Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Mol Pharmacol ; 99(3): 184-196, 2021 03.
Article in English | MEDLINE | ID: mdl-33483427

ABSTRACT

The HIV protease inhibitor nelfinavir is currently being analyzed for repurposing as an anticancer drug for many different cancers because it exerts manifold off-target protein interactions, finally resulting in cancer cell death. Xenosensing pregnane X receptor (PXR), which also participates in the control of cancer cell proliferation and apoptosis, was previously shown to be activated by nelfinavir; however, the exact molecular mechanism is still unknown. The present study addresses the effects of nelfinavir and its major and pharmacologically active metabolite nelfinavir hydroxy-tert-butylamide (M8) on PXR to elucidate the underlying molecular mechanism. Molecular docking suggested direct binding to the PXR ligand-binding domain, which was confirmed experimentally by limited proteolytic digestion and competitive ligand-binding assays. Concentration-response analyses using cellular transactivation assays identified nelfinavir and M8 as partial agonists with EC50 values of 0.9 and 7.3 µM and competitive antagonists of rifampin-dependent induction with IC50 values of 7.5 and 25.3 µM, respectively. Antagonism exclusively resulted from binding into the PXR ligand-binding pocket. Impaired coactivator recruitment by nelfinavir as compared with the full agonist rifampin proved to be the underlying mechanism of both effects on PXR. Physiologic relevance of nelfinavir-dependent modulation of PXR activity was investigated in respectively treated primary human hepatocytes, which showed differential induction of PXR target genes and antagonism of rifampin-induced ABCB1 and CYP3A4 gene expression. In conclusion, we elucidate here the molecular mechanism of nelfinavir interaction with PXR. It is hypothesized that modulation of PXR activity may impact the anticancer effects of nelfinavir. SIGNIFICANCE STATEMENT: Nelfinavir, which is being investigated for repurposing as an anticancer medication, is shown here to directly bind to human pregnane X receptor (PXR) and thereby act as a partial agonist and competitive antagonist. Its major metabolite nelfinavir hydroxy-tert-butylamide exerts the same effects, which are based on impaired coactivator recruitment. Nelfinavir anticancer activity may involve modulation of PXR, which itself is discussed as a therapeutic target in cancer therapy and for the reversal of chemoresistance.


Subject(s)
Hepatocytes/metabolism , Nelfinavir/analogs & derivatives , Nelfinavir/pharmacology , Pregnane X Receptor/metabolism , ATP Binding Cassette Transporter, Subfamily B/genetics , Binding Sites , Cytochrome P-450 CYP3A/genetics , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Hep G2 Cells , Hepatocytes/cytology , Hepatocytes/drug effects , Humans , Models, Molecular , Molecular Conformation , Molecular Docking Simulation , Nelfinavir/chemistry , Pregnane X Receptor/agonists , Pregnane X Receptor/antagonists & inhibitors , Pregnane X Receptor/chemistry , Primary Cell Culture
2.
Cancer Immunol Immunother ; 68(9): 1479-1492, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31463653

ABSTRACT

RIG-I is a cytosolic RNA sensor that recognizes short 5' triphosphate RNA, commonly generated during virus infection. Upon activation, RIG-I initiates antiviral immunity, and in some circumstances, induces cell death. Because of this dual capacity, RIG-I has emerged as a promising target for cancer immunotherapy. Previously, a sequence-optimized RIG-I agonist (termed M8) was generated and shown to stimulate a robust immune response capable of blocking viral infection and to function as an adjuvant in vaccination strategies. Here, we investigated the potential of M8 as an anti-cancer agent by analyzing its ability to induce cell death and activate the immune response. In multiple cancer cell lines, M8 treatment strongly activated caspase 3-dependent apoptosis, that relied on an intrinsic NOXA and PUMA-driven pathway that was dependent on IFN-I signaling. Additionally, cell death induced by M8 was characterized by the expression of markers of immunogenic cell death-related damage-associated molecular patterns (ICD-DAMP)-calreticulin, HMGB1 and ATP-and high levels of ICD-related cytokines CXCL10, IFNß, CCL2 and CXCL1. Moreover, M8 increased the levels of HLA-ABC expression on the tumor cell surface, as well as up-regulation of genes involved in antigen processing and presentation. M8 induction of the RIG-I pathway in cancer cells favored dendritic cell phagocytosis and induction of co-stimulatory molecules CD80 and CD86, together with increased expression of IL12 and CXCL10. Altogether, these results highlight the potential of M8 in cancer immunotherapy, with the capacity to induce ICD-DAMP on tumor cells and activate immunostimulatory signals that synergize with current therapies.


Subject(s)
Antineoplastic Agents/therapeutic use , Dendritic Cells/immunology , Melanoma/drug therapy , Nelfinavir/analogs & derivatives , Alarmins/immunology , Antigen Presentation/drug effects , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Calreticulin/metabolism , Caspase 3/metabolism , Cell Differentiation , Cell Line, Tumor , DEAD Box Protein 58/antagonists & inhibitors , HMGB1 Protein/metabolism , Humans , Immunization , Interferons/metabolism , Molecular Targeted Therapy , Nelfinavir/pharmacology , Nelfinavir/therapeutic use , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptors, Immunologic , Signal Transduction
3.
J Clin Pharmacol ; 59(3): 386-393, 2019 03.
Article in English | MEDLINE | ID: mdl-30358179

