under erect conditions.
Collapse
Affiliation(s)
- James Hayes
- Animal Resources Branch, Division of Scientific Resources, National Center for Emerging Zoonotic and Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Nathaniel Powell
- Animal Resources Branch, Division of Scientific Resources, National Center for Emerging Zoonotic and Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - George Lathrop
- Animal Resources Branch, Division of Scientific Resources, National Center for Emerging Zoonotic and Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Walid Heneine
- Laboratory Branch, Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Charles W Dobard
- Laboratory Branch, Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, GA, USA
| |
Collapse
|
18
|
Abstract
PURPOSE OF REVIEW To discuss nondaily preexposure prophylaxis (PrEP) modalities that may provide advantages compared with daily PrEP in cost and cumulative toxicity, but may have lower adherence forgiveness. RECENT FINDINGS Animal models have informed our understanding of early viral transmission events, which help guide event-driven PrEP dosing strategies. These models indicate early establishment of viral replication in rectal or cervicovaginal tissues, so event-driven PrEP should rapidly deliver high mucosal drug concentrations within hours of the potential exposure event. Macaque models have demonstrated the high biological efficacy for event-driven dosing of oral tenofovir disoproxil fumarate (TDF) and emtricitabine (FTC) against both vaginal and rectal virus transmission. In humans, the IPERGAY study demonstrated 86% efficacy for event-driven oral TDF/FTC dosing among men who have sex with men (MSM), while no similar efficacy data are available on women or heterosexual men. The HPTN 067 study showed that certain MSM populations adhere well to nondaily PrEP, whereas other populations of women adhere more poorly to nondaily versus daily regimens. Pharmacokinetic studies following oral TDF/FTC dosing in humans indicate that TFV-diphosphate (the active form of TFV) accumulates to higher concentrations in rectal versus cervicovaginal tissue, but nonadherence in trials complicates the interpretation of differential mucosal drug concentrations. SUMMARY Event-driven dosing for TFV-based PrEP has promise for HIV prevention in MSM. Future research of event-driven PrEP in women and heterosexual men should be guided by a better understanding of the importance of mucosal drug concentrations for PrEP efficacy and its sensitivity to adherence.
Collapse
Affiliation(s)
- Peter L Anderson
- aDepartment of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado bLaboratory Branch, Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | | | | |
Collapse
|
19
|
Animal and human mucosal tissue models to study HIV biomedical interventions: can we predict success? J Int AIDS Soc 2015; 18:20301. [PMID: 26530077 PMCID: PMC4631705 DOI: 10.7448/ias.18.1.20301] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 09/10/2015] [Accepted: 09/28/2015] [Indexed: 12/20/2022] Open
Abstract
Introduction Preclinical testing plays an integral role in the development of HIV prevention modalities. Several models are used including humanized mice, non-human primates and human mucosal tissue cultures. Discussion Pharmaceutical development traditionally uses preclinical models to evaluate product safety. The HIV prevention field has extended this paradigm to include models of efficacy, encompassing humanized mice, non-human primates (typically Asian macaques) and human mucosal tissue (such as cervical and colorectal). As our understanding of the biology of HIV transmission improves and includes the influence of human behaviour/biology and co-pathogens, these models have evolved as well to address more complex questions. These three models have demonstrated the effectiveness of systemic (oral) and topical use of antiretroviral drugs. Importantly, pharmacokinetic/pharmacodynamic relationships are being developed and linked to information gathered from human clinical trials. The models are incorporating co-pathogens (bacterial and viral) and the effects of coitus (mucosal fluids) on drug distribution and efficacy. Humanized mice are being tailored in their immune reconstitution to better represent humans. Importantly, human mucosal tissue cultures are now being used in early clinical trials to provide information on product efficacy to more accurately characterize efficacious products to advance to larger clinical trials. While all of these models have made advancements in product development, each has limitations and the data need to be interpreted by keeping these limitations in mind. Conclusions Development and refinement of each of these models has been an iterative process and linkages to data generated among each of them and from human clinical trials are needed to determine their reliability. Preclinical testing has evolved from simply identifying products that demonstrate efficacy prior to clinical trials to defining essential pharmacokinetic/pharmacodynamic relationships under a variety of conditions and has the potential to improve product selection prior to the initiation of large-scale human clinical trials. The goal is to provide researchers with ample information to make conversant decisions that guide optimized and efficient product development.
