Next Article in Journal
Self-Assembled Ru(II)-Coumarin Complexes for Selective Cell Membrane Imaging
Next Article in Special Issue
Dengue, West Nile, and Zika Viruses: Potential Novel Antiviral Biologics Drugs Currently at Discovery and Preclinical Development Stages
Previous Article in Journal
The Improved Brain-Targeted Drug Delivery of Edaravone Temperature-Sensitive Gels by Ultrasound for γ-ray Radiation-Induced Brain Injury
Previous Article in Special Issue
The MEK1/2 Inhibitor ATR-002 (Zapnometinib) Synergistically Potentiates the Antiviral Effect of Direct-Acting Anti-SARS-CoV-2 Drugs
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Pharmacological Validation of Long-Term Treatment with Antiretroviral Drugs in a Model of SIV-Infected Non-Human Primates

by
Thibaut Gelé
1,
Hélène Gouget
1,
Nathalie Dereuddre-Bosquet
1,
Valérie Furlan
2,
Roger Le Grand
1,
Olivier Lambotte
3,
Delphine Desjardins
1 and
Aurélie Barrail-Tran
4,*
1
Immunologie des Maladies Virales, Auto-Immunes, Hématologiques et Bactériennes, Université Paris-Saclay, Inserm, CEA, 92265 Fontenay-aux-Roses, France
2
Service de Pharmacologie-Toxicologie, Hôpital Bicêtre, AP-HP. Université Paris-Saclay, 94275 Le Kremlin-Bicêtre, France
3
Immunologie des Maladies Virales, Auto-Immunes, Hématologiques et Bactériennes, Service de Médecine Interne Immunologie Clinique, Hôpital Bicêtre, Université Paris-Saclay, AP-HP, Inserm, CEA, 94275 Le Kremlin-Bicêtre, France
4
Immunologie des Maladies Virales, Auto-Immunes, Hématologiques et Bactériennes, Service de Pharmacie, Hôpital Bicêtre, Université Paris-Saclay, AP-HP, Inserm, CEA, 94275 Le Kremlin-Bicêtre, France
*
Author to whom correspondence should be addressed.
Pharmaceutics 2022, 14(11), 2282; https://doi.org/10.3390/pharmaceutics14112282
Submission received: 23 September 2022 / Revised: 17 October 2022 / Accepted: 20 October 2022 / Published: 25 October 2022
(This article belongs to the Special Issue Drugs for Antiviral Combination Therapy)

Abstract

:
The development of animal models undergoing long-term antiretroviral treatment (ART) makes it possible to understand a number of immunological, virological, and pharmacological issues, key factors in the management of HIV infection. We aimed to pharmacologically validate a non-human primate (NHP) model treated in the long term with antiretroviral drugs after infection with the pathogenic SIVmac251 strain. A single-dose pharmacokinetic study of tenofovir disoproxil fumarate, emtricitabine, and dolutegravir was first conducted on 13 non-infected macaques to compare three different routes of administration. Then, 12 simian immunodeficiency virus (SIV)-infected (SIV+) macaques were treated with the same regimen for two years. Drug monitoring, virological efficacy, and safety were evaluated throughout the study. For the single-dose pharmacokinetic study, 24-h post-dose plasma concentrations for all macaques were above or close to 90% inhibitory concentrations and consistent with human data. During the two-year follow-up, the pharmacological data were consistent with those observed in humans, with low inter- and intra-individual variability. Rapid and sustained virological efficacy was observed for all macaques, with a good safety profile. Overall, our SIV+ NHP model treated with the ART combination over a two-year period is suitable for investigating the question of pharmacological sanctuaries in HIV infection and exploring strategies for an HIV cure.

1. Introduction

In the management of people living with HIV (PLWHIV), current antiretroviral treatment (ART) is effective in achieving and maintaining undetectable plasma HIV-RNA levels [1]. However, it does not eradicate the virus from reservoirs [2]. Among the factors of viral persistence is the presence of long-lived cells harbouring replication-competent HIV in a latent state coupled with insufficient tissular diffusion of current antiretroviral drugs (ARVs), leading to subtherapeutic concentrations and the establishment of pharmacological sanctuaries [3,4,5,6]. A better understanding of the mechanisms of HIV persistence is crucial for the optimisation of strategies to achieve an HIV cure [7]. Studies to investigate determinants of HIV reservoirs, such as the distribution of antiretroviral drugs in tissues or immunological responses, require access to tissues and cells [8,9,10].
Animal models are critical for tissue-based studies. Non-human primates (NHPs) can be infected with the pathogenic simian immunodeficiency virus (SIV), which recapitulates many of the characteristics of HIV infection and AIDS in humans [11,12]. The use of this animal model over the last several decades has allowed a better understanding of the disease [13]. Comparisons of the results obtained in NHP models with those observed in human studies will lead to further improvement in our scientific knowledge [14,15,16], especially for the development of therapeutic strategies towards an HIV cure.
Access to NHP models that show plasma exposure similar to that in humans and robust suppression of viremia is a prerequisite for understanding the mechanisms of viral persistence under suppressive ART. We selected tenofovir (TNF) disoproxil fumarate (TDF), emtricitabine (FTC), and dolutegravir (DTG), as they are among the preferred first-line treatment options worldwide [17,18,19,20].
This study had two main objectives: (1) to estimate and compare plasma pharmacokinetic (PK) parameters of the three ARVs administrated by three different routes of administration to determine whether the selected doses result in plasma exposure equivalent to that observed in humans and (2) to determine the systemic levels of ARV drugs in our SIV-infected (SIV+) NHP model on long-term ART.

2. Materials and Methods

2.1. Animals

Cynomolgus macaques (Macaca fascicularis) were imported from Mauritius and housed at the IDMIT animal facility at the Commissariat à l’Energie Atomique et aux Energies Alternatives (CEA, Fontenay-aux-Roses, France). All NHP studies at the CEA are conducted in accordance with French National Regulations under the supervision of National Veterinary Inspectors (CEA accreditation number D92-032-02). The CEA complies with the Standards for Human Care and Use of Laboratory Animals of the Office for Laboratory Animal Welfare (OLAW, Bethesda, MD, USA) under OLAW Assurance number #A5826-01. All experimental procedures were conducted according to European Directive 2010/63 (Recommendation number 9). The study was approved by the ethics committee “Comité d’éthique en expérimentation animale dn°44” and the French Ministry of Education and Research under reference APAFIS#2453-2015102713323361.
Two groups of macaques were used in this study: 13 non-infected macaques and 12 macaques infected with the pathogenic SIVmac251 strain.

