Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 192
Filter
1.
Colloids Surf B Biointerfaces ; 176: 288-299, 2019 Apr 01.
Article in English | MEDLINE | ID: mdl-30634155

ABSTRACT

The current endeavor was aimed to fabricate spray dried Nelfinavir Mesylate particles (SDNPs) that upon reconstitution with body fluids releases amorphous Nelfinavir Mesylate with enhanced oral bioavailability. For this purpose, feed mixture was prepared containing solubilized NFM, solid substrate and solvent system. The NFM was solubilized in a mixture of Maisine 35-1 (200 mg), Tween80 (500 mg) and Transcutol HP (300 mg). Three different solid substrates with high specific surface area were used: Neusilin® UFL2, (magnesium aluminometasilicate), Aerosil 200® (colloidal silica) and Syloid 244 FP® (porous silicon dioxide). Central composite design-response surface methodology (CCD-RSM) with three-factor (two numeric and one categorical) at three-level was used to select the appropriate solid substrate and to develop optimized SDNPs. The solid characterization by scanning electron microscopy, differential scanning calorimetry and powder X-ray diffraction studies indicated the absence of crystalline NFM in the formulations. The drug distribution analysis using raman spectroscopy suggested uniform distribution of amorphous NFM in optimized SDNPs-1 formulation. The transmission electron microscopy revealed the spherical structure of reconstituted SDNPs that releases amorphous NFM with globules size less than 110 nm. The solid substrate had significant and positive effect in drug dissolution; the mean dissolution time of NFM loaded in SDNPs was considerably improved. The bioavailability study resulted in enhanced magnitude of Cmax and AUC for SDNPs. The tissue distribution studies exhibited significantly higher brain and lymph nodes distribution as compared to putative form. Overall, the results pointed towards the overwhelming response of the SDNPs to be used for HIV treatment.


Subject(s)
Chemistry, Pharmaceutical/methods , Desiccation , Nelfinavir/administration & dosage , Administration, Oral , Animals , Biological Availability , Calorimetry, Differential Scanning , Drug Liberation , Excipients/chemistry , Male , Nelfinavir/pharmacokinetics , Numerical Analysis, Computer-Assisted , Particle Size , Rats, Wistar , Risk Assessment , Spectrum Analysis, Raman , Tissue Distribution
2.
J Pharm Sci ; 108(1): 584-591, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30423339

ABSTRACT

The aim of the current research was to develop an in silico oral absorption model coupled with an in vitro dissolution/precipitation testing to predict gastric pH-dependent drug-drug interactions for weakly basic drugs. The effects of elevated gastric pH on the plasma profiles of dipyridamole, prasugrel, and nelfinavir were simulated and compared with pharmacokinetic data reported in humans with or without use of proton pump inhibitors or histamine H2 receptor antagonists. The in vitro dissolution and precipitation data for the weakly basic drugs in biorelevant media were obtained using paddle apparatus. An in silico prediction model based on the STELLA software was designed and simulations were conducted to predict the oral pharmacokinetic profiles of the 3 drugs under both usual (low) and elevated gastric pH conditions. The changes in oral absorption of dipyridamole and prasugrel in subjects with elevated gastric pH compared with those with low stomach pH were predicted well using the in vitro-in silico-in vivo approach. The proposed approach could become a powerful tool in the formulation development of poorly soluble weak base drugs.


Subject(s)
Dipyridamole/pharmacokinetics , Nelfinavir/pharmacokinetics , Prasugrel Hydrochloride/metabolism , Administration, Oral , Computer Simulation , Gastric Emptying/physiology , Humans , Hydrogen-Ion Concentration , Intestinal Absorption/physiology , Models, Biological , Permeability/drug effects , Solubility , Stomach/physiology
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.
AAPS PharmSciTech ; 19(8): 3584-3598, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30209788

ABSTRACT

Nelfinavir mesylate (NFV), a human immunodeficiency virus (HIV) protease inhibitor, is an integral component of highly active anti retro viral therapy (HAART) for management of AIDS. NFV possesses pH-dependent solubility and has low and variable bioavailability hampering its use in therapeutics. Lipid-based particulates have shown to improve solubility of poorly water soluble drugs and oral absorption, thereby aiding in improved bioavailability. The current study compares potential of vesicular and solid lipid nanocarriers of NFV with drug nanocrystallites and microvesicular systems like cochleates in improving bioavailability of NFV. The paper outlines investigation of systems using in vitro models like in vitro lipolysis, in vitro release, and permeation through cell lines to predict the in vivo potential of nanocarriers. Finally, in vivo pharmacokinetic study is reported which provided proof of concept in sync with results from in vitro studies. Graphical Abstract ᅟ.


