Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
1.
AIDS ; 34(9): 1331-1338, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32205723

ABSTRACT

OBJECTIVE: Pretreatment HIV-drug resistance (PDR, HIVDR) to non-nucleoside reverse transcriptase inhibitors (NNRTIs) is increasing globally. NNRTIs continue to be used as first-line antiretroviral therapy (ART) in some communities due to the cost of dolutegravir-based ART or dolutegravir-associated adverse events. A simplified version of the oligonucleotide ligation assay (OLA) - 'OLA-Simple' - is a low-cost, near point-of-care assay that provides ready-to-use lyophilized reagents and reports HIVDR mutations as colored lines on lateral flow strips. Our objective was to design and validate OLA-Simple for a Mexican cohort. DESIGN: OLA-Simple probes to detect K65R, K103N/S, Y181C, M184V, and G190A were optimized for HIV Mexican sequences. Sixty clinical plasma specimens were analyzed by OLA-Simple by technicians blinded to Illumina-MiSeq sequences, and HIVDR results were compared. METHODS: Plasma RNA was tested using OLA-Simple kits. OLA-Simple lateral flow strips were read by in-house software, and were classified as mutant or wild-type at each codon. The comparison of results by OLA-Simple and Miseq was used to generate receiver-operating characteristic curves. RESULTS: OLA-Simple PCR amplified 59 of 60 specimens and successfully genotyped 287 of 295 codons, with eight of 295 (2.7%) indeterminate results. Compared to MiSeq, OLA-Simple gave five of 295 (1.7%) false-positive and four of 295 (1.4%) false-negative results. Excluding indeterminate results, OLA-Simple classified mutant with an accuracy of 97.4 and 98.8% when using thresholds at 10 and 25% mutant within an individual's HIV quasispecies, respectively. CONCLUSIONS: Compared to MiSeq, OLA-Simple detected HIVDR with high sensitivity and accuracy. OLA-Simple could expand access to affordable and rapid HIVDR testing to guide appropriate ART choices in populations using NNRTI-based ART.


Subject(s)
Antiretroviral Therapy, Highly Active , Drug Resistance, Viral/genetics , HIV Infections/drug therapy , HIV-1/drug effects , HIV-1/genetics , Mutation/drug effects , Point-of-Care Systems/statistics & numerical data , Precision Medicine , Reverse Transcriptase Inhibitors/pharmacology , Anti-HIV Agents/administration & dosage , Anti-HIV Agents/pharmacology , Anti-HIV Agents/therapeutic use , Genotype , HIV Infections/blood , HIV Infections/virology , HIV-1/isolation & purification , Humans , Mexico , Microbial Sensitivity Tests/methods , Oligonucleotide Probes , Polymerase Chain Reaction , RNA-Directed DNA Polymerase/genetics , Reproducibility of Results , Reverse Transcriptase Inhibitors/administration & dosage , Sensitivity and Specificity , Sequence Analysis, DNA
2.
PLoS One ; 14(6): e0217502, 2019.
Article in English | MEDLINE | ID: mdl-31206536

ABSTRACT

BACKGROUND: Residual HIV-1 replication among individuals under antiretroviral therapy (ART) relates to HIV micro-inflammation. OBJECTIVES: To determine the levels of residual HIV replication markers among distinct subgroups of antiretroviral-treated individuals. METHODS: One hundred sixteen patients were distributed into 5 treatment groups: first-line suppressive ART with a non-nucleoside analog reverse-transcriptase inhibitor (NNRTI) (n = 26), first-line suppressive ART with boosted protease inhibitors (PI-r) (n = 25), salvage therapy using PI-r (n = 27), salvage therapy with PI-r and raltegravir (n = 22) and virologic failure (n = 16). Episomal and total DNA quantitation was evaluated. ELISA was used for HIV antibody and LPS quantitation. RESULTS: Episomal DNA was positive in 26% to 38% of individuals under suppressive ART, and it was higher among individuals experiencing ART virologic failure (p = 0.04). The HIV proviral load was higher among patients with detectable episomal DNA (p = 0.01). Individuals receiving initial PI-r treatment presented lower HIV antibody (p = 0.027) and LPS (p = 0.029) levels than individuals receiving NNRTI. There was a negative correlation between episomal DNA quantitation and the duration of suppressive ART (p = 0.04), CD4+ T-cell count (p = 0.08), and CD8+ T-cell count (p = 0.07). CONCLUSIONS: Residual HIV replication has been inferred among individuals under suppressive ART according to episomal DNA detection. Residual replication may decrease with longer periods of suppressive ART and higher levels of CD4+ and CD8+ T cells. The relationship between episomal DNA and total DNA suggests there is a replenishment of the proviral reservoir with impacts on HIV persistence. Lower antibody and LPS levels among patients with initial PI-r ART suggest these regimens may more effectively suppress HIV and have a higher capacity to decrease the HIV antigenic component.


Subject(s)
HIV Antibodies/blood , HIV Infections , HIV Protease Inhibitors/administration & dosage , HIV-1/physiology , Plasmids/metabolism , Reverse Transcriptase Inhibitors/administration & dosage , Virus Replication/drug effects , Biomarkers/blood , Brazil , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/virology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/virology , Cells, Cultured , Female , HIV Infections/blood , HIV Infections/drug therapy , Humans , Lipopolysaccharides/pharmacology , Male
3.
Antivir Ther ; 24(2): 95-103, 2019.
Article in English | MEDLINE | ID: mdl-30530925