2.2. Antiretroviral Drug Formulations

The combined antiretroviral therapy (cART) consisted of a three-drug regimen comprised of TDF, FTC, and DTG. Three different routes of administration were compared: oral (PO), subcutaneous (SC), and intravenous (IV).
Treatment for the PO route was prepared by crushing commercially available 300 mg TDF and 50 mg DTG tablets and opening commercially available 200 mg FTC capsules and resuspending the powder in the flavoured oral suspending vehicle Ora-Blend (Perrigo Company plc, Allegan, MI, USA).
Solutions for the SC and IV routes were prepared by dissolving TDF, FTC, and DTG in sterile water using parenteral grade Kleptose HPB (Roquette Pharma, Lestrem, France), as previously described [21], to enhance the solubility and stability of the molecules.

2.3. Single-Dose Antiretroviral Drug Plasma Pharmacokinetic Study

To compare the pharmacokinetic parameters, 13 non-infected male cynomolgus macaques were assigned to three groups according to the route of administration. ARV doses were selected to approximate the human equivalent dose [22]. The oral formulation of TDF (44 mg/kg), FTC (50 mg/kg), and DTG (20 mg/kg) was administered once by gavage to two cynomolgus macaques. A single SC or IV injection of TDF (5.1 mg/kg), FTC (40.0 mg/kg), and DTG (2.5 mg/kg) was administered to seven and four cynomolgus macaques, respectively.

2.4. Repeated Antiretroviral Doses in Long-Term ART-Treated SIV-Infected NHP

To investigate the long-term administration of ARVs, 12 cynomolgus macaques were inoculated by the intravenous route with 1000 50% animal infectious doses (AID50) of cell-free SIVmac251 (kindly provided by A.M. Aubertin, Université Louis Pasteur, Strasbourg, France). The pathogenicity of this strain without antiretroviral treatment has already been shown, hence the absence of a control group [23].
The treatment was initiated 17 weeks post-infection, during the chronic phase of infection, for all NHPs [23]. Infected animals were assigned into two groups according to the route of ART administration at the time of initiation: six animals treated by the SC route and six animals by the PO route for the first eight weeks. For the PO route, ART was administered using an oral formulation of TDF (30 mg/kg), FTC (40 mg/kg), and DTG (20 mg/kg). For the SC route, the doses were 5.1/40.0/2.5 mg/kg for TDF/FTC/DTG, respectively. After eight weeks, all macaques were treated by the SC route for two years (Figure S1).

2.5. Blood Samples and Antiretroviral Drug Assays

For the single-dose pharmacokinetic study, blood samples were collected 0.25 (only for IV injection), 1, 2, 4, 6, and 24 h after ART administration.
For the long-term study, blood samples were collected just before ART administration and on days 1, 3, 8, 15, 21, 28, 43, and 58 and then every one or two months up to two years after treatment initiation. All animals were euthanised at the last sample time.
Blood samples were centrifuged at 4000× g for 10 min at +4 °C and the resulting plasma samples collected and stored at −80 °C until analysis. The concentrations of TNF, FTC, and DTG were measured simultaneously in plasma samples using an LC-MS/MS method, as previously described [24]. Briefly, 100 μL plasma was mixed with 300 μL acetonitrile containing deuterated internal standards. The samples were then vortexed and centrifuged and 50 μL of the supernatant mixed with 750 μL of an aqueous solution containing 0.1% formic acid. Finally, 20 μL was injected into the LC-MS/MS system (TSQ ALTIS, Thermo Fisher Scientific, Villebon- sur-Yvette, France).

2.6. Efficacy and Safety Assessments

Quantitative RT-PCR was performed on the plasma of ART-treated SIV+ NHPs to monitor plasma viral load (VL) using a SuperScript III Platinum One-Step qRT-PCR Kit (Thermo Fisher Scientific, Villebon-sur-Yvette, France) with a CFX96 Touch Real-Time PCR Detection System (Bio-Rad, Marnes-la-Coquette, France). Primary virological efficacy was assessed based on the change from pre-cART plasma VL to the VL at week 8 of cART, the last sampling before switching from the oral treatment to the SC route for the oral group. The secondary virological efficacy endpoint was the percentage of macaques with a plasma viral load <40 copies/mL at month 6 and at the end of the study.
The treatment-emergent adverse events were recorded to evaluate safety, both in the single-dose study and for the ART-treated SIV+ NHPs. In addition, given the known risk of a progressive decline in the estimated glomerular filtration rate (eGFR) linked to TDF-based regimens [25,26], renal safety was assessed by monitoring creatinine levels.

2.7. Pharmacokinetic Analysis

For the single-dose pharmacokinetic study, a non-compartmental analysis using Phoenix WinNonlin 8.1 (Certara, St. Louis, MO, USA) was performed to estimate the pharmacokinetic parameters. The observed parameters were the maximal concentration (Cmax), 24-h post-dose concentration (C24h), and time required to reach Cmax (Tmax). The half-life (t½) was estimated from the log-linear terminal portion of the phase of decrease in concentration according to the formula t½ = 0.693/λz, where λz is the slope of the decrease in concentration. Areas under the curve (AUCs) were estimated using the linear up log down trapezoidal method until 24 h post dose (AUC0→24h).
For the repeated long-term pharmacokinetic study, the observed parameter was the trough concentration (Ct).
Plasma C24h or Ct were compared to wild-type HIV-1 90% inhibitory concentrations (IC90) for TNF (2.98 ng/mL) [27] and FTC (51 ng/mL) [28] and to the protein-adjusted IC90 (PA-IC90 = 64 ng/mL) for DTG [29].

2.8. Statistical Analysis

Intra-individual variability was calculated as the coefficient of variation of the Ct for each NHP from day 3 to the end of the study. For NHPs first treated via the oral route, intra-individual variability was calculated from day 3 to day 58 for the oral data and then from day 85 to the end of the study for the subcutaneous data. Inter-individual variability was calculated as the coefficient of variation of Ct at each time point from day 3 to the end of the study.
The data were analysed using GraphPad Prism (version 9.0.0 for Windows, GraphPad Software, San Diego, CA, USA). A Mann–Whitney test was performed to compare the inter- and intra-individual variability between the different routes of administration for the three ARVs. The threshold for statistical significance was set to p < 0.05. All numerical variables are expressed as medians and interquartile ranges (IQR) unless otherwise indicated.