Collapse
|
20
|
Massud I, Martin A, Dinh C, Mitchell J, Jenkins L, Heneine W, Pau CP, García-Lerma JG. Pharmacokinetic profile of raltegravir, elvitegravir and dolutegravir in plasma and mucosal secretions in rhesus macaques. J Antimicrob Chemother 2015; 70:1473-81. [PMID: 25630643 DOI: 10.1093/jac/dku556] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 12/12/2014] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVES Pharmacokinetic studies in animal models are important for assessing the prophylactic potential of antiretroviral drugs for HIV prevention. This study sought to identify clinically relevant doses of the marketed integrase inhibitors raltegravir, elvitegravir and dolutegravir in macaques and investigate drug penetration and antiviral activity in mucosal secretions. METHODS Macaques received one oral dose of raltegravir, elvitegravir or dolutegravir alone or in combination with emtricitabine and tenofovir disoproxil fumarate followed by drug level measurements in blood and rectal and vaginal secretions. Antiviral activity was investigated in TZM-bl cells exposed to SHIV162p3 in the presence of rectal secretions collected from treated animals. RESULTS Plasma drug concentrations with 50 mg/kg raltegravir or elvitegravir were within the range seen in humans receiving 400-800 mg of raltegravir or 800 mg of unboosted elvitegravir but lower than with 150 mg of elvitegravir boosted with cobicistat. AUC0-24 values for dolutegravir increased proportionally with the dose, with a calculated human-equivalent dose of 20 mg/kg. Elvitegravir showed the highest penetration in rectal and vaginal fluids despite the absence of pharmacological boosting, followed by raltegravir and dolutegravir. Rectal secretions collected at 24 h from treated macaques blocked infection of TZM-bl cells by 50% at dilutions of 1/1000 (raltegravir), 1/800 (dolutegravir) and >1/30 000 (elvitegravir). CONCLUSIONS We defined macaque doses of HIV integrase inhibitors that recapitulate human clinical doses, which will facilitate efficacy and dose escalation studies in macaques. High and sustained drug concentrations and activity in mucosal secretions suggest that integrase inhibitors are promising candidates for HIV prevention.
Collapse
Affiliation(s)
- Ivana Massud
- Laboratory Branch, Division of HIV/AIDS Prevention, National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, 1600 Clifton Road, Atlanta, GA 30329, USA
| | - Amy Martin
- Laboratory Branch, Division of HIV/AIDS Prevention, National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, 1600 Clifton Road, Atlanta, GA 30329, USA
| | - Chuong Dinh
- Laboratory Branch, Division of HIV/AIDS Prevention, National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, 1600 Clifton Road, Atlanta, GA 30329, USA
| | - James Mitchell
- Laboratory Branch, Division of HIV/AIDS Prevention, National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, 1600 Clifton Road, Atlanta, GA 30329, USA
| | - Leecresia Jenkins
- Laboratory Branch, Division of HIV/AIDS Prevention, National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, 1600 Clifton Road, Atlanta, GA 30329, USA
| | - Walid Heneine
- Laboratory Branch, Division of HIV/AIDS Prevention, National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, 1600 Clifton Road, Atlanta, GA 30329, USA
| | - Chou-Pong Pau
- Laboratory Branch, Division of HIV/AIDS Prevention, National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, 1600 Clifton Road, Atlanta, GA 30329, USA
| | - J Gerardo García-Lerma
- Laboratory Branch, Division of HIV/AIDS Prevention, National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, 1600 Clifton Road, Atlanta, GA 30329, USA
| |
Collapse
|