ABSTRACT

BACKGROUND: Limited data exist on efavirenz pharmacokinetics in HIV-positive pregnant women and neonatal washout. METHODS: HIV-infected pregnant women receiving 600 mg efavirenz once daily had intensive steady-state 24-h pharmacokinetics profiles during the second trimester (2T), third trimester (3T) and 6-12 weeks postpartum (PP). Maternal and umbilical cord blood samples were drawn at delivery and neonatal washout pharmacokinetics were determined. Therapeutic targets were the estimated 10th percentile efavirenz area under the concentration-time curve (AUC) in non-pregnant historical controls (40.0 µg•h/ml) and a trough concentration (C24 h) of 1 µg/ml. Data were prospectively collected within two trials: IMPAACT P1026s (United States) and PANNA (Europe). RESULTS: Among 42 women studied, 15, 42 and 40 had efavirenz pharmacokinetic data available in 2T, 3T and PP, respectively. Median (range) 3T age 33 (20.7-43.5) years, weight 74 (50-132) kg and gestational age 33.4 (28.4-37.9 weeks). Efavirenz AUC during the 3T (60 µg•h/ml) was similar to that reported in non-pregnant adults (58 µg•h/ml). Exposure in the 2T was lower, but within the 0.80-1.25 range. C24 concentrations during pregnancy were lower compared to historical controls on 600 mg efavirenz, however, they were similar to the C24 concentrations after equally potent dose of 400 mg efavirenz. Cord blood/maternal plasma concentration ratio (range) was 0.67 (0.36-0.95). Among 23 infants with washout data available, median (interquartile range) elimination half-life was 65.6 h (40.6-129). HIV RNA viral loads at delivery were <400 and <50 copies/ml for 96.7% and 86.7% of women, respectively. In 3T and PP, respectively, 8/41 (19%) and 6/40 (15%) had AUC below target; 7/41 (17%) and 3/39 (8%) had C24 below target. CONCLUSIONS: Efavirenz exposure was similar during pregnancy compared with PP, C24 was in line with C24 after 400 mg equipotent efavirenz dosing. Efavirenz readily crossed the placenta and infant elimination half-life was over twice that of maternal participants. Clincaltrials.gov identifiers: NCT00825929 and NCT00042289.


Subject(s)
Benzoxazines/pharmacokinetics , HIV Infections/drug therapy , Reverse Transcriptase Inhibitors/pharmacokinetics , Adolescent , Adult , Alkynes , Antiretroviral Therapy, Highly Active , Benzoxazines/administration & dosage , Benzoxazines/adverse effects , Child , Child, Preschool , Cyclopropanes , Drug Monitoring , Female , HIV Infections/blood , HIV Infections/diagnosis , HIV Infections/virology , Humans , Infant , Infant, Newborn , Pregnancy , Pregnancy Complications, Infectious/drug therapy , Reverse Transcriptase Inhibitors/administration & dosage , Reverse Transcriptase Inhibitors/adverse effects , Treatment Outcome , Young Adult
5.
J Antimicrob Chemother ; 73(9): 2460-2467, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29868865

ABSTRACT

Objectives: There are limited data regarding efavirenz pharmacogenetics in admixed populations. The Brazilian population is highly admixed. In a Brazilian cohort, we sought to characterize associations between efavirenz adverse effects (all-cause and CNS) and polymorphisms in seven genes known or suspected to affect efavirenz metabolism and transport. Methods: We studied 225 HIV-positive individuals who had been prescribed efavirenz-containing regimens at a hospital in Rio de Janeiro, Brazil. Eighty-nine cases had efavirenz adverse effects, including 43 with CNS adverse effects, while 136 controls had no adverse effect of any antiretroviral after treatment for at least 6 months. A total of 67 candidate polymorphisms in ABCB1, CYP2A6, CYP2B6, CYP3A4, CYP3A5, NR1I2 and NR1I3 genes were selected for association analysis. Admixture was assessed using 28 ancestry-informative polymorphisms previously validated for the Brazilian population. Associations were evaluated with logistic regression models adjusted for sex and genetic ancestry. Results: There was extensive African, European and Native American admixture in the cohort. Increased all-cause adverse effects were associated with the CYP2B6 genotype combination 15582CC-516TT-983TT (OR = 7.26, P = 0.003) and with the CYP2B6 slow metabolizer group 516TT or 516GT-983CT (OR = 3.10, P = 0.04). CNS adverse effects were nominally associated with CYP3A4 rs4646437 (OR = 4.63, P = 0.014), but not after adjusting for multiple comparisons. Conclusions: In a highly admixed Brazilian cohort, the CYP2B6 slow metabolizer genotype was associated with an increased risk of efavirenz adverse effects.


Subject(s)
Benzoxazines/adverse effects , Benzoxazines/metabolism , Drug-Related Side Effects and Adverse Reactions/genetics , HIV Infections/drug therapy , Inactivation, Metabolic/genetics , Reverse Transcriptase Inhibitors/adverse effects , Reverse Transcriptase Inhibitors/metabolism , Adult , Aged , Alkynes , Benzoxazines/administration & dosage , Brazil , Constitutive Androstane Receptor , Cyclopropanes , Drug-Related Side Effects and Adverse Reactions/pathology , Female , Genetic Association Studies , Humans , Male , Middle Aged , Polymorphism, Genetic , Reverse Transcriptase Inhibitors/administration & dosage
6.
Eur J Clin Pharmacol ; 74(8): 1077-1088, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29740676

ABSTRACT

OBJECTIVE: To evaluate the prevalence of adverse drug reactions (ADR) and associated factors during the use of Highly Active Antiretroviral Therapy (HAART) in patients initiating treatment. METHODS: This is a cross-sectional analysis of a prospective study conducted in three public referral services specialized in HIV/AIDS care in Belo Horizonte, Brazil. Self-reported ADR and explanatory variables were obtained from face-to-face interview and from Information Systems. Associated factors with ADR were evaluated by logistic regression in SPSS software v.22. RESULTS: We included 399 patients, of which 85.5% reported at least one and 72.7% up to 5 ADRs after HAART initiation. Neurological reactions were the most frequent, with self-reported ADRs being distinct according to HAART regimen used. The global model showed higher chance of ADRs among females (OR = 3.52) and illicit drug users (OR = 2.28). Lower chance of ADRs was found for patients aged > 33 years (OR = 0.37), DTG/TDF/3TC users (OR = 0.41), and higher physical domain of quality of life (OR = 0.78). The model restricted to patients using the single-tablet regimen EFV/TDF/3TC showed lower ADRs among patients with CD4+ T lymphocyte count > 200 cells/mm3 (OR = 0.23) and higher independence domain of quality of life (OR = 0.74). The model restricted to DTG/TDF/3TC and to other regimens showed lower ADRs with higher physical domain of quality of life (OR = 0.74 and OR = 0.55, respectively). CONCLUSIONS: The prevalence of self-reported ADRs to first-line antiretroviral regimens was high and patients using DTG/TDF/3TC had a smaller number of ADRs. In addition to HAART regimen, sociodemographic, clinical, and quality of life characteristics were associated with ADRs.