3. Results

3.1. Single-Dose Antiretroviral Drug Plasma Pharmacokinetic Study

Plots of the concentration against the time after IV, PO, and SC administration of TDF, FTC, and DTG are shown in Figure 1. Antiretroviral pharmacokinetic parameters are shown in Table 1.
The PO route showed large differences between the two macaques for the three drugs. The C24h was above the IC90 for TNF and the PA-IC90 for DTG, regardless of the route of administration. For FTC, the C24h was above the IC90, except for three macaques that received the ART by the SC route. TNF C24h inter-individual variability was similar for the IV and SC routes (58%). DTG and FTC C24h inter-individual variability was lower for the IV than the SC route: 108% vs. 59% for FTC and 26% vs. 13% for DTG.
For TNF and FTC, the t½ was similar for both the IV and SC routes. However, the t½ was higher for DTG given by the SC or PO route than by the IV route, suggesting a flip-flop phenomenon.
Except for DTG given by the SC route, the three drugs showed AUCs similar to or higher than those observed for humans. AUC inter-individual variability was lower for the IV than the SC route: 64% vs. 20% for TNF, 25% vs. 5% for FTC, and 19% vs. 6% for DTG. No adverse effects were recorded in this single-dose study.

3.2. Pharmacokinetics, Efficacy, and Safety in the Long-Term Treatment Model

The Ct for TNF, FTC, and DTG over two years of treatment is shown in Figure 2. Steady-state concentrations were reached for the three drugs for all animals from day 3. At steady state, all TNF and DTG Ct values (except one concentration of 49 ng/mL by the PO route on day 43) were above the IC90 or PA-IC90, respectively, regardless of the route of administration, PO or SC.
For FTC by the PO route, the Ct (except one concentration of 40 ng/mL on day 28 and one concentration of 28 ng/mL on day 43) was above the IC90. By the SC route, the Ct was close to the IC90 during the first 43 days and then above the IC90 throughout the rest of the treatment, except at month 9. At this time point, three macaques (25%) had exceptionally low Ct values (below 10 ng/mL).
The intra-individual and inter-individual variability of the Ct at steady state is shown in Figure 3. Ct values were significantly lower for the PO than the SC route for TNF (51 vs. 66 ng/mL, p = 0.007) and DTG (221 vs. 727 ng/mL, p < 0.001), but were significantly higher for FTC (86 vs. 100 ng/mL, p = 0.012).
Intra- and inter-individual variability were relatively low, with only one intra-individual coefficient of variation above 100%. There were no significant differences in the intra-individual variability between the SC and PO routes for TNF (p = 0.067) or FTC (p = 0.892) but it was significantly lower for the SC than the PO route for DTG (p = 0.003). The inter-individual variability was significantly lower for the SC than the PO route for TNF (p < 0.001) and DTG (p = 0.001) but was not significantly different for FTC (p = 0.079).
Plasma viral loads declined in all animals after the initiation of treatment. We observed a significant reduction in plasma viral load from baseline (before treatment initiation, median [IQR]: 4.3 [4.8] log10 copies/mL) to month 2. After two months under ART, all macaques had plasma viral loads <40 copies/mL, except one animal. This macaque belonged to the oral group and showed high viremia prior to treatment. Its plasma viral load became quickly undetectable after switching to the SC route. Antiviral drug pressure was then sustained without any viral rebound between month 6 and the end of the treatment phase for all NHPs, including the three macaques with FTC Ct values below the IC90 at month 9 (Table S1).
In terms of safety, few drug-related adverse events were reported during the entire follow-up period. Two adverse events were related to the treatment administration procedure (SC route for the majority of the study): induration or abscess and bleeding or hematomas. Diarrhoea was also observed, but even more rarely. None required treatment interruption, and all were self-limiting. After two years of treatment, changes in the level of the renal biomarker creatinine were small, with a median [IQR] increase of 2 [2] mg/L, i.e., 16.0% [23.8%]. The median [IQR] creatinine concentration was 13 [3] mg/L and was still within the usual biological reference values, except for two macaques with concentrations of 17 and 18 mg/L.