Subject(s)
HIV Protease Inhibitors/chemistry , Lipids/chemistry , Nelfinavir/chemistry , Animals , Biological Availability , Caco-2 Cells , Female , Humans , Nelfinavir/pharmacokinetics , Rats , Rats, Sprague-Dawley , Solubility
5.
J Neurovirol ; 23(4): 603-614, 2017 08.
Article in English | MEDLINE | ID: mdl-28762183

ABSTRACT

Drug abuse (e.g., methamphetamine-Meth or cocaine-Coc) is one of the major risk factors for becoming infected with HIV-1, and studies show that in combination, drug abuse and HIV-1 lead to significantly greater damage to CNS. To overcome these issues, we have developed a novel nanoformulation (NF) for drug-abusing population infected with HIV-1. In this work, a novel approach was developed for the co-encapsulation of Nelfinavir (Nel) and Rimcazole (Rico) using layer-by-layer (LbL) assembled magnetic nanoformulation for the cure of neuroAIDS. Developed NF was evaluated for blood-brain barrier (BBB) transmigration, cell uptake, cytotoxicity and efficacy (p24 assay) in HIV-1 infected primary astrocyte (HA) in presence or absence of Coc and Meth. Developed magnetic nanoformulation (NF) fabricated using the LbL approach exhibited higher amounts of drug loading (Nel and Rico) with 100% release of both the therapeutic agents in a sustained manner for 8 days. NF efficacy studies indicated a dose-dependent decrease in p24 levels in HIV-1-infected HA (~55%) compared to Coc + Meth treated (~50%). The results showed that Rico significantly subdued the effect of drugs of abuse on HIV infectivity. NF successfully transmigrated (38.8 ± 6.5%) across in vitro BBB model on the application of an external magnetic field and showed >90% of cell viability with efficient cell uptake. In conclusion, our proof of concept study revealed that sustained and concurrent release of sigma σ1 antagonist and anti-HIV drug from the developed novel sustained release NF can overcome the exacerbated effects of drugs of abuse in HIV infection and may solve the issue of medication adherence in the drug-abusing HIV-1 infected population.


Subject(s)
Carbazoles/pharmacokinetics , Cocaine/pharmacokinetics , Delayed-Action Preparations/pharmacokinetics , Illicit Drugs/pharmacokinetics , Methamphetamine/pharmacokinetics , Nelfinavir/pharmacokinetics , AIDS Dementia Complex/drug therapy , Astrocytes/cytology , Astrocytes/drug effects , Astrocytes/metabolism , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Cocaine/chemistry , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , HIV Protease Inhibitors/pharmacokinetics , HIV-1/drug effects , HIV-1/growth & development , Humans , Illicit Drugs/chemistry , Magnets/chemistry , Methamphetamine/chemistry , Nanostructures/chemistry , Neuroprotective Agents/pharmacokinetics , Primary Cell Culture , Substance Abuse, Intravenous/prevention & control
6.
Clin Pharmacol Ther ; 101(4): 501-509, 2017 04.
Article in English | MEDLINE | ID: mdl-28074467

ABSTRACT

Elevations in serum bilirubin during drug treatment may indicate global liver dysfunction and a high risk of liver failure. However, drugs also can increase serum bilirubin in the absence of hepatic injury by inhibiting specific enzymes/transporters. We constructed a mechanistic model of bilirubin disposition based on known functional polymorphisms in bilirubin metabolism/transport. Using physiologically based pharmacokinetic (PBPK) model-predicted drug exposure and enzyme/transporter inhibition constants determined in vitro, our model correctly predicted indinavir-mediated hyperbilirubinemia in humans and rats. Nelfinavir was predicted not to cause hyperbilirubinemia, consistent with clinical observations. We next examined a new drug candidate that caused both elevations in serum bilirubin and biochemical evidence of liver injury in rats. Simulations suggest that bilirubin elevation primarily resulted from inhibition of transporters rather than global liver dysfunction. We conclude that mechanistic modeling of bilirubin can help elucidate underlying mechanisms of drug-induced hyperbilirubinemia, and thereby distinguish benign from clinically important elevations in serum bilirubin.


Subject(s)
Carrier Proteins/antagonists & inhibitors , Chemical and Drug Induced Liver Injury/diagnosis , Enzyme Inhibitors/adverse effects , Hyperbilirubinemia/chemically induced , Hyperbilirubinemia/enzymology , Liver/pathology , Animals , Bilirubin/blood , Bilirubin/metabolism , Chemical and Drug Induced Liver Injury/pathology , Computer Simulation , HIV Protease Inhibitors/pharmacokinetics , HIV Protease Inhibitors/toxicity , Humans , Hyperbilirubinemia/pathology , Indinavir/pharmacokinetics , Indinavir/toxicity , Mice , Mice, Knockout , Models, Biological , Nelfinavir/pharmacokinetics , Nelfinavir/toxicity , Pharmacokinetics , Rats , Rats, Gunn , Receptors, Chemokine/antagonists & inhibitors , Systems Biology
7.
Int J Pharm ; 501(1-2): 311-25, 2016 Mar 30.
Article in English | MEDLINE | ID: mdl-26854426

ABSTRACT

Poor aqueous solubility and moderate permeability of Nelfinavir mesylate (NFM) leads to high variability in absorption after oral administration. To improve the solubility and bioavailability of NFM, the self microemulsifying drug delivery system (SMEDDS) was developed. For this purpose, Quality by design (QbD) approach employing D-optimal mixture design was used to prepare SMEDDS of NFM. Further, the software generated numerically optimized SMEDDS were developed by utilizing desirability function. Maisine 35-1, Tween 80, and Transcutol HP were identified as oil, surfactant, and co-surfactant that had best solubility for NFM. Ternary phase diagrams were plotted to identify the self-emulsification region. Dissolution of putative NFM in simulated fasted or fed small intestinal conditions, respectively, predicted that there is a positive food effect. However, NFM loaded SMEDDS showed absence of food effect with no significant difference in dissolution performance either in Fasted or fed state simulated intestinal fluid (FaSSIF or FeSSIF) biorelevent dissolution media. The prepared SMEDDS were thermodynamically stable with droplet size (121 nm), poly dispersity index (PDI) (0.198) and emulsification time (<1 min). Transmission electron microscopy (TEM) analysis confirmed the spherical shape of the reconstituted SMEDDS droplets. The ex vivo performance revealed 4.57 fold enhancement in the apparent permeability of NFM as compared to NFM suspension. The animal pharmacokinetic analysis in New Zealand strain rabbits indicated food effect on pure NFM suspension. However, absence of food effect and 3.5-3.6 fold enhancement in the oral bioavailability was observed when NFM was formulated into SMEDDS. Thus, it could be envisaged that development of SMEDDS formulation of NFM could be one of the best alternative to enhance oral bioavailability of NFM.


Subject(s)
Drug Delivery Systems , Nelfinavir/administration & dosage , Animals , Biological Availability , Chemistry, Pharmaceutical , Emulsions , Ethylene Glycols/administration & dosage , Ethylene Glycols/chemistry , Fasting/metabolism , Food-Drug Interactions , Gastrointestinal Tract/metabolism , HIV Protease Inhibitors/administration & dosage , HIV Protease Inhibitors/blood , HIV Protease Inhibitors/chemistry , HIV Protease Inhibitors/pharmacokinetics , Intestinal Absorption/drug effects , Linoleic Acids/administration & dosage , Linoleic Acids/chemistry , Male , Nelfinavir/blood , Nelfinavir/chemistry , Nelfinavir/pharmacokinetics , Permeability/drug effects , Polysorbates/administration & dosage , Polysorbates/chemistry , Rabbits , Rats , Solubility , Surface-Active Agents/administration & dosage , Surface-Active Agents/chemistry
8.
Haematologica ; 101(3): 346-55, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26659919

ABSTRACT

Downregulation of the unfolded protein response mediates proteasome inhibitor resistance in multiple myeloma. The Human Immunodeficieny Virus protease inhibitor nelfinavir activates the unfolded protein response in vitro. We determined dose-limiting toxicity and recommended dose for phase II of nelfinavir in combination with the proteasome inhibitor bortezomib. Twelve patients with advanced hematologic malignancies were treated with nelfinavir (2500-5000 mg/day p.o., days 1-14, 3+3 dose escalation) and bortezomib (1.3 mg/m(2), days 1, 4, 8, 11; 21-day cycles). A run in phase with nelfinavir monotherapy allowed pharmakokinetic/pharmakodynamic assessment of nelfinavir in the presence or absence of concomittant bortezomib. End points included dose-limiting toxicity, activation of the unfolded protein response, proteasome activity, toxicity and response to trial treatment. Nelfinavir 2×2500 mg was the recommended phase II dose identified. Nelfinavir alone significantly up-regulated expression of proteins related to the unfolded protein response in peripheral blood mononuclear cells and inhibited proteasome activity. Of 10 evaluable patients in the dose escalation cohort, 3 achieved a partial response, 4 stable disease for 2 cycles or more, while 3 had progressive disease as best response. In an exploratory extension cohort with 6 relapsed, bortezomib-refractory, lenalidomide-resistant myeloma patients treated at the recommended phase II dose, 3 reached a partial response, 2 a minor response, and one progressive disease. The combination of nelfinavir with bortezomib is safe and shows promising activity in advanced, bortezomib-refractory multiple myeloma. Induction of the unfolded protein response by nelfinavir may overcome the biological features of proteasome inhibitor resistance. (clinicaltrials.gov identifier: 01164709).


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bortezomib/therapeutic use , Leukemia/drug therapy , Lymphoma/drug therapy , Multiple Myeloma/drug therapy , Nelfinavir/therapeutic use , Aged , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Bortezomib/pharmacokinetics , Drug Administration Schedule , Drug Combinations , Drug Resistance, Neoplasm/drug effects , Female , HIV Protease Inhibitors/pharmacokinetics , HIV Protease Inhibitors/therapeutic use , Humans , Leukemia/diagnosis , Leukemia/genetics , Leukemia/pathology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/pathology , Lymphoma/diagnosis , Lymphoma/genetics , Lymphoma/pathology , Male , Middle Aged , Multiple Myeloma/diagnosis , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Nelfinavir/pharmacokinetics , Proteasome Endopeptidase Complex/drug effects , Treatment Outcome , Unfolded Protein Response/drug effects
9.
Br J Clin Pharmacol ; 80(2): 267-75, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25752914

ABSTRACT

AIM: This study evaluated the influence of CYP2C19 polymorphisms on the pharmacokinetics of nelfinavir and its metabolite M8 in patients with pancreatic cancer. METHODS: Nelfinavir was administered orally to patients for over 10 days. The plasma concentrations of nelfinavir and M8 were measured by HPLC. The genotypes of CYP2C19*1, CYP2C19*2 and CYP2C19*3 were determined by the polymerase chain reaction-restriction fragment length polymorphism method. RESULTS: Pharmacokinetic profiles of nelfinavir and M8 were characterized by wide interindividual variability. The mean Cmax of nelfinavir in CYP2C19*1/*1 patients was 3.89 ± 0.40 (n = 3) and 5.12 ± 0.41 (n = 30) µg ml(-1) , while that of CYP2C19*1/*2 patients was 3.60 (n = 1) and 6.14 ± 0.31 (n = 5) µg ml(-1) at the doses of 625 and 1250 mg nelfinavir twice daily, respectively. For the M8 metabolite, the mean Cmax of CYP2C19*1/*1 patients was 1.06 ± 0.06 (n = 3) and 1.58 ± 0.27 (n = 30) µg ml(-1) , while those of CYP2C19*1/*2 patients were 1.01 (n = 1) and 1.23 ± 0.15 (n = 5) µg ml(-1) at the doses of 625 and 1250 mg nelfinavir twice daily, respectively. The area under the plasma concentration-time curve (AUC(0,12 h)) values of nelfinavir for CYP2C19*1/*1 patients were 28.90 ± 1.27 and 38.90 ± 4.99 µg ml(-1) ·h and for CYP2C19*1/*2 patients, AUC(0,12 h) was 28.20 (n = 1) and 40.22 ± 3.17 (n = 5) µg ml(-1) ·h at the doses of 625 and 1250 mg nelfinavir twice daily, respectively. The Cmax of nelfinavir was significantly higher (P <0.05) in CYP2C19*1/*2 patients but there was no statistical difference in AUC(0,12 h). CONCLUSION: CYP2C19*1/*2 genotype modestly affected the pharmacokinetic profiles of nelfinavir and M8 in patients with locally advanced pancreatic cancer.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Cytochrome P-450 CYP2C19/genetics , Nelfinavir/pharmacokinetics , Pancreatic Neoplasms/drug therapy , Polymorphism, Restriction Fragment Length , Adult , Aged , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/blood , Antineoplastic Agents/therapeutic use , Area Under Curve , Dose-Response Relationship, Drug , Female , Humans , Male , Middle Aged , Nelfinavir/administration & dosage , Nelfinavir/blood , Nelfinavir/therapeutic use , Pancreatic Neoplasms/blood , Pancreatic Neoplasms/enzymology
10.
Biochem J ; 465(3): 479-88, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25377919

ABSTRACT

The relative contribution of hepatic compared with intestinal oxidative metabolism is a crucial factor in drug oral bioavailability and therapeutic efficacy. Oxidative metabolism is mediated by the cytochrome P450 mono-oxygenase system to which cytochrome P450 reductase (POR) is the essential electron donor. In order to study the relative importance of these pathways in drug disposition, we have generated a novel mouse line where Cre recombinase is driven off the endogenous Cyp1a1 gene promoter; this line was then crossed on to a floxed POR mouse. A 40 mg/kg dose of the Cyp1a1 inducer 3-methylcholanthrene (3MC) eliminated POR expression in both liver and small intestine, whereas treatment at 4 mg/kg led to a more targeted deletion in the liver. Using this approach, we have studied the pharmacokinetics of three probe drugs--paroxetine, midazolam, nelfinavir--and show that intestinal metabolism is a determinant of oral bioavailability for the two latter compounds. The Endogenous Reductase Locus (ERL) mouse represents a significant advance on previous POR deletion models as it allows direct comparison of hepatic and intestinal effects on drug and xenobiotic clearance using lower doses of a single Cre inducing agent, and in addition minimizes any cytotoxic effects, which may compromise interpretation of the experimental data.


Subject(s)
Integrases/physiology , Intestinal Mucosa/metabolism , Microsomes, Liver/metabolism , Midazolam/metabolism , Nelfinavir/metabolism , Paroxetine/metabolism , Administration, Oral , Animals , Biological Availability , Female , Intestines/drug effects , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microsomes, Liver/drug effects , Midazolam/pharmacokinetics , Nelfinavir/pharmacokinetics , Paroxetine/pharmacokinetics
11.
Oncotarget ; 5(18): 8161-72, 2014 Sep 30.
Article in English | MEDLINE | ID: mdl-25327558

ABSTRACT

Nelfinavir is an HIV protease inhibitor being repurposed as an anti-cancer agent in preclinical models and in small oncology trials, yet the MTD of nelfinavir has not been determined. Therefore, we conducted a Phase Ia study to establish the maximum tolerated dose (MTD) and dose limiting toxicities (DLT) of nelfinavir in subjects with advanced solid tumors. Adults with refractory cancers were given oral nelfinavir twice daily with pharmacokinetic and pharmacodynamic analyses. Twenty-eight subjects were enrolled. Nelfinavir was generally well tolerated. Common adverse events included diarrhea, anemia, and lymphopenia, which were mostly mild. The DLT was rapid-onset neutropenia that was reversible. The MTD was established at 3125 mg twice daily. In an expansion cohort at the MTD, one of 11 (9%) evaluable subjects had a confirmed partial response. This, plus two minor responses, occurred in subjects with neuroendocrine tumors of the midgut or pancreatic origin. Thirty-six percent of subjects had stable disease for more than 6 months. In peripheral blood mononuclear cells, Nelfinavir inhibited AKT and induced markers of ER stress. In summary, nelfinavir is well tolerated in cancer patients at doses 2.5 times the FDA-approved dose for HIV management and showed preliminary activity in tumors of neuroendocrine origin.


Subject(s)
Antineoplastic Agents/therapeutic use , HIV Protease Inhibitors/therapeutic use , Nelfinavir/therapeutic use , Neoplasms/drug therapy , Administration, Oral , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Drug Administration Schedule , Female , HIV Protease Inhibitors/administration & dosage , HIV Protease Inhibitors/adverse effects , HIV Protease Inhibitors/pharmacokinetics , Humans , Male , Maryland , Maximum Tolerated Dose , Middle Aged , Nelfinavir/administration & dosage , Nelfinavir/adverse effects , Nelfinavir/pharmacokinetics , Neoplasms/pathology , Time Factors , Tomography, X-Ray Computed , Treatment Outcome , Young Adult
12.
Article in English | MEDLINE | ID: mdl-25571293

ABSTRACT

BACKGROUND: Simvastatin, a commonly used HMG-CoA reductase inhibitor, is extensively metabolized by CYP3A4. Therefore, co-administration of simvastatin and CYP3A4 inhibitor can affect simvastatin pharmacokinetics. Nelfinavir, a protease inhibitor, and its major metabolite (M8) are known to be potent CYP3A4 inhibitors. When simvastatin and nelfinavir are co-administered, simvastatin pharmacokinetics is significantly altered and may result in an increased risk of rhabdomyolysis. OBJECTIVE: To develop a mathematical model describing a drug-drug interaction between simvastatin and nelfinavir in humans. METHODS: Eligible pharmacokinetic studies were selected from Pubmed database and concentration time course data were digitally extracted and used for model development. Compartmental pharmacokinetic models for simvastatin and nelfinavir were developed separately. A drug-drug interaction model of simvastatin and nelfinavir was subsequently developed using the prior information. Finally, the final drug-drug interaction modeled was validated against observed simvastatin concentrations. RESULTS: Three compartmental pharmacokinetic models were successfully developed. Simvastatin pharmacokinetics was best described by a one compartment model for simvastatin linked to its active form, simvastatin hydroxy acid. Nelfinavir pharmacokinetics could be adequately described by a one compartment parent-metabolite model. Our final drug-drug interaction model predicted an increase in simvastatin exposure which is in line with clinical observations linking the simvastatin-nelfinavir combination to an increased risk of rhabdomyolysis. CONCLUSION: Simvastatin-nelfinavir pharmacokinetic interaction can be explained by our final model. This model framework will be useful in further advanced developing other mechanism based drug-drug interaction model used to predict the risk of rhabdomyolysis occurrence in patients prescribed simvastatin and nelfinavir concurrently.


Subject(s)
Cytochrome P-450 CYP3A Inhibitors/pharmacokinetics , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Nelfinavir/pharmacokinetics , Simvastatin/pharmacokinetics , Adolescent , Adult , Area Under Curve , Cytochrome P-450 CYP3A Inhibitors/adverse effects , Drug Incompatibility , Drug Interactions , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/adverse effects , Middle Aged , Models, Biological , Nelfinavir/adverse effects , Rhabdomyolysis/chemically induced , Simvastatin/adverse effects , Young Adult
13.
Mol Pharm ; 11(2): 436-44, 2014 Feb 03.
Article in English | MEDLINE | ID: mdl-24364805

ABSTRACT

Using positron emission tomography (PET), (11)C-verapamil as the P-gp substrate, and cyclosporine A (CsA) as the P-gp inhibitor, we showed that the magnitude of P-gp-based drug interactions at the human blood-brain barrier (BBB) is modest. However, such interactions at clinically relevant CsA blood concentrations may be greater for substrates where P-gp plays an even larger role (fractional contribution of P-gp, ft > 0.97) in preventing the CNS entry of the drug (e.g., nelfinavir). Since we have shown that the rat is an excellent predictor of the verapamil-CsA interaction at the human BBB, we determined the magnitude of drug interaction at the rat BBB between nelfinavir and CsA. Under isoflurane anesthesia, male Sprague-Dawley rats were coadministered IV infusions of nelfinavir and escalating doses of CsA to achieve pseudo steady-state plasma/blood and brain concentrations of both drugs (blood CsA ranged 0-264.9 µM, n = 3-6/group). The percent increase in the brain:blood nelfinavir concentration ratio (determined by LC/MS) was described by the Hill equation with Emax = 6481%, EC50 = 12.3 µM, and γ = 1.6. Then, using these data, as well as in vitro data in LLCPK1 cells expressing the human P-gp, we predicted that CsA (at clinically relevant blood concentration of 1.5 µM) will increase the distribution of nelfinavir into the human brain by 236%. Collectively, our data suggest that clinically significant P-gp based drug interactions at the human BBB are possible for P-gp substrates highly excluded from the brain (ft > 0.97) and should be investigated using noninvasive approaches (e.g., PET).


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/metabolism , Blood-Brain Barrier/metabolism , Animals , Biological Transport , Cells, Cultured , Cyclosporine/chemistry , Cyclosporine/metabolism , Cyclosporine/pharmacokinetics , Humans , Male , Nelfinavir/blood , Nelfinavir/metabolism , Nelfinavir/pharmacokinetics , Rats , Rats, Sprague-Dawley , Verapamil/blood , Verapamil/metabolism , Verapamil/pharmacokinetics
14.
Cancer Chemother Pharmacol ; 70(6): 791-9, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22983015

ABSTRACT

PURPOSE: HIV protease inhibitors are associated with HIV protease inhibitor-related lipodystrophy syndrome. We hypothesized that liposarcomas would be similarly susceptible to the apoptotic effects of an HIV protease inhibitor, nelfinavir. METHODS: We conducted a phase I trial of nelfinavir for liposarcomas. There was no limit to prior chemotherapy. The starting dose was 1,250 mg twice daily (Level 1). Doses were escalated in cohorts of three to a maximally evaluated dose of 4,250 mg (Level 5). One cycle was 28 days. Steady-state pharmacokinetics (PKs) for nelfinavir and its primary active metabolite, M8, were determined at Levels 4 (3,000 mg) and 5. RESULTS: Twenty subjects (13 males) were enrolled. Median (range) age was 64 years (37-81). One subject at Level 1 experienced reversible, grade 3 pancreatitis after 1 week and was replaced. No other dose-limiting toxicities were observed. Median (range) number of cycles was 3 (0.6-13.5). Overall best responses observed were 1 partial response, 1 minor response, 4 stable disease, and 13 progressive disease. Mean peak plasma levels and AUCs for nelfinavir were higher at Level 4 (7.3 mg/L; 60.9 mg/L × h) than 5 (6.3 mg/L; 37.7 mg/L × h). The mean ratio of M8:nelfinavir AUCs for both levels was ~1:3. CONCLUSIONS: PKs demonstrate auto-induction of nelfinavir clearance at the doses studied, although the mechanism remains unclear. Peak plasma concentrations were within range where anticancer activity was demonstrated in vitro. M8 metabolite is present at ~1/3 the level of nelfinavir and may also contribute to the anticancer activity observed.


Subject(s)
HIV Protease Inhibitors/pharmacokinetics , HIV Protease Inhibitors/therapeutic use , Liposarcoma/drug therapy , Nelfinavir/pharmacokinetics , Nelfinavir/therapeutic use , Adult , Aged , Aged, 80 and over , Area Under Curve , Drug Administration Schedule , Female , HIV Protease Inhibitors/administration & dosage , HIV Protease Inhibitors/blood , HIV-Associated Lipodystrophy Syndrome/drug therapy , Humans , Liposarcoma/blood , Male , Middle Aged , Nelfinavir/administration & dosage , Nelfinavir/blood , Research Design , Treatment Outcome
15.
HIV Clin Trials ; 13(1): 46-59, 2012.
Article in English | MEDLINE | ID: mdl-22306587

ABSTRACT

PURPOSE: Evaluate the safety, tolerability, and pharmacokinetics (PK) of nelfinavir during pregnancy and postpartum in HIV-infected women. METHODS: Phase IV, non-randomized, open-label study of nelfinavir 625 mg tablets (1250 mg) in combination with lamivudine/zidovudine twice daily. Primary endpoint was treatment-related or possibly treatment-related gastrointestinal or hepatic adverse events (AEs). Selected maternal and infant outcomes were recorded. Frequent plasma samples were collected for PK studies during the 2nd and 3rd trimesters, and 6 weeks postpartum, to analyze total and free nelfinavir and M8 concentrations. RESULTS: Sixteen HIV+ pregnant women were enrolled. Six mild treatment-related AEs and 3 serious AEs occurred; 1 serious AE (elevated AST) met the primary endpoint. Compared with 6 weeks postpartum, levels of total nelfinavir were reduced by 44% and 46%, total M8 by 82% and 83%, free nelfinavir by 48% and 39%, and free M8 by 83% and 79% in the 2nd and 3rd trimesters, respectively. At 6 weeks postpartum, 75% and 50% of subjects maintained HIV-1 RNA levels <400 and <50 copies/mL, respectively. All pregnancies resulted in live births without transmission in 15 infants. CONCLUSIONS: Nelfinavir in combination with lamivudine/zidovudine was generally well tolerated. Total and free nelfinavir and M8 exposure were reduced in late pregnancy.


Subject(s)
HIV Infections/drug therapy , HIV Protease Inhibitors/adverse effects , Nelfinavir/adverse effects , Pregnancy Complications, Infectious/drug therapy , Adult , CD4 Lymphocyte Count , Female , HIV-1/genetics , HIV-1/isolation & purification , Humans , Nelfinavir/pharmacokinetics , Postpartum Period , Pregnancy , Pregnancy Trimester, Second , Pregnancy Trimester, Third , RNA, Viral/blood
16.
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
17.
Pediatr Infect Dis J ; 30(9): 769-72, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21666540

ABSTRACT

BACKGROUND: There are no previous data describing nelfinavir and lamivudine pharmacokinetics in neonates treated with weight-band dosing regimens. DESIGN: Pharmacokinetic study of nelfinavir and lamivudine pharmacokinetics in infants during the first 2 weeks of life treated with weight-band dosing regimens. METHODS: Intensive 12-hour pharmacokinetic profiles were performed between either days 4-7 or days 10-14 of life in 26 Brazilian infants. RESULTS: Pharmacokinetic data were obtained from 26 infants who received median (range) per kg doses of 58.8 (48.4-79.0) mg/kg for nelfinavir and 2.0 (1.5-3.2) mg/kg for lamivudine. Median nelfinavir 12-hour AUC (AUC0-12) was 25.5 (1.7-183.5) µg*h/mL and median 12-hour concentration (C12h) was 1.09 (<0.04-14.44) µg/mL. AUC0-12 was less than 15 µg*h/mL (the 10% for adults) in 12 infants (46%). Median lamivudine AUC0-12 was 7.8 (2.7-15.6) µg*h/mL and median C12h was 0.23 (<0.04-0.74) µg/mL. CONCLUSIONS: : Lamivudine pharmacokinetic parameters observed in this study were consistent with those seen in other studies of neonates. While median nelfinavir AUC and C12h in these neonates were above the exposure targets, interindividual variability in nelfinavir exposure was large and nelfinavir exposure failed to meet the exposure targets in 46% of infants.


Subject(s)
Anti-HIV Agents/pharmacokinetics , Lamivudine/pharmacokinetics , Nelfinavir/pharmacokinetics , Anti-HIV Agents/therapeutic use , Area Under Curve , Female , HIV Infections/drug therapy , HIV Infections/virology , HIV-1/physiology , Humans , Infant, Newborn , Infectious Disease Transmission, Vertical , Lamivudine/administration & dosage , Lamivudine/therapeutic use , Male , Nelfinavir/administration & dosage , Nelfinavir/therapeutic use , Viral Load
18.
PLoS Comput Biol ; 7(4): e1002037, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21552547

ABSTRACT

Nelfinavir is a potent HIV-protease inhibitor with pleiotropic effects in cancer cells. Experimental studies connect its anti-cancer effects to the suppression of the Akt signaling pathway, but the actual molecular targets remain unknown. Using a structural proteome-wide off-target pipeline, which integrates molecular dynamics simulation and MM/GBSA free energy calculations with ligand binding site comparison and biological network analysis, we identified putative human off-targets of Nelfinavir and analyzed the impact on the associated biological processes. Our results suggest that Nelfinavir is able to inhibit multiple members of the protein kinase-like superfamily, which are involved in the regulation of cellular processes vital for carcinogenesis and metastasis. The computational predictions are supported by kinase activity assays and are consistent with existing experimental and clinical evidence. This finding provides a molecular basis to explain the broad-spectrum anti-cancer effect of Nelfinavir and presents opportunities to optimize the drug as a targeted polypharmacology agent.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Discovery/methods , Nelfinavir/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Kinases/metabolism , Systems Biology/methods , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Binding Sites , Drug Interactions , Humans , Molecular Dynamics Simulation , Nelfinavir/chemistry , Nelfinavir/pharmacokinetics , Protein Binding , Protein Kinase Inhibitors/chemistry , Thermodynamics
19.
AAPS PharmSciTech ; 12(2): 534-43, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21519984

ABSTRACT

Many nasally applied compounds gain access to the brain and the central nervous system (CNS) with varying degree. Direct nose-to-brain access is believed to be achieved through nervous connections which travel from the CNS across the cribriform plate into the olfactory region of the nasal cavity. However, current delivery strategies are not targeted to preferentially deposit drugs to the olfactory at cribriform. Therefore, we have developed a pressurized olfactory delivery (POD) device which consistently and non-invasively deposited a majority of drug to the olfactory region of the nasal cavity in rats. Using both a hydrophobic drug, mannitol (log P = -3.1), and a hydrophobic drug, nelfinavir (log P = 6.0), and POD device, we compared brain and blood levels after nasal deposition primarily on the olfactory region with POD or nose drops which deposited primarily on the respiratory region in rats. POD administration of mannitol in rats provided a 3.6-fold (p < 0.05) increase in cortex-to-blood ratio, compared to respiratory epithelium deposition with nose drop. Administration of nelfinavir provided a 13.6-fold (p < 0.05) advantage in cortex-to-blood ratio with POD administration, compared to nose drops. These results suggest that increasing the fraction of drug deposited on the olfactory region of the nasal cavity will result in increased direct nose-to-brain transport.


Subject(s)
Brain/metabolism , Drug Delivery Systems/methods , Hydrophobic and Hydrophilic Interactions , Mannitol/administration & dosage , Nelfinavir/administration & dosage , Olfactory Mucosa/metabolism , Administration, Intranasal/standards , Animals , Brain/drug effects , Drug Delivery Systems/standards , Male , Mannitol/pharmacokinetics , Mannitol/standards , Nelfinavir/pharmacokinetics , Nelfinavir/standards , Olfactory Mucosa/drug effects , Rats , Rats, Sprague-Dawley
20.
Eur J Pharm Biopharm ; 79(2): 349-56, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21527341

ABSTRACT

The aim of this study was to evaluate the utility of biorelevant dissolution tests coupled with in silico simulation technology to forecast in vivo bioperformance of poorly water-soluble bases, using nelfinavir mesylate as a model compound. An in silico physiologically based pharmacokinetic (PBPK) model for poorly water-soluble, weakly basic drugs was used to generate plasma profiles of nelfinavir by coupling dissolution results and estimates of precipitation with standard gastrointestinal (GI) parameters and the disposition pharmacokinetics of nelfinavir. In vitro dissolution of nelfinavir mesylate film-coated tablets was measured in biorelevant and compendial media. Drug precipitation in the small intestine was estimated from crystal growth theory. GI parameters (gastric emptying rate and fluid volume) appropriate to the dosing conditions (fasting and fed states) were used in the PBPK model. The disposition parameters of nelfinavir were estimated by fitting compartmental models to the in vivo oral PK data. The in vivo performance in each prandial state was simulated with the PBPK model, and predicted values for AUC and C(max) were compared to observed values. Dissolution results in FaSSIF-V2 and FeSSIF-V2, simulating the fasting and fed small intestinal conditions, respectively, correctly predicted that there would be a positive food effect for nelfinavir mesylate, but overestimated the food effect observed in healthy human volunteers. In order to better predict the food effect, an in silico PBPK simulation model using STELLA® software was evolved. Results with the model indicated that invoking drug precipitation in the small intestine is necessary to describe the in vivo performance of nelfinavir mesylate in the fasted state, whereas a good prediction under fed state conditions is obtained without assuming any precipitation. In vitro-in silico-in vivo relationships (IVISIV-R) may thus be a helpful tool in understanding the critical parameters that affect the oral absorption of poorly soluble weak bases.


Subject(s)
Fasting/metabolism , Food-Drug Interactions , Intestine, Small/metabolism , Nelfinavir/pharmacokinetics , Administration, Oral , Adolescent , Adult , Area Under Curve , Chemical Precipitation , Computer Simulation , Cross-Over Studies , Gastric Emptying , HIV Protease Inhibitors/administration & dosage , HIV Protease Inhibitors/pharmacokinetics , Humans , Intestinal Absorption/physiology , Male , Models, Biological , Nelfinavir/administration & dosage , Solubility , Tablets/administration & dosage , Tablets/pharmacokinetics , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...