Subject(s)
Antiretroviral Therapy, Highly Active/adverse effects , Reverse Transcriptase Inhibitors/adverse effects , Adolescent , Adult , Aged , Alkynes , Benzoxazines/adverse effects , Brazil , Cross-Sectional Studies , Cyclopropanes , Drug-Related Side Effects and Adverse Reactions , Female , HIV Infections/drug therapy , HIV Integrase Inhibitors/administration & dosage , HIV Integrase Inhibitors/adverse effects , Heterocyclic Compounds, 3-Ring/adverse effects , Humans , Lamivudine/adverse effects , Male , Middle Aged , Oxazines , Piperazines , Prospective Studies , Pyridones , Quality of Life , Reverse Transcriptase Inhibitors/administration & dosage , Tenofovir/adverse effects , Young Adult
7.
Lancet HIV ; 3(6): e247-58, 2016 06.
Article in English | MEDLINE | ID: mdl-27240787

ABSTRACT

BACKGROUND: For second-line antiretroviral therapy, WHO recommends a boosted protease inhibitor plus nucleoside or nucleotide reverse transcriptase inhibitors (NRTIs). However, concerns about toxicity and cross-resistance motivated a search for regimens that do not contain NRTIs. We aimed to assess whether boosted lopinavir plus raltegravir would be non-inferior to boosted lopinavir plus NRTIs for virological suppression in resource-limited settings. METHODS: A5273 was a randomised, open-label, phase 3, non-inferiority study at 15 AIDS Clinical Trials Group (ACTG) research sites in nine resource-limited countries (three sites each in India and South Africa, two each in Malawi and Peru, and one each in Brazil, Kenya, Tanzania, Thailand, and Zimbabwe). Adults with plasma HIV-1 RNA concentrations of at least 1000 copies per mL after at least 24 weeks on a regimen based on a non-NRTI inhibitor were randomly assigned (1:1) to receive oral ritonavir-boosted lopinavir (100 mg ritonavir, 400 mg lopinavir) plus 400 mg raltegravir twice a day (raltegravir group) or to ritonavir-boosted lopinavir plus two or three NRTIs selected from an algorithm (eg, zidovudine after failure with tenofovir and vice versa; NRTI group). Randomised group assignment was done with a computer algorithm concealed to site personnel, and stratified by HIV-1 RNA viral load, CD4 cell count, and intention to use zidovudine, with the groups balanced by each site. The primary endpoint was time to confirmed virological failure (two measurements of HIV-1 RNA viral load >400 copies per mL) at or after week 24 in the intention-to-treat population. Non-inferiority (10% margin) was assessed by comparing the cumulative probability of virological failure by 48 weeks. This trial was registered with ClinicalTrials.gov, NCT01352715. FINDINGS: Between March 13, 2012, and Oct 2, 2013, we randomly assigned 515 participants: 260 to the raltegravir group and 255 to the NRTI group; two participants in the raltegravir group and one in the NRTI group were excluded from analyses because of ineligibility. By the end of follow-up (October, 2014), 96 participants had virological failure (46 in the raltegravir group and 50 in the NRTI group). By 48 weeks, the cumulative probability of virological failure was 10·3% (95% CI 6·5-14·0) in the raltegravir group and 12·4% (8·3-16·5) in the NRTI group, with a weighted difference of -3·4% (-8·4 to 1·5), indicating that raltegravir was non-inferior, but not superior, to NRTIs. 62 (24%) participants in the raltegravir group and 81 (32%) in the NRTI group had grade 3 or higher adverse events; 19 (7%) and 29 (11%), respectively, had serious adverse events. Three participants in each group died, all from HIV-related causes. INTERPRETATION: In settings with extensive NRTI resistance but no available resistance testing, our data support WHO's recommendation for ritonavir-boosted lopinavir plus NRTI for second-line antiretroviral therapy. Ritonavir-boosted lopinavir plus raltegravir is an appropriate alternative, especially if NRTI use is limited by toxicity. FUNDING: National Institutes of Health.


Subject(s)
Anti-HIV Agents/therapeutic use , HIV Infections/drug therapy , Health Resources/economics , Raltegravir Potassium/therapeutic use , Adult , Africa South of the Sahara , Anti-HIV Agents/administration & dosage , Anti-HIV Agents/adverse effects , Antiretroviral Therapy, Highly Active , Brazil/epidemiology , CD4 Lymphocyte Count , Drug Resistance, Viral , HIV Infections/epidemiology , HIV Infections/virology , HIV-1/genetics , Health Resources/supply & distribution , Health Services Accessibility/economics , Humans , India/epidemiology , Lopinavir/administration & dosage , Lopinavir/adverse effects , Lopinavir/therapeutic use , Malawi/epidemiology , Male , Medically Underserved Area , Peru/epidemiology , RNA, Viral/blood , Raltegravir Potassium/administration & dosage , Raltegravir Potassium/adverse effects , Reverse Transcriptase Inhibitors/administration & dosage , Reverse Transcriptase Inhibitors/adverse effects , Reverse Transcriptase Inhibitors/therapeutic use , Thailand/epidemiology , Viral Load
8.
Rev Invest Clin ; 67(4): 273-4, 2015.
Article in English | MEDLINE | ID: mdl-26426594

ABSTRACT

Dear Editor, The prevalence of type 2 diabetes (DM-2) in HIV-infected patients and the concomitant use of metformin (MTF) and non-nucleoside reverse transcriptase inhibitors (NRTI) is rising. Through inhibition of NADH dehydrogenase and DNA pol-γ, both drugs hinder oxidative phosphorylation that may lead to lactic acidosis (LA). Among NRTIs, abacavir and tenofovir have the lowest mitochondrial toxicity, with only a few LA cases reported2-4. We describe here a case of MTF-associated LA (MALA) secondary to the interaction with NRTI.


