Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Biotechnol J ; 18(10): e2300008, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37300817

ABSTRACT

Biofilm-associated infections are a critical element in infectious diseases and play an important role in antibiotic resistance. Biosynthesized gold nanoparticles (AuNPs) using ethanolic extract of Musa sapientum unripe fruit were performed. The nanoparticles demonstrated an absorption peak at 554 nm with particle sizes ranging from 5.45 to 104.44 nm. High negative zeta potential value of -33.97 mV confirmed the high stability of AuNPs. The presence of bioconstituents responsible for capping and stabilization was indicated by intensity changes of several peaks from Fourier-transform infrared spectroscopy analysis. The minimum inhibitory concentrations (MIC) of the biosynthesized AuNPs against important pathogens ranged from 10 to 40 µg mL-1 . Synthesized nanoparticles at 0.062 to 0.5 × MIC significantly inhibited biofilm formation in all the tested microorganisms (p < 0.05). Scanning electron microscopy and confocal scanning laser microscopy images clearly illustrated in disruption and architectural changes of microbial biofilms at sub-MIC of biosynthesized AuNPs. Excellent antioxidant and antityrosinase activities of AuNPs were observed. The biosynthesized AuNPs at 20 µg mL-1 significantly inhibited nitric oxide production by 93% in lipopolysaccharide-stimulated RAW 264.7 cells, compared with control (p < 0.05). The biosynthesized AuNPs at 0.6 to 40 µg mL-1 demonstrated no toxic effects on L929 fibroblast cells.

2.
J Proteome Res ; 20(1): 154-163, 2021 01 01.
Article in English | MEDLINE | ID: mdl-32911932

ABSTRACT

It is well known that most microbial populations develop their intrinsic antibiotics resistance at low concentrations in antibiotics environments, but the factors influencing spontaneous resistance are still largely unknown. In this study, Aeromonas hydrophila strains with different resistance levels to oxytetracycline (OXY) were induced by sublethal antibiotic selection pressure, and differential expression of proteins was compared among them using iTRAQ-based quantitative proteomics. Our following bioinformatic analysis showed that energy metabolism-related proteins were downregulated, while several iron-related proteins were upregulated in high OXY-resistant strains. To further investigate the role of spontaneous OXY resistance evolution, four TonB-dependent receptor-coded genes were deleted, and their OXY susceptibility capabilities and antibiotic evolutionary assays were performed, respectively. Our results showed that the deletion of these genes did not affect the susceptibility to OXY, but showed different evolution rates in the spontaneous OXY evolution compared with wild-type strain, especially for AHA_0971 and AHA_4251. Therefore, this study indicates the important role of TonB-dependent receptor proteins during the bacterial antibiotics resistance evolution and may provide a new prophylactic strategy against the development of antibiotic resistance.


Subject(s)
Aeromonas hydrophila , Oxytetracycline , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , Drug Resistance, Bacterial/genetics , Oxytetracycline/pharmacology
3.
Front Microbiol ; 11: 577376, 2020.
Article in English | MEDLINE | ID: mdl-33013815

ABSTRACT

Aeromonas hydrophila is an aquatic pathogen of freshwater fish. The emergence of widespread antimicrobial-resistance strains of this pathogen has caused increasing rates of fish infections. Our previous research reported that A. hydrophila yeeY, a LysR-type transcriptional regulator (LTTR), negatively regulated furazolidone (FZ) resistance. Although, it's intrinsic regulatory mechanism is still unclear. In this study, a data-independent acquisition (DIA) quantitative proteomics method was used to compare the differentially expressed proteins (DEPs) between the ΔyeeY and wild-type strain under FZ treatment. When compared to the control, a total of 594 DEPs were identified in ΔyeeY. Among which, 293 and 301 proteins were substantially increased and decreased in abundance, respectively. Bioinformatics analysis showed that several biological pathways such as the secretion system and protein transport were mainly involved in FZ resistance. Subsequently, the antibiotics susceptibility assays of several gene deletion strains identified from the proteomics results showed that YeeY may regulate some important genes such as cysD, AHA_2766, AHA_3195, and AHA_4275, which affects the FZ resistance in A. hydrophila. Furthermore, 34 antimicrobial resistance genes (ARGs) from the bacterial drug resistance gene database (CARD) were found to be directly or indirectly regulated by YeeY. A subsequent assay of several ARGs mutants showed that ΔAHA_3222 increased the susceptibility of A. hydrophila to FZ, while ΔcysN and ΔAHA_3753 decreased the susceptibility rate. Finally, the chromatin immunoprecipitation (ChIP) PCR and an electrophoretic mobility shift assay (EMSA) have revealed that the genes such as AHA_3222 and AHA_4275 were directly and transcriptionally regulated by YeeY. Taken together, our findings demonstrated that YeeY may participate in antimicrobial resistance of A. hydrophila to FZ, which provides a new target for the development of novel antimicrobial agents in the future.

4.
Microb Pathog ; 140: 103952, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31899323

ABSTRACT

Acyl-homoserine-lactone synthase (AhyI) of Aeromonas hydrophila can produce quorum sensing (QS) auto-inducer 1 (AI-1) type signal molecule, which plays important roles in various biological phenomenons such as biofilm formation, hemolysin production and motility. Previous research revealed that the AhyI of A. hydrophila has acetylation modification on lysine 7 site, but its intrinsic biological function is still largely unknown. To study the effect of AhyI protein and its acetylation modification on the physiological traits of A. hydrophila, the site-directed mutagenesis strains including ΔahyI::ahyI-K7Q and ΔahyI::ahyI-K7R were made in this study. The mutation at K7 site of lysine acetylation in AhyI protein decreased the protease production, but the lysine acetylations do not affect the biofilm formation and hemolysin production. To further study the effect of lysine acetylation on AI-1 signal molecule production, the acyl-homoserine lactones (AHLs) extraction and bioluminescence quantification were performed. Compared with the rescue strain, the acetylation on K7 of AhyI resulted in a decreased level of AHLs and bioluminescence production. It indicated that the lysine acetylation modification on the AhyI protein can regulate the production of signalling molecules. Overall, the obtained data in this study provide a theoretical basis for further understanding the role of lysine acetylation of AhyI protein and lay a foundation to systematically study the regulatory mechanism of QS.


Subject(s)
Aeromonas hydrophila/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Lysine/metabolism , Acetylation , Aeromonas hydrophila/chemistry , Aeromonas hydrophila/genetics , Aeromonas hydrophila/growth & development , Amino Acid Motifs , Bacterial Proteins/genetics , Biofilms , Gene Expression Regulation, Bacterial , Lysine/genetics
5.
Front Cell Infect Microbiol ; 10: 626574, 2020.
Article in English | MEDLINE | ID: mdl-33614530

ABSTRACT

Recent studies have shown that a key strategy of many pathogens is to use post-translational modification (PTMs) to modulate host factors critical for infection. Lysine succinylation (Ksuc) is a major PTM widespread in prokaryotic and eukaryotic cells, and is associated with the regulation of numerous important cellular processes. Vibrio alginolyticus is a common pathogen that causes serious disease problems in aquaculture. Here we used the affinity enrichment method with LC-MS/MS to report the first identification of 2082 lysine succinylation sites on 671 proteins in V. alginolyticus, and compared this with the lysine acetylation of V. alginolyticus in our previous work. The Ksuc modification of SodB and PEPCK proteins were further validated by Co-immunoprecipitation combined with Western blotting. Bioinformatics analysis showed that the identified lysine succinylated proteins are involved in various biological processes and central metabolism pathways. Moreover, a total of 1,005 (25.4%) succinyl sites on 502 (37.3%) proteins were also found to be acetylated, which indicated that an extensive crosstalk between acetylation and succinylation in V. alginolyticus occurs, especially in three central metabolic pathways: glycolysis/gluconeogenesis, TCA cycle, and pyruvate metabolism. Furthermore, we found at least 50 (7.45%) succinylated virulence factors, including LuxS, Tdh, SodB, PEPCK, ClpP, and the Sec system to play an important role in bacterial virulence. Taken together, this systematic analysis provides a basis for further study on the pathophysiological role of lysine succinylation in V. alginolyticus and provides targets for the development of attenuated vaccines.


Subject(s)
Lysine , Vibrio alginolyticus , Acetylation , Chromatography, Liquid , Lysine/metabolism , Protein Processing, Post-Translational , Tandem Mass Spectrometry , Vibrio alginolyticus/metabolism , Virulence
6.
Pathog Dis ; 76(5)2018 07 01.
Article in English | MEDLINE | ID: mdl-29893828

ABSTRACT

Staphylococcal infections associated with indwelling medical devices are difficult to eradicate owing to its recalcitrant nature of biofilms to conventional antibiotics. In our earlier study, we reported the efficacy of geraniol (GE) in inhibiting the in vitro biofilm formation of Staphylococcus epidermidis and adaptive resistant development. To examine the in vivo potential of GE in eradicating the in vivo colonization of S. epidermidis, an implanted rat jugular vein catheter model was developed. Oral supplementation of GE (GE at 200 mg/kg bw for three days) in rats infected with S. epidermidis exhibited a significant reduction of the bacterial burden in catheter, blood, heart and kidney, when compared to the untreated infection control. In addition, GE supplemented animals showed significantly reduced level of inflammatory markers such as nitric oxide and malondialdehyde in heart and kidney tissues. Furthermore, in contrast to the infection control, histopathology analysis of the heart and kidney tissues of the GE-treated group showed a normal histoarchitecture similar to animal control. Thus, the outcome of the present study exhibits the potential of GE as antibiofilm and anti-inflammatory agent against S. epidermidis infections. Furthermore, elucidating the molecular mechanism of GE is important to exploit the therapeutic efficacy of GE.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Catheter-Related Infections/prevention & control , Catheterization, Central Venous/adverse effects , Catheters/microbiology , Staphylococcal Infections/prevention & control , Staphylococcus epidermidis/drug effects , Terpenes/administration & dosage , Acyclic Monoterpenes , Administration, Oral , Animal Structures/microbiology , Animal Structures/pathology , Animals , Catheter-Related Infections/microbiology , Catheter-Related Infections/pathology , Disease Models, Animal , Histocytochemistry , Jugular Veins , Rats , Staphylococcal Infections/microbiology , Staphylococcal Infections/pathology
7.
Antonie Van Leeuwenhoek ; 111(4): 501-515, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29101490

ABSTRACT

The aim of this study was to evaluate the anti-biofilm and anti-virulence properties of petroselinic acid (PSA) against the environmental pathogen Serratia marcescens. PSA significantly inhibited the quorum sensing (QS)-dependent virulence factors such as prodigiosin, protease productions, and biofilm formation in S. marcescens. The antibiofilm potential of PSA was also confirmed through light, confocal laser scanning, and scanning electron microscopic analyses. Furthermore, PSA effectively inhibited the biofilm-related phenomena such as exopolysaccharide production, hydrophobicity production, swimming, and swarming motility without affecting the bacterial growth. In FT-IR analysis, the PSA treated S. marcescens cells displayed a reduction in cellular components compared to the untreated controls. The real-time analysis revealed the downregulation of QS-controlled virulence genes such as bsmB, fimA, fimC, and flhD in S. marcescens on treatment with PSA. The obtained results strongly suggested that PSA could be further explored as an antipathogenic drug to treat QS-mediated infections caused by S. marcescens.


Subject(s)
Biofilms/drug effects , Gene Expression Regulation, Bacterial/drug effects , Oleic Acids/pharmacology , Quorum Sensing/genetics , Serratia marcescens/drug effects , Serratia marcescens/physiology , Virulence/drug effects , Anti-Bacterial Agents/pharmacology , Polysaccharides, Bacterial/genetics , Spectroscopy, Fourier Transform Infrared , Virulence/genetics , Virulence Factors/genetics
8.
Antimicrob Agents Chemother ; 60(9): 5135-40, 2016 09.
Article in English | MEDLINE | ID: mdl-27216057

ABSTRACT

Tenofovir alafenamide (TAF) is an oral prodrug of tenofovir (TFV) that has greater stability in plasma than TFV disoproxil fumarate (TDF) and circulates as intact TAF, resulting in the direct and higher lymphatic loading of and exposure to TFV diphosphate, the active moiety. Unlike TFV, TAF is minimally eliminated in urine. The pharmacokinetics (PK) of TAF and TFV in HIV-uninfected subjects with severe renal impairment and matched healthy controls were evaluated. Subjects with severe renal impairment (RI; estimated glomerular filtration rate [eGFR], 15 to 29 ml/min) and controls (eGFR, ≥90 ml/min) matched for age, gender, and body mass index received a single dose of TAF at 25 mg. Blood and urine samples for TAF and TFV PK determinations were collected over 7 days postdosing, and subjects were followed up at 14 days. A total of 14 renally impaired subjects and 13 control subjects enrolled and completed the study. The TAF maximum observed concentration in plasma (Cmax) and the area under the concentration-versus-time curve (AUC) extrapolated to infinite time (AUCinf) were 79% and 92% higher, respectively, in subjects with severe RI than the controls, primarily due to higher absorption. The TFV Cmax and AUCinf were 2.8-fold and 5.7-fold higher, respectively, in subjects with severe RI than the controls. In subjects with severe RI, TAF at 25 mg provided a TFV AUC 10 to 40% lower than that from historical TDF-based TFV exposures in subjects with normal renal function. There were no discontinuations due to adverse events. In subjects with severe RI receiving TAF at 25 mg, TAF exposures were higher than those for the controls; these differences are unlikely to be clinically meaningful. TFV exposures were higher than those for the controls but lower than the exposures in nonrenally impaired subjects on TDF-based regimens.


Subject(s)
Adenine/analogs & derivatives , Anti-HIV Agents/blood , Renal Insufficiency, Chronic/blood , Adenine/blood , Adenine/pharmacokinetics , Aged , Alanine , Anti-HIV Agents/pharmacokinetics , Area Under Curve , Case-Control Studies , Female , Glomerular Filtration Rate , HIV Infections , Humans , Male , Middle Aged , Renal Insufficiency, Chronic/physiopathology , Tenofovir/analogs & derivatives
9.
Clin Pharmacokinet ; 55(1): 33-45, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26242379

ABSTRACT

Idelalisib is a potent and selective phosphatidylinositol 3-kinase-δ inhibitor, which is a first-in-class agent to be approved for the treatment of relapsed chronic lymphocytic leukaemia, follicular B cell non-Hodgkin's lymphoma and small lymphocytic lymphoma. In dose-ranging studies, idelalisib exposure increased in a less than dose-proportional manner, likely because of solubility-limited absorption. The approved starting dose of 150 mg twice daily was supported by extensive exposure-response evaluations, with dose reduction to 100 mg twice daily being allowed for specific toxicities. Idelalisib may be administered without regard to food on the basis of the absence of clinically relevant food effects, and was accordingly dosed in primary efficacy/safety studies. Idelalisib is metabolized primarily via aldehyde oxidase (AO) and, to a lesser extent, via cytochrome P450 (CYP) 3A. Coadministration with the strong CYP3A inhibitor ketoconazole 400 mg once daily resulted in a ~79 % increase in the idelalisib area under the plasma concentration-time curve (AUC). Administration with the potent inducer rifampin resulted in a 75 % decrease in idelalisib exposure (AUC) and, as such, coadministration with strong inducers should be avoided. GS-563117 is an inactive primary circulating metabolite of idelalisib formed mainly via AO. Unlike idelalisib, GS-563117 is a mechanism-based inhibitor of CYP3A. Accordingly, idelalisib 150 mg twice-daily dosing increases the midazolam AUC 5.4-fold. Clinically, idelalisib is not an inhibitor of the transporters P-glycoprotein, breast cancer resistance protein, organic anion-transporting polypeptide (OATP) 1B1 or OAPT1B3. In a population pharmacokinetic model, no meaningful impact on idelalisib pharmacokinetics was noted for any of the covariates tested. Idelalisib exposure was ~60 % higher with moderate/severe hepatic impairment; no relevant changes were observed with severe renal impairment. This article reviews a comprehensive pharmacology programme, including drug-drug interaction studies and mechanistic and special population studies, which has allowed a thorough understanding of idelalisib clinical pharmacokinetics and their impact on clinical safety and efficacy.


Subject(s)
Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Purines/administration & dosage , Purines/pharmacokinetics , Quinazolinones/administration & dosage , Quinazolinones/pharmacokinetics , Administration, Oral , Dose-Response Relationship, Drug , Drug Interactions , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Lymphoma, Non-Hodgkin/drug therapy , Lymphoma, Non-Hodgkin/metabolism
10.
Antimicrob Agents Chemother ; 58(5): 2564-9, 2014 May.
Article in English | MEDLINE | ID: mdl-24550332

ABSTRACT

Elvitegravir (EVG), an HIV strand transfer integrase inhibitor, is metabolized primarily via cytochrome P450 3A4 (CYP3A) and secondarily via glucuronidation. The pharmacokinetics (PK) and safety of cobicistat (COBI)-boosted EVG (EVG/co) were evaluated in subjects with impaired liver function. The enrolled subjects had stable moderate liver impairment (n = 10; Child-Pugh-Turcotte [CPT] class B) or were healthy controls (n = 10) matched for age (±5 years), gender, and body mass index (±15%). EVG/co (150/150 mg) was administered once daily for 10 days, followed by pharmacokinetic (PK) sampling. Safety was assessed throughout the study. EVG and COBI exposures were compared between the impairment and control groups, with a ≥100% increase considered clinically relevant. EVG and COBI protein binding was also measured. All enrolled subjects completed the study. The treatment-emergent adverse event (AE) incidences were comparable between the groups; all study drug-related AEs were mild. The geometric mean ratio (90% confidence interval [CI]) for EVG area under the concentration-time curve over the dosing interval (AUCtau) and maximum observed plasma concentration (Cmax) were 135% (103%, 177%) and 141% (109%, 183%), respectively. The corresponding values for COBI were 99.8% (76.0%, 131%) and 86.1% (65.4%, 113%), respectively, indicating no clinically relevant change in exposure. No correlations were observed between the EVG and COBI exposures versus CPT score. The EVG- and COBI-free fractions were similar between groups. EVG and COBI do not require dose adjustment in moderate or mild liver impairment, as no clinically relevant PK changes were observed for EVG or COBI in this special population. No PK or safety data are available for EVG or COBI in subjects with severe hepatic impairment.


Subject(s)
Anti-HIV Agents/adverse effects , Anti-HIV Agents/pharmacokinetics , Liver Diseases/metabolism , Quinolones/adverse effects , Quinolones/pharmacokinetics , Adolescent , Adult , Aged , Anti-HIV Agents/blood , Female , Humans , Liver Diseases/blood , Male , Middle Aged , Quinolones/blood , Young Adult
11.
J Clin Pharmacol ; 54(4): 378-85, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24142299

ABSTRACT

The effect of food on rilpivirine/emtricitabine/tenofovir disoproxil fumarate single-tablet regimen (STR) was evaluated in healthy subjects. Subjects (N = 24) received rilpivirine/emtricitabine/tenofovir disoproxil fumarate (25/200/300 mg) under fasted or fed conditions (light [390 kcal, 12 g fat]; standard [540 kcal, 21 g fat]) followed by pharmacokinetic (PK) sampling. The 90% confidence interval (CI) of the geometric mean ratio for rilpivirine, emtricitabine, tenofovir exposure was estimated for fed versus fasted dosing and light versus standard meal, with equivalence boundaries of 80 - 125%. Safety was assessed throughout study. Twenty-three subjects completed the study; one discontinued due to protocol violation. Adverse events were mild to moderate. Emtricitabine PK was unaffected. Tenofovir AUCinf was 38% and 28% higher, respectively, with standard and light meal versus fasted. Rilpivirine AUCinf and Cmax were 16% and 26% higher with a standard, and 9% and 34% with a light meal, respectively, versus fasted. Compared to standard meal, the lower limit of rilpivirine AUClast and AUCinf when taken with the light meal were narrowly below the equivalence bounds (79.9 and 79.2, respectively), rilpivirine Cmax was narrowly above (129). Rilpivirine/emtricitabine/tenofovir disoproxil fumarate should be administered with food, which can be a standard or light meal.


Subject(s)
Adenine/analogs & derivatives , Anti-HIV Agents/pharmacokinetics , Deoxycytidine/analogs & derivatives , Food-Drug Interactions , HIV Infections/metabolism , Nitriles/pharmacokinetics , Organophosphonates/pharmacokinetics , Pyrimidines/pharmacokinetics , Adenine/administration & dosage , Adenine/adverse effects , Adenine/pharmacokinetics , Adult , Anti-HIV Agents/administration & dosage , Anti-HIV Agents/adverse effects , Cross-Over Studies , Deoxycytidine/administration & dosage , Deoxycytidine/adverse effects , Deoxycytidine/pharmacokinetics , Drug Combinations , Emtricitabine , Fasting/metabolism , Female , HIV Infections/drug therapy , Humans , Male , Middle Aged , Nitriles/administration & dosage , Nitriles/adverse effects , Organophosphonates/administration & dosage , Organophosphonates/adverse effects , Pyrimidines/administration & dosage , Pyrimidines/adverse effects , Rilpivirine , Tablets , Tenofovir , Young Adult
12.
J Clin Pharmacol ; 54(6): 649-56, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24375014

ABSTRACT

Statins are commonly used medications by HIV-1 patients. Elvitegravir/cobicistat/emtricitabine/tenofovir DF is a single tablet regimen for the treatment of HIV. The pharmacokinetic interaction between cobicistat-boosted elvitegravir (EVG/co) and rosuvastatin was evaluated. Breast cancer resistance protein (BCRP) and organic anion transporting polypeptide (OATP) 1B1 and 1B3 inhibition were assessed in vitro. Healthy subjects (N = 12) received a single dose of rosuvastatin 10 mg alone and in combination with EVG/co. Intensive pharmacokinetic sampling was conducted and safety was assessed throughout the study. Rosuvastatin pharmacokinetic exposure parameters were evaluated using 90% confidence intervals (CI) of the geometric mean ratio (GMR) of the test (combination) versus reference (rosuvastatin alone) using equivalence boundaries of 70-143% for AUCinf and 70-175% for Cmax . Elvitegravir and cobicistat inhibited BCRP and OATP in vitro, emtricitabine and TDF did not. Clinically, study treatments were well tolerated, with adverse events generally mild. Upon coadministration, rosuvastatin plasma concentrations increased (Cmax 89% higher), while AUCinf changes were modest (38% higher) and clinically nonrelevant, potentially driven by moderate inhibition of intestinal efflux by BCRP, and/or hepatic uptake by OATPs by EVG/co. Elvitegravir and cobicistat pharmacokinetics were comparable to historical data. Rosuvastatin may be coadministered with EVG/COBI/FTC/TDF without dose adjustment.


Subject(s)
Anti-HIV Agents/pharmacokinetics , Carbamates/pharmacokinetics , Fluorobenzenes/pharmacokinetics , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Pyrimidines/pharmacokinetics , Quinolones/pharmacokinetics , Sulfonamides/pharmacokinetics , Thiazoles/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/antagonists & inhibitors , ATP-Binding Cassette Transporters/metabolism , Adult , Animals , Anti-HIV Agents/blood , Anti-HIV Agents/pharmacology , CHO Cells , Carbamates/blood , Carbamates/pharmacology , Cobicistat , Cricetulus , Dogs , Drug Combinations , Drug Interactions , Drug Therapy, Combination , Female , Fluorobenzenes/blood , Fluorobenzenes/pharmacology , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/blood , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Liver-Specific Organic Anion Transporter 1 , Madin Darby Canine Kidney Cells , Male , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Organic Anion Transporters/antagonists & inhibitors , Organic Anion Transporters/metabolism , Organic Anion Transporters, Sodium-Independent/antagonists & inhibitors , Organic Anion Transporters, Sodium-Independent/metabolism , Pyrimidines/blood , Pyrimidines/pharmacology , Quinolones/blood , Quinolones/pharmacology , Rosuvastatin Calcium , Solute Carrier Organic Anion Transporter Family Member 1B3 , Sulfonamides/blood , Sulfonamides/pharmacology , Thiazoles/blood , Thiazoles/pharmacology
13.
J Acquir Immune Defic Syndr ; 64(1): 45-50, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23774876

ABSTRACT

BACKGROUND: Acid-reducing agents are commonly used co-medications by HIV-1-infected patients receiving antiretroviral treatment. The effects of various representative acid-reducing agents on the pharmacokinetics (PK) of boosted elvitegravir were evaluated by 1-way interaction in 4 studies. METHODS: Healthy subjects received ritonavir-boosted elvitegravir (EVG/r; 50/100 mg QD) administered alone or with antacid simultaneously in Study 1, staggered (±2 or ±4 hours) or with omeprazole in Study 2; Studies 3 and 4 evaluated cobicistat-boosted elvitegravir (EVG/co; 150/150 mg QD) administered simultaneously or staggered (+12 hours) with famotidine or omeprazole. Lack of PK alteration was defined as 90% confidence intervals about the geometric least squares means ratio (coadministration:alone) being within 70%-143% for elvitegravir Cmax (maximum concentration), Ctau (trough), and AUCtau (area under plasma concentration-time curve; 0-24 hours); cobicistat PK were explored. RESULTS: EVG exposures were 40%-50% lower upon simultaneous dosing of EVG/r and antacids, probably due to local complexation with cations in gastrointestinal tract, and were unaffected with ≥2 hours staggered dosing. No relevant drug interactions were observed between EVG/co and famotidine or between EVG/r or EVG/co and omeprazole, indicating the absence of a broader pH effect on boosted EVG PK. In all studies, study treatments were well tolerated, with adverse events being generally mild to moderate in severity and primarily gastrointestinal disorders. CONCLUSIONS: There are no clinically relevant interactions between boosted elvitegravir, and thus elvitegravir/cobicistat/emtricitabine/tenofovir DF single-tablet regimen, and H2-receptor antagonists or proton pump inhibitors; staggered antacid administration by ≥2 hours is recommended.


Subject(s)
Antacids/pharmacology , Anti-HIV Agents/pharmacology , Carbamates/pharmacology , Quinolones/pharmacokinetics , Thiazoles/pharmacology , Adolescent , Adult , Antacids/administration & dosage , Anti-HIV Agents/administration & dosage , Area Under Curve , Carbamates/administration & dosage , Cobicistat , Cross-Over Studies , Drug Administration Schedule , Drug Interactions , Drug Therapy, Combination , Female , Humans , Male , Middle Aged , Quinolones/administration & dosage , Thiazoles/administration & dosage
14.
J Acquir Immune Defic Syndr ; 63(4): 480-4, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23599011

ABSTRACT

BACKGROUND: Interactions between HIV and opioid-dependence therapies are known to occur. We sought to determine if such interactions occurred between buprenorphine/naloxone and elvitegravir boosted with cobicistat. METHODS: We performed a within-subject open-labeled pharmacokinetic and pharmacodynamic study in 17 HIV-seronegative subjects stabilized on at least 2 weeks of buprenorphine/naloxone therapy. Subjects underwent baseline and steady state evaluation of the effect of elvitegravir 150 mg once daily boosted with 150 mg once daily of cobicistat (EVG/COBI) on buprenorphine/naloxone parameters. Safety was monitored throughout the study. RESULTS: Compared with baseline values, buprenorphine mean AUCtau (69.0 versus 95.6 hr*ng/mL) and mean Cmax (8.4 versus 9.3 ng/mL) increased significantly in the presence of EVG/COBI. Compared with baseline values, norbuprenorphine mean AUCtau (103.4 versus 163.4 hr*ng/mL) and mean Cmax (6.9 versus 9 ng/mL) also increased significantly after achieving steady state EVG/COBI. Naloxone mean AUCtau (0.57 versus 0.45 hr*ng/mL) and mean Cmax (0.25 versus 0.16 ng/mL) decreased after the addition of EVG/COBI. The AUCtau, Cmax and Ctau of EVG and cobicistat did not significantly differ from historical controls. Opioid withdrawal or overdose was not observed among subjects in this study. CONCLUSION: The addition of EVG/COBI to stabilized patients receiving buprenorphine/naloxone modestly increased buprenorphine and norbuprenorphine levels without affecting the opioid pharmacodynamics.


Subject(s)
Anti-Retroviral Agents/pharmacology , Buprenorphine/pharmacokinetics , Carbamates/pharmacology , Naloxone/pharmacokinetics , Narcotic Antagonists/pharmacokinetics , Quinolones/pharmacology , Thiazoles/pharmacology , Adult , Area Under Curve , Buprenorphine/pharmacology , Buprenorphine/therapeutic use , Cobicistat , Drug Interactions , Female , HIV Infections/complications , HIV Infections/drug therapy , HIV Seronegativity , Humans , Male , Middle Aged , Naloxone/pharmacology , Naloxone/therapeutic use , Narcotic Antagonists/pharmacology , Narcotic Antagonists/therapeutic use , Opioid-Related Disorders/complications , Opioid-Related Disorders/drug therapy
15.
J Acquir Immune Defic Syndr ; 62(5): 483-6, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23337366

ABSTRACT

This ongoing, randomized, double-blind, active-controlled phase 3 international trial demonstrated the noninferior efficacy of elvitegravir/cobicistat/emtricitabine/tenofovir DF (EVG/COBI/FTC/TDF) compared with atazanavir boosted by ritonavir (ATV/RTV) plus emtricitabine/tenofovir disoproxil fumarate (FTC/TDF) at 48 weeks. Here, we report the week 96 results. Of 708 treated subjects, virological success (Food and Drug Administration snapshot) was maintained at week 96 with EVG/COBI/FTC/TDF and ATV/RTV + FTC/TDF (83% vs 82%, difference 1.1%, 95% confidence interval -4.5% to 6.7%). Study drug discontinuations due to adverse events were low (4% vs 6%). Median increases from baseline in serum Cr (mg/dL) in EVG/COBI/FTC/TDF vs ATV/RTV + FTC/TDF at week 96 (0.12 vs 0.08) were similar to those at week 48 (0.12 vs 0.08). EVG/COBI/FTC/TDF showed similar mean decreases (%) in bone mineral density from baseline vs ATV/RTV + FTC/TDF (hip: -3.16 vs -4.19, P = 0.069; spine: -1.96 vs -3.54, P = 0.049). Overall, week 96 results support durable efficacy and safety of EVG/COBI/FTC/TDF in HIV-1-infected patients.


Subject(s)
Anti-HIV Agents/therapeutic use , HIV Infections/drug therapy , HIV-1/isolation & purification , Adenine/analogs & derivatives , Adenine/therapeutic use , Adult , Alanine Transaminase/blood , Aspartate Aminotransferases/blood , Atazanavir Sulfate , Bilirubin/blood , Bone Density/drug effects , Carbamates/therapeutic use , Cobicistat , Confidence Intervals , Creatinine/blood , Deoxycytidine/analogs & derivatives , Deoxycytidine/therapeutic use , Double-Blind Method , Drug Combinations , Emtricitabine , Female , HIV Infections/blood , HIV Infections/virology , HIV-1/genetics , Humans , Male , Oligopeptides/therapeutic use , Organophosphonates/therapeutic use , Pyridines/therapeutic use , Quinolones/therapeutic use , RNA, Viral/blood , Ritonavir/therapeutic use , Tenofovir , Thiazoles/therapeutic use , Triglycerides/blood
16.
Lancet ; 379(9835): 2429-2438, 2012 Jun 30.
Article in English | MEDLINE | ID: mdl-22748590

ABSTRACT

BACKGROUND: The HIV integrase strand transfer inhibitor elvitegravir (EVG) has been co-formulated with the CYP3A4 inhibitor cobicistat (COBI), emtricitabine (FTC), and tenofovir disoproxil fumarate (TDF) into a once-daily, single tablet. We compared EVG/COBI/FTC/TDF with a ritonavir-boosted (RTV) protease inhibitor regimen of atazanavir (ATV)/RTV+FTC/TDF as initial therapy for HIV-1 infection. METHODS: This phase 3, non-inferiority study enrolled treatment-naive patients with an HIV-1 RNA concentration of 5000 copies per mL or more and susceptibility to atazanavir, emtricitabine, and tenofovir. Patients were randomly assigned (1:1) to receive EVG/COBI/FTC/TDF or ATV/RTV+FTC/TDF plus matching placebos, administered once daily. Randomisation was by a computer-generated random sequence, accessed via an interactive telephone and web response system. Patients, and investigators and study staff who gave treatments, assessed outcomes, or analysed data were masked to the assignment. The primary endpoint was HIV RNA concentration of 50 copies per mL or less after 48 weeks (according to the US FDA snapshot algorithm), with a 12% non-inferiority margin. This trial is registered with ClinicalTrials.gov, number NCT01106586. FINDINGS: 1017 patients were screened, 715 were enrolled, and 708 were treated (353 with EVG/COBI/FTC/TDF and 355 with ATV/RTV+FTC/TDF). EVG/COBI/FTC/TDF was non-inferior to ATV/RTV+FTC/TDF for the primary outcome (316 patients [89·5%] vs 308 patients [86·8%], adjusted difference 3·0%, 95% CI -1·9% to 7·8%). Both regimens had favourable safety and tolerability; 13 (3·7%) versus 18 (5·1%) patients discontinued treatment because of adverse events. Fewer patients receiving EVG/COBI/FTC/TDF had abnormal results in liver function tests than did those receiving ATV/RTV+FTC/TDF and had smaller median increases in fasting triglyceride concentration (90 µmol/L vs 260 µmol/L, p=0·006). Small median increases in serum creatinine concentration with accompanying decreases in estimated glomerular filtration rate occurred in both study groups by week 2; they generally stabilised by week 8 and did not change up to week 48 (median change 11 µmol/L vs 7 µmol/L). INTERPRETATION: If regulatory approval is given, EVG/COBI/FTC/TDF would be the first integrase-inhibitor-based regimen given once daily and the only one formulated as a single tablet for initial HIV treatment. FUNDING: Gilead Sciences.


Subject(s)
Anti-HIV Agents/administration & dosage , HIV Infections/drug therapy , HIV-1 , Adenine/administration & dosage , Adenine/analogs & derivatives , Adult , Atazanavir Sulfate , Carbamates/administration & dosage , Cobicistat , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Double-Blind Method , Drug Combinations , Emtricitabine , Female , Humans , Male , Middle Aged , Oligopeptides/administration & dosage , Organophosphonates/administration & dosage , Pyridines/administration & dosage , Quinolones/administration & dosage , Ritonavir/administration & dosage , Tenofovir , Thiazoles/administration & dosage
17.
AIDS ; 25(6): F7-12, 2011 Mar 27.
Article in English | MEDLINE | ID: mdl-21412057

ABSTRACT

OBJECTIVE: To assess the safety and efficacy of two, single-tablet regimens for the initial treatment of HIV infection. DESIGN: Phase 2, randomized, double-blind, double-dummy, multicenter, active-controlled study. METHODS: Antiretroviral treatment-naive adults with a screening HIV-1 RNA at least 5000 copies/ml and a CD4 cell count more than 50 cells/µl were randomized 2: 1 to receive fixed-dose combination tablets of elvitegravir/cobicistat/emtricitabine/tenofovir disoproxil fumarate (EVG/COBI/FTC/TDF; N = 48) or efavirenz/emtricitabine/tenofovir disoproxil fumarate (EFV/FTC/TDF; n = 23) for 48 weeks. The primary endpoint was proportion of participants with HIV-1 RNA less than 50 copies/ml at week 24. RESULTS: Participants receiving EVG/COBI/FTC/TDF exhibited a more rapid decline in HIV-1 RNA and a greater proportion suppressed viral load to less than 50 copies/ml than participants receiving EFV/FTC/TDF. Both EVG/COBI/FTC/TDF and EFV/FTC/TDF resulted in high rates of viral suppression and increases in CD4 cell count. Ninety and 83% of participants suppressed HIV-1 RNA to less than 50 copies/ml both at the 24-week and 48-week visits for EVG/COBI/FTC/TDF and EFV/FTC/TDF, respectively. Once-daily administration of EVG/COBI/FTC/TDF provided a mean EVG trough concentration 10-fold over its protein binding-adjusted IC(95) across study visits. EVG/FTC/TDF/GS-9350 was generally well tolerated with a lower rate of drug-related central nervous system (17%) and psychiatric (10%) adverse events versus EFV/FTC/TDF (26 and 44%, respectively). Decreases in estimated glomerular filtration rate occurred within the first few weeks of dosing in participants receiving EVG/COBI/FTC/TDF, remained within the normal range and did not progress at week 24 or 48; no participant experienced a clinical adverse event or discontinued study drug due to changes in serum creatinine or renal function. CONCLUSION: Once-daily EVG/COBI/FTC/TDF achieved and maintained a high rate of virologic suppression with fewer central nervous system and psychiatric adverse events compared to a current standard-of-care regimen of EFV/FTC/TDF.


Subject(s)
Adenine/analogs & derivatives , Benzoxazines/administration & dosage , Deoxycytidine/analogs & derivatives , HIV Infections/drug therapy , HIV-1 , Organophosphonates/administration & dosage , Adenine/administration & dosage , Adenine/pharmacology , Adult , Alkynes , Anti-HIV Agents , Benzoxazines/pharmacology , CD4 Lymphocyte Count , Carbamates/administration & dosage , Cobicistat , Cyclopropanes , Deoxycytidine/administration & dosage , Deoxycytidine/pharmacology , Drug Combinations , Emtricitabine , Female , Humans , Male , Organophosphonates/pharmacology , Patient Compliance , Patient Selection , Quinolones/administration & dosage , Tenofovir , Thiazoles/administration & dosage , Treatment Outcome
18.
Clin Pharmacokinet ; 50(4): 229-44, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21348537

ABSTRACT

Elvitegravir is a potent, boosted, once-daily, HIV integrase inhibitor with antiviral activity against wild-type and drug-resistant strains of HIV. Because elvitegravir is metabolized primarily by cytochrome P450 (CYP) 3A enzymes, coadministration with a strong CYP3A inhibitor such as ritonavir or cobicistat (also known as GS-9350), an investigational pharmacoenhancer, substantially increases (boosts) elvitegravir plasma exposures and prolongs its elimination half-life to ∼9.5 hours, allowing once-daily administration of a low 150 mg dose. Boosting also results in low intra- and intersubject pharmacokinetic variability and high elvitegravir trough concentrations (∼6- to 10-fold above the concentration producing 95% inhibition of wild-type HIV-1 virus [IC95] of 45 ng/mL [protein binding-adjusted]), which is the pharmacokinetic parameter best associated with its antiviral activity. Data from extensive evaluation of the potential for boosted elvitegravir to undergo drug-drug interactions with other antiretroviral agents or concomitant medications indicate the absence of clinically relevant interactions or the need for dose modification in several cases, except for dose reduction of elvitegravir from 150 to 85 mg when coadministered with atazanavir/ritonavir or lopinavir/ritonavir. Dose adjustments for maraviroc and rifabutin, when each is coadministered with boosted elvitegravir, are consistent with their observed interactions with other ritonavir-boosted agents. The presence of a strong CYP3A inhibitor such as ritonavir or cobicistat renders the potential for increase in systemic exposures of CYP3A substrates coadministered with boosted elvitegravir. This article reviews a comprehensive pharmacology programme, including drug-drug interaction studies, mechanistic and special population studies, that has allowed a thorough understanding of elvitegravir clinical pharmacokinetics and its impact on pharmacodynamics.


Subject(s)
HIV Integrase Inhibitors/pharmacokinetics , Quinolones/pharmacokinetics , Animals , Drug Interactions , Drug Therapy, Combination , HIV Infections/drug therapy , HIV Integrase Inhibitors/adverse effects , HIV Integrase Inhibitors/pharmacology , HIV Integrase Inhibitors/therapeutic use , Humans , Quinolones/adverse effects , Quinolones/pharmacology , Quinolones/therapeutic use
19.
J Acquir Immune Defic Syndr ; 53(2): 209-14, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19851115

ABSTRACT

BACKGROUND: The pharmacokinetic (PK) interaction between ritonavir-boosted elvitegravir (elvitegravir/r) and maraviroc was evaluated. METHODS: Healthy subjects were randomized to receive elvitegravir/r (150/100 mg once daily) before or after elvitegravir/r plus maraviroc (150 mg twice daily) (group 1; n = 20) or receive maraviroc before or after maraviroc plus elvitegravir/r (group 2; n = 16). All regimens were administered for 10 days and elvitegravir, ritonavir, and maraviroc PK determined. Lack of PK alteration was defined as 90% confidence intervals for ratio of geometric least squares means ratio (coadministration:alone) between 70% and 143% for elvitegravir and ritonavir Cmax (maximum concentration), Ctau (trough), and AUCtau (area under plasma concentration-time curve; 0-24 hours); for maraviroc, given a 100% increase in Cmax and AUCtau (0-12 hours); the predicted 90% confidence intervals were 162% to 247% and 136% to 295%, respectively. RESULTS: Twenty-eight of 36 enrolled subjects completed the study; one discontinuation was due to an adverse event. The most common adverse event across treatments was headache. Upon coadministration, elvitegravir and ritonavir PK were unaltered, but maraviroc exposures were 2-fold to 4-fold higher presumably due to ritonavir-mediated CYP3A-/Pgp inhibition. CONCLUSIONS: During elvitegravir/r plus maraviroc administration, no elvitegravir or ritonavir dose change and a reduced 150-mg dose of maraviroc are recommended.


Subject(s)
Anti-HIV Agents/pharmacokinetics , Cyclohexanes/pharmacokinetics , Quinolones/pharmacokinetics , Ritonavir/pharmacokinetics , Triazoles/pharmacokinetics , Adolescent , Adult , Anti-HIV Agents/administration & dosage , Anti-HIV Agents/blood , Area Under Curve , Cross-Over Studies , Cyclohexanes/administration & dosage , Cyclohexanes/blood , Drug Interactions , Drug Therapy, Combination , Half-Life , Humans , Maraviroc , Quinolones/administration & dosage , Quinolones/blood , Ritonavir/administration & dosage , Ritonavir/blood , Triazoles/administration & dosage , Triazoles/blood , Young Adult
20.
Antivir Ther ; 13(8): 1011-7, 2008.
Article in English | MEDLINE | ID: mdl-19195326

ABSTRACT

BACKGROUND: This crossover, open-label clinical study evaluated the potential for clinically relevant drug interactions between ritonavir-boosted elvitegravir (elvitegravir/r), an HIV integrase inhibitor, and etravirine, a non-nucleoside reverse transcriptase inhibitor. METHODS: Healthy volunteers were randomized into one of two groups, each with two arms. Group 1 (n = 20) followed a sequence of 10-day dosing of elvitegravir/r (150/100 mg once daily) and elvitegravir/r plus etravirine (200 mg twice daily) or the reverse (n = 10 per sequence). Group 2 (n = 14) followed a sequence of 10-day dosing of etravirine and etravirine plus elvitegravir/r or the reverse (n = 7 per sequence), all under fed conditions. Elvitegravir, ritonavir and etravirine pharmacokinetics were determined on days 10 and 20 using non-compartmental analyses. Lack of pharmacokinetic alteration bounds for 90% confidence intervals (CI) about the geometric mean ratio (GMR; coadministration versus alone) were 70-143% for elvitegravir and ritonavir pharmacokinetics (maximum concentration [C(max)], concentration at the end of the dosing interval [C(tau)] and area under the plasma concentration-time curve [AUC(tau); 0-24 h] and 80-125% for etravirine pharmacokinetics (AUC(tau) 0-12 h). RESULTS: Of the 34 enrolled participants, 31 completed the study. There were three discontinuations, but none were caused by adverse events (AEs). The most common treatment-emergent AE was headache. Elvitegravir pharmacokinetic GMR was 6-7% higher following elvitegravir/r plus etravirine dosing versus elvitegravir/r. The GMR for etravirine and ritonavir AUC(tau) were 2.4% and 12.3% lower, respectively. Importantly, the 90% CI for elvitegravir and etravirine pharmacokinetics and AUC(tau) and C(max) for ritonavir were within the lack of alteration bounds. CONCLUSIONS: Elvitegravir/r and etravirine do not undergo clinically relevant drug interactions and can be coadministered without dose adjustment.


Subject(s)
HIV Protease Inhibitors/pharmacokinetics , Pyridazines/pharmacokinetics , Quinolones/pharmacokinetics , Reverse Transcriptase Inhibitors/pharmacokinetics , Ritonavir/administration & dosage , Adolescent , Adult , Cross-Over Studies , Drug Therapy, Combination , Female , HIV Protease Inhibitors/administration & dosage , Humans , Male , Middle Aged , Nitriles , Pyridazines/administration & dosage , Pyridazines/adverse effects , Pyridazines/blood , Pyrimidines , Quinolones/administration & dosage , Quinolones/adverse effects , Quinolones/blood , Reverse Transcriptase Inhibitors/administration & dosage , Ritonavir/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL
...