4. Discussion

In this study, we evaluated the plasma pharmacokinetics of TDF/FTC/DTG given simultaneously to healthy NHPs by three different routes of administration. We also reported the long-term plasma pharmacokinetics of these molecules in SIV+ NHPs. We thus demonstrated that it is possible to simultaneously and easily administer a three-antiretroviral-drug formulation at doses that enable a level of exposure close to that observed in humans over a two-year period with virological efficacy.
Developing and validating an NHP model for the long-term administration of three combined ARVs (two years) is of particular interest for further immunological, virological, and pharmacological studies.
Before validating our SIV+ long-term ART-treated NHP model, we determined the single-dose pharmacokinetics of the three selected drugs to validate the doses for the three routes of administration. These routes included that used clinically (PO), that used in preclinical studies in NHPs for long-term treatment (SC), and that used as the reference in pharmacokinetic studies (IV). They were tested to evaluate the difference between each main pharmacokinetic parameter. As ARVs may be considered as time-dependant anti-infective agents, the most important pharmacokinetic parameter is the C24h, which should be above the (PA-)IC90. The C24h was above or close to the (PA-)IC90 for all three ARVs. For TNF and DTG, the C24h was slightly lower than that observed in humans but was, nonetheless, above the respective IC90 or PA-IC90 [30,31]. For FTC, the C24h was close to both that observed in humans and the IC90 [32]. For TNF and FTC, the high AUCs or Cmax observed could be attributed to higher-weighted doses than used in humans [30,33]. However, no dose-effect toxicity was observed in our single-dose study. For DTG, the use of hydroxypropyl-beta-cyclodextrin and the SC route could be responsible for the observed flip-flop phenomenon, leading to a lower Cmax and AUC and a higher t½ than expected and compared to those observed in humans [34,35,36,37].
The inter-individual variability for the AUC and C24h was relatively low but higher for the oral than the SC and IV routes, the IV route showing the lowest inter-individual variability for the three ARVs studied. This can be explained by the different absorption mechanisms involved. Indeed, the oral route requires multiple mechanisms involved in drug absorption, whereas the SC route only requires that the drugs cross the capillary barrier without an intestinal or hepatic first-pass effect. For the IV route, the entire dose is immediately delivered to the systemic circulation without limiting factors. Overall, based on the C24h, the main pharmacokinetic parameter, all NHPs had concentrations within the expected therapeutic range for all three ARVs and the three routes of administration. Thus, the pharmaceutical formulations were validated. The C24h for TDF and FTC was slightly higher for the oral route than that observed in humans and higher than that for the SC or IV routes in our study. Thus, the doses of these two ARVs were slightly reduced for the long-term study of the ART of SIV+ macaques.
We developed a model of long-term ART in macaques chronically infected with SIVmac251. At the time of ART initiation, we compared two routes of administration: the oral route, with the ARV administered in fruit, and the SC route. The IV route was not used for obvious reasons of ease of administration and the requirement of sedation. Compared to the administration by the oral route, the SC route has the advantage that the exact amount of drug intake is known and that no flavour adjustment is required. Flavour was a key point in the acceptance of oral treatment by the cynomolgus macaques. The oral suspending vehicle used had a sweet citrus-berry flavour, which appeared to be suitable.
Our data show rapid and sustained virological efficacy and good tolerance of the ARV treatments throughout the study. The slight increase in creatinine levels after two years under ART, although still within the usual biological reference values [38], is consistent with observed data in treatment-naive patients after 96 weeks [39,40]. This increase can be explained by two phenomena: the inhibition of the renal transporters SCL22A2 and SLC47A1 by DTG [41], both involved in the tubular secretion of creatinine [42,43], and the potential renal toxicity of tenofovir due to accumulation in the proximal tubular cells [25,26].
The novelty of our study consists of the pharmacological validation of the NHP model. In contrast to many studies that reported little or even no pharmacological data, we carried out regular long-term monitoring of the trough concentrations of the three ARVs used. Unlike the single-dose study, the Ct values of TNF and DTG were statistically higher following administration by the SC than the PO route. This can be explained by the use of hydroxypropyl-beta-cyclodextrin, an excipient for the SC formulation, which may be responsible for the flip-flop phenomenon and therefore produce a delayed-release form, increasing the drug accumulation ratio. However, despite these statistical differences between routes, all concentrations sampled 24 h after the previous administration were above the (PA-)IC90 throughout the study and consistent with clinical data. At month 9, three macaques had FTC Ct values below the IC90. However, a delay in the administration (median = 29 h 27 min, IQR = 1 min, after the previous one) and, therefore, in the residual sampling for this time point, could explain these low Ct values, also observed for TNF and DTG but with Ct values still above the (PA-)IC90. We observed no viral rebound around this sampling time for any of the three macaques. Therefore, these lower concentrations had no impact on the plasma viral load.
Only one previous study has reported long-term treatment and follow-up in NHPs [44]. The study was performed in rhesus macaques (Macaca mulatta) treated solely with TNF by the SC route and reported no trough concentrations, making a comparison with our results difficult. In addition, as for our single-dose study, the inter-individual variability was greater by the oral than the SC route. The potential impact of high doses on intestinal absorption, the hepatic first-pass effect, and the administration via the fruit with which the treatment is given may explain such differences.
Overall, our pharmacological data, associated with the robust viral suppression observed in the animals, allowed us to validate our long-term ART-treated SIV-infected macaque model. This model will be used for further pharmacokinetic/pharmacodynamic studies. We will characterize the diffusion of the three ARVs into different tissues to identify pharmacological sanctuaries. This model will also be a valuable tool to evaluate the impact of ARVs on viral reservoirs and on immunological parameters [45,46].
Our study had several limitations. First, the SC route is an injectable route. It is not the route used in humans for these ARVs and it may cause safety problems, such as injection-site pain, injection-site reactions, or a greater risk of infection. Nevertheless, only a few injection-site reactions and no infections arose during our two-year follow-up study. Moreover, being an injectable route, the SC route provides the advantage of lower intra- and inter-individual variability relative to the oral route and the precise dose administered is known. Second, the follow-up of the oral route arm was only carried out two months before the switch to the SC route. However, considering that the concentrations were close to those of the SC route arm and were virologically effective, the oral route also appears to be a potential route of administration for long-term follow-up.
Given that antiretroviral drugs are a long-term treatment modality for PLWHIV, our study, which highlights the pharmacokinetic and other advantages of oral and SC administration, makes an important contribution. This study provides the first description of long-term pharmacological exposure of a first-line worldwide treatment option administered simultaneously in an NHP model. Understanding the virus/drug interactions will be necessary to optimize newly developed ARV combinations. These data will have important implications towards an HIV cure.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/pharmaceutics14112282/s1, Figure S1: Repeated-dose study design, Table S1: Virological follow-up.

Author Contributions

Conceptualization, R.L.G., O.L., D.D. and A.B.-T.; methodology, T.G., H.G., V.F. and A.B.-T.; formal analysis, T.G. and A.B.-T.; investigation, T.G., H.G., D.D. and A.B.-T.; resources, N.D.-B., R.L.G. and D.D.; data curation, T.G., H.G. and A.B.-T.; writing—original draft preparation, T.G. and A.B.-T.; writing—review and editing, O.L. and D.D.; visualization, T.G. and A.B.-T.; supervision, R.L.G., O.L., D.D. and A.B.-T.; project administration, N.D.-B. and D.D.; funding acquisition, R.L.G. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by the UMR1184 research centre. This work was supported by the “Programme Investissements d’Avenir” (PIA), managed by the ANR under reference ANR-11-INBS-0008, funding the Infectious Disease Models and Innovative Therapies (IDMIT, Fontenay-aux-Roses, France) infrastructure, and ANR-10-EQPX-02-01, funding the FlowCyTech facility (IDMIT, Fontenay-aux-Roses, France). The animal studies were supported by the ANRS, as part of the SIVART ANRS-IDMIT CO1 research program.

Institutional Review Board Statement

The animal study protocol was approved by the ethics committee “Comité d’éthique en expérimentation animale dn°44” and the French Ministry of Education and Research under reference APAFIS#2453-2015102713323361.

Data Availability Statement

The data presented in this study are available on request from the corresponding author. The data are not publicly available due to privacy.

Acknowledgments

We thank Gilead and ViiV for providing the antiretroviral drugs.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Perelson, A.S.; Essunger, P.; Cao, Y.; Vesanen, M.; Hurley, A.; Saksela, K.; Markowitz, M.; Ho, D.D. Decay Characteristics of HIV-1-Infected Compartments during Combination Therapy. Nature 1997, 387, 188–191. [Google Scholar] [CrossRef] [PubMed]
  2. Henderson, L.J.; Reoma, L.B.; Kovacs, J.A.; Nath, A. Advances toward Curing HIV-1 Infection in Tissue Reservoirs. J. Virol. 2020, 94, e00375-19. [Google Scholar] [CrossRef] [PubMed]
  3. Cory, T.J.; Schacker, T.W.; Stevenson, M.; Fletcher, C.V. Overcoming Pharmacologic Sanctuaries. Curr. Opin. HIV AIDS 2013, 8, 190–195. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Fletcher, C.V.; Staskus, K.; Wietgrefe, S.W.; Rothenberger, M.; Reilly, C.; Chipman, J.G.; Beilman, G.J.; Khoruts, A.; Thorkelson, A.; Schmidt, T.E.; et al. Persistent HIV-1 Replication Is Associated with Lower Antiretroviral Drug Concentrations in Lymphatic Tissues. Proc. Natl. Acad. Sci. USA 2014, 111, 2307–2312. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Lorenzo-Redondo, R.; Fryer, H.R.; Bedford, T.; Kim, E.-Y.; Archer, J.; Kosakovsky Pond, S.L.; Chung, Y.-S.; Penugonda, S.; Chipman, J.G.; Fletcher, C.V.; et al. Persistent HIV-1 Replication Maintains the Tissue Reservoir during Therapy. Nature 2016, 530, 51–56. [Google Scholar] [CrossRef] [Green Version]
  6. Mzingwane, M.L.; Tiemessen, C.T. Mechanisms of HIV Persistence in HIV Reservoirs. Rev. Med. Virol. 2017, 27, e1924. [Google Scholar] [CrossRef]
  7. Deeks, S.G.; Autran, B.; Berkhout, B.; Benkirane, M.; Cairns, S.; Chomont, N.; Chun, T.-W.; Churchill, M.; Mascio, M.D.; Katlama, C.; et al. Towards an HIV Cure: A Global Scientific Strategy. Nat. Rev. Immunol. 2012, 12, 607–614. [Google Scholar] [CrossRef]
  8. Kline, C.; Ndjomou, J.; Franks, T.; Kiser, R.; Coalter, V.; Smedley, J.; Piatak, M.; Mellors, J.W.; Lifson, J.D.; Ambrose, Z. Persistence of Viral Reservoirs in Multiple Tissues after Antiretroviral Therapy Suppression in a Macaque RT-SHIV Model. PLoS ONE 2013, 8, e84275. [Google Scholar] [CrossRef] [Green Version]
  9. Wong, J.K.; Yukl, S.A. Tissue Reservoirs of HIV. Curr. Opin. HIV AIDS 2016, 11, 362–370. [Google Scholar] [CrossRef] [Green Version]
  10. Labarthe, L.; Gelé, T.; Gouget, H.; Benzemrane, M.-S.; Le Calvez, P.; Legrand, N.; Lambotte, O.; Le Grand, R.; Bourgeois, C.; Barrail-Tran, A. Pharmacokinetics and Tissue Distribution of Tenofovir, Emtricitabine and Dolutegravir in Mice. J. Antimicrob. Chemother. 2022, 77, 1094–1101. [Google Scholar] [CrossRef]
  11. Bruel, T.; Hamimi, C.; Dereuddre-Bosquet, N.; Cosma, A.; Shin, S.Y.; Corneau, A.; Versmisse, P.; Karlsson, I.; Malleret, B.; Targat, B.; et al. Long-Term Control of Simian Immunodeficiency Virus (SIV) in Cynomolgus Macaques Not Associated with Efficient SIV-Specific CD8+ T-Cell Responses. J. Virol. 2015, 89, 3542–3556. [Google Scholar] [CrossRef] [Green Version]
  12. Terrade, G.; Huot, N.; Petitdemange, C.; Lazzerini, M.; Orta Resendiz, A.; Jacquelin, B.; Müller-Trutwin, M. Interests of the Non-Human Primate Models for HIV Cure Research. Vaccines 2021, 9, 958. [Google Scholar] [CrossRef]
  13. Harding, J.D. Nonhuman Primates and Translational Research: Progress, Opportunities, and Challenges. ILAR J. 2017, 58, 141–150. [Google Scholar] [CrossRef] [Green Version]
  14. Bender, A.M.; Simonetti, F.R.; Kumar, M.R.; Fray, E.J.; Bruner, K.M.; Timmons, A.E.; Tai, K.Y.; Jenike, K.M.; Antar, A.A.R.; Liu, P.-T.; et al. The Landscape of Persistent Viral Genomes in ART Treated SIV, SHIV, and HIV-2 Infections. Cell Host Microbe 2019, 26, 73–85.e4. [Google Scholar] [CrossRef]
  15. Burgunder, E.; Fallon, J.K.; White, N.; Schauer, A.P.; Sykes, C.; Remling-Mulder, L.; Kovarova, M.; Adamson, L.; Luciw, P.; Garcia, J.V.; et al. Antiretroviral Drug Concentrations in Lymph Nodes: A Cross-Species Comparison of the Effect of Drug Transporter Expression, Viral Infection, and Sex in Humanized Mice, Nonhuman Primates, and Humans. J. Pharmacol. Exp. Ther. 2019, 370, 360–368. [Google Scholar] [CrossRef]
  16. Harper, J.; Ribeiro, S.P.; Chan, C.N.; Aid, M.; Deleage, C.; Micci, L.; Pino, M.; Cervasi, B.; Raghunathan, G.; Rimmer, E.; et al. Interleukin-10 Contributes to Reservoir Establishment and Persistence in SIV-Infected Macaques Treated with Antiretroviral Therapy. J. Clin. Investig. 2022, 132, e155251. [Google Scholar] [CrossRef]
  17. Saag, M.S.; Gandhi, R.T.; Hoy, J.F.; Landovitz, R.J.; Thompson, M.A.; Sax, P.E.; Smith, D.M.; Benson, C.A.; Buchbinder, S.P.; del Rio, C.; et al. Antiretroviral Drugs for Treatment and Prevention of HIV Infection in Adults: 2020 Recommendations of the International Antiviral Society–USA Panel. JAMA 2020, 324, 1651. [Google Scholar] [CrossRef]