Subject(s)
Acidosis, Lactic/chemically induced , Hypoglycemic Agents/adverse effects , Metformin/adverse effects , Reverse Transcriptase Inhibitors/adverse effects , Adult , Antiretroviral Therapy, Highly Active/adverse effects , Antiretroviral Therapy, Highly Active/methods , Diabetes Mellitus, Type 2/drug therapy , Drug Interactions , HIV Infections/drug therapy , Humans , Hypoglycemic Agents/administration & dosage , Male , Metformin/administration & dosage , Reverse Transcriptase Inhibitors/administration & dosage
9.
Molecules ; 20(6): 11474-89, 2015 Jun 22.
Article in English | MEDLINE | ID: mdl-26111177

ABSTRACT

Ipecac alkaloids are secondary metabolites produced in the medicinal plant Psychotria ipecacuanha. Emetine is the main alkaloid of ipecac and one of the active compounds in syrup of Ipecac with emetic property. Here we evaluated emetine's potential as an antiviral agent against Human Immunodeficiency Virus. We performed in vitro Reverse Transcriptase (RT) Assay and Natural Endogenous Reverse Transcriptase Activity Assay (NERT) to evaluate HIV RT inhibition. Emetine molecular docking on HIV-1 RT was also analyzed. Phenotypic assays were performed in non-lymphocytic and in Peripheral Blood Mononuclear Cells (PBMC) with HIV-1 wild-type and HIV-harboring RT-resistant mutation to Nucleoside Reverse Transcriptase Inhibitors (M184V). Our results showed that HIV-1 RT was blocked in the presence of emetine in both models: in vitro reactions with isolated HIV-1 RT and intravirion, measured by NERT. Emetine revealed a strong potential of inhibiting HIV-1 replication in both cellular models, reaching 80% of reduction in HIV-1 infection, with low cytotoxic effect. Emetine also blocked HIV-1 infection of RT M184V mutant. These results suggest that emetine is able to penetrate in intact HIV particles, and bind and block reverse transcription reaction, suggesting that it can be used as anti-HIV microbicide. Taken together, our findings provide additional pharmacological information on the potential therapeutic effects of emetine.


Subject(s)
Alkaloids/administration & dosage , Emetine/administration & dosage , HIV Infections/drug therapy , HIV Reverse Transcriptase/antagonists & inhibitors , HIV-1/drug effects , Alkaloids/chemistry , Anti-HIV Agents/administration & dosage , Anti-HIV Agents/chemistry , Emetine/chemistry , HIV Infections/enzymology , HIV Infections/virology , HIV Reverse Transcriptase/chemistry , HIV-1/pathogenicity , Humans , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/virology , Mutation , Reverse Transcriptase Inhibitors/administration & dosage , Reverse Transcriptase Inhibitors/chemistry , Virus Replication/drug effects
10.
Biomed Pharmacother ; 70: 46-52, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25776478

ABSTRACT

Didanosine-loaded chitosan microspheres were developed applying a surface-response methodology and using a modified Maximum Likelihood Classification. The operational conditions were optimized with the aim of maintaining the active form of didanosine (ddI), which is sensitive to acid pH, and to develop a modified and mucoadhesive formulation. The loading of the drug within the chitosan microspheres was carried out by ionotropic gelation technique with sodium tripolyphosphate (TPP) as cross-linking agent and magnesium hydroxide (Mg(OH)2) to assure the stability of ddI. The optimization conditions were set using a surface-response methodology and applying the "Maximum Likelihood Classification", where the initial chitosan concentration, TPP and ddI concentration were set as the independent variables. The maximum ddI-loaded in microspheres (i.e. 1433 mg of ddI/g chitosan), was obtained with 2% (w/v) chitosan and 10% TPP. The microspheres depicted an average diameter of 11.42 µm and ddI was gradually released during 2 h in simulated enteric fluid.


Subject(s)
Chitosan/chemistry , Didanosine/chemistry , Microspheres , Reverse Transcriptase Inhibitors/chemistry , Didanosine/administration & dosage , Dosage Forms , Reverse Transcriptase Inhibitors/administration & dosage , Surface Properties , Technology, Pharmaceutical
11.
Enferm Infecc Microbiol Clin ; 32 Suppl 3: 7-11, 2014 Nov.
Article in Spanish | MEDLINE | ID: mdl-25542869

ABSTRACT

According to evidence from randomized controlled trials and epidemiological data, the antiretroviral treatment (ART) of choice has consisted of the combination of 2 nucleoside analog reverse-transcriptase inhibitors (NRTI) plus 1 non-nucleoside analog reverse-transcriptase inhibitor (NNRTI) or a protease inhibitor (PI) for more than 17 years. There are several unresolved issues, notably the toxocity associated with NRTI, especially thymidine analogs, and the possibility of cross resistance, which may affect subsequent treatment. The development of new antiretroviral drugs with simpler dosing regimens and lower toxicity has led to evaluation of innovative strategies such as dual therapy for initial ART in treatment-naive, with the aim of preventing long-term toxicity and increasing treatment adherence. Despite encouraging results, some combinations have proven unsatisfactory. The strategies with favorable results to date consist of twice-daily lopinavir/ritonavir (LPV/r)-based regimens, those in the PROGRESS (LPV/r + raltegravir) and GARDEL (LPV/r + lamivudine) trials, and the combination of darunavir and raltegravir (NEAT 001 trial), although the latter observed a higher tendency (statistically nonsignificant) to virological failure in the dual combination arm. These trials were based on the use of NRTI-sparing regimens consisting of 2-3 fully- active agents for highly-active ART in treatment-naïve HIV-positive patients. Recent studies provide evidence supporting the use of NRTI-sparing regimens in HIV-infected patients with failure to an initial NNRTI-based ART regimen. The present review will discuss only LPV/r-based innovative strategies in initial ART regimens.