ABSTRACT

This study aims to evaluate the safety, acceptability, and pharmacokinetics (PK) of an increased dose of nelfinavir (NFV) during the third trimester of pregnancy. The study was registered as part of the International Maternal Pediatric Adolescent AIDS Clinical Trials network (IMPAACT-P1026s), an ongoing multicenter prospective cohort study of antiretroviral PK during pregnancy (NCT00042289). NFV intensive PK evaluations were performed at steady state during the third trimester of pregnancy and 2-3 weeks postpartum. Plasma concentrations of NFV and its active metabolite, hydroxyl-tert-butylamide (M8) were measured using high-performance liquid chromatography with ultraviolet detection. A total of 18 women are included in the analysis. NFV area under the concentration-time curve (AUC) with the increased dose during the third trimester was nearly identical to the standard dose postpartum, with a geometric mean ratio for third trimester to postpartum AUC of 0.98 (90%CI 0.71-1.35). Despite the increased dose, M8 AUC was lower during the third trimester compared to postpartum (0.53, IQR [0.38-0.75]), as was the M8/NFV AUC ratio (0.51, IQR [0.42-0.63]). NFV AUC0-12 was above target in 15 of 18 (83%) of participants during the third trimester compared to 14 of 16 (88%) postpartum. No major safety concerns were noted. Increasing the NFV dose to 1875 mg twice daily during the third trimester achieved similar concentrations postpartum compared to standard dosing (1250 mg twice daily). Increased NFV dose regimens may still have some benefit to human immunodeficiency virus (HIV)-positive pregnant women living in countries where novel protease inhibitors are currently unavailable or in individuals who are intolerant to ritonavir-boosted HIV medications.


Subject(s)
Anti-HIV Agents/pharmacokinetics , HIV Protease Inhibitors/pharmacokinetics , Nelfinavir/pharmacokinetics , Adult , Anti-HIV Agents/blood , Female , HIV Protease Inhibitors/blood , Humans , Nelfinavir/analogs & derivatives , Nelfinavir/blood , Postpartum Period , Pregnancy , Pregnancy Complications, Infectious , Prospective Studies
4.
Molecules ; 17(1): 688-702, 2012 Jan 12.
Article in English | MEDLINE | ID: mdl-22241465

ABSTRACT

ß-Carotene supplements are often taken by individuals living with HIV-1. Contradictory results from in vitro studies suggest that ß-carotene may inhibit or induce cytochrome P450 enzymes and transporters. The study objective was to investigate the effect of ß-carotene on the steady-state pharmacokinetics of nelfinavir and its active metabolite M8 in HIV-1 infected individuals. Twelve hour nelfinavir pharmacokinetic analysis was conducted at baseline and after 28 days of ß-carotene supplementation (25,000 IU twice daily). Nelfinavir and M8 concentrations were measured with validated assays. Non-compartmental methods were used to calculate the pharmacokinetic parameters. Geometric mean ratios comparing day 28 to day 1 area under the plasma concentration-time curve (AUC(0-12 h)), maximum (C(max)) and minimum (C(min)) concentrations of nelfinavir and M8 are presented with 90% confidence intervals. Eleven subjects completed the study and were included in the analysis. There were no significant differences in nelfinavir AUC(0-12 h) and C(min) (-10%, +4%) after ß-carotene supplementation. The M8 C(min) was increased by 31% while the M8 AUC(0-12 h) and C(max) were unchanged. During the 28 day period, mean CD4+ % and CD4+:CD8+ ratio increased significantly (p < 0.01). ß-carotene supplementation increased serum carotene levels but did not cause any clinically significant difference in the nelfinavir and M8 exposure.


Subject(s)
Dietary Supplements , HIV Infections/drug therapy , HIV Protease Inhibitors/pharmacokinetics , HIV-1 , Nelfinavir/analogs & derivatives , Nelfinavir/pharmacokinetics , beta Carotene/administration & dosage , Adult , Area Under Curve , Drug Stability , Female , HIV Infections/virology , Humans , Male , Middle Aged , Viral Load , beta Carotene/pharmacokinetics
5.
Antimicrob Agents Chemother ; 55(11): 5168-71, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21876052

ABSTRACT

Antiretroviral drugs cross from maternal plasma to breast milk and from breast milk to the infant in different concentrations. We measured concentrations of nelfinavir and its active metabolite (M8) in maternal plasma and breast milk from women and in dried blood spots collected from their infants at delivery and postnatal weeks 2, 6, 14, and 24 in the Kisumu Breastfeeding Study, Kisumu, Kenya. Nelfinavir-based antiretroviral regimens given to mothers as prevention of mother-to-child HIV transmission (PMTCT) do not expose the breast-feeding infant to biologically significant concentrations of nelfinavir or M8.


Subject(s)
Anti-HIV Agents/blood , Anti-HIV Agents/metabolism , Milk, Human/metabolism , Nelfinavir/analogs & derivatives , Nelfinavir/blood , Nelfinavir/metabolism , Adult , Breast Feeding , Female , Humans , Pregnancy , Young Adult
6.
Clin Cancer Res ; 17(7): 1796-806, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21355074

ABSTRACT

PURPOSE: We previously reported that nelfinavir (NFV) induces G(1) cell-cycle block and apoptosis selectively in liposarcoma cell lines due to increased SREBP-1 (sterol regulatory element binding protein-1) expression in the absence of increased transcription. We postulate that NFV interferes with regulated intramembrane proteolysis of SREBP-1 and ATF6 (activating transcription factor 6). EXPERIMENTAL DESIGN: Time-lapse, confocal microscopic studies show that NFV inhibits the nuclear translocation of full-length SREBP-1-EGFP and ATF6-EGFP fusion proteins. siRNA-mediated knockdown of site-1 protease (S1P) and/or site-2 protease (S2P) leads to inhibition of SREBP-1 intracellular trafficking to the nucleus and reduces liposarcoma cell proliferation. Treatment of LiSa-2 liposarcoma cells with 3,4-dichloroisocoumarin, a serine protease inhibitor of S1P, did not affect SREBP-1 processing. In contrast, 1,10-phenanthroline, an S2P-specific inhibitor, reproduces the molecular and biological phenotypes observed in NFV-treated cells, which implicates S2P as a target of NFV. In vivo evaluation of NFV in a murine liposarcoma xenograft model leads to inhibition of tumor growth without significant toxicity. RESULTS: NFV-induced upregulation of SREBP-1 and ATF6 results from inhibition of S2P, which together with S1P mediates regulated intramembrane proteolysis from their precursor to their transcriptionally active forms. The resulting endoplasmic reticulum (ER) stress and concurrent inhibition of the unfolded protein response induce caspase-mediated apoptosis. CONCLUSIONS: These results provide new insight into the mechanism of NFV-mediated induction of ER stress and cell death in liposarcomas and are the first to report targeting S2P for cancer therapy.


Subject(s)
Activating Transcription Factor 6/metabolism , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Liposarcoma/pathology , Metalloendopeptidases/antagonists & inhibitors , Nelfinavir/pharmacology , Sterol Regulatory Element Binding Protein 1/metabolism , Animals , Antineoplastic Agents/pharmacokinetics , Caspase 6/metabolism , Caspase 9/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Coumarins/pharmacology , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/physiology , Enzyme Activation , Fatty Acid Synthase, Type I/genetics , Fatty Acid Synthase, Type I/metabolism , Female , Humans , Isocoumarins , Liposarcoma/drug therapy , Metalloendopeptidases/genetics , Mice , Mice, SCID , Nelfinavir/analogs & derivatives , Nelfinavir/pharmacokinetics , Neoplasm Transplantation , Phenanthrolines/pharmacology , Proprotein Convertases/genetics , Proprotein Convertases/metabolism , Protease Inhibitors/pharmacology , Protein Transport , RNA Interference , RNA, Small Interfering/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Transcription, Genetic , Transplantation, Heterologous , Tumor Burden/drug effects
7.
J Med Chem ; 53(4): 1451-7, 2010 Feb 25.
Article in English | MEDLINE | ID: mdl-20108932

ABSTRACT

A series of new thienyl ring containing analogues of nelfinavir and saquinavir with different substitution patterns were synthesized from suitable enantiopure diols. Their inhibitory activity against wild type recombinant HIV-1 protease was evaluated. In general thienyl groups spaced from the core by a methylene group gave products showing IC(50) in the nanomolar range, irrespective of the type and the substitution pattern of the heterocycle. The range of activity of the two most active compounds is substantially maintained or even increased against two commonly selected mutants, under drug pressure, such as V32I and V82A.


Subject(s)
Asparagine/analogs & derivatives , HIV Protease Inhibitors/chemical synthesis , HIV Protease/chemistry , Nelfinavir/analogs & derivatives , Nelfinavir/chemical synthesis , Quinolines/chemical synthesis , Saquinavir/analogs & derivatives , Saquinavir/chemical synthesis , Asparagine/chemical synthesis , Asparagine/chemistry , HIV Protease/genetics , HIV Protease Inhibitors/chemistry , Mutation , Nelfinavir/chemistry , Quinolines/chemistry , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/chemistry , Saquinavir/chemistry , Stereoisomerism , Structure-Activity Relationship
8.
Br J Clin Pharmacol ; 68(5): 682-9, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19916992

ABSTRACT

AIMS: This study reports the pharmacokinetics of nelfinavir, its active metabolite, M8, and active moiety (nelfinavir + M8) in volunteers genotyped for CYP2C19 as extensive metabolizer (*1*1; n = 38), heterozygous poor metabolizer (PM) (*1*2; n = 22) and homozygous PM (*2*2; n = 6). METHODS: Subjects received nelfinavir at normal dose (3.5 days of 1250 mg q12h) or high dose (1250 mg q12h for 3 days and single dose of 3125 mg on day 4). Steady-state plasma samples were analysed by high-performance liquid chromatography/ultraviolet assay to determine pharmacokinetics. RESULTS: At steady state, the mean C(max) was 42% [95% confidence interval (CI) 19, 69] and 63% (95% CI 20, 122) higher, and mean AUC was 51% (95% CI 24, 83) and 85% (95% CI 32, 159) higher for *1*2 and *2*2 compared with *1*1 subjects, respectively. For M8, the mean C(max) and AUC were 35% (95% CI 6, 55) and 33% (95% CI -3, 56), respectively, lower for *1*2 compared with *1*1 subjects. M8 was not detectable in *2*2 subjects. The mean C(max) and AUC values for the active moiety were higher by 30-35% for the *1*2 and *2*2 compared with *1*1 subjects. CONCLUSIONS: Mutation in CYP2C19 increased the systemic exposure of nelfinavir and reduced the exposure of M8. No significant differences were noted among the heterozygous (*1*2) and homozygous (*2*2) PMs. These changes are not considered to be clinically relevant and hence the use of nelfinavir does not require prior assessment of CYP2C19 genotype.


Subject(s)
Aryl Hydrocarbon Hydroxylases/pharmacokinetics , HIV Protease Inhibitors/pharmacokinetics , Nelfinavir/analogs & derivatives , Nelfinavir/pharmacokinetics , Adult , Aryl Hydrocarbon Hydroxylases/genetics , Cytochrome P-450 CYP2C19 , Female , Humans , Male , Metabolic Clearance Rate/genetics , Middle Aged , Polymorphism, Genetic , Young Adult
9.
Curr HIV Res ; 7(3): 293-301, 2009 May.
Article in English | MEDLINE | ID: mdl-19442125

ABSTRACT

Virakinetics II was designed as an observational, multicenter cohort study conducted in HIV-positive patients treated with NFV-based combinations. Trough (pre-dose) concentrations of NFV+M8 in plasma were determined using a novel ELISA test (NFV TDM-ELISA) and analyzed using clinical and laboratory parameters. Drug levels were sorted as below, within or above a given interval (<0.8 microg/mL, 0.8-3.5 microg/mL and >3.5 microg/mL, respectively). Longitudinal analysis was performed in a subset of patients who underwent two or more determinations. Ninety patients on NFV-containing HAART were enrolled and 43 were coinfected with HCV and/or HBV. Among coinfected patients, 10 subjects had a clinical or histological diagnosis of cirrhosis. Compared to the HIV-monoinfected, the coinfected patients were significantly older, more treatment-experienced, with higher frequency of lipodystrophy and altered liver function test values (all p values: <0.05). Coinfected patients were also more likely to be on a reduced dose of NFV than monoinfected (p=0.03). No significant difference was observed between the two groups with regard to NFV+M8 trough values and concentration range distribution. Median NFV+M8 C(trough) concentrations were higher in coinfected patients, but without reaching statistical significance (p=0.2). This new ELISA test proved to be a rapid, convenient and reliable tool for assessing NFV+M8 plasma levels in HIV-positive patients. It could be suitable for use within the framework of routine clinical practice even in peripheral centers without specialized laboratories.


Subject(s)
Anti-HIV Agents/pharmacokinetics , Anti-HIV Agents/therapeutic use , HIV Infections/drug therapy , Hepatitis B, Chronic/complications , Hepatitis C, Chronic/complications , Nelfinavir/analogs & derivatives , Adult , Antiretroviral Therapy, Highly Active , Enzyme-Linked Immunosorbent Assay/methods , Female , HIV Infections/complications , Humans , Male , Middle Aged , Nelfinavir/pharmacokinetics , Nelfinavir/therapeutic use , Plasma/chemistry
10.
Br J Clin Pharmacol ; 65(4): 548-57, 2008 Apr.
Article in English | MEDLINE | ID: mdl-17922881

ABSTRACT

WHAT IS ALREADY KNOWN ABOUT THIS SUBJECT: * Nelfinavir is an HIV protease inhibitor, substrate of the transporter P-glycoprotein and metabolized via CYP2C19, CYP3A4 and CYP3A5 enzymes. * Pharmacokinetic studies have shown wide interindividual variability of nelfinavir concentrations, some of this variability perhaps caused by variant drug metabolism or transporter genes. * For CYP3A4*1B and CYP3A5*3 polymorphism, results from three studies are in agreement, showing no difference in nelfinavir concentrations between patients with these different genotypes. * However, for MDR1 and CYP2C19 polymorphism, there have been contradictory studies, showing either no impact on nelfinavir concentration or modified concentrations which could influence virological response. WHAT THIS STUDY ADDS: * Patients with an *1/*2 or *2/*2 genotype for CYP2C19 had a nelfinavir to M8 biotransformation divided by 2 compared with *1/*1 patients. * No evidence of any influence of MDR1 polymorphism on nelfinavir absorption could be detected. AIMS: To evaluate the effect of CYP2C19 polymorphism on nelfinavir and M8 pharmacokinetic variability in human immunodeficiency virus-infected patients and to study the link between pharmacokinetic exposure and short-term efficacy and toxicity. METHODS: Nelfinavir (n = 120) and M8 (n = 119) concentrations were measured in 34 protease inhibitor-naive patients. Two weeks after initiating the treatment, blood samples were taken before, 1, 3 and 6 h after drug administration. Genotyping for CYP3A4, 3A5, 2C19 and MDR1 was performed. A population pharmacokinetic model was developed to describe nelfinavir-M8 concentration time-courses and to estimate interpatient variability. The influence of individual characteristics and genotypes were tested using a likelihood ratio test. Estimated mean (C(mean)), maximal (C(max)) and trough (C(trough)) nelfinavir and M8 concentrations were correlated to short-term virological efficacy and tolerance using Spearman nonparametric correlation tests. RESULTS: A one-compartment model with first-order absorption, elimination and metabolism to M8 best described nelfinavir data. M8 was modelled by an additional compartment. Mean pharmacokinetic estimates and the corresponding intersubject variabilities were: absorption rate 0.17 h(-1) (99%), absorption lag time 0.82 h, apparent nelfinavir total clearance 52 l h(-1) (49%), apparent nelfinavir volume of distribution 191 l, M8 elimination rate constant 1.76 h(-1) and nelfinavir to M8 0.39 h(-1) (59%) in *1/*1 patients and 0.20 h(-1) in *1/*2 or *2/*2 patients for CYP2C19*2. Nelfinavir C(mean) was positively correlated to glycaemia and triglyceride increases (P = 0.02 and P = 0.04, respectively). CONCLUSIONS: The rate of metabolism of nelfinavir to M8 was reduced by 50% in patients with *1/*2 or *2/*2 genotype for CYP2C19 compared with those with *1/*1 genotype.


Subject(s)
Aryl Hydrocarbon Hydroxylases/genetics , HIV Infections/drug therapy , HIV Protease Inhibitors/pharmacokinetics , Mixed Function Oxygenases/genetics , Nelfinavir/analogs & derivatives , Polymorphism, Genetic , Adult , Aryl Hydrocarbon Hydroxylases/drug effects , Biotransformation/drug effects , Body Weight/drug effects , Body Weight/physiology , Cytochrome P-450 CYP2C19 , Cytochrome P-450 CYP3A/drug effects , Dose-Response Relationship, Drug , Female , Genotype , HIV Infections/blood , HIV Protease Inhibitors/blood , Humans , Male , Middle Aged , Mixed Function Oxygenases/drug effects , Nelfinavir/metabolism , Nelfinavir/pharmacokinetics , Treatment Outcome , Viral Load/statistics & numerical data
11.
New Microbiol ; 30(3): 318-20, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17802918

ABSTRACT

Therapeutic drug monitoring (TDM) is an important tool in the management of antiretroviral (ARV) therapy. The gold standard for measuring drugs plasma levels is High-Performance Liquid Chromatographic Assay (HPLC) however it is technically-demanding and time-consuming. We evaluated a new immunoenzymatic test (TDM-ELISA, Biostrands, Trieste, Italy) for nelfinavir and its active metabolite M8 in comparison with HPLC. A statistically significant difference in Ctrough between the two different tests was demonstrated but this difference was no longer significant when a value of 29% due to M8 aliquot was deleted. This faster TDM-ELISA may have an important role for TDM in HIV patients taking ARVs.


Subject(s)
Enzyme-Linked Immunosorbent Assay , HIV Infections/diagnosis , HIV Protease Inhibitors/blood , Nelfinavir/blood , Reagent Kits, Diagnostic , Anti-Retroviral Agents/therapeutic use , Antiretroviral Therapy, Highly Active , Drug Monitoring , Female , HIV Infections/blood , HIV Infections/drug therapy , HIV Protease Inhibitors/pharmacokinetics , Humans , Male , Nelfinavir/analogs & derivatives , Nelfinavir/pharmacokinetics , Sensitivity and Specificity
12.
Pharmacotherapy ; 27(6): 793-800, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17542762

ABSTRACT

STUDY OBJECTIVE: To characterize the bidirectional interaction between twice-daily nelfinavir and twice-weekly rifabutin and isoniazid in patients with tuberculosis and human immunodeficiency virus (HIV) infection. DESIGN: Prospective cohort study. SETTING: Three clinical research centers. PATIENTS: Seven patients with HIV-related tuberculosis. INTERVENTION: Rifabutin 300 mg and isoniazid 15 mg/kg (maximum dose 900 mg) twice/week were administered for at least 2 weeks during the continuation phase of tuberculosis treatment. Antiretroviral therapy with nelfinavir 1250 mg twice/day and two nucleoside reverse transcriptase inhibitors was then added. MEASUREMENTS AND MAIN RESULTS: Patients underwent blood sampling for pharmacokinetic analysis during the continuation phase of tuberculosis therapy and after a median of 21 days after the addition of antiretroviral treatment. When rifabutin was coadministered with nelfinavir, its area under the concentration-time curve from 0-21 hours (AUC(0-21)) increased 22% (geometric mean 5.01 microg.hr/ml [90% confidence interval (CI) 3.25-7.71] with nelfinavir vs 4.10 microg.hr/ml [90% CI 3.18-5.27] without nelfinavir; geometric mean ratio 1.22 [90% CI 0.78-1.92]). Also, the AUC(0-21) for the active metabolite, desacetylrifabutin, increased significantly (geometric mean ratio 3.46, 90% CI 1.84-6.47, p=0.009). In the presence of rifabutin, the pharmacokinetic parameters of nelfinavir and its principal metabolite M8 were similar to those of patients not taking rifabutin. No drug interaction between nelfinavir and isoniazid was detected. CONCLUSIONS: Coadministration of rifabutin and isoniazid without dosage adjustment during twice-weekly tuberculosis therapy with nelfinavir-based antiretroviral therapy resulted in rifabutin exposures within the acceptable ranges for safety and efficacy. Therefore, this combination is an appropriate option for the simultaneous treatment of tuberculosis and HIV infection when tuberculosis therapy is given twice weekly.


Subject(s)
Antibiotics, Antitubercular/pharmacokinetics , HIV Infections/drug therapy , HIV Protease Inhibitors/pharmacokinetics , Nelfinavir/pharmacokinetics , Rifabutin/pharmacokinetics , Tuberculosis/drug therapy , Antibiotics, Antitubercular/adverse effects , Antibiotics, Antitubercular/therapeutic use , Antitubercular Agents/therapeutic use , Area Under Curve , Cohort Studies , Drug Interactions , Drug Therapy, Combination , Female , HIV Infections/complications , HIV Protease Inhibitors/adverse effects , HIV Protease Inhibitors/therapeutic use , Humans , Isoniazid/therapeutic use , Male , Nelfinavir/adverse effects , Nelfinavir/analogs & derivatives , Nelfinavir/blood , Nelfinavir/therapeutic use , Prospective Studies , Rifabutin/adverse effects , Rifabutin/analogs & derivatives , Rifabutin/blood , Rifabutin/therapeutic use , Tuberculosis/complications
13.
J Antimicrob Chemother ; 59(3): 560-4, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17255144

ABSTRACT

OBJECTIVES: This study evaluated the steady-state pharmacokinetic interaction between ritonavir-boosted saquinavir and nelfinavir. METHODS: Open label, multiple-dose, two parallel-groups, single crossover study conducted in 24 HIV-infected patients (12 in each group). Patients in the nelfinavir group added saquinavir/ritonavir, 1000/100 mg twice daily to their ongoing stable treatment regimen consisting of nelfinavir, 1250 mg twice daily and two nucleoside reverse transcriptase inhibitors (NRTIs). Patients in the saquinavir group added nelfinavir, 1250 mg twice daily to their ongoing stable treatment regimen consisting of saquinavir/ritonavir, 1000/100 mg twice daily and two NRTIs. Pharmacokinetic assessments were performed before and 7 days after the start of combined treatment with nelfinavir/saquinavir/ritonavir. Blood samples were collected before and 1, 2, 3, 4, 6, 8, 10 and 12 h after dosing for measurement of nelfinavir, the nelfinavir metabolite M8 and saquinavir using liquid chromatography tandem mass spectrometry (LC-MS/MS). RESULTS: The addition of saquinavir/ritonavir to the nelfinavir-containing regimen resulted in significant increases in the M8 pharmacokinetic parameters AUC(0-12), Cmax and C12; geometric mean ratios (90% confidence intervals) of 2.25 ng.h/mL (1.47-3.44), 1.74 ng/mL (1.25-2.40) and 4.21 ng/mL (2.10-8.47), respectively. The intra-individual changes in nelfinavir and saquinavir concentrations were highly variable. Statistical analysis could not discard a relevant interaction but includes the possibility that some parameters may be halved, others more than doubled. At the same time the analysis failed to show any directed change. CONCLUSIONS: The co-administration of nelfinavir and saquinavir/ritonavir leads to unpredictable changes in concentrations of both drugs. It is unclear whether the increased concentrations of M8 are associated with a clinical benefit.


Subject(s)
Anti-HIV Agents/pharmacokinetics , HIV Infections/drug therapy , Nelfinavir/analogs & derivatives , Nelfinavir/pharmacokinetics , Saquinavir/pharmacokinetics , Adult , Cross-Over Studies , Drug Administration Schedule , Drug Interactions , Drug Therapy, Combination , Humans , Male , Middle Aged , Nelfinavir/administration & dosage , Ritonavir/administration & dosage , Saquinavir/administration & dosage
14.
Article in English | MEDLINE | ID: mdl-16997640

ABSTRACT

A simple, accurate and fast method was developed for determination of the commonly used HIV protease inhibitors (PIs) amprenavir, indinavir, atazanavir, ritonavir, lopinavir, nelfinavir, M8-nelfinavir metabolite and saquinavir in human plasma. Liquid-liquid extraction was used with hexane/ethylacetate from buffered plasma samples with a borate buffer pH 9.0. Isocratic chromatographic separation of all components was performed on an Allsphere hexyl HPLC column with combined UV and fluorescence detection. Calibration curves were constructed in the range of 0.025-10 mg/l. Accuracy and precision of the standards were all below 15% and the lowest limit of quantitation was 0.025 mg/l. Stability of quality control samples at different temperature conditions was found to be below 20% of nominal values. The advantages of this method are: (1) inclusion and determination of the newly approved atazanavir, (2) simultaneous isocratic HPLC separation of all compounds and (3) increased specificity and sensitivity for amprenavir by using fluorescence detection. This method can be used for therapeutic drug monitoring of all PIs currently commercialised and is now part of current clinical practice.


Subject(s)
Chromatography, High Pressure Liquid/methods , HIV Protease Inhibitors/blood , Atazanavir Sulfate , Calibration , Carbamates/blood , Drug Monitoring , Drug Stability , Fluorescence , Furans , Humans , Indinavir/blood , Lopinavir , Nelfinavir/analogs & derivatives , Nelfinavir/blood , Oligopeptides/blood , Pyridines/blood , Pyrimidinones/blood , Reproducibility of Results , Ritonavir/blood , Saquinavir/blood , Sensitivity and Specificity , Sulfonamides/blood , Ultraviolet Rays
15.
Antimicrob Agents Chemother ; 50(6): 2079-86, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16723569

ABSTRACT

A relationship between nelfinavir antiretroviral efficacy and plasma concentrations has been previously established. As physiological changes associated with pregnancy have a large impact on the pharmacokinetics of many drugs, a nelfinavir population study with women was developed, and the large intersubject variability was analyzed in order to optimize individual treatment schedules for this drug during pregnancy. A population pharmacokinetic model was developed in order to describe the concentration time course of nelfinavir and its metabolite M8 in pregnant and nonpregnant women. Individual characteristics, such as age, body weight, and weeks of gestation or delivery, which may influence nelfinavir-M8 pharmacokinetics were investigated. Data from therapeutic drug monitoring in 133 women treated with nelfinavir were retrospectively analyzed with NONMEM. Nelfinavir pharmacokinetics was described by a one-compartment model with linear absorption and elimination and M8 produced from the nelfinavir central compartment. Mean pharmacokinetic estimates and the corresponding intersubject percent variabilities for a nonpregnant woman were the following: absorption rate, 0.83 h(-1); absorption lag time, 0.85 h; apparent nelfinavir elimination clearance (CL(10)/F), 35.5 liters/h (50%); apparent volume of distribution (V/F), 596 liters (118%); apparent formation clearance to M8 (CL(1M)/F), 0.65 liters/h (69%); and M8 elimination rate constant (k(M0)), 3.3 h(-1) (59%). During pregnancy, we observed significant increases in nelfinavir (44.4 liters/h) and M8 (5 h(-1)) elimination but unchanged nelfinavir transformation clearance to M8, suggesting an induction of CYP3A4 but no effect on CYP2C19. Apparent nelfinavir clearance and volume showed a twofold increase on the day of delivery, suggesting a decrease in bioavailability on this day. The M8 elimination rate was increased by concomitant administration of nonnucleoside reverse transcriptase inhibitors. A trough nelfinavir plasma concentration above 1 mg/liter was previously shown to improve the antiretroviral response. The Bayesian individual pharmacokinetic estimates suggested that the dosage should not be changed in pregnant women but may be doubled on the day of delivery.


Subject(s)
HIV Protease Inhibitors/administration & dosage , HIV Protease Inhibitors/pharmacokinetics , Nelfinavir/analogs & derivatives , Nelfinavir/administration & dosage , Nelfinavir/pharmacokinetics , Adult , Bayes Theorem , Delivery, Obstetric , Dose-Response Relationship, Drug , Drug Monitoring , Female , HIV Protease Inhibitors/blood , Humans , Models, Statistical , Nelfinavir/blood , Pregnancy , Retrospective Studies
16.
Antimicrob Agents Chemother ; 50(3): 910-6, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16495250

ABSTRACT

As a relationship between nelfinavir antiretroviral efficacy and plasma concentrations has been previously established, nelfinavir pharmacokinetics was investigated in order to optimize the individual treatment schedule in a pediatric population. A population pharmacokinetic model was developed to describe the concentration-time course of nelfinavir and its active metabolite M8. Individual characteristics were used to explain the large interindividual variability in children. Data from therapeutic drug monitoring in 182 children treated with nelfinavir were analyzed with NONMEM. Then Food and Drug Administration (FDA) current recommendations were evaluated estimating the percentage of children who reached the target minimum plasma concentration (0.8 mg/liter) by using Bayesian estimates. Nelfinavir pharmacokinetics was described by a one- compartment model with linear absorption and elimination. Pharmacokinetic estimates and the corresponding intersubject variabilities for the model were as follows: nelfinavir total clearance, 0.93 liters/h/kg (39%); volume of distribution, 6.9 liters/kg (109%); absorption rate, 0.5 h(-1); formation clearance fraction to hydroxy-tert-butylamide (M8), 0.025; M8 elimination rate, 1.88 h(-1) (49%). Apparent nelfinavir total clearance and volume of distribution decreased as a function of age. M8 elimination rate was increased by concomitant administration of nevirapine or efavirenz. Our data confirm that the FDA recommendations for children from 2 to 13 years are optimal and that the dose recommended for children younger than 2 years is adequate for the children from 2 months to 2 years old. However, in children younger than 2 months, the proposed nelfinavir newborn dose of 40 mg/kg of body weight twice daily is inadequate and we suggest increasing the dose to 50 to 60 mg/kg administered thrice daily. This assumption should be further evaluated.


Subject(s)
Aging , HIV Protease Inhibitors/administration & dosage , HIV Protease Inhibitors/pharmacokinetics , Nelfinavir/analogs & derivatives , Nelfinavir/administration & dosage , Nelfinavir/pharmacokinetics , Bayes Theorem , Child , Child, Preschool , Dose-Response Relationship, Drug , Female , HIV Protease Inhibitors/blood , HIV-1 , Humans , Infant , Male , Models, Biological , Models, Statistical , Nelfinavir/blood
17.
HIV Med ; 7(2): 122-8, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16420257

ABSTRACT

OBJECTIVES: To determine the characteristics of the binding of nelfinavir and active M8 to alpha1-acid glycoprotein (AAG) and human serum albumin (HSA), and to examine the displacement effects of drugs binding extensively to AAG (ritonavir and saquinavir) or to HSA (salicylic acid and valproic acid). METHODS: Free drugs were separated by equilibrium dialysis after incubation with human plasma or purified plasma proteins and after co-incubation with potential displacers. Association constants were estimated from double-reciprocal plots of the data. RESULTS: Nelfinavir and M8 free fractions [fractions of unbound drug (fus)] were 0.42+/-0.08% (mean+/-standard deviation) and 0.64+/-0.07%, respectively. For the two analytes, respectively, association constants were 7.25 x 10(7)/m and 3.33 x 10(7)/m for AAG and 1.11 x 10(6)/m and 7.92 x 10(5)/m for HSA. Nelfinavir fu in an AAG solution was significantly (P < 0.01) increased by the addition of ritonavir or saquinavir, whereas it was unaltered by addition of these drugs to whole plasma. Similarly, fu in an HSA solution was significantly increased (P < 0.01) by the addition of salicylic acid or valproic acid, whereas there was no difference in the free fraction in plasma. CONCLUSIONS: The affinity of nelfinavir for human plasma proteins was higher than that of M8, and both nelfinavir and M8 showed higher affinity to AAG than to HSA. The free fraction of nelfinavir was not affected by drugs that bind extensively to AAG or albumin when these drugs were added to whole plasma in combination, suggesting a compensatory effect of alternate binding proteins.


Subject(s)
Blood Proteins/metabolism , HIV Protease Inhibitors/blood , Nelfinavir/blood , Binding, Competitive , Dose-Response Relationship, Drug , Humans , In Vitro Techniques , Nelfinavir/analogs & derivatives , Orosomucoid/metabolism , Protein Binding , Serum Albumin/metabolism
18.
Clin Pharmacokinet ; 44(12): 1267-78, 2005.
Article in English | MEDLINE | ID: mdl-16372824

ABSTRACT

OBJECTIVES: To describe plasma concentrations of indinavir alone or combined with ritonavir, and of nelfinavir and its active metabolite M8, and to measure their variabilities in HIV-infected patients treated with a stable antiretroviral regimen and experiencing a sustained virological response for at least 12 months. PATIENTS AND METHODS: In this prospective trial, blood samples were drawn during a 6-hour time interval between two doses at enrolment to assess protease inhibitor (PI) pharmacokinetic parameters, and 4 months later to assess plasma trough and peak concentrations. Safety and adherence assessments and laboratory data were collected during an 8-month period. PI pharmacokinetic characteristics were analysed using a non-compartmental approach. Inter- and intrapatient variabilities were estimated using a linear mixed-effect model. The impact of different covariates on plasma trough concentrations was investigated. Eighty-eight patients were analysed: 42 treated with indinavir and 46 with nelfinavir. RESULTS: The interquartile range (IQR) of the plasma trough concentration corrected for the sampling time (Ccalc) was 116-374 microg/L for indinavir alone and 163-508 microg/L for indinavir/ritonavir. Ritonavir significantly increased indinavir elimination half-life and plasma exposure. For nelfinavir, the IQR of Ccalc was 896-2059 microg/L for three-times-daily administration and 998-2124 microg/L for twice-daily administration. Variabilities were high for both PIs. Intrapatient variability for indinavir alone (and indinavir + ritonavir) was 76% (107%) and interpatient variability was 58% (10%) in adherent patients. Intrapatient variability for nelfinavir three times daily (and twice daily) was 41% (74%) and interpatient variability was 62% (50%). Intrapatient variability was lowered in patients with a high adherence level. CONCLUSION: Although performed in a homogeneous population, this study documented a high interpatient but also intrapatient variability of indinavir and nelfinavir pharmacokinetics, which should be taken into account when interpreting therapeutic drug monitoring. Once patients have reached a sustained virological response, plasma PI monitoring may have a limited impact.


Subject(s)
HIV Infections/metabolism , HIV Protease Inhibitors/pharmacokinetics , Indinavir/pharmacokinetics , Nelfinavir/pharmacokinetics , Ritonavir/pharmacokinetics , Adult , Aged , Antiretroviral Therapy, Highly Active , Female , HIV Infections/drug therapy , HIV Infections/virology , HIV Protease Inhibitors/blood , HIV Protease Inhibitors/therapeutic use , Humans , Indinavir/blood , Indinavir/therapeutic use , Male , Middle Aged , Multicenter Studies as Topic , Nelfinavir/analogs & derivatives , Nelfinavir/blood , Nelfinavir/therapeutic use , Patient Compliance , Prospective Studies , Ritonavir/blood , Ritonavir/therapeutic use , Viral Load
19.
Br J Clin Pharmacol ; 60(4): 390-403, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16187971

ABSTRACT

AIMS: To describe the pharmacokinetics of nelfinavir and its main metabolite M8 in HIV-infected patients with a sustained virological response, to characterize the effect of covariates and to estimate inter- and intra-individual variability in the pharmacokinetics. METHODS: Three hundred and twenty concentrations of both nelfinavir and M8 were measured in 46 patients enrolled in the COPHAR 1-ANRS 102 study. Blood samples were taken at a first visit (one sample before drug administration and four samples at fixed times after) and at a second visit 1 to 3 months later (one before and one 3 h after drug administration). The data from both visits on nelfinavir and M8 were modelled jointly in all patients using a population approach. RESULTS: A one-compartment model with first-order absorption and elimination best described nelfinavir data, with an additional compartment incorporating a first order rate-constant describing the metabolism of the drug to M8. For nelfinavir, the apparent volume of distribution (V/F ) (95% confidence interval for the mean), was 309 l (185, 516), the absorption rate constant (k(a)) was 0.4 h(-1) (0.2, 0.8), and the apparent clearance (CL/F ) was 37.3 l h(-1) (32, 44). For M8, V(m) /(Fk(m)) and CL(m)/(Fk(m)) were 866 l h(-1) (351, 2161) and 1670 l (965, 2894), respectively. The interindividual variabilities were 34.9%, 34.3% and 62.2% for V/F, CL/F and CL(m)/(Fk(m)), respectively. The interoccasion variability was 27.8% for CL/F. The mean half-lives were 05.38 h and 00.44 h for nelfinavir and M8, respectively. Significant but opposite effects of comedication with zidovudine were found on nelfinavir CL/F and M8 CL(m)/(Fk(m)), but they were not considered to be clinically relevant. CONCLUSIONS: A joint model was found to describe adequately nelfinavir and M8 concentrations and was used to estimate pharmacokinetic parameters for M8. The model can be used to build reference pharmacokinetic profiles for therapeutic drug monitoring of the drug.


Subject(s)
Anti-HIV Agents/pharmacokinetics , HIV Infections/drug therapy , HIV Protease Inhibitors/pharmacokinetics , Nelfinavir/analogs & derivatives , Nelfinavir/pharmacokinetics , Adult , Aged , Anti-HIV Agents/blood , Antiretroviral Therapy, Highly Active , Female , HIV Infections/blood , HIV Protease Inhibitors/blood , Humans , Male , Middle Aged , Models, Chemical , Nelfinavir/blood , Prospective Studies
20.
Antivir Ther ; 10(2): 349-55, 2005.
Article in English | MEDLINE | ID: mdl-15865230

ABSTRACT

BACKGROUND: The nucleotide analogue, tenofovir, has been shown to lower plasma atazanavir levels in pharmacokinetic trials, an interaction that may be partly reversed by the addition of ritonavir, whereas plasma tenofovir levels are themselves raised when the drug is combined with lopinavir/ritonavir. OBJECTIVE: To investigate the effect of tenofovir coadministration on the steady-state pharmacokinetics of nelfinavir in HIV-infected patients. METHODS: Eighteen patients received nelfinavir 1250 mg twice daily plus prescribed nucleoside reverse transcriptase inhibitors for at least 14 days, with pharmacokinetic measurements performed on day 15. Treatment with nelfinavir was continued for another 7 days with the addition of 300 mg tenofovir once daily. Pharmacokinetic measurements were repeated on day 22. Plasma samples were analysed by liquid chromatography-tandem mass spectrometry for nelfinavir, its primary metabolite, M8, and tenofovir. The parameters AUC0-12, C0, Cmax and Tmax were compared for nelfinavir with and without tenofovir by calculating geometric mean ratios (GMRs) of the pharmacokinetic parameters with associated 95% confidence intervals (95% CIs). Safety was assessed throughout the study. RESULTS: The addition of tenofovir to the nelfinavir-based regimen had no effect on the pharmacokinetics of nelfinavir. The GMR of the nelfinavir AUC0-12 values was 0.97 (95% CI: 0.80-1.17). There was a slight decrease in M8 metabolite (AUC0-12 ratio, 0.87; 95% CI: 0.68-1.11) but this was not significant. No serious adverse events occurred through the study period. CONCLUSION: Nelfinavir does not require dose adjustment when coadministered with tenofovir and appears to be well-tolerated by HIV-infected patients.


Subject(s)
Adenine/analogs & derivatives , Adenine/pharmacokinetics , HIV Infections/metabolism , HIV Protease Inhibitors/pharmacology , Nelfinavir/analogs & derivatives , Nelfinavir/pharmacokinetics , Organophosphonates/pharmacokinetics , Reverse Transcriptase Inhibitors/pharmacokinetics , Adenine/therapeutic use , Adult , Aged , Drug Therapy, Combination , Female , HIV Infections/drug therapy , HIV Protease Inhibitors/therapeutic use , Humans , Male , Middle Aged , Nelfinavir/blood , Nelfinavir/therapeutic use , Organophosphonates/therapeutic use , Reverse Transcriptase Inhibitors/therapeutic use , Tenofovir , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...