Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Infect Dis ; 215(1): 95-104, 2017 Jan 01.
Article in English | MEDLINE | ID: mdl-28077588

ABSTRACT

BACKGROUND: We report the first-in-human safety and immunogenicity assessment of a prototype intranasally administered, replication-competent Sendai virus (SeV)-vectored, human immunodeficiency virus type 1 (HIV-1) vaccine. METHODS: Sixty-five HIV-1-uninfected adults in Kenya, Rwanda, and the United Kingdom were assigned to receive 1 of 4 prime-boost regimens (administered at 0 and 4 months, respectively; ratio of vaccine to placebo recipients, 12:4): priming with a lower-dose SeV-Gag given intranasally, followed by boosting with an adenovirus 35-vectored vaccine encoding HIV-1 Gag, reverse transcriptase, integrase, and Nef (Ad35-GRIN) given intramuscularly (SLA); priming with a higher-dose SeV-Gag given intranasally, followed by boosting with Ad35-GRIN given intramuscularly (SHA); priming with Ad35-GRIN given intramuscularly, followed by boosting with a higher-dose SeV-Gag given intranasally (ASH); and priming and boosting with a higher-dose SeV-Gag given intranasally (SHSH). RESULTS: All vaccine regimens were well tolerated. Gag-specific IFN-γ enzyme-linked immunospot-determined response rates and geometric mean responses were higher (96% and 248 spot-forming units, respectively) in groups primed with SeV-Gag and boosted with Ad35-GRIN (SLA and SHA) than those after a single dose of Ad35-GRIN (56% and 54 spot-forming units, respectively) or SeV-Gag (55% and 59 spot-forming units, respectively); responses persisted for ≥8 months after completion of the prime-boost regimen. Functional CD8+ T-cell responses with greater breadth, magnitude, and frequency in a viral inhibition assay were also seen in the SLA and SHA groups after Ad35-GRIN boost, compared with those who received either vaccine alone. SeV-Gag did not boost T-cell counts in the ASH group. In contrast, the highest Gag-specific antibody titers were seen in the ASH group. Mucosal antibody responses were sporadic. CONCLUSIONS: SeV-Gag primed functional, durable HIV-specific T-cell responses and boosted antibody responses. The prime-boost sequence appears to determine which arm of the immune response is stimulated. CLINICAL TRIALS REGISTRATION: NCT01705990.


Subject(s)
AIDS Vaccines/adverse effects , AIDS Vaccines/immunology , CD8-Positive T-Lymphocytes/immunology , HIV Infections/prevention & control , HIV-1/immunology , Sendai virus/genetics , Vaccines, DNA/adverse effects , Vaccines, DNA/immunology , AIDS Vaccines/administration & dosage , AIDS Vaccines/genetics , Administration, Intranasal , Adult , Female , Genes, Viral/immunology , Genetic Vectors , HIV Antibodies/blood , HIV Antibodies/immunology , HIV Infections/immunology , HIV-1/genetics , Humans , Immunity, Cellular , Immunity, Humoral , Immunization, Secondary , Immunogenicity, Vaccine , Kenya , Male , Middle Aged , Rwanda , Sendai virus/immunology , Sendai virus/physiology , United Kingdom , Vaccines, DNA/administration & dosage , Virus Replication
2.
J Infect Dis ; 213(6): 1013-7, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26563240

ABSTRACT

The injectable long-acting formulation of rilpivirine (TMC278LA) is a promising preexposure prophylaxis (PrEP) candidate for prevention of human immunodeficiency virus type 1 (HIV-1) infection. We evaluated HIV-1 in plasma obtained from an unexpected seroconverter in the 300-mg arm of the SSAT040 TMC278LA pharmacokinetic study for rilpivirine (RPV) resistance. Infection with wild-type HIV-1 was confirmed on day 84 after TMC278LA injection, and the K101E mutation was detected on day 115. Plasma-derived HIV-1 clones containing K101E had 4-fold increased resistance to RPV and 4-8-fold increased cross-resistance to etravirine, nevirapine, and efavirenz compared with wild type HIV-1 plasma-derived clones from the same individual. This case is a unique instance of infection with wild-type HIV-1 and subsequent selection of resistant virus by persistent exposure to long-acting PrEP.


Subject(s)
Anti-HIV Agents/therapeutic use , HIV Infections/drug therapy , HIV Infections/virology , HIV-1/drug effects , Rilpivirine/therapeutic use , Adult , Anti-HIV Agents/administration & dosage , Anti-HIV Agents/pharmacology , Delayed-Action Preparations , Drug Resistance, Viral , Female , Humans , Rilpivirine/administration & dosage , Rilpivirine/pharmacology , Selection, Genetic
3.
J Antimicrob Chemother ; 71(4): 1031-6, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26679246

ABSTRACT

OBJECTIVES: To evaluate dolutegravir and elvitegravir/cobicistat pharmacokinetics in HIV-negative volunteers up to 10 days after drug cessation. METHODS: Healthy volunteers received 50 mg of dolutegravir once-daily for 10 days, then underwent a 9 day wash-out period, and then received elvitegravir/cobicistat as part of Stribild(®) (245 mg of tenofovir, 200 mg of emtricitabine, 150 mg of elvitegravir and 150 mg of cobicistat) for 10 days. Serial pharmacokinetic (PK) sampling occurred prior to the final dose of each course and at regular intervals for up to 216 h (10 days) after drug cessation. Concentrations were determined by LC-MS/MS, and PK parameters were illustrated as geometric mean and 90% CI. RESULTS: Seventeen volunteers completed the study. For dolutegravir, plasma terminal elimination t1/2 to the last measurable concentration (within 216 h) was longer than its t1/2 within the dosing interval (0-24 h): 14.3 h (12.9-15.7 h) versus 23.1 h (19.7-26.6 h); conversely, the terminal elimination t1/2 for elvitegravir was lower than its t1/2 within the dosing interval (0-24 h): 10.8 h (9.7-13.0 h) versus 5.2 h (4.7-6.1 h). Dolutegravir concentrations were above the protein-adjusted (PA) IC90 (64 ng/mL) in 100% of subjects after 36 and 48 h and in 94% after 60 and 72 h. All subjects had detectable dolutegravir concentrations at 96 h, a mean of 23.5% above the IC90. Elvitegravir concentrations were above the PA IC95 (45 ng/mL) in 100% of subjects at 24 h, 65% at 36 h but 0% after 48 h. CONCLUSIONS: Our data show marked differences in the elimination rates of dolutegravir and elvitegravir following treatment interruption, which is likely to impact the extent to which drug doses can be delayed or missed. They suggest that clinical differences may emerge in patients who have suboptimal adherence.