Subject(s)
HIV Infections/drug therapy , HIV Protease Inhibitors/therapeutic use , Lopinavir/therapeutic use , Ritonavir/therapeutic use , Therapies, Investigational , Anti-HIV Agents/administration & dosage , Anti-HIV Agents/adverse effects , Anti-HIV Agents/therapeutic use , Antiretroviral Therapy, Highly Active , Drug Combinations , HIV Integrase Inhibitors/administration & dosage , HIV Integrase Inhibitors/adverse effects , HIV Integrase Inhibitors/therapeutic use , HIV Protease Inhibitors/administration & dosage , Humans , Lopinavir/administration & dosage , Multicenter Studies as Topic , Randomized Controlled Trials as Topic , Reverse Transcriptase Inhibitors/administration & dosage , Reverse Transcriptase Inhibitors/adverse effects , Reverse Transcriptase Inhibitors/therapeutic use , Ritonavir/administration & dosage
12.
Colloids Surf B Biointerfaces ; 122: 56-65, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25016545

ABSTRACT

In this work, Nevirapine (NVP) was encapsulated within three derivatives of poly(ethylene oxide)-poly(propylene oxide)-poly(ethylene oxide) (PEO-PPO-PEO) block copolymers (Tetronic(®) 904, 1107 and Pluronic(®) F127) with and without the addition of three pharmaceutical cosolvents (glycerin, propylene glycol and polyethylene glycol 400) over a wider range of concentrations (0-40% v/v). Also, we evaluated the effect of addition of the cosolvents on the micellar size as determined by dynamic light scattering (DLS) measurements and transmission electron microscopy (TEM). The solubilization capacity of the systems was investigated by UV-spectrophotometry (282nm) and the systems stability was evaluated for 1 month at 25°C. Finally, oral bioavailability of the NVP-loaded micellar systems (2mg/mL) was assessed in male Wistar rats (8mg/kg) and compared with a pediatric commercially available formulation (Viramune(®)). The present study demonstrates that PEO-PPO-PEO polymeric micelles were able to enhance apparent aqueous solubility of NVP with the addition of cosolvents. Moreover, micellar nanocarriers significantly (p<0.05) improved the oral bioavailability of the drug versus Viramune(®). Overall results support the suitability of the strategy toward the development of an optimized NVP aqueous formulation to prevent HIV/AIDS mother-to-child transmission.


Subject(s)
Drug Carriers , Micelles , Nevirapine/administration & dosage , Nevirapine/pharmacokinetics , Reverse Transcriptase Inhibitors/pharmacokinetics , Administration, Oral , Animals , Biological Availability , Male , Microscopy, Electron, Transmission , Rats , Rats, Wistar , Reverse Transcriptase Inhibitors/administration & dosage , Spectrophotometry, Ultraviolet
13.
AIDS ; 27(15): 2403-11, 2013 Sep 24.
Article in English | MEDLINE | ID: mdl-23921615

ABSTRACT

OBJECTIVE: To compare changes over 48 weeks in bone mineral density (BMD) between participants randomized to lopinavir/ritonavir (LPV/r) + raltegravir (RAL) or LPV/r + 2-3 nucleoside/nucleotide reverse transcriptase inhibitors (N(t)RTIs) as second line therapy. DESIGN: 48-week open-label sub-study of the Second Line trial conducted in South Africa, India, Thailand, Malaysia and Argentina. METHODS: Dual energy X-ray absorptiometry scans of proximal femur and lumbar spine were performed at baseline and week 48. Linear regression was used to compare means of differences between arms. McNemars test compared osteopenia and osteoporosis. Associations between percentage BMD changes and baseline variables were assessed by multivariate linear regression. RESULTS: Two hundred and ten participants were randomized. Analyses were adjusted for sex, BMI and smoking status. Mean (95% CI) proximal femur BMD% reduced over 48 weeks by -5.2% (-6.7 to -3.8%) in the LPV/r+2-3N(t)RTIs arm and by -2.9% (-4.3 to -1.5%) in the LPV/r+RAL arm (P = 0.0001). Lumbar spine BMD reduced by -4.2% (-5.7 to -2.7%) in the LPV/r+2-3N(t)RTIs arm and by -2.0% (-3.5 to -0.6%) in the LPV/r+RAL arm (P = 0.0006). The incidence of osteopenia (7.6%) and osteoporosis (2.0%) assessed over 48 weeks were similar between arms. Reduced BMD over 48 weeks was significantly associated with longer duration of tenofovir on study [% change (SE) -1.58 (0.38) femur, -1.65 (0.38) spine, P = 0.0001] and low baseline BMI [% change (SE) 0.5 (0.13) femur, 0.17 (0.07) spine; P < 0.01]. CONCLUSION: An N(t)RTI-sparing antiretroviral regimen of LPV/r and raltegravir as second line therapy is associated with less bone loss than a LPV/r regimen containing N(t)RTIs.


Subject(s)
Bone Density/drug effects , Bone Diseases, Metabolic/epidemiology , HIV Infections/drug therapy , HIV Protease Inhibitors/adverse effects , Osteoporosis/epidemiology , Reverse Transcriptase Inhibitors/adverse effects , Absorptiometry, Photon , Adult , Argentina/epidemiology , Bone Diseases, Metabolic/complications , Female , Femur/diagnostic imaging , HIV Infections/complications , HIV Protease Inhibitors/administration & dosage , Humans , India/epidemiology , Lopinavir/adverse effects , Lumbar Vertebrae/diagnostic imaging , Malaysia/epidemiology , Male , Osteoporosis/complications , Prevalence , Pyrrolidinones/adverse effects , Raltegravir Potassium , Reverse Transcriptase Inhibitors/administration & dosage , Risk Factors , Ritonavir/adverse effects , South Africa/epidemiology , Thailand/epidemiology , Treatment Outcome
14.
J Acquir Immune Defic Syndr ; 63(1): 59-66, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23392467

ABSTRACT

BACKGROUND: Reduced atazanavir exposure has been demonstrated during pregnancy with standard atazanavir/ritonavir dosing. We studied an increased dose during the third trimester of pregnancy. METHODS: International Maternal Pediatric Adolescent AIDS Clinical Trials Group 1026s is a prospective, nonblinded, pharmacokinetic study of HIV-infected pregnant women taking antiretrovirals for clinical indications, including 2 cohorts (with or without tenofovir) receiving atazanavir/ritonavir 300/100 mg once daily during the second trimester, 400/100 mg during the third trimester, and 300/100 mg postpartum (PP). Intensive steady-state 24-hour pharmacokinetic profiles were performed. Atazanavir concentrations were measured by high-performance liquid chromatography. Pharmacokinetic targets were the 10th percentile atazanavir area under the concentration versus time curve (AUC) (29.4 µg·hr·mL·) in nonpregnant adults on standard dose and 0.15 µg/mL, minimum trough concentration. RESULTS: Atazanavir pharmacokinetic data were available for 37 women without tenofovir, 35 with tenofovir; median (range) pharmacokinetic parameters are presented for second trimester, third trimester, and PP and number who met target/total. ATAZANAVIR WITHOUT TENOFOVIR: AUC 30.5 (9.19-93.8), 45.7 (11-88.3), and 48.8 (9.9-112.2) µg·hr·mL, and 8/14, 29/37, and 27/34 met target. C24 h was 0.49 (0.09-4.09), 0.71 (0.14-2.09), and 0.90 (0.05-2.73) µg/mL; 13/14, 36/37, and 29/34 met target. ATAZANAVIR WITH TENOFOVIR: AUC 26.2 (6.8-60.9) (P < 0.05 compared with PP), 37.7 (0.72-88.2) (P < 0.05 compared with PP), and 58.6 (6-149) µg·hr·mL, and 7/17, 23/32, and 27/29 met target. C24 h was 0.44 (0.12-1.06) (P < 0.05 compared with PP), 0.57 (0.02-2.06) (P < 0.05 compared with PP), and 1.26 (0.09-5.43) µg/mL; 7/17, 23/32, and 27/29 met target. Atazanavir/ritonavir was well tolerated with no unanticipated adverse events. CONCLUSIONS: Atazanavir/ritonavir increased to 400/100 mg provides adequate atazanavir exposure during the third trimester and should be considered during the second trimester.


Subject(s)
Adenine/analogs & derivatives , Anti-HIV Agents/pharmacokinetics , HIV Infections/drug therapy , HIV Protease Inhibitors/pharmacokinetics , Oligopeptides/pharmacokinetics , Organophosphonates/pharmacokinetics , Pregnancy Complications, Infectious/drug therapy , Pregnancy Trimester, Third , Pyridines/pharmacokinetics , Reverse Transcriptase Inhibitors/pharmacokinetics , Adenine/administration & dosage , Adenine/pharmacokinetics , Adenine/therapeutic use , Adolescent , Adult , Anti-HIV Agents/administration & dosage , Anti-HIV Agents/therapeutic use , Atazanavir Sulfate , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Therapy, Combination , Female , HIV Infections/virology , HIV Protease Inhibitors/administration & dosage , HIV-1/drug effects , Humans , Oligopeptides/administration & dosage , Oligopeptides/therapeutic use , Organophosphonates/administration & dosage , Organophosphonates/therapeutic use , Postpartum Period , Pregnancy , Pregnancy Complications, Infectious/virology , Pregnancy Trimester, Second , Prospective Studies , Pyridines/administration & dosage , Pyridines/therapeutic use , Reverse Transcriptase Inhibitors/administration & dosage , Reverse Transcriptase Inhibitors/therapeutic use , Tenofovir , Treatment Outcome , Young Adult
15.
Nanomedicine (Lond) ; 8(2): 223-37, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23173734

ABSTRACT

AIM: To investigate the intranasal administration of poly(ethylene oxide)-poly(propylene oxide) polymeric micelles loaded with high payloads of the first-line antiretroviral drug efavirenz for targeting to the CNS. METHODS & MATERIALS: The effect of micellar size and composition and drug payload was assessed, employing simple micelles made of a highly hydrophilic copolymer, poloxamer F127, loaded with 20 mg/ml drug and mixed micelles containing 75% of a poloxamine of intermediate hydrophobicity, T904, and 25% F127 loaded with 20 and 30 mg/ml drug. F127 confers high physical stability, while T904 substantially improves the encapsulation capacity of the micelles. RESULTS: The bioavailability of the drug in the CNS was increased fourfold and the relative exposure index (ratio between the area under the curve in the CNS and plasma) was increased fivefold with respect to the same system administered intravenously. CONCLUSION: These findings demonstrate the potential of this scalable and cost-viable strategy to address the HIV sanctuary in the CNS.


Subject(s)
Benzoxazines/administration & dosage , Benzoxazines/therapeutic use , HIV Infections/drug therapy , Micelles , Administration, Intranasal , Alkynes , Animals , Cyclopropanes , Male , Polyethylenes/chemistry , Polypropylenes/chemistry , Rats , Rats, Wistar , Reverse Transcriptase Inhibitors/administration & dosage , Reverse Transcriptase Inhibitors/therapeutic use
16.
Pediatr Infect Dis J ; 31(5): 469-73, 2012 May.
Article in English | MEDLINE | ID: mdl-22301477

ABSTRACT

BACKGROUND: There are few data on the safety and antiviral activity of tenofovir disoproxil fumarate (TDF) in HIV-1 infected adolescents. METHODS: A randomized, double-blinded, placebo-controlled study was conducted. Ninety adolescents (12 to <18 years) who were viremic while receiving antiretroviral treatment were randomized to receive TDF 300 mg (mean, 216.8 mg/m(2)) or placebo in combination with an optimized background regimen (OBR) for 48 weeks. The primary efficacy endpoint was time-weighted average change in plasma HIV-1 RNA from baseline at week 24 RESULTS: Eighty-seven subjects (45 TDF, 42 placebo) received the study drug. Through week 24, the median time-weighted average change in plasma HIV-1 RNA was not different between the TDF and placebo groups (-1.6 versus -1.6 log(10)copies/mL, P = 0.55). The percentages of subjects who achieved HIV-1 RNA <400 copies/mL were similar at week 24 (40.9 versus 41.5%). One fourth of subjects in the TDF and placebo groups (24.4 versus 28.6%) had at least 3 active agents in the OBR. Many subjects in both groups had baseline genotypic resistance to TDF (48.9 versus 33.3%). TDF was generally safe and well tolerated. There were no statistically significant differences in changes of renal function and bone mineral density between the 2 groups. CONCLUSION: This study of TDF in combination with an OBR in antiretroviral-experienced adolescents did not meet its primary or secondary efficacy endpoints. The effectiveness of the OBR and baseline genotypic resistance to TDF in both groups may have confounded the efficacy findings. No clinically relevant TDF-related renal or bone toxicities were observed in this adolescent population.


Subject(s)
Adenine/analogs & derivatives , Anti-HIV Agents , HIV Infections/drug therapy , HIV-1/drug effects , Organophosphonates , Reverse Transcriptase Inhibitors , Adenine/administration & dosage , Adenine/adverse effects , Adenine/therapeutic use , Adolescent , Anti-HIV Agents/administration & dosage , Anti-HIV Agents/adverse effects , Anti-HIV Agents/therapeutic use , Child , Double-Blind Method , Drug Resistance, Viral , Drug Therapy, Combination , Female , HIV Infections/virology , HIV-1/physiology , Humans , Male , Organophosphonates/administration & dosage , Organophosphonates/adverse effects , Organophosphonates/therapeutic use , RNA, Viral/blood , Reverse Transcriptase Inhibitors/administration & dosage , Reverse Transcriptase Inhibitors/adverse effects , Reverse Transcriptase Inhibitors/therapeutic use , Tenofovir , Treatment Outcome , Viremia/drug therapy , Viremia/virology
17.
Antimicrob Agents Chemother ; 56(5): 2570-5, 2012 May.
Article in English | MEDLINE | ID: mdl-22314532

ABSTRACT

GSK2248761 is a novel, once-daily (QD), next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI) with activity against efavirenz-resistant strains. Two phase I/IIa, double-blind, randomized, placebo-controlled studies investigated the antiviral activity, safety, and pharmacokinetics (PK) of several doses of GSK2248761 monotherapy in treatment-naive HIV-infected subjects. In the initial study, 10 subjects (8 active and 2 placebo) per dose received sequentially descending GSK2248761 monotherapy regimens of 800, 400, 200, and 100 mg QD for 7 days. Because a dose-response relationship was not identified, a second study examined a lower, 30-mg QD dose in 8 subjects (6 active and 2 placebo). Adverse events, viral load (VL), PK, and reverse transcriptase mutations were assessed and combined for analysis. Treatment with GSK2248761 for 7 days was well tolerated with no serious adverse events or discontinuations. The mean VL reductions from baseline on day 8 were 0.97, 1.87, 1.84, 1.81, and 1.78 log(10) copies/ml for GSK2248761 doses of 30, 100, 200, 400, and 800 mg QD, respectively. GSK2248761 PK (maximum drug concentration in serum [C(max)], area under the plasma concentration-time curve from 0 h to the end of the dosing interval [AUC(0-τ)], and concentration at the end of the dosing interval [C(τ)]) increased proportionally over the dose range of 30 to 800 mg QD. The relationship between short-term VL change and GSK2248761 PK was best described by a maximum-effect (E(max)) model using C(τ) (E(max) = 2.0; 50% effective concentration [EC(50)] = 36.9 ng/ml). No NNRTI resistance mutations emerged during the study. GSK2248761 at 100 to 800 mg QD for 7 days was well tolerated, demonstrated potent antiviral activity in treatment-naive HIV-infected subjects, and had favorable PK and resistance profiles. GSK2248761 is no longer in clinical development.


Subject(s)
Anti-HIV Agents/therapeutic use , HIV Infections/drug therapy , HIV Reverse Transcriptase/antagonists & inhibitors , HIV-1/drug effects , Indoles/therapeutic use , Phosphinic Acids/therapeutic use , Reverse Transcriptase Inhibitors/therapeutic use , Adult , Alkynes , Anti-HIV Agents/administration & dosage , Anti-HIV Agents/chemical synthesis , Argentina , Benzoxazines , Cyclopropanes , Double-Blind Method , Drug Administration Schedule , Drug Resistance, Viral , Drug-Related Side Effects and Adverse Reactions , Female , HIV Infections/virology , HIV-1/enzymology , HIV-1/genetics , Humans , Indoles/administration & dosage , Indoles/chemical synthesis , Male , Mutation , Phosphinic Acids/administration & dosage , Phosphinic Acids/chemical synthesis , Placebos , Reverse Transcriptase Inhibitors/administration & dosage , Reverse Transcriptase Inhibitors/chemical synthesis , Viral Load/drug effects
18.
AIDS Rev ; 13(4): 198-213, 2011.
Article in English | MEDLINE | ID: mdl-21975356

ABSTRACT

The global prevalence of HIV infection in the female population presents a significant healthcare burden in terms of mother-to-child transmission (MTCT) of the disease. This review aims to discuss current trends and treatment guidelines for the use of antiretroviral therapy during pregnancy and associated complications in this population. Historically, antiretroviral monotherapy with zidovudine was commonly used for preventing MTCT, and monotherapy with single-dose nevirapine is still used for prevention in resource-limited settings. Evidence suggests that combination therapy with HAART is a more effective treatment option than monotherapy when managing HIV in pregnant women. Current treatment guidelines recommend the use of HAART with a protease inhibitor (PI) or a nonnucleoside reverse transcriptase inhibitor (NNRTI) plus two nucleoside reverse transcriptase inhibitors (NRTI) as first-line therapy for the management of HIV infection in pregnant women and for preventing MTCT. Complications associated with the use of antiretroviral therapy during pregnancy should be taken into consideration when selecting a new antiretroviral regimen, or when continuing certain antiretroviral regimens in HIV-infected women who become pregnant while on therapy. NNRTI have been associated with severe and sometimes fatal hepatoxicity in some pregnant women and potentially teratogenic side effects in the fetus, and their use raises concerns regarding the development of drug- and class-resistant mutations. PI-based HAART has been associated with an increased risk of adverse effects such as premature delivery, low birth weight, dyslipidemia, glucose intolerance, and lipodystrophy. Despite this, initiating antiretroviral therapy with a PI plus two NRTI may become the preferred treatment option in pregnant women. Many of the side effects associated with PI were more prevalent when older PI and PI-based regimens that included those in combination with thymidine analog NRTI were used. An individual's history and baseline clinical and laboratory parameters should also be taken into consideration when choosing the most appropriate antiretroviral regimen during pregnancy.


Subject(s)
Anti-HIV Agents/therapeutic use , Antiretroviral Therapy, Highly Active/trends , HIV Infections/drug therapy , Infectious Disease Transmission, Vertical/prevention & control , Pregnancy Complications, Infectious/drug therapy , Anti-HIV Agents/adverse effects , Antiretroviral Therapy, Highly Active/adverse effects , Drug Therapy, Combination , Female , HIV Infections/prevention & control , HIV Infections/transmission , Humans , Infant, Newborn , Nevirapine/adverse effects , Nevirapine/therapeutic use , Pregnancy , Protease Inhibitors/administration & dosage , Protease Inhibitors/therapeutic use , Reverse Transcriptase Inhibitors/administration & dosage , Reverse Transcriptase Inhibitors/adverse effects , Reverse Transcriptase Inhibitors/therapeutic use , Zidovudine/administration & dosage , Zidovudine/adverse effects , Zidovudine/therapeutic use
19.
J Antimicrob Chemother ; 66(11): 2601-4, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21890538

ABSTRACT

BACKGROUND: Efavirenz is among the most widely used antiretroviral drugs. Increased efavirenz exposure has been associated with CNS side effects and also with the chance of emergence of resistance upon treatment interruptions. The objective of this study was to evaluate factors associated with efavirenz plasma concentrations in a cohort of HIV-infected individuals. METHODS: From July 2009 to March 2010, HIV-infected patients with efavirenz as part of antiretroviral therapy (600 mg at night), undetectable viral load for at least 1 year and CD4 cell count >200 cells/mm(3) were consecutively enrolled at the HIV/AIDS ambulatory care unit in southern Brazil. Plasma samples were taken 18-23 h after efavirenz last dose and analysed by validated ultra-performance liquid chromatography. RESULTS: Forty-one subjects were included (21 females). Mean age and weight were 45.4 years and 70.9 kg, respectively. Mean efavirenz plasma concentration was 2.20 ±â€Š2.17 mg/L. Most plasma concentrations (73%) were within the therapeutic window (1-4 mg/L); 17% were below and 10% above the limits. There were no significant associations between efavirenz concentration and age, CD4 cell count, time on antiretroviral treatment and gender. There was significant and inverse correlation between efavirenz concentrations and body weight (P = 0.013) and body mass index (P = 0.001). CONCLUSIONS: In this cohort of well-controlled HIV-positive individuals, patients with lower weight or body mass index had a higher chance of presenting elevated plasma concentrations of efavirenz. Therapeutic drug monitoring to adjust dose might be a helpful tool to decrease efavirenz dose in order to minimize costs and adverse effects.


Subject(s)
Anti-HIV Agents/blood , Benzoxazines/blood , Body Weight , HIV Infections/drug therapy , Reverse Transcriptase Inhibitors/blood , Adult , Aged , Aging , Alkynes , Anti-HIV Agents/administration & dosage , Anti-HIV Agents/adverse effects , Benzoxazines/administration & dosage , Benzoxazines/adverse effects , CD4 Lymphocyte Count , Cyclopropanes , Drug Monitoring , Female , HIV Infections/blood , HIV Infections/virology , HIV-1/drug effects , Humans , Male , Middle Aged , Reverse Transcriptase Inhibitors/administration & dosage , Reverse Transcriptase Inhibitors/adverse effects , Viral Load
20.
Arzneimittelforschung ; 61(1): 55-60, 2011.
Article in English | MEDLINE | ID: mdl-21355446

ABSTRACT

Tenofovir disoproxil fumarate (TDF, CAS 147127-20-6) is a nucleotide reverse transcriptase inhibitor which is indicated in combination with other antiretroviral agents for the management of HIV-1 infection. The objective of this study was to compare the rate and extent of absorption and to assess the bioequivalence between a new pharmaceutical equivalent tablet formulation containing 300 mg of TDF and the innovator product. A randomized, single-center, open-label, single-dose, two-way crossover bioequivalence study in 40 healthy adult subjects was conducted. Dosing was separated by a wash-out period of 14 days. Blood samples were collected over 48 h and plasma levels of tenofovir (TFV) were determined by a validated HPLC assay. Rate and extent of absorption were similar between products. The 90% confidence interval (CI) of the ratio of the geometric means for log-transformed C(max), AUC(last) and AUC(inf) values were used to assess bioequivalence between the two formulations using the equivalence interval of 80 and 125%. In healthy subjects, the point estimate and 90% CI of the ratios of C(max), AUC(last) and AUC(inf) values were 0.99 (0.92-1.02), 0.99 (0.95-1.03) and 0.93 (0.85-1.02), respectively. Both treatments exhibited similar tolerability and safety. It was concluded that the new pharmaceutical product was bioequivalent to the innovator.


Subject(s)
Adenine/analogs & derivatives , Organophosphonates/pharmacokinetics , Reverse Transcriptase Inhibitors/pharmacokinetics , Adenine/administration & dosage , Adenine/adverse effects , Adenine/pharmacokinetics , Adult , Blood Cell Count , Blood Chemical Analysis , Chemistry, Pharmaceutical , Chromatography, High Pressure Liquid , Cross-Over Studies , Female , Food-Drug Interactions , Humans , Intestinal Absorption , Male , Middle Aged , Organophosphonates/administration & dosage , Organophosphonates/adverse effects , Reverse Transcriptase Inhibitors/administration & dosage , Reverse Transcriptase Inhibitors/adverse effects , Spectrometry, Fluorescence , Tablets , Tenofovir , Therapeutic Equivalency , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL