Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 90
1.
Biopharm Drug Dispos ; 42(8): 351-358, 2021 Sep.
Article En | MEDLINE | ID: mdl-34191301

Gamma hydroxybutyric acid (GHB) has been approved clinically to treat excessive daytime sleepiness and cataplexy in patients with narcolepsy, alcohol and opioid withdrawal, and as an anesthetic. The use of GHB clinically is limited due to its high abuse potential. The absorption, clearance and tissue uptake of GHB is mediated by proton-dependent and sodium-coupled monocarboxylate transporters (MCTs and SMCTs) and inhibition of these transporters may result in a change in GHB pharmacokinetics and pharmacodynamics. Previous studies have reported that non-steroidal anti-inflammatory drugs (NSAIDs) may inhibit these monocarboxylate transporters. Therefore, the purpose of this work was to analyze the interaction between GHB (at a dose of 600 mg/kg i. v.) and the NSAID, diclofenac, by examining the effects of this drug on the in vivo pharmacokinetics and pharmacodynamics in rat studies. The pharmacodynamic effect evaluated was respiratory depression, a measure of toxicity observed by GHB at this dose. There was an improvement in the respiratory rate with diclofenac administration suggesting an effect of diclofenac on GHB toxicity. In vitro studies with rat blood brain endothelial cells (RBE4) that express MCT1 indicated that diclofenac can inhibit GHB transport with an IC50 of 10.6 µM at pH 7.4. In vivo studies found a decrease in brain GHB concentrations and a decrease in the brain-to-plasma concentration ratio following diclofenac treatment. With this study we can conclude that diclofenac and potentially other NSAIDs can inhibit the transport of GHB into the brain, therefore decreasing GHB's pharmacodynamic effects and toxicity.


Brain , Diclofenac/pharmacokinetics , Drug Interactions , Hydroxybutyrates/pharmacokinetics , Monocarboxylic Acid Transporters , Respiratory Insufficiency , Symporters , Anesthetics/pharmacokinetics , Anesthetics/toxicity , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Biological Transport, Active/drug effects , Brain/drug effects , Brain/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Endothelial Cells/metabolism , Hydroxybutyrates/toxicity , Monocarboxylic Acid Transporters/antagonists & inhibitors , Monocarboxylic Acid Transporters/metabolism , Rats , Rats, Sprague-Dawley , Respiratory Insufficiency/chemically induced , Respiratory Insufficiency/drug therapy , Sodium Oxybate/pharmacokinetics , Symporters/antagonists & inhibitors , Symporters/metabolism
2.
AAPS J ; 23(1): 22, 2021 01 08.
Article En | MEDLINE | ID: mdl-33417072

Gamma-hydroxybutyrate (GHB) is a short-chain fatty acid present endogenously in the brain and used therapeutically for the treatment of narcolepsy, as sodium oxybate, and for alcohol abuse/withdrawal. GHB is better known however as a drug of abuse and is commonly referred to as the "date-rape drug"; current use in popular culture includes recreational "chemsex," due to its properties of euphoria, loss of inhibition, amnesia, and drowsiness. Due to the steep concentration-effect curve for GHB, overdoses occur commonly and symptoms include sedation, respiratory depression, coma, and death. GHB binds to both GHB and GABAB receptors in the brain, with pharmacological/toxicological effects mainly due to GABAB agonist effects. The pharmacokinetics of GHB are complex and include nonlinear absorption, metabolism, tissue uptake, and renal elimination processes. GHB is a substrate for monocarboxylate transporters, including both sodium-dependent transporters (SMCT1, 2; SLC5A8; SLC5A12) and proton-dependent transporters (MCT1-4; SLC16A1, 7, 8, and 3), which represent significant determinants of absorption, renal reabsorption, and brain and tissue uptake. This review will provide current information of the pharmacology, therapeutic effects, and pharmacokinetics/pharmacodynamics of GHB, as well as therapeutic strategies for the treatment of overdoses. Graphical abstract.


Drug Overdose/therapy , Hydroxybutyrates/pharmacokinetics , Sodium Oxybate/pharmacokinetics , Substance Abuse, Oral/therapy , Alcoholism/complications , Alcoholism/drug therapy , Animals , Disease Models, Animal , Drug Evaluation, Preclinical , Drug Overdose/etiology , Humans , Hydroxybutyrates/administration & dosage , Hydroxybutyrates/toxicity , Metabolic Clearance Rate , Narcolepsy/drug therapy , Sodium Oxybate/administration & dosage , Sodium Oxybate/toxicity , Substance Abuse, Oral/etiology , Substance Withdrawal Syndrome/drug therapy
3.
Arthritis Care Res (Hoboken) ; 73(7): 983-989, 2021 07.
Article En | MEDLINE | ID: mdl-32339392