  18. Department of Health and Human Services. Guidelines for the Use of Antiretroviral Agents in Adults and Adolescents with HIV; Department of Health and Human Services: Washington, DC, USA, 2021.
  19. European AIDS Clinical Society. European AIDS Clinical Society Guidelines V11; European AIDS Clinical Society: Brussels, Belgium, 2021. [Google Scholar]
  20. Organisation Mondiale de la Santé. Consolidated Guidelines on HIV Prevention, Testing, Treatment, Service Delivery and Monitoring: Recommendations for a Public Health Approach; Organisation Mondiale de la Santé: Geneva, Switzerland, 2021; p. 592. [Google Scholar]
  21. Del Prete, G.Q.; Smedley, J.; Macallister, R.; Jones, G.S.; Li, B.; Hattersley, J.; Zheng, J.; Piatak, M.; Keele, B.F.; Hesselgesser, J.; et al. Short Communication: Comparative Evaluation of Coformulated Injectable Combination Antiretroviral Therapy Regimens in Simian Immunodeficiency Virus-Infected Rhesus Macaques. AIDS Res. Hum. Retrovir. 2016, 32, 163–168. [Google Scholar] [CrossRef] [Green Version]
  22. U.S. Department of Health and Human Services; Food and Drug Administration; Center for Drug Evaluation and Research (CDER). Guidance for Industry|Estimating the Maximum Safe Starting Dose in Initial Clinical Trials for Therapeutics in Adult Healthy Volunteers; Food and Drug Administration: Silver Spring, MD, USA, 2005.
  23. Karlsson, I.; Malleret, B.; Brochard, P.; Delache, B.; Calvo, J.; Le Grand, R.; Vaslin, B. Dynamics of T-Cell Responses and Memory T Cells during Primary Simian Immunodeficiency Virus Infection in Cynomolgus Macaques. J. Virol. 2007, 81, 13456–13468. [Google Scholar] [CrossRef] [Green Version]
  24. Gouget, H.; Noé, G.; Barrail-Tran, A.; Furlan, V. UPLC–MS/MS Method for the Simultaneous Quantification of Bictegravir and 13 Others Antiretroviral Drugs plus Cobicistat and Ritonavir Boosters in Human Plasma. J. Pharm. Biomed. Anal. 2020, 181, 113057. [Google Scholar] [CrossRef]
  25. Cooper, R.D.; Wiebe, N.; Smith, N.; Keiser, P.; Naicker, S.; Tonelli, M. Systematic Review and Meta-Analysis: Renal Safety of Tenofovir Disoproxil Fumarate in HIV-Infected Patients. Clin. Infect. Dis. 2010, 51, 496–505. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Moss, D.M.; Neary, M.; Owen, A. The Role of Drug Transporters in the Kidney: Lessons from Tenofovir. Front. Pharmacol. 2014, 5, 248. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Thompson, C.G.; Rosen, E.P.; Prince, H.M.A.; White, N.; Sykes, C.; de la Cruz, G.; Mathews, M.; Deleage, C.; Estes, J.D.; Charlins, P.; et al. Heterogeneous Antiretroviral Drug Distribution and HIV/SHIV Detection in the Gut of Three Species. Sci. Transl. Med. 2019, 11, eaap8758. [Google Scholar] [CrossRef] [PubMed]
  28. Flynn, P.M.; Mirochnick, M.; Shapiro, D.E.; Bardeguez, A.; Rodman, J.; Robbins, B.; Huang, S.; Fiscus, S.A.; Van Rompay, K.K.A.; Rooney, J.F.; et al. Pharmacokinetics and Safety of Single-Dose Tenofovir Disoproxil Fumarate and Emtricitabine in HIV-1-Infected Pregnant Women and Their Infants. Antimicrob. Agents Chemother. 2011, 55, 5914–5922. [Google Scholar] [CrossRef] [Green Version]
  29. Min, S.; Song, I.; Borland, J.; Chen, S.; Lou, Y.; Fujiwara, T.; Piscitelli, S.C. Pharmacokinetics and Safety of S/GSK1349572, a Next-Generation HIV Integrase Inhibitor, in Healthy Volunteers. Antimicrob. Agents Chemother. 2010, 54, 254–258. [Google Scholar] [CrossRef] [Green Version]
  30. Barditch-Crovo, P.; Deeks, S.G.; Collier, A.; Safrin, S.; Coakley, D.F.; Miller, M.; Kearney, B.P.; Coleman, R.L.; Lamy, P.D.; Kahn, J.O.; et al. Phase i/Ii Trial of the Pharmacokinetics, Safety, and Antiretroviral Activity of Tenofovir Disoproxil Fumarate in Human Immunodeficiency Virus-Infected Adults. Antimicrob. Agents Chemother. 2001, 45, 2733–2739. [Google Scholar] [CrossRef] [Green Version]
  31. Min, S.; Sloan, L.; DeJesus, E.; Hawkins, T.; McCurdy, L.; Song, I.; Stroder, R.; Chen, S.; Underwood, M.; Fujiwara, T.; et al. Antiviral Activity, Safety, and Pharmacokinetics/Pharmacodynamics of Dolutegravir as 10-Day Monotherapy in HIV-1-Infected Adults. AIDS 2011, 25, 1737–1745. [Google Scholar] [CrossRef]
  32. Zong, J.; Chittick, G.E.; Wang, L.H.; Hui, J.; Begley, J.A.; Blum, M.R. Pharmacokinetic Evaluation of Emtricitabine in Combination With Other Nucleoside Antivirals in Healthy Volunteers. J. Clin. Pharmacol. 2007, 47, 877–889. [Google Scholar] [CrossRef]
  33. Han, W.-L.; Shang, J.-C.; Yan, B.; Tan, R.; Huang, W.-X.; Zhong, X.-N.; Yang, J.-Q.; Huang, A.-L. Pharmacokinetics of Single- and Multiple-Dose Emtricitabine in Healthy Male Chinese Volunteers. PHA 2014, 93, 166–171. [Google Scholar] [CrossRef]
  34. Loftsson, T.; Jarho, P.; Másson, M.; Järvinen, T. Cyclodextrins in Drug Delivery. Expert Opin. Drug Deliv. 2005, 2, 335–351. [Google Scholar] [CrossRef]