Subject(s)
Anti-HIV Agents/pharmacokinetics , Heterocyclic Compounds, 3-Ring/pharmacokinetics , Quinolones/pharmacokinetics , Adult , Anti-HIV Agents/administration & dosage , Drug Monitoring , Female , Heterocyclic Compounds, 3-Ring/administration & dosage , Humans , Male , Middle Aged , Oxazines , Piperazines , Pyridones , Quinolones/administration & dosage , Time Factors
4.
Antimicrob Agents Chemother ; 59(10): 6080-6, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26195515

ABSTRACT

Pharmacokinetic (PK) data describing a prolonged time course of antiretrovirals in plasma and peripheral blood mononuclear cells (PBMCs) are important for understanding and managing late or missed doses and to assess the appropriateness of compounds for preexposure prophylaxis (PrEP). This study aimed to evaluate the PK of coformulated tenofovir disoproxil fumarate (DF), emtricitabine, and rilpivirine in plasma and of the intracellular (IC) anabolites tenofovir diphosphate (TFV-DP) and emtricitabine triphosphate (FTC-TP) in healthy volunteers up to 9 days after drug cessation. Individuals received daily tenofovir DF-emtricitabine-rilpivirine (245/200/25 mg) for 14 days. Drug intake was stopped, and serial sampling occurred prior to the final dose and up to 216 h (9 days) after stopping drug intake. Concentrations were quantified and PK parameters calculated. Eighteen volunteers completed the study. The terminal elimination plasma half-lives for tenofovir and emtricitabine over 216 h (geometric mean [90% confidence interval]) were higher than those seen over 0 to 24 h (for tenofovir, 31 h [27 to 40 h] versus 13.3 h [12.5 to 15.1 h]; for emtricitabine, 41 h [36 to 54 h] versus 6.4 h (5.9 to 7.6 h]). Model-predicted IC half-lives (0 to 168 h) were 116 h (TFV-DP) and 37 h (FTC-TP). The plasma rilpivirine concentration at 216 h was 4.5 ng/ml (4.2 to 6.2 ng/ml), and half-lives over 0 to 216 h and 0 to 24 h were 47 h (41 to 59 h) and 35 h (28 to 46 h), respectively. These data contribute to our understanding of drug behavior following treatment interruption; however, adherence to therapy should be promoted. Validated plasma and IC target concentrations are necessary to allow interpretation with respect to sustained virus suppression or HIV prevention. (The trial was conducted in accordance with the Declaration of Helsinki [EudraCT 2012-002781-13].).


Subject(s)
Anti-HIV Agents/blood , Emtricitabine/blood , Rilpivirine/blood , Tenofovir/blood , Adenine/analogs & derivatives , Adenine/blood , Adenine/pharmacokinetics , Adenine/therapeutic use , Adolescent , Adult , Aged , Anti-HIV Agents/pharmacokinetics , Anti-HIV Agents/therapeutic use , Emtricitabine/pharmacokinetics , Emtricitabine/therapeutic use , Female , HIV Infections/drug therapy , HIV Infections/prevention & control , Humans , Middle Aged , Organophosphates/blood , Organophosphates/pharmacokinetics , Organophosphates/therapeutic use , Rilpivirine/pharmacokinetics , Rilpivirine/therapeutic use , Tenofovir/pharmacokinetics , Tenofovir/therapeutic use , Young Adult
5.
HIV Clin Trials ; 16(1): 39-42, 2015.
Article in English | MEDLINE | ID: mdl-25777188

ABSTRACT

BACKGROUND: Antiretroviral safety and efficacy and may differ in older versus younger HIV-infected patients. The objective of this study was to assess the pharmacokinetic (PK) profile in older HIV-infected subjects (>60 years) switching combination antiretroviral therapy (cART) to a raltegravir (RAL) containing regimen. METHODS: Nineteen HIV-infected patients over 60 years of age on effective cART (HIV-RNA < 50 copies/ml) were enrolled in this prospective 24-week study. On day 1, patients switched to tenofovir/emtricitabine (245/200 mg once daily) and RAL (400 mg twice daily). On day 28, intensive PK sampling was undertaken in a fasted state and RAL plasma concentrations determined. Neurocognitive function was assessed at baseline and week 24 using a neuropsychological battery. RAL PK parameters were compared to those of two younger historical HIV-infected control groups that received twice-daily RAL co-administered with darunavir/ritonavir (DRV/r) 800/100 once daily by nonlinear mixed effects modelling. RESULTS: In HIV-infected subjects over the age of 60 (mean ± SD age: 66 ± 3.4 years, n = 19) switching to a RAL containing regimen, we observed no safety concerns, no plasma virological rebounds, and no differences in RAL apparent oral clearance when compared to younger HIV-infected populations (mean ± SD age: 41 ± 9.2 years, n = 38) based on population pharmacokinetic analysis. After 24 weeks of study therapy a decline in cognitive function was observed [change in (SD) global score of (0.91 (1.3), P = 0.018]. CONCLUSIONS: No significant changes in RAL exposure associated with age were observed.


Subject(s)
Anti-HIV Agents/pharmacokinetics , Emtricitabine/pharmacokinetics , HIV Infections/drug therapy , Raltegravir Potassium/pharmacokinetics , Tenofovir/pharmacokinetics , Adult , Anti-HIV Agents/pharmacology , Area Under Curve , CD4 Lymphocyte Count , Drug Therapy, Combination , Emtricitabine/pharmacology , Female , HIV Infections/virology , HIV-1/drug effects , Humans , Male , Mental Status and Dementia Tests , Middle Aged , Prospective Studies , Raltegravir Potassium/pharmacology , Tenofovir/pharmacology , United Kingdom , Viral Load/drug effects
6.
Clin Infect Dis ; 59 Suppl 1: S52-4, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24926035

ABSTRACT

Pharmacological studies in the context of preexposure prophylaxis (PrEP) for human immunodeficiency virus (HIV) are fundamental to inform on different drug pharmacokinetics, pharmacodynamics, and pharmacogenetics in view of the absence of easily measurable surrogate markers of efficacy. Although the combination of tenofovir and emtricitabine is the only PrEP agent that was studied and showed efficacy in preventing HIV transmission, prospective randomized clinical trials have reported varying efficacy due to poor adherence to the drug. Importantly, this could be overcome by the introduction of long-acting injectable PrEP agents, which may be administered monthly and ensure optimal and prolonged drug exposure in HIV target tissues. Notably, clinical pharmacology studies play a central role in interpreting drug concentration-responses and optimal drug exposure achievement.


Subject(s)
Anti-HIV Agents/therapeutic use , Chemoprevention , HIV Infections/drug therapy , Humans
7.
J Antimicrob Chemother ; 69(7): 1911-5, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24610312

ABSTRACT

BACKGROUND: St John's wort (SJW; Hypericum perforatum) induces CYP3A4 that is involved in the metabolism of the hepatitis C virus (HCV) protease inhibitor boceprevir. Reduced boceprevir exposure and efficacy would contribute to therapeutic failure and increase the risk for resistance development. Boceprevir is co-administered with interferon/ribavirin, and depression has been described frequently in patients undergoing HCV treatment. Patients may purchase over-the-counter herbals to manage depression, and knowing the interaction between SJW and boceprevir is desirable. METHODS: This Phase I, open-label, three-period, cross-over pharmacokinetic study enrolled healthy males and females who, following consent and screening procedures, were randomized to receive SJW on days 1-14, SJW plus boceprevir (SJW on days 22-35 and together on days 31-35) and boceprevir on days 52-56, separated by 7 day washout periods, or the same treatment in the opposite order. Pharmacokinetic sampling was performed at the end of each phase. RESULTS: Seventeen (11 female) subjects completed the study and no serious adverse events were reported. Geometric mean ratios (GMRs) and 90% CIs for boceprevir (with SJW versus alone) AUC(0-8), C(max) and C8 were 0.91 (0.87-0.96), 0.94 (0.82-1.07) and 1.00 (0.79-1.27), respectively. GMRs and 90% CIs for hypericin, the active component of SJW, (with boceprevir versus alone) AUC(0-8), C(max) and C(8) were 1.23 (1.10-1.38), 1.32 (1.16-1.52) and 1.37 (1.19-1.58), respectively. CONCLUSIONS: SJW did not have a clinically significant effect on boceprevir plasma concentrations (or those of its metabolite), suggesting that SJW and boceprevir can be safely co-administered.


Subject(s)
Antiviral Agents/pharmacokinetics , Enzyme Activation , Hypericum , Plant Extracts/pharmacokinetics , Proline/analogs & derivatives , Adult , Antiviral Agents/administration & dosage , Cross-Over Studies , Drug Interactions , Female , Healthy Volunteers , Humans , Male , Middle Aged , Plant Extracts/administration & dosage , Plasma/chemistry , Proline/administration & dosage , Proline/pharmacokinetics
8.
Front Immunol ; 15: 1400553, 2024.
Article in English | MEDLINE | ID: mdl-38817615

ABSTRACT

Background and aims: Metabolic dysfunction-associated steatohepatitis (MASH) is a significant health concern with limited treatment options. AXL, a receptor tyrosine kinase activated by the GAS6 ligand, promotes MASH through activation of hepatic stellate cells and inflammatory macrophages. This study identified cell subsets affected by MASH progression and the effect of AXL inhibition. Methods: Mice were fed chow or different fat-enriched diets to induce MASH, and small molecule AXL kinase inhibition with bemcentinib was evaluated. Gene expression was measured by qPCR. Time-of-flight mass cytometry (CyTOF) used single cells from dissociated livers, acquired on the Fluidigm Helios, and cell populations were studied using machine learning. Results: In mice fed different fat-enriched diets, liver steatosis alone was insufficient to elevate plasma soluble AXL (sAXL) levels. However, in conjunction with inflammation, sAXL increases, serving as an early indicator of steatohepatitis progression. Bemcentinib, an AXL inhibitor, effectively reduced proinflammatory responses in MASH models, even before fibrosis appearance. Utilizing CyTOF analysis, we detected a decreased population of Kupffer cells during MASH while promoting infiltration of monocytes/macrophages and CD8+ T cells. Bemcentinib partially restored Kupffer cells, reduced pDCs and GzmB- NK cells, and increased GzmB+CD8+ T cells and LSECs. Additionally, AXL inhibition enhanced a subtype of GzmB+CD8+ tissue-resident memory T cells characterized by CX3CR1 expression. Furthermore, bemcentinib altered the transcriptomic landscape associated with MASH progression, particularly in TLR signaling and inflammatory response, exhibiting differential cytokine expression in the plasma, consistent with liver repair and decreased inflammation. Conclusion: Our findings highlight sAXL as a biomarker for monitoring MASH progression and demonstrate that AXL targeting shifted liver macrophages and CD8+ T-cell subsets away from an inflammatory phenotype toward fibrotic resolution and organ healing, presenting a promising strategy for MASH treatment.