OBJECTIVE: Leflunomide is a commonly used disease-modifying drug in the treatment of rheumatoid arthritis (RA). Its effects are mediated via inhibition of dihydroorotate dehydrogenase (DHODH) by its active metabolite teriflunomide, and the pharmacokinetics of teriflunomide are highly variable. Our objective was to examine the association between the DHODH haplotype and plasma teriflunomide concentration with response to leflunomide in patients with RA where leflunomide was added to an existing disease-modifying drug regimen after failure to achieve an adequate response with conventional triple therapy. METHODS: Patients with RA who were taking, or were about to initiate, leflunomide were included. Participant characteristics, including the DHODH haplotype, were determined. Up to 5 plasma samples were collected after leflunomide was initiated for assays of total and free teriflunomide concentration. Disease activity was determined via the 28-joint Disease Activity Score (DAS28). The association between DAS28 scores and patient covariates was determined by linear mixed-effects modeling. RESULTS: A total of 67 patients were included in the study. The DAS28 score after initiation of leflunomide was associated with the baseline DAS28 score (ß = 0.70, P < 0.001) and was higher in those who carried the DHODH haplotype 2 (ß = 0.56. P = 0.01) and did not carry the shared epitope (ß = 0.56, P = 0.013). As total and free plasma teriflunomide concentration increased, the DAS28 score was significantly lower (P < 0.001 and P = 0.001, respectively). When considering threshold concentrations, teriflunomide concentrations >16 mg/liter were associated with a DAS28 score that was 0.33 lower, and when free teriflunomide concentration was >35 µg/liter, the DAS28 score was 0.32 lower. CONCLUSION: Teriflunomide concentration and carriage of the DHODH haplotype 2 are associated with response to leflunomide in patients with RA, and a total plasma teriflunomide concentration of at least 16 mg/liter is needed to maximize the likelihood of response.


Arthritis, Rheumatoid/drug therapy , Crotonates/pharmacokinetics , Enzyme Inhibitors/pharmacokinetics , Hydroxybutyrates/pharmacokinetics , Immunosuppressive Agents/pharmacokinetics , Leflunomide/pharmacokinetics , Nitriles/pharmacokinetics , Oxidoreductases Acting on CH-CH Group Donors/antagonists & inhibitors , Oxidoreductases Acting on CH-CH Group Donors/genetics , Pharmacogenomic Variants , Polymorphism, Single Nucleotide , Toluidines/pharmacokinetics , Adult , Arthritis, Rheumatoid/diagnosis , Arthritis, Rheumatoid/genetics , Crotonates/blood , Dihydroorotate Dehydrogenase , Drug Monitoring , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/blood , Female , Haplotypes , Humans , Hydroxybutyrates/blood , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/blood , Leflunomide/administration & dosage , Leflunomide/blood , Male , Middle Aged , Nitriles/blood , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Pharmacogenetics , Precision Medicine , Recovery of Function , Remission Induction , Toluidines/blood , Treatment Outcome
4.
Biomed Chromatogr ; 34(3): e4781, 2020 Mar.
Article En | MEDLINE | ID: mdl-31845359

A volumetric microsampling (VAMS) device (20 µl) was evaluated and validated for the analysis of γ-hydroxybutyric acid (GHB) in venous blood using a simple ultra-high-pressure liquid chromatography-tandem mass spectrometry (UHPLC-MS/MS) method. GHB was extracted from VAMS device by acetonitrile, after a re-hydration step in a temperature-controlled ultrasonic bath at 60°C for 10 min. Chromatographic analysis was carried out on a Kinetex C18 column using 0.1% formic acid in water and acetonitrile as binary gradient mobile phase (from 5 to 95% of acetonitrile from 1 to 2.5 min) at a flow rate of 0.3 ml/min. The VAMS method was fully validated according to current guidelines with satisfactory results in terms of linearity, selectivity, precision, absolute recovery, matrix effect and stability. The linearity was determined from 0.5 to 200 µg/ml and the lower limit of quantitation was 0.5 µg/ml. The novel VAMS-UHPLC-MS/MS method was successfully compared with plasma-based method in a GHB-treated patient as a proof of concept.


Chromatography, High Pressure Liquid/methods , Hydroxybutyrates/blood , Tandem Mass Spectrometry/methods , Blood Specimen Collection , Humans , Hydroxybutyrates/pharmacokinetics , Hydroxybutyrates/therapeutic use , Limit of Detection , Linear Models , Narcolepsy/drug therapy , Reproducibility of Results
5.
Nucl Med Biol ; 71: 11-18, 2019 04.
Article En | MEDLINE | ID: mdl-31108463

BACKGROUND: [166Ho]Ho-acetylacetonate-poly(L-lactic acid) microspheres were used in radioembolization of liver malignancies by intra-arterial administration. The primary aim of this study was to assess the stability and biodistribution of these microspheres. MATERIALS AND METHODS: Peripheral blood and urine samples were obtained from two clinical studies. Patient and in vitro experiment samples were analyzed using inductively coupled plasma mass spectrometry (ICP-MS), gamma-ray spectroscopy, light microscopy, Coulter particle counting, and high performance liquid chromatography (HPLC). RESULTS: The median percentage holmium compared to the total amount injected into the hepatic artery was 0.19% (range 0.08-2.8%) and 0.32% (range 0.03-1.8%) in the 1 h blood plasma and 24 h urine, respectively. Both the blood plasma and urine were correlated with the neutron irradiation exposure required for [166Ho]Ho-AcAc-PLLA microsphere production (ρ = 0.616, p = 0.002). After a temporary interruption of the phase 2 clinical study, the resuspension medium was replaced to precipitate [166Ho]Ho3+ pre-administration using phosphate. The in vitro near-maximum neutron irradiation experiments showed significant [166Ho]Ho-AcAc-PLLA microsphere damage. CONCLUSION: The amount of holmium in the peripheral blood and urine samples after [166Ho]Ho-AcAc-PLLA microsphere intrahepatic infusion was low. A further decrease was observed after reformulation of the resuspension solution but minimization of production damage is necessary.


Embolization, Therapeutic , Hydroxybutyrates/chemistry , Hydroxybutyrates/therapeutic use , Lactates/chemistry , Lactates/therapeutic use , Lactic Acid/chemistry , Lactic Acid/therapeutic use , Liver Neoplasms/radiotherapy , Microspheres , Pentanones/chemistry , Pentanones/therapeutic use , Drug Stability , Humans , Hydroxybutyrates/pharmacokinetics , Lactates/pharmacokinetics , Lactic Acid/pharmacokinetics , Liver Neoplasms/blood , Liver Neoplasms/urine , Pentanones/pharmacokinetics , Tissue Distribution
6.
J Pharmacol Exp Ther ; 370(1): 84-91, 2019 07.
Article En | MEDLINE | ID: mdl-31010842

The illicit use of γ-hydroxybutyric acid (GHB), and its prodrug, γ-butyrolactone (GBL), results in severe adverse effects including sedation, coma, respiratory depression, and death. Current treatment of GHB/GBL overdose is limited to supportive care. Recent reports indicate that GHB-related deaths are on the rise; a specific treatment may reduce lethality associated with GHB/GBL. Pretreatment with inhibitors of monocarboxylate transporter 1 (MCT1), a transporter that mediates many of the processes involved in the absorption, distribution (including brain uptake), and elimination of GHB/GBL, has been shown to prevent GHB-induced respiratory depression by increasing the renal clearance of GHB. To identify whether MCT1 inhibition is an effective treatment of GHB overdose, the impact of two MCT1 inhibitors, (S)-5-(4-hydroxy-4-methylisoxazolidine-2-carbonyl)-1-isopropyl-3-methyl-6-((3-methyl-5-(trifluoromethyl)-1H-pyrazol-4-yl)methyl)thieno[2,3-day]pyrimidine-2,4(1H,3H)-dione (AZD3965) and 6-[(3,5-dimethyl-1H-pyrazol-4-yl)methyl]-5-[[(4S)-4-hydroxy-2-isoxazolidinyl]carbonyl]-3-methyl-1-(2-methylpropyl)thieno[2,3-day]pyrimidine2,4(1H,3H)-dione (AR-C155858), on the toxicokinetics and toxicodynamics of GHB/GBL was assessed when the administration of the inhibitor was delayed 60 and 120 minutes (post-treatment) after administration of GHB/GBL. AR-C155858 and AZD3965 reduced the toxicodynamic effects of GHB when GHB was administered intravenously, orally, or orally as the prodrug GBL. The impact of these inhibitors on GHB toxicokinetics was dependent on the route of GHB administration and the delay between GHB/GBL administration and administration of the MCT1 inhibitor. The reduction in GHB plasma exposure did not explain the observed effect of MCT1 inhibition on GHB-induced respiratory depression. The efficacy of MCT1 inhibition on GHB toxicodynamics is likely driven by the pronounced reduction in GHB brain concentrations. Overall, this study indicates that inhibition of MCT1 is an effective treatment of GHB/GBL overdose.


4-Butyrolactone/toxicity , Drug Overdose/drug therapy , Hydroxybutyrates/toxicity , Monocarboxylic Acid Transporters/antagonists & inhibitors , Prodrugs/pharmacology , Pyrimidinones/pharmacology , Symporters/antagonists & inhibitors , Thiophenes/pharmacology , Uracil/analogs & derivatives , 4-Butyrolactone/administration & dosage , Administration, Intravenous , Administration, Oral , Animals , Brain/drug effects , Brain/metabolism , Drug Overdose/blood , Drug Overdose/metabolism , Hydroxybutyrates/administration & dosage , Hydroxybutyrates/blood , Hydroxybutyrates/pharmacokinetics , Male , Pyrimidinones/therapeutic use , Rats , Rats, Sprague-Dawley , Thiophenes/therapeutic use , Uracil/pharmacology , Uracil/therapeutic use
7.
Biopharm Drug Dispos ; 39(4): 218-231, 2018 Apr.
Article En | MEDLINE | ID: mdl-29635775