  35. Yáñez, J.A.; Remsberg, C.M.; Sayre, C.L.; Forrest, M.L.; Davies, N.M. Flip-Flop Pharmacokinetics—Delivering a Reversal of Disposition: Challenges and Opportunities during Drug Development. Ther. Deliv. 2011, 2, 643–672. [Google Scholar] [CrossRef] [Green Version]
  36. Adams, J.L.; Patterson, K.B.; Prince, H.M.A.; Sykes, C.; Greener, B.N.; Dumond, J.B.; Kashuba, A.D.M. Single and Multiple Dose Pharmacokinetics of Dolutegravir in the Genital Tract of HIV-Negative Women. Antivir. Ther. 2013, 18, 1005–1013. [Google Scholar] [CrossRef] [Green Version]
  37. Greener, B.N.; Patterson, K.B.; Prince, H.M.A.; Sykes, C.S.; Adams, J.L.; Dumond, J.B.; Shaheen, N.J.; Madanick, R.D.; Dellon, E.S.; Cohen, M.S.; et al. Dolutegravir Pharmacokinetics in the Genital Tract and Colorectum of HIV-Negative Men after Single and Multiple Dosing. J. Acquir. Immune Defic. Syndr. 2013, 64, 39–44. [Google Scholar] [CrossRef] [Green Version]
  38. Koo, B.-S.; Lee, D.-H.; Kang, P.; Jeong, K.-J.; Lee, S.; Kim, K.; Lee, Y.; Huh, J.-W.; Kim, Y.-H.; Park, S.-J.; et al. Reference Values of Hematological and Biochemical Parameters in Young-Adult Cynomolgus Monkey (Macaca Fascicularis) and Rhesus Monkey (Macaca Mulatta) Anesthetized with Ketamine Hydrochloride. Lab. Anim. Res. 2019, 35, 7. [Google Scholar] [CrossRef] [Green Version]
  39. van Lunzen, J.; Maggiolo, F.; Arribas, J.R.; Rakhmanova, A.; Yeni, P.; Young, B.; Rockstroh, J.K.; Almond, S.; Song, I.; Brothers, C.; et al. Once Daily Dolutegravir (S/GSK1349572) in Combination Therapy in Antiretroviral-Naive Adults with HIV: Planned Interim 48 Week Results from SPRING-1, a Dose-Ranging, Randomised, Phase 2b Trial. Lancet Infect. Dis. 2012, 12, 111–118. [Google Scholar] [CrossRef]
  40. Cahn, P.; Madero, J.S.; Arribas, J.R.; Antinori, A.; Ortiz, R.; Clarke, A.E.; Hung, C.-C.; Rockstroh, J.K.; Girard, P.-M.; Sievers, J.; et al. Dolutegravir plus Lamivudine versus Dolutegravir plus Tenofovir Disoproxil Fumarate and Emtricitabine in Antiretroviral-Naive Adults with HIV-1 Infection (GEMINI-1 and GEMINI-2): Week 48 Results from Two Multicentre, Double-Blind, Randomised, Non-Inferiority, Phase 3 Trials. Lancet 2019, 393, 143–155. [Google Scholar] [CrossRef]
  41. Reese, M.J.; Savina, P.M.; Generaux, G.T.; Tracey, H.; Humphreys, J.E.; Kanaoka, E.; Webster, L.O.; Harmon, K.A.; Clarke, J.D.; Polli, J.W. In Vitro Investigations into the Roles of Drug Transporters and Metabolizing Enzymes in the Disposition and Drug Interactions of Dolutegravir, a HIV Integrase Inhibitor. Drug Metab. Dispos. 2013, 41, 353–361. [Google Scholar] [CrossRef] [Green Version]
  42. Urakami, Y.; Kimura, N.; Okuda, M.; Inui, K. Creatinine Transport by Basolateral Organic Cation Transporter HOCT2 in the Human Kidney. Pharm. Res. 2004, 21, 976–981. [Google Scholar] [CrossRef]
  43. Tanihara, Y.; Masuda, S.; Sato, T.; Katsura, T.; Ogawa, O.; Inui, K.-I. Substrate Specificity of MATE1 and MATE2-K, Human Multidrug and Toxin Extrusions/H+-Organic Cation Antiporters. Biochem. Pharmacol. 2007, 74, 359–371. [Google Scholar] [CrossRef] [Green Version]
  44. Van Rompay, K.K.A.; Durand-Gasselin, L.; Brignolo, L.L.; Ray, A.S.; Abel, K.; Cihlar, T.; Spinner, A.; Jerome, C.; Moore, J.; Kearney, B.P.; et al. Chronic Administration of Tenofovir to Rhesus Macaques from Infancy through Adulthood and Pregnancy: Summary of Pharmacokinetics and Biological and Virological Effects. Antimicrob. Agents Chemother. 2008, 52, 3144–3160. [Google Scholar] [CrossRef]
  45. Lemaitre, J.; Desjardins, D.; Gallouët, A.-S.; Gomez-Pacheco, M.; Bourgeois, C.; Favier, B.; Sáez-Cirión, A.; Le Grand, R.; Lambotte, O. Expansion of Immature Neutrophils During SIV Infection Is Associated With Their Capacity to Modulate T-Cell Function. Front. Immunol. 2022, 13, 781356. [Google Scholar] [CrossRef] [PubMed]
  46. Mausoléo, A.; Olivo, A.; Desjardins, D.; Sáez-Cirión, A.; Barrail-Tran, A.; Avettand-Fenoel, V.; Noël, N.; Lagathu, C.; Béréziat, V.; Le Grand, R.; et al. Prolonged Antiretroviral Treatment Induces Adipose Tissue Remodelling Associated with Mild Inflammation in SIV-Infected Macaques. Cells 2022, 11, 3104. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Tenofovir, emtricitabine, and dolutegravir total plasma pharmacokinetic profiles after a single dose. For the subcutaneous (SC, red) and intravenous (IV, blue) routes, the continuous lines and coloured areas represent the median and interquartile range, respectively. Individual data are represented for the oral (PO, green) route. Abbreviations: IC90: wild-type HIV-1 90% inhibitory concentration, PA-IC90: protein-adjusted wild-type HIV-1 90% inhibitory concentration.
Figure 1. Tenofovir, emtricitabine, and dolutegravir total plasma pharmacokinetic profiles after a single dose. For the subcutaneous (SC, red) and intravenous (IV, blue) routes, the continuous lines and coloured areas represent the median and interquartile range, respectively. Individual data are represented for the oral (PO, green) route. Abbreviations: IC90: wild-type HIV-1 90% inhibitory concentration, PA-IC90: protein-adjusted wild-type HIV-1 90% inhibitory concentration.