Subject(s)
Axl Receptor Tyrosine Kinase , Liver Cirrhosis , Proto-Oncogene Proteins , Receptor Protein-Tyrosine Kinases , Animals , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/metabolism , Mice , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/metabolism , Liver Cirrhosis/immunology , Liver Cirrhosis/drug therapy , Liver Cirrhosis/pathology , Liver Cirrhosis/metabolism , Male , Disease Models, Animal , Mice, Inbred C57BL , Benzocycloheptenes/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Liver/pathology , Liver/immunology , Liver/metabolism , Liver/drug effects , Macrophages/immunology , Macrophages/metabolism , Macrophages/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , Triazoles
9.
Antimicrob Agents Chemother ; 57(8): 3640-4, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23689708

ABSTRACT

Atazanavir (ATV) causes an elevation of unconjugated hyperbilirubinemia (HBR) as a result of UDP glucuronyltransferase (UGT) 1A1 inhibition. Zinc sulfate (ZnSO4) reduces unconjugated hyperbilirubinemia in individuals with Gilbert's syndrome. We assessed the changes in total, conjugated, and unconjugated bilirubin and the effect on ATV pharmacokinetics (PK) after single and 14-day dosing of ZnSO(4). HIV patients, stable on ATV/ritonavir (ATV/r)-containing regimens with a total bilirubin level of >25 mmol/liter received 125 mg daily of ZnSO(4) as Solvazinc tablets for 14 days. ATV/r and bilirubin concentrations were measured pre-ATV/r dose and 2, 4, 6, 8, and 24 h post-ATV/r dose; before ZnSO4 initiation (phase 1), after a single dose (phase 2) and after 14 days (phase 3). Changes in bilirubin and ATV/r concentrations in the absence or presence of ZnSO4 were evaluated by geometric mean ratios (GMRs) and 90% confidence intervals (CIs; we used phase 1 as a reference). Sixteen male patients completed the study maintaining virologic suppression; ZnSO(4) was well tolerated. Statistically significant declines in total bilirubin C(max) and AUC(0-24) of 16 and 17% were seen in phase2 and 20% in phase 3. Although there were no significant changes in conjugated bilirubin, unconjugated bilirubin Cmax and AUC(0-24) of were lower (17 and 19%, phase 2; 20 and 23% during phase 3). The ATV GMRs (90% CI) for C(trough), C(max), and AUC(0-24) were 0.74 (0.62 to 0.89), 0.82 (0.70 to 0.97), and 0.78 (0.70 to 0.88). Intake of ZnSO(4) decreases total and unconjugated bilirubin and causes modest declines in ATV exposure. ZnSO(4) supplementation may be useful in management of ATV-related HBR in selected patients.


Subject(s)
HIV Infections/drug therapy , Hyperbilirubinemia/prevention & control , Oligopeptides/administration & dosage , Pyridines/administration & dosage , Ritonavir/pharmacokinetics , Zinc Sulfate/therapeutic use , Adolescent , Adult , Aged , Anti-HIV Agents/administration & dosage , Anti-HIV Agents/pharmacokinetics , Area Under Curve , Atazanavir Sulfate , Bilirubin/analysis , Confidence Intervals , Cross-Over Studies , Drug Administration Schedule , Drug Therapy, Combination , Drug Tolerance , Female , HIV-1 , Humans , Hyperbilirubinemia/drug therapy , Male , Middle Aged , Oligopeptides/pharmacokinetics , Pyridines/pharmacokinetics , Ritonavir/administration & dosage , Young Adult , Zinc Sulfate/administration & dosage
10.
Antimicrob Agents Chemother ; 56(3): 1427-33, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22183172

ABSTRACT

There is interest in evaluating the efficacy of lower doses of certain antiretrovirals for clinical care. We determined here the bioequivalence of plasma lamivudine (3TC) and intracellular 3TC-triphosphate (3TC-TP) concentrations after the administration of two different doses. ENCORE 2 was a randomized crossover study. Subjects received 3TC at 300 and 150 mg once daily for 10 days (arm 1; n = 13) or vice versa (arm 2; n = 11), separated by a 10-day washout. Pharmacokinetic (PK) profiles (0 to 24 h) were assessed on days 10 and 30. Plasma 3TC and 3TC-TP levels in peripheral blood mononuclear cells were quantified by high-performance liquid chromatography-tandem mass spectrometry. Within-subject changes in PK parameters (the area under the concentration-time curve from 0 to 24 h [AUC(0-24)], the trough concentration of drug in plasma at 24 h [C(24)], and the maximum concentration of drug in plasma [C(max)]) were evaluated by determining the geometric mean ratios (GMRs) adjusted for study arm, period, and intra-individual variation. Regimens were considered bioequivalent if the 90% confidence interval (90% CI) fell within the range of 0.8 to 1.25. A total of 24 subjects completed the study. The GM (90% CI) 3TC AUC(0-24)), expressed as ng·h/ml, for the 300- and 150-mg doses were 8,354 (7,609 to 9,172) and 4,773 (4,408 to 5,169), respectively. Bioequivalence in 3TC PK following the administration of 300 and 150 mg was not demonstrated: the GMRs for AUC(0-24), C(24), and C(max) were 0.57 (0.55 to 0.60), 0.63 (0.59 to 0.67), and 0.56 (0.53 to 0.60), respectively. The GM (90% CI) 3TC-TP AUC(0-24) values (pmol·h/10(6) cells) for the 300- and 150-mg doses were 59.5 (51.8 to 68.3) and 44.0 (38.0 to 51.0), respectively. Bioequivalence in 3TC-TP PK following the administration of 300 and 150 mg was not demonstrated: the GMRs for AUC(0-24), C(24), and C(max) were 0.73 (0.64 to 0.83), 0.82 (0.68 to 0.99), and 0.70 (0.61 to 0.82), respectively. We found that 3TC at 150 mg is not bioequivalent to the standard regimen of 300 mg, indicating that saturation of cytosine phosphorylation pathways is not achieved at a dose of 150 mg.


Subject(s)
Cytidine Triphosphate/analogs & derivatives , Dideoxynucleotides/pharmacokinetics , Lamivudine/analogs & derivatives , Reverse Transcriptase Inhibitors/pharmacokinetics , Administration, Oral , Adolescent , Adult , Aged , Area Under Curve , Biological Availability , Chromatography, High Pressure Liquid , Cross-Over Studies , Cytidine Triphosphate/blood , Cytidine Triphosphate/pharmacokinetics , Dideoxynucleotides/blood , Drug Administration Schedule , Female , Humans , Lamivudine/blood , Lamivudine/pharmacokinetics , Leukocytes, Mononuclear/metabolism , Male , Middle Aged , Reverse Transcriptase Inhibitors/blood , Tandem Mass Spectrometry , Therapeutic Equivalency , United Kingdom , Young Adult
11.
Mol Med ; 18: 1240-8, 2012 Oct 24.
Article in English | MEDLINE | ID: mdl-22875102

ABSTRACT

Maraviroc (MVC) is the first licensed antiretroviral therapeutic agent to target a host cell surface molecule, and successful HIV-1 entry blockade by this C-C chemokine receptor type 5 (CCR5)-antagonist potentiates immunomodulation. We hypothesized that MVC intensification impacts immunization responses, T-cell phenotype, function and delayed type hypersensitivity (DTH) in HIV-1(+) subjects. A 24-wk, double-blinded, placebo-controlled study of the addition of MVC to suppressive antiretroviral therapy in HIV-1(+) persons was performed. Subjects received DTH tests, intramuscular tetanus, meningococcal and oral cholera immunizations. Antibody titers, T-cell function and phenotype were assessed. Of 157 patients referred, 47 were randomized 1:1; MVC:placebo. MVC enhanced meningococcal neo-immunization, blunted cholera response and expedited lymphoproliferation to tetanus boost, without affecting recall humoral response. Anti-HIV-1 group-specific antigen (Gag) and tetanus toxoid (TTox) function improved significantly, HIV-1-associated CD8 T-cell skewing normalized, and the percentage of late-stage and major histocompatibility complex (MHC) class II expressing CD4 T-cells increased. Activated CD4(+) CD38(+) human leukocyte antigen (HLA)-DR(+) T-cells declined, and costimulation shifted to coinhibition. DTH was unchanged. Maraviroc intensification, through antagonism of the cell surface molecule CCR5, favorably influences immune profiles of HIV-1(+) patients, supporting its immunomodulatory use in HIV-1 infection and potentially in other immunologically relevant settings.


Subject(s)
AIDS Vaccines/immunology , CCR5 Receptor Antagonists , HIV Infections/immunology , HIV Infections/prevention & control , HIV-1/immunology , Immunity/immunology , Vaccination , Cyclohexanes/adverse effects , Cyclohexanes/pharmacology , Female , HIV-1/drug effects , Humans , Immunity/drug effects , Immunity, Cellular/drug effects , Immunity, Cellular/immunology , Immunity, Humoral/drug effects , Immunity, Humoral/immunology , Immunophenotyping , Male , Maraviroc , Middle Aged , Receptors, CCR5/metabolism , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Treatment Outcome , Triazoles/adverse effects , Triazoles/pharmacology , Vaccination/adverse effects
12.
Antimicrob Agents Chemother ; 55(9): 4218-23, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21709075

ABSTRACT

The object of this study was to investigate the pharmacokinetics of darunavir-ritonavir and atazanavir-ritonavir once-daily dosing over 72 h (h) following drug intake cessation. Volunteers received darunavir-ritonavir at 800 and 100 mg, respectively, once daily for 10 days, followed by a 7-day washout period, and atazanavir-ritonavir at 300 and 100 mg, respectively, once daily for 10 days. Full pharmacokinetic profiles were assessed for each phase for the 72 h following day 10. Pharmacokinetic parameters were determined over 24 h and to the last measurable concentration by noncompartmental methods. Seventeen subjects completed the study. The geometric mean (GM) terminal elimination half-life to 72 h of darunavir was 6.48 h, which was lower than the 0- to 24-h half-life (10.70 h). The terminal elimination half-life of atazanavir was 6.74 h, which was lower than the 0- to 24-h half-life (13.72 h). All subjects but one had darunavir concentrations higher than the target of 550 ng/ml for protease-resistant HIV isolates (equivalent to 10 times the protein-binding-corrected 50% inhibitory concentration [IC(50)] for wild-type virus) at 24 h postdose, and 14 out of 17 had concentrations higher than the target at 30 h postdose (GM of 1,088 and 851 ng/ml). All subjects had atazanavir concentrations above the suggested minimum effective concentration of 150 ng/ml (equivalent to 10 times the protein-binding-corrected IC(50) for wild-type virus) at 24 and 30 h postdose (GM of 693 and 392 ng/ml). Two of 17 and 5 of 17 subjects were above target at 48 h postdose while on darunavir-ritonavir and atazanavir-ritonavir. Ritonavir half-life to 72 h was 6.84 h with darunavir and 6.07 with atazanavir. This study investigated the pharmacokinetic forgiveness of two boosted protease inhibitors. Although the rates of decline of darunavir and atazanavir slightly increased as ritonavir concentrations declined, most individuals had concentrations 6 h after the end of the ideal dosing interval of 24 h which were above the cutoff used to define therapeutic concentrations.


Subject(s)
Oligopeptides/pharmacokinetics , Pyridines/pharmacokinetics , Ritonavir/pharmacokinetics , Sulfonamides/pharmacokinetics , Adolescent , Adult , Aged , Atazanavir Sulfate , Darunavir , Female , Humans , Male , Middle Aged , Oligopeptides/administration & dosage , Oligopeptides/blood , Pyridines/administration & dosage , Pyridines/blood , Ritonavir/administration & dosage , Ritonavir/blood , Sulfonamides/administration & dosage , Sulfonamides/blood , Young Adult
13.
J Antimicrob Chemother ; 66(4): 885-9, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21393207

ABSTRACT

OBJECTIVES: To assess the pharmacokinetics (PK) of raltegravir and ezetimibe when co-administered to healthy volunteers. METHODS: This was a prospective, open-label, crossover study, with subjects randomly assigned to group 1 (raltegravir 400 mg twice daily, raltegravir plus ezetimibe 10 mg once daily, wash-out period, ezetimibe) or group 2 (ezetimibe, raltegravir plus ezetimibe, wash-out period, raltegravir); all phases lasted for 10 days. Steady-state full PK sampling was performed at days 10, 20 and 40. Raltegravir and ezetimibe PK parameters were determined by non-compartmental methods and comparisons in the presence of the potentially interactive drug measured by geometric mean ratio (GMR) and 95% confidence intervals (CIs). RESULTS: Twenty subjects (10 females) completed the study. Raltegravir PK parameters did not change significantly in the presence of ezetimibe: GMRs (95% CI) were 1.16 (0.89-1.51) for AUC(0-12), 1.13 (0.81-1.58) for maximum plasma concentration (Cmax) and 1.12 (0.72-1.74) for trough concentration (Ctrough). Ezetimibe AUC0-24 and Ctrough were lower in the presence of raltegravir [GMRs (95% CI) were 0.79 (0.68-0.91) for AUC0-24 and 0.78 (0.60-0.99) for Ctrough], while ezetimibe glucuronide Cmax was 40% higher (90% CI 1.17-1.66). There was marked inter-individual variability in the PK of the two drugs, especially during co-administration. CONCLUSIONS: There were no significant changes in raltegravir PK parameters with or without ezetimibe. However, in the presence of raltegravir, ezetimibe AUC0-24 and Ctrough were significantly lower (>20%) and ezetimibe glucuronide Cmax was higher. Clinical data to assess the importance of the change in ezetimibe concentrations are warranted.


Subject(s)
Azetidines/adverse effects , Azetidines/pharmacokinetics , Pyrrolidinones/adverse effects , Pyrrolidinones/pharmacokinetics , Adult , Anticholesteremic Agents/administration & dosage , Anticholesteremic Agents/adverse effects , Anticholesteremic Agents/pharmacokinetics , Antiviral Agents/administration & dosage , Antiviral Agents/adverse effects , Antiviral Agents/pharmacokinetics , Azetidines/administration & dosage , Cross-Over Studies , Drug Therapy, Combination/adverse effects , Drug Therapy, Combination/methods , Ezetimibe , Female , Healthy Volunteers , Humans , Male , Middle Aged , Prospective Studies , Pyrrolidinones/administration & dosage , Raltegravir Potassium
14.
J Antimicrob Chemother ; 66(3): 635-40, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21172791

ABSTRACT

OBJECTIVES: Data suggest that some licensed antiretroviral doses could be reduced. We assessed the safety, tolerability and pharmacokinetics of lopinavir/ritonavir at doses of 400/100, 200/150 and 200/50 mg twice daily in HIV-negative volunteers (http://clinicaltrials.gov/ct2/show/NCT00985543). METHODS: Male and female volunteers were administered lopinavir/ritonavir at doses of 400/100 mg (two lopinavir/ritonavir Meltrex 200/50 mg tablets, Regimen 1), 200/150 mg (one Meltrex tablet, one 100 mg ritonavir capsule, Regimen 2) and 200/50 mg (one Meltrex tablet, Regimen 3). Each dose was given twice daily for 7 days sequentially, separated by a 7 day wash-out period. Lopinavir/ritonavir steady-state pharmacokinetics was assessed over 12 h at the end of each phase (days 7, 21 and 35). Pharmacokinetic parameters were compared using the 400/100 mg twice daily dose as reference, by determining geometric mean ratios (GMRs) and 90% confidence intervals. RESULTS: Twenty-two subjects (eight females) completed the study. Lopinavir AUC(0-12) (ng h/mL), C(max) (ng/mL) and the minimum concentration (C(trough)) (ng/mL) for the 400/100, 200/150 and 200/50 mg twice daily doses, respectively, were as follows: 99,599, 73,603 and 45,146; 11,965, 8939 and 6404; and 5776, 4293 and 1749. Lopinavir pharmacokinetic parameters were significantly lower for Regimens 2 and 3: GMR (90% CI) AUC(0-12), 0.74 (0.65-0.84) and 0.45 (0.40-0.51); C(max), 0.75 (0.66-0.85) and 0.54 (0.40-0.60); and C(trough), 0.74 (0.62-0.89) and 0.30 (0.25-0.36), respectively. All subjects taking the 400/100 and 200/150 mg twice daily doses, and 19 (86%) subjects taking 200/50 mg twice daily had lopinavir concentrations above the suggested minimum effective concentration of 1000 ng/mL. CONCLUSIONS: These pharmacokinetic data show that therapeutic plasma concentrations of lopinavir can be achieved with 200/150 mg of lopinavir/ritonavir twice daily (one Meltrex tablet and one 100 mg ritonavir capsule twice daily).


Subject(s)
Anti-HIV Agents/administration & dosage , Anti-HIV Agents/pharmacokinetics , Plasma/chemistry , Pyrimidinones/administration & dosage , Pyrimidinones/pharmacokinetics , Ritonavir/administration & dosage , Ritonavir/pharmacokinetics , Administration, Oral , Adult , Anti-HIV Agents/adverse effects , Drug Therapy, Combination/methods , Female , Human Experimentation , Humans , Lopinavir , Male , Middle Aged , Pyrimidinones/adverse effects , Ritonavir/adverse effects
15.
Br J Nutr ; 102(7): 1038-46, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19480729

ABSTRACT

Mechanisms underlying the lipodystrophy syndrome associated with antiretroviral therapy (ART) for HIV infection are not completely understood. We investigated the effect of ART on blood lipid concentrations in the fasting state and after consumption of a meal containing [1-13C]palmitic acid in HIV-positive men receiving nucleoside reverse transcriptase inhibitors (NRTI, n 7), NRTI combined with protease inhibitors (PI; NRTIPI, n 6), in HIV-positive but therapy-naïve men (noART, n 5) and in HIV-seronegative men (controls, n 6). HIV-positive subjects had higher fasting TAG concentrations and resting energy expenditure than controls. Subjects receiving NRTIPI therapy had higher fasting NEFA concentrations than the other groups. There were no significant differences in postprandial lipid metabolism between noART subjects and controls. NRTI therapy impaired hydrolysis of meal-derived TAG, most evidently when combined with PI (the NRTIPI group). Accumulation of 13C-label in the NEFA fraction was not different between groups. In the NRTIPI group, fasting and postprandial NEFA concentrations were significantly higher than other groups. Postprandial glucose and insulin responses in HIV-positive subjects did not differ from controls. These findings suggest that ART dyslipidaemia is associated with impaired postprandial TAG clearance, which is exacerbated by NRTIPI therapy. If dyslipidaemia is to be minimised in ART, the specific adverse effects of particular combinations during the fed state should be considered.


Subject(s)
Anti-HIV Agents/adverse effects , HIV Infections/drug therapy , HIV-1 , Hypertriglyceridemia/chemically induced , Adult , Anti-HIV Agents/therapeutic use , Antiretroviral Therapy, Highly Active/adverse effects , Antiretroviral Therapy, Highly Active/methods , Blood Glucose/metabolism , Energy Metabolism/drug effects , HIV Infections/blood , HIV Protease Inhibitors/adverse effects , HIV Protease Inhibitors/therapeutic use , Humans , Hydrolysis/drug effects , Hypertriglyceridemia/blood , Insulin/blood , Male , Middle Aged , Oxidation-Reduction/drug effects , Postprandial Period/physiology , Reverse Transcriptase Inhibitors/adverse effects , Reverse Transcriptase Inhibitors/therapeutic use , Triglycerides/blood , Young Adult
16.
HIV Clin Trials ; 19(1): 31-37, 2018 02.
Article in English | MEDLINE | ID: mdl-29189101

ABSTRACT

BACKGROUND: We aimed at investigating the antiviral activity and the pharmacokinetics of the dual antiretroviral (ARV) combination of rilpivirine plus darunavir/ritonavir 25/800/100 mg once-daily in naïve HIV-1-infected individuals (NHII) with different baseline viral loads. SETTINGS: Pharmacokinetic/pharmacodynamics study in ARV-naïve HIV-infected individuals. METHODS: The primary endpoint was the number of NHII with HIV-RNA < 40 copies/mL at week 48. Secondary endpoints included rilpivirine/darunavir/ritonavir pharmacokinetics, HIV-RNA decay, and changes in ECG QT interval. RESULTS: Thirty-six individuals were enrolled, 18 with a baseline viral load < 100,000 copies/mL (group A) and 18 with a baseline viral load > 100,000 copies/mL (group B). All but 1 (HIV-RNA = 63 copies/mL) subjects achieved viral load < 50 copies/mL by week 36, and all at week 48. Median (range) HIV-RNA reduction (Log10 copies/mL) was 1.3 (0.6-1.9) over the first week, with no differences between groups A and B. Geometric mean and 95%CI rilpivirine Cmax, Ctrough, AUC were 183 (165-239), 114 (104-109) ng/mL, 2966 (2704-3820) ng h/mL. No QTcF interval changes were recorded. CONCLUSIONS: rilpivirine/darunavir/ritonavir could be efficacious, with limited short-term toxicity in ARV-naïve patients. Although rilpivirine was co-administered with ritonavir, its exposure was within ranges measured during phase III trials.


Subject(s)
Anti-HIV Agents/pharmacokinetics , Darunavir/pharmacokinetics , HIV Infections/drug therapy , Rilpivirine/pharmacokinetics , Ritonavir/pharmacokinetics , Adult , Anti-HIV Agents/administration & dosage , Anti-HIV Agents/therapeutic use , Area Under Curve , Darunavir/administration & dosage , Darunavir/blood , Darunavir/therapeutic use , Drug Therapy, Combination , Female , HIV Protease Inhibitors/administration & dosage , HIV Protease Inhibitors/pharmacokinetics , HIV Protease Inhibitors/therapeutic use , HIV-1 , Humans , Male , Middle Aged , Rilpivirine/administration & dosage , Rilpivirine/blood , Rilpivirine/therapeutic use , Ritonavir/administration & dosage , Ritonavir/blood , Ritonavir/therapeutic use , Viral Load , Young Adult
17.
AIDS ; 21(11): 1449-55, 2007 Jul 11.
Article in English | MEDLINE | ID: mdl-17589191

ABSTRACT

BACKGROUND: Cumulative antiretroviral exposure can result in multiclass HIV drug resistance. Experimental antiretroviral agents offer limited therapeutic benefit as resistance quickly develops after their introduction as a sole new agent. OBJECTIVE: To assess the pharmacokinetic profile, safety and virological response of two novel investigational antiretroviral agents when used in combination in HIV-1-infected subjects with multidrug-resistant virus. METHODS: HIV-1-infected subjects, with current virological failure on a stable antiretroviral regimen with no viable treatment options were assigned to a regimen comprising two new investigational agents, etravirine, a novel nonnucleoside reverse transcriptase inhibitor, and darunavir, a novel protease inhibitor, plus nucleoside reverse transcriptase inhibitors (and enfuvirtide in selected patients) for 24 weeks. Virological, immunological and safety parameters were collected. Detailed pharmacokinetic assessments of darunavir and etravirine were determined on days 7 and 28. RESULTS: Follow up of 24 weeks was achieved by 10/12 patients. Median reduction in HIV RNA was 2.7 log10 copies/ml (range, 2.3-3.9) and increase in CD4 lymphocytes was 113 cells/microl (range, 41-268). HIV RNA was < 40 copies/ml in nine. No serious adverse events were recorded. Plasma exposure to darunavir was similar to historic control data and exposure to etravirine similar to historic data when etravirine was administered with a boosted protease inhibitor. CONCLUSION: This first study to assess the use of etravirine and darunavir in HIV-1-infected subjects with no treatment options showed highly effective virological and immunological responses over 24 weeks of therapy with no new safety concerns or unexpected pharmacokinetic interactions.


Subject(s)
HIV Infections/drug therapy , HIV Protease Inhibitors/pharmacokinetics , HIV-1 , Pyridazines/pharmacokinetics , Ritonavir/pharmacokinetics , Sulfonamides/pharmacokinetics , Adult , Anti-Retroviral Agents/therapeutic use , Area Under Curve , CD4 Lymphocyte Count , Darunavir , Drug Resistance, Viral , Female , HIV Infections/immunology , HIV Protease Inhibitors/therapeutic use , HIV-1/genetics , Humans , Male , Middle Aged , Nitriles , Pyridazines/therapeutic use , Pyrimidines , RNA, Viral/analysis , Ritonavir/therapeutic use , Sulfonamides/therapeutic use , Viral Load
18.
Antivir Ther ; 22(2): 145-151, 2017.
Article in English | MEDLINE | ID: mdl-27708251

ABSTRACT

BACKGROUND: We aimed to measure the effect of raltegravir (RAL) on insulin sensitivity and surrogates of cardiovascular risk in healthy HIV-seronegative volunteers compared to that of lopinavir/r (LPV/r), a positive control. METHODS: An open-label, two phase crossover study in HIV-negative male subjects randomized 1:1 to receive either 2 weeks of LPV/r followed by a 2-week washout period and 2 weeks of RAL, or RAL initially followed by LPV/r. A hyperinsulinaemic euglycaemic clamp was performed prior to and following each 2-week dosing phase. Fasting samples for lipids, adiponectin, leptin, vascular inflammatory biomarkers and CD36 were also taken. RESULTS: A total of 16 subjects completed the study. At the baseline visit the mean insulin-stimulated glucose disposal per unit insulin (M/I) was 7.97 and 8.30 for LPV/r and RAL, respectively. The mean (sem) percentage change from baseline was -16.10% (3.84) after 2 weeks of LPV/r and -0.43% (4.83) after 2 weeks of RAL. Absolute M/I was 25% lower for LPV/r than for RAL (P=0.001). Triglycerides and total cholesterol rose significantly with LPV/r (+0.5 mmol/l, P=0.002 and +0.4 mmol/l, P<0.0001), but were unchanged with RAL. Proathrogenic lipid subfractions of low-density lipoprotein (LDL) cholesterol increased with LPV/r and were unaffected with RAL. LDL peak and mean particle diameter and LDL I significantly decreased with LPV/r (P<0.05), and trend of increased LDL III was detected. High-sensitivity C-reactive protein declined with RAL (-0.2 mg/l, P=0.043) but was elevated after LPV/r (+0.25 mg/l, P=0.03). CONCLUSIONS: RAL was not associated with measurable change in glycaemic, metabolic or inflammatory effects.


Subject(s)
Anti-HIV Agents/administration & dosage , Lopinavir/administration & dosage , Raltegravir Potassium/administration & dosage , Ritonavir/administration & dosage , Adiponectin/blood , Adolescent , Adult , C-Reactive Protein/metabolism , CD36 Antigens/blood , Cholesterol/blood , Cholesterol, LDL/blood , Cross-Over Studies , Glucose Clamp Technique , Healthy Volunteers , Humans , Insulin Resistance , Leptin/blood , Male , Middle Aged , Random Allocation , Triglycerides/blood
19.
J Acquir Immune Defic Syndr ; 73(3): 252-257, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27727157

ABSTRACT

To investigate the pharmacokinetics/pharmacodynamics of single-dose maraviroc 300 mg in HIV-1 exposure compartments. Maraviroc concentrations in blood, secretions (vaginal, urethral, oral, and rectal), and tissue (vaginal and rectal) were measured, and ex vivo challenge was performed in 54 healthy volunteers to study protection from HIV infection. Maraviroc Cmax occurred within 4 hours in most compartments. Concentrations from 4 to 72 hours were above intracellular (IC) IC90 in all compartments, range 15-8095 ng/mL. Mean AUC0-72 compartment-to-plasma ratios were highest in the rectum (45-819) and urethra (144) compared with the female genital tract (1.6-4.8) and saliva (0.2). No sex differences in AUC0-72 or Cmax were observed. No ex vivo protection from HIV-1BaL occurred in rectal or vaginal tissue. Despite high and sustained concentrations, single-dose maraviroc was not protective against ex vivo challenge of vaginal/rectal tissue.


Subject(s)
CCR5 Receptor Antagonists/pharmacokinetics , Cyclohexanes/pharmacokinetics , HIV Fusion Inhibitors/pharmacokinetics , HIV Infections/prevention & control , HIV Infections/virology , Pre-Exposure Prophylaxis , Triazoles/pharmacokinetics , Administration, Oral , Adult , CCR5 Receptor Antagonists/administration & dosage , CCR5 Receptor Antagonists/pharmacology , Cyclohexanes/administration & dosage , Cyclohexanes/pharmacology , Female , HIV Fusion Inhibitors/administration & dosage , HIV Fusion Inhibitors/pharmacology , HIV-1/drug effects , Healthy Volunteers , Humans , Intestinal Mucosa/drug effects , Intestinal Mucosa/virology , Male , Maraviroc , Models, Biological , Randomized Controlled Trials as Topic , Rectum/drug effects , Rectum/virology , Saliva/drug effects , Saliva/virology , Treatment Outcome , Triazoles/administration & dosage , Triazoles/pharmacology , Urethra/drug effects , Urethra/virology , Vagina/drug effects , Vagina/virology
20.
AIDS Rev ; 6(4): 208-17, 2004.
Article in English | MEDLINE | ID: mdl-15700619

ABSTRACT

Therapeutic agents used to inhibit the HIV-1 protease are integral to the effective suppression of HIV virus replication by use of synergistic combination therapy. This is inherently dependent on the achievement of effective plasma concentrations of the drug in its active form, and sustaining such levels for the duration of a dosing interval without exceeding thresholds of toxicity. The issues determining the absorption, biotransformation, distribution to and activity at the intended site, and elimination, are myriad and complex. Studies at molecular, cell, and tissue levels are useful for predicting the possible fate of these agents at in vivo, but the wide interindividual variability shown in whole-body pharmacokinetic studies is illustrative of the difficulty in making general statements rather than more guarded recommendations. This paper summarizes the current state of understanding of the major interactions between protease inhibitors themselves and other important commonly used agents in the management of HIV disease, based on data from clinical pharmacokinetic studies.


Subject(s)
HIV Protease Inhibitors/pharmacology , HIV Protease Inhibitors/pharmacokinetics , Cytochrome P-450 Enzyme System/metabolism , Drug Interactions , HIV Infections/drug therapy , Humans
SELECTION OF CITATIONS
SEARCH DETAIL