Renal impairment (RI) significantly impacts the clearance of drugs through changes in the glomerular filtration rate, protein binding and alterations in the expression of renal drug transport proteins and hepatic metabolizing enzymes. The objectives of this study were to evaluate quantitatively the effects of renal impairment on the pharmacokinetics of drugs undergoing renal transporter-mediated reabsorption. A previously published semi-mechanistic kidney model incorporating physiologically relevant fluid reabsorption and transporter-mediated active renal reabsorption (PMID: 26341876) was utilized in this study. The probe drug/transporter pair utilized was γ-hydroxybutyric acid (GHB) and monocarboxylate transporter 1 (SCL16A1, MCT1). γ-Hydroxybutyric acid concentrations in the blood and amount excreted into urine were simulated using ADAPT 5 for the i.v. dose range of 200-1500 mg/kg in rats and the impact of renal impairment on CLR and AUC was evaluated. A 90% decrease in GFR resulted in a > 100-fold decrease in GHB CLR . When expression of reabsorptive transporters was decreased and fu was increased, CLR approached GFR. The effect of renal impairment on CLR was reduced when the expression of drug metabolizing enzymes (DME) was increased as a result of increased metabolic clearance; the converse held true when the DME expression was decreased. In conclusion, this study quantitatively demonstrated that the effects of renal insufficiency on the clearance of drugs is modulated by transporter expression, contribution of renal clearance to overall clearance, expression of drug metabolizing enzymes, fraction unbound and drug-drug interactions with inhibitors of renal transporters that may be increased in the presence of renal impairment.


Hydroxybutyrates/pharmacokinetics , Monocarboxylic Acid Transporters/metabolism , Renal Insufficiency/metabolism , Symporters/metabolism , Animals , Computer Simulation , Drug Interactions , Hydroxybutyrates/blood , Hydroxybutyrates/urine , Kidney/metabolism , Rats
8.
AAPS J ; 20(1): 21, 2017 12 26.
Article En | MEDLINE | ID: mdl-29280004

The drug of abuse γ-hydroxybutyric acid (GHB) demonstrates complex toxicokinetics with dose-dependent metabolic and renal clearance. GHB is a substrate of monocarboxylate transporters (MCTs) which are responsible for the saturable renal reabsorption of GHB. MCT expression is observed in many tissues and therefore may impact the tissue distribution of GHB. The objective of the present study was to evaluate the tissue distribution kinetics of GHB at supratherapeutic doses. GHB (400, 600, and 800 mg/kg iv) or GHB 600 mg/kg plus L-lactate (330 mg/kg iv bolus followed by 121 mg/kg/h infusion) was administered to rats and blood and tissues were collected for up to 330 min post-dose. K p values for GHB varied in both a tissue- and dose-dependent manner and were less than 0.5 (except in the kidney). Nonlinear partitioning was observed in the liver (0.06 at 400 mg/kg to 0.30 at 800 mg/kg), kidney (0.62 at 400 mg/kg to 0.98 at 800 mg/kg), and heart (0.15 at 400 mg/kg to 0.29 at 800 mg/kg), with K p values increasing with dose consistent with saturation of transporter-mediated efflux. In contrast, lung partitioning decreased in a dose-dependent manner (0.43 at 400 mg/kg to 0.25 at 800 mg/kg) suggesting saturation of active uptake. L-lactate administration decreased K p values in liver, striatum, and hippocampus and increased K p values in lung and spleen. GHB demonstrates tissue-specific nonlinear distribution consistent with the involvement of monocarboxylate transporters. These observed complexities are likely due to the involvement of MCT1 and 4 with different affinities and directionality for GHB transport.


Hydroxybutyrates/pharmacokinetics , Illicit Drugs/pharmacokinetics , Monocarboxylic Acid Transporters/metabolism , Muscle Proteins/metabolism , Symporters/metabolism , Administration, Intravenous , Animals , Drug Overdose/drug therapy , Drug Overdose/etiology , Hydroxybutyrates/adverse effects , Hydroxybutyrates/metabolism , Illicit Drugs/adverse effects , Illicit Drugs/metabolism , Male , Models, Animal , Monocarboxylic Acid Transporters/antagonists & inhibitors , Muscle Proteins/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Symporters/antagonists & inhibitors , Tissue Distribution , Toxicokinetics
9.
Article En | MEDLINE | ID: mdl-28703776

Research on the uptake and effects of bioplastics by aquatic organisms is still in its infancy. Here, we aim to advance the field by comparing uptake and effects of microplastic particles (MPP) of a biodegradable bioMPP (polyhydroxybutyrate (PHB)) and petroleum-based MPP (polymethylmethacrylate (PMMA)) in the freshwater amphipod Gammarus fossarum. Ingestion of both MPP in different particle sizes (32-250 µm) occurred after 24 h, with highest ingestion of particles in the range 32-63 µm and almost complete egestion after 64 h. A four-week effect-experiment showed a significant decrease of the assimilation efficiency in amphipods exposed to the petroleum-based MPP from week two onwards. The petroleum-based PMMA affected assimilation efficiency significantly in contrast to the biodegradable PHB, but overall differences in direct comparison of MPP types were small. Both MPP types led to a significantly lower wet weight gain relative to the control treatments. After four weeks, differences between both MPP types and silica, used as a natural particle control, were detected. In summary, these results suggest that both MPP types provoke digestive constraints on the amphipods, which go beyond those of natural non-palatable particles. This highlights the need for more detailed research comparing environmental effects of biodegradable and petroleum-based MPP and testing those against naturally occurring particle loads.


Amphipoda/drug effects , Hydroxybutyrates/toxicity , Polyesters/toxicity , Polymethyl Methacrylate/toxicity , Amphipoda/metabolism , Animals , Digestion/drug effects , Eating , Fresh Water , Hydroxybutyrates/pharmacokinetics , Particle Size , Petroleum , Polyesters/pharmacokinetics , Polymethyl Methacrylate/pharmacokinetics
10.
AAPS J ; 19(5): 1449-1460, 2017 09.
Article En | MEDLINE | ID: mdl-28653244

An overdose of γ-hydroxybutyric acid (GHB), a drug of abuse, results in fatality caused by severe respiratory depression. In this study, a semi-mechanistic pharmacokinetic/pharmacodynamic (PK/PD) model was developed to characterize monocarboxylate transporter 1 (MCT1)-mediated transport of GHB, as well as effects of GHB on respiration frequency, for IV doses of 200, 600, and 1500 mg/kg in rats. The proposed PK/PD model for GHB consists of nonlinear metabolism of GHB in the liver, MCT1-mediated renal reabsorption with physiologically relevant concurrent fluid reabsorption, MCT1-mediated uptake into the brain, and direct effects of binding of GHB to GABAB receptors on the PD parameter, respiration frequency. Michaelis-Menten affinity constants for metabolism, renal reabsorption, and uptake into and efflux from the brain were fixed to the observed in vitro values. The IC 50 value for the effect of GHB on respiration frequency was fixed to a reported value for binding of GHB to GABAB receptors. All physiological parameters were fixed to the reported values for a 300-g rat. The model successfully captured the GHB PK/PD data and was further validated using the data for a 600-mg/kg dose of GHB after IV bolus administration. Unbound GHB brain ECF/blood partition coefficient (Kp u,u ) values obtained from the model agreed well with values calculated using experimental ECF concentrations obtained with brain microdialysis, demonstrating the physiological relevance of this model. Sensitivity analysis indicated that the PK/PD model was stable. In conclusion, we developed a semi-mechanistic and physiologically relevant PK/PD model of GHB using in vitro drug-transporter kinetics and in vivo PK/PD data in rats.


Hydroxybutyrates/pharmacology , Hydroxybutyrates/pharmacokinetics , Brain/metabolism , Humans , Models, Biological
11.
AAPS J ; 18(3): 678-88, 2016 05.
Article En | MEDLINE | ID: mdl-26893218

The administration of ketones to induce a mild ketosis is of interest for the alleviation of symptoms associated with various neurological disorders. This study aimed to understand the pharmacokinetics (PK) of D-ß-hydroxybutyrate (BHB) and quantify the sources of variability following a dose of (R)-3-hydroxybutyl (R)-3-hydroxybutyrate (ketone monoester). Healthy volunteers (n = 37) were given a single drink of the ketone monoester, following which, 833 blood BHB concentrations were measured. Two formulations and five dose levels of ketone monoester were used. A nonlinear mixed effect modelling approach was used to develop a population PK model. A one compartment disposition model with negative feedback effect on endogenous BHB production provided the best description of the data. Absorption was best described by two consecutive first-order inputs and elimination by dual processes involving first-order (CL = 10.9 L/h) and capacity limited elimination (V max = 4520 mg/h). Covariates identified were formulation (on relative oral bioavailable fraction and absorption rate constant) and dose (on relative oral bioavailable fraction). Lean body weight (on first-order clearance) and sex (on apparent volume of distribution) were also significant covariates. The PK of BHB is complicated by complex absorption process, endogenous production and nonlinear elimination. Formulation and dose appear to strongly influence the kinetic profile following ketone monoester administration. Further work is needed to quantify mechanisms of absorption and elimination of ketones for therapeutic use in the form of ketone monoester.


Hydroxybutyrates/pharmacokinetics , Female , Humans , Hydroxybutyrates/administration & dosage , Male , Models, Theoretical
12.
Nanoscale ; 8(3): 1332-7, 2016 Jan 21.
Article En | MEDLINE | ID: mdl-26692041

A star polymer of poly[(R,S)-3-hydroxybutyrate] (PHB) with adamantyl end-terminals extended from an α-cyclodextrin (α-CD) core is designed. It subsequently self-assembles to form controllable and uniform nanovesicles induced by host-guest interactions between heptakis(2,6-di-O-methyl)-ß-CD and the adamantyl ends. The nanovesicles are suitable for loading and intracellular delivery of the anticancer drug doxorubicin.


Antibiotics, Antineoplastic , Doxorubicin , Drug Carriers , Hydroxybutyrates , Nanoparticles/chemistry , Polyesters , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/pharmacokinetics , Antibiotics, Antineoplastic/pharmacology , Doxorubicin/chemistry , Doxorubicin/pharmacokinetics , Doxorubicin/pharmacology , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Drug Carriers/pharmacology , HeLa Cells , Humans , Hydroxybutyrates/chemistry , Hydroxybutyrates/pharmacokinetics , Hydroxybutyrates/pharmacology , Polyesters/chemistry , Polyesters/pharmacokinetics , Polyesters/pharmacology , Prohibitins
13.
J Biomed Mater Res B Appl Biomater ; 104(1): 106-15, 2016 Jan.
Article En | MEDLINE | ID: mdl-25655488

Guided tissue regeneration is a technique used for periodontium reconstruction. This technique uses barrier membranes, which prevent epithelial growth in the wound site and may also be used to release antibiotics, to protect the wound against opportunistic infections. Periodontal poly(3-hydroxybutyrate) membranes containing metronidazole (a drug used to help in infection control) were produced and characterized. The kinetic mechanism of the metronidazole delivery of leached and nonleached membrane as well as its cytotoxicity and structural integrity were evaluated. Poly(3-hydroxybutyrate) membranes containing 0.5-2 wt % of the drug and 20 wt % of the plasticizer were manufactured via compression molding. Based on morphological analysis, membranes loaded with 2% metronidazole were considered for detailed studies. The results revealed that metronidazole delivery by the leached membranes seemed to follow the Fick's law. Membranes were noncytotoxic. The amount of metronidazole delivered was in the range of the minimal inhibitory concentration for Porphyromonas gingivalis, and the membranes inhibited the proliferation of these bacteria. Besides, they maintained their mechanical resistance after 30 days of immersion in phosphate buffer at pH 7.4.


Anti-Bacterial Agents , Hydroxybutyrates , Membranes, Artificial , Metronidazole , Periodontitis/therapy , Polyesters , Porphyromonas gingivalis/growth & development , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacokinetics , Drug Evaluation, Preclinical , Guided Tissue Regeneration, Periodontal/methods , Humans , Hydroxybutyrates/chemistry , Hydroxybutyrates/pharmacokinetics , Hydroxybutyrates/pharmacology , Metronidazole/chemistry , Metronidazole/pharmacokinetics , Metronidazole/pharmacology , Periodontitis/microbiology , Polyesters/chemistry , Polyesters/pharmacokinetics , Polyesters/pharmacology
14.
J Pharmacokinet Pharmacodyn ; 42(5): 497-513, 2015 Oct.
Article En | MEDLINE | ID: mdl-26341876

This study developed a semi-mechanistic kidney model incorporating physiologically-relevant fluid reabsorption and transporter-mediated active reabsorption. The model was applied to data for the drug of abuse γ-hydroxybutyric acid (GHB), which exhibits monocarboxylate transporter (MCT1/SMCT1)-mediated renal reabsorption. The kidney model consists of various nephron segments--proximal tubules, Loop-of-Henle, distal tubules, and collecting ducts--where the segmental fluid flow rates, volumes, and sequential reabsorption were incorporated as functions of the glomerular filtration rate. The active renal reabsorption was modeled as vectorial transport across proximal tubule cells. In addition, the model included physiological blood, liver, and remainder compartments. The population pharmacokinetic modeling was performed using ADAPT5 for GHB blood concentration-time data and cumulative amount excreted unchanged into urine data (200-1000 mg/kg IV bolus doses) from rats [Felmlee et al (PMID: 20461486)]. Simulations assessed the effects of inhibition (R = [I]/KI = 0-100) of renal reabsorption on systemic exposure (AUC) and renal clearance of GHB. Visual predictive checks and other model diagnostic plots indicated that the model reasonably captured GHB concentrations. Simulations demonstrated that the inhibition of renal reabsorption significantly increased GHB renal clearance and decreased AUC. Model validation was performed using a separate dataset. Furthermore, our model successfully evaluated the pharmacokinetics of L-lactate using data obtained from Morse et al (PMID: 24854892). In conclusion, we developed a semi-mechanistic kidney model that can be used to evaluate transporter-mediated active renal reabsorption of drugs by the kidney.


Body Fluids/metabolism , Hydroxybutyrates/pharmacokinetics , Kidney/metabolism , Lactic Acid/pharmacokinetics , Renal Reabsorption/drug effects , Animals , Biological Transport/drug effects , Dose-Response Relationship, Drug , Hydrogen-Ion Concentration , Injections, Intravenous/methods , Male , Membrane Transport Proteins/metabolism , Rats , Rats, Sprague-Dawley
15.
Int Rev Neurobiol ; 120: 205-33, 2015.
Article En | MEDLINE | ID: mdl-26070759

Eating, drinking, sexual activity, and parenting invoke pleasure, an emotion that promotes repetition of these behaviors, are essential for survival. Euphoria, a feeling or state of intense excitement and happiness, is an amplification of pleasure, aspired to one's essential biological needs that are satisfied. People use party drugs as a shortcut to euphoria. Ecstasy (3,4-methylenedioxymethamphetamine), γ-hydroxybutyric acid, and ketamine fall under the umbrella of the term "party drugs," each with differing neuropharmacological and physiological actions. This chapter seeks to survey the history and epidemiology of party drug use; we will then discuss the pharmacological characteristics of each drug to provide a platform for understanding the difficulties that party drug users encounter through intoxication, harmful use, dependence, and withdrawal and how these should be clinically managed.


Euphoria/drug effects , Hydroxybutyrates/adverse effects , Illicit Drugs/adverse effects , Ketamine/adverse effects , N-Methyl-3,4-methylenedioxyamphetamine/adverse effects , Substance-Related Disorders/drug therapy , History, 20th Century , Humans , Hydroxybutyrates/pharmacokinetics , Hydroxybutyrates/pharmacology , Illicit Drugs/history , Ketamine/pharmacokinetics , Ketamine/pharmacology , N-Methyl-3,4-methylenedioxyamphetamine/pharmacokinetics , N-Methyl-3,4-methylenedioxyamphetamine/pharmacology
16.
Biochem Pharmacol ; 92(3): 499-505, 2014 Dec 01.
Article En | MEDLINE | ID: mdl-25256836

Gamma hydroxybutyrate (GHB) is a recreational and date-rape drug, for which the detection following ingestion is hampered by rapid metabolism and its endogenous presence. GHB catabolism occurs mainly by its oxidation to succinic semialdehyde (SSA), which converts to succinate and enters the tricarboxylic acid cycle. A high Km aldehyde reductase has previously been reported to catalyse the NADP-dependent oxidation of GHB at high concentrations. It is assumed that this enzyme is identical to the aldo-keto reductase AKR1A1, but its role in GHB oxidation has not been fully evaluated. In this study, the extent of AKR1A1 in GHB metabolism has been determined in HepG2 cells using RNA-interference technology. The gene encoding AKR1A1 was targeted by siRNA. Results demonstrate a successful knock-down of the AKR1A1 gene with 92% reduction in total mRNA and 93% reduction in protein expression. Demolishing AKR1A1 expression in HepG2 cells leads to significant 82% decrease in NADP-dependent GHB-dehydrogenase activity at high concentration (10mM) of GHB. Moreover, when exposing the cells to 50 µM of GHB for 24h, and measuring intracellular and extracellular GHB levels by GC/MS, a significant two-fold increase was observed on GHB intracellular level in silenced cells. In contrast, measuring SSA-reductase activity in silenced cells indicated that AKR1A1 is not involved in endogenous GHB production. These findings describe a pathway for GHB metabolism in the liver which should be useful in GHB exposure cases, and will enable a better understanding of the enzymes participating in its metabolism at natural and overexposed levels.


Aldehyde Reductase/metabolism , Hydroxybutyrates/pharmacokinetics , Aldehyde Reductase/genetics , Gas Chromatography-Mass Spectrometry , Gene Knockdown Techniques , Hep G2 Cells/drug effects , Humans , Hydroxybutyrates/analysis , Hydroxybutyrates/metabolism , Inactivation, Metabolic , Liver/drug effects , Liver/metabolism
17.
J Pharm Sci ; 103(10): 3310-5, 2014 Oct.
Article En | MEDLINE | ID: mdl-25174723

γ-Hydroxybutyric acid (GHB) is widely abused in combination with other club drugs such as 3,4-methylenedioxymethamphetamine (MDMA). The objectives of this study were to characterize the effects of MDMA on GHB toxicokinetics/toxicodynamics (TK/TD) and evaluate the use of monocarboxylate transporter (MCT) inhibition as a potential treatment strategy for GHB overdose when GHB is abused with MDMA. Rats were administered GHB 400 mg/kg i.v. alone or with MDMA (5 mg/kg i.v). Effects of MDMA and of the MCT inhibitor, l-lactate, on GHB TK and sedative effects were evaluated. The results of this study demonstrated no significant effect of MDMA on GHB TK or TD. GHB plasma concentrations were unchanged, and GHB concentration-effect relationships, based on plasma and brain concentrations and the return-to-righting reflex (RRR), were similar in the presence and absence of MDMA. l-Lactate administration resulted in a significant decrease in the sedative effect (RRR) of GHB when it was coadministered with MDMA. Our results indicate that MDMA does not affect the TK/TD of GHB at the doses used in this study, and MCT inhibition using l-lactate, an effective overdose treatment strategy for GHB alone, is also effective for GHB overdose when GHB is coingested with MDMA.


Hydroxybutyrates/pharmacokinetics , Hydroxybutyrates/toxicity , Hypnotics and Sedatives/pharmacokinetics , Hypnotics and Sedatives/toxicity , N-Methyl-3,4-methylenedioxyamphetamine/pharmacology , Animals , Hydroxybutyrates/pharmacology , Male , Rats , Rats, Sprague-Dawley
18.
Curr Pharm Des ; 20(25): 4076-85, 2014.
Article En | MEDLINE | ID: mdl-24001290

A growing body of evidence shows that gamma-hydroxybutyric acid (GHB) is an addictive substance. Its precursors gammabutyrolactone (GBL) and 1,4-butanediol (1,4-BD) show the same properties and may pose even more risks due to different pharmacokinetics. There are indications that problematic GHB use is increasing in the European Union. This review investigates the existing literature on the neurochemistry of GHB and its precursors, their acute toxicity, addiction potential and withdrawal, the proposed molecular mechanism underlying addiction and the treatment of withdrawal and addiction. Current evidence shows that GHB and its precursors are highly addictive, both in humans and animals, probably through a GABAB receptor related mechanism. Severity of withdrawal symptoms can be considered as a medical emergency. Recent studies suggest that benzodiazepines are not very effective, showing a high treatment resistance, whereas detoxification with pharmaceutical GHB proved to be successful. However, relapse in GHB use is frequent and more research is warranted on relapse prevention. This might aid medical practitioners in the field and improve general understanding of the severity of addiction to GHB, GBL and 1,4-BD.


4-Butyrolactone/adverse effects , Butylene Glycols/adverse effects , Hydroxybutyrates/adverse effects , Substance-Related Disorders/drug therapy , 4-Butyrolactone/metabolism , 4-Butyrolactone/pharmacokinetics , Animals , Butylene Glycols/metabolism , Butylene Glycols/pharmacokinetics , Humans , Hydroxybutyrates/metabolism , Hydroxybutyrates/pharmacokinetics , Substance-Related Disorders/prevention & control
19.
Invest New Drugs ; 32(1): 160-70, 2014 Feb.
Article En | MEDLINE | ID: mdl-23817974

INTRODUCTION: This phase 1 study assessed safety, maximum tolerated dose (MTD), pharmacokinetics, cerebrospinal fluid (CSF) distribution, and preliminary clinical activity of the receptor tyrosine kinase inhibitor TAK-285. METHODS: Patients with advanced, histologically confirmed solid tumors and Eastern Cooperative Oncology Group performance status ≤2 received daily oral TAK-285; daily dose was escalated within defined cohorts until MTD and recommended phase 2 dose (RP2D) were determined. Eleven patients were enrolled into an RP2D cohort. Blood samples were collected from all cohorts; CSF was collected at pharmacokinetic steady-state from RP2D patients. Tumor responses were assessed every 8 weeks per Response Evaluation Criteria in Solid Tumors. RESULTS: Fifty-four patients were enrolled (median age 60; range, 35-76 years). The most common diagnoses were cancers of the colon (28 %), breast (17 %), and pancreas (9 %). Escalation cohorts evaluated doses from 50 mg daily to 500 mg twice daily; the MTD/RP2D was 400 mg twice daily. Dose-limiting toxicities included diarrhea, hypokalemia, and fatigue. Drug absorption was fast (median time of maximum concentration was 2-3 h), and mean half-life was 9 h. Steady-state average unbound CSF concentration (geometric mean 1.54 [range, 0.51-4.27] ng/mL; n = 5) at the RP2D was below the 50 % inhibitory concentration (9.3 ng/mL) for inhibition of tyrosine kinase activity in cells expressing recombinant HER2. Best response was stable disease (12 weeks of nonprogression) in 13 patients. CONCLUSIONS: TAK-285 was generally well tolerated at the RP2D. Distribution in human CSF was confirmed, but the free concentration of the drug was below that associated with biologically relevant target inhibition.


Antineoplastic Agents/cerebrospinal fluid , Antineoplastic Agents/pharmacokinetics , Bridged Bicyclo Compounds, Heterocyclic/cerebrospinal fluid , Bridged Bicyclo Compounds, Heterocyclic/pharmacokinetics , ErbB Receptors/antagonists & inhibitors , Hydroxybutyrates/cerebrospinal fluid , Hydroxybutyrates/pharmacokinetics , Neoplasms/cerebrospinal fluid , Neoplasms/metabolism , Receptor, ErbB-2/antagonists & inhibitors , Adult , Aged , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Bridged Bicyclo Compounds, Heterocyclic/administration & dosage , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Demography , Dose-Response Relationship, Drug , ErbB Receptors/metabolism , Female , Humans , Hydroxybutyrates/administration & dosage , Hydroxybutyrates/therapeutic use , Male , Middle Aged , Neoplasms/drug therapy , Receptor, ErbB-2/metabolism , Treatment Outcome
20.
Biomaterials ; 34(30): 7552-62, 2013 Oct.
Article En | MEDLINE | ID: mdl-23849878

Alzheimer's disease (AD) is induced by many reasons, including decreased cellular utilization of glucose and brain cell mitochondrial damages. Degradation product of microbially synthesized polyhydroxybutyrate (PHB), namely, 3-hydroxybutyrate (3HB), can be an alternative to glucose during sustained hypoglycemia. In this study, the derivative of 3HB, 3-hydroxybutyrate methyl ester (HBME), was used by cells as an alternative to glucose. HBME inhibited cell apoptosis under glucose deprivation, rescued activities of mitochondrial respiratory chain complexes that were impaired in AD patients and decreased the generation of ROS. Meanwhile, HBME stabilized the mitochondrial membrane potential. In vivo studies showed that HBME crossed the blood brain barrier easier compared with charged 3HB, resulting in a better bioavailability. AD mice treated with HBME performed significantly better (p < 0.05) in the Morris water maze compared with other groups, demonstrating that HBME has a positive in vivo pharmaceutical effect to improve the spatial learning and working memory of mice. A reduced amyloid-