Pharmaceutics 14 02282 g001
Figure 2. Tenofovir, emtricitabine, and dolutegravir trough plasma concentrations during long-term treatment of SIV+ macaques. The continuous lines and coloured areas represent the median and interquartile range, respectively. Oral (PO) route: green, subcutaneous (SC) route: red. Abbreviations: IC90: wild-type HIV-1 90% inhibitory concentration, PA-IC90: protein-adjusted wild-type HIV-1 90% inhibitory concentration.
Figure 2. Tenofovir, emtricitabine, and dolutegravir trough plasma concentrations during long-term treatment of SIV+ macaques. The continuous lines and coloured areas represent the median and interquartile range, respectively. Oral (PO) route: green, subcutaneous (SC) route: red. Abbreviations: IC90: wild-type HIV-1 90% inhibitory concentration, PA-IC90: protein-adjusted wild-type HIV-1 90% inhibitory concentration.
Pharmaceutics 14 02282 g002
Figure 3. Steady-state trough concentrations depending on the route of administration and the intra- and inter-individual variability. The boxplot graphs represent trough concentrations (left), intra-individual variability (middle), and inter-individual variability (right); continuous lines represent the median and interquartile range values. Statistically significant differences are indicated as follows: * p < 0.05, ** p < 0.01, *** p < 0.001, ns: non-significant. Abbreviations: TNF: tenofovir, FTC: emtricitabine, DTG: dolutegravir, PO: oral route, SC: subcutaneous route.
Figure 3. Steady-state trough concentrations depending on the route of administration and the intra- and inter-individual variability. The boxplot graphs represent trough concentrations (left), intra-individual variability (middle), and inter-individual variability (right); continuous lines represent the median and interquartile range values. Statistically significant differences are indicated as follows: * p < 0.05, ** p < 0.01, *** p < 0.001, ns: non-significant. Abbreviations: TNF: tenofovir, FTC: emtricitabine, DTG: dolutegravir, PO: oral route, SC: subcutaneous route.
Pharmaceutics 14 02282 g003
Table 1. Tenofovir/emtricitabine/dolutegravir single-dose pharmacokinetic parameters.
Table 1. Tenofovir/emtricitabine/dolutegravir single-dose pharmacokinetic parameters.
Cmax (ng/mL)Tmax (h)C24h (ng/mL)t½ (h)AUC0→24h (ng·h/mL)
Tenofovir
PO
(n = 2)
1668
231
1.0
6.0
272
88
31.0
14.2
11,115
3857
SC
(n = 7)
1103
(954–1654)
1.0
(1.0–2.0)
32
(16–50)
4.7
(1.2–6.1)
4391
(2326–8598)
IV
(n = 4)
2096
(1583–2352)
0.25
(0.25–0.25)
21
(12–38)
5.1
(1.4–14.1)
1983
(1825–2570)
Emtricitabine
PO
(n = 2)
5062
2957
1.0
4.0
632
87
9.6
4.8
42,768
19,280
SC
(n = 7)
25,882
(19,607–28,634)
1.0
(1.0–1.0)
44
(29–61)
3.1
(2.6–3.2)
79,578
(76,106–96,001)
IV
(n = 4)
67,732
(62,920–70,569)
0.25
(0.25–0.25)
58
(54–127)
3.0
(2.8–3.2)
111,530
(106,512–116,484)
Dolutegravir
PO
(n = 2)
5105
2550
6.0
4.0
1109
113
8.2
4.1
72,426
23,956
SC
(n = 7)
1229
(1082–1467)
1.0
(1.0–2.0)
224
(180–281)
9.2
(8.8–9.7)
15,352
(11,401–16,540)
IV
(n = 4)
11,643
(10,995–12,583)
0.25
(0.25–0.25)
86
(82–102)
3.8
(3.7–4.2)
39,601
(37,865–42,241)
Variables are expressed as medians and interquartile ranges (IQR) when n ≥ 3. For the oral (PO) route, individual values are expressed. Abbreviations: PO: oral route, SC: subcutaneous route, IV: intravenous route, Cmax: maximal concentration, Cmax values for intravenous administration were observations at 0.25 h (as first point of blood sampling), Tmax: time to reach Cmax, C24h: 24-h post-dose concentration, t½: half-life, AUC0→24h: area under the curve until 24 h post dose.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Gelé, T.; Gouget, H.; Dereuddre-Bosquet, N.; Furlan, V.; Le Grand, R.; Lambotte, O.; Desjardins, D.; Barrail-Tran, A. Pharmacological Validation of Long-Term Treatment with Antiretroviral Drugs in a Model of SIV-Infected Non-Human Primates. Pharmaceutics 2022, 14, 2282. https://doi.org/10.3390/pharmaceutics14112282

AMA Style

Gelé T, Gouget H, Dereuddre-Bosquet N, Furlan V, Le Grand R, Lambotte O, Desjardins D, Barrail-Tran A. Pharmacological Validation of Long-Term Treatment with Antiretroviral Drugs in a Model of SIV-Infected Non-Human Primates. Pharmaceutics. 2022; 14(11):2282. https://doi.org/10.3390/pharmaceutics14112282

Chicago/Turabian Style

Gelé, Thibaut, Hélène Gouget, Nathalie Dereuddre-Bosquet, Valérie Furlan, Roger Le Grand, Olivier Lambotte, Delphine Desjardins, and Aurélie Barrail-Tran. 2022. "Pharmacological Validation of Long-Term Treatment with Antiretroviral Drugs in a Model of SIV-Infected Non-Human Primates" Pharmaceutics 14, no. 11: 2282. https://doi.org/10.3390/pharmaceutics14112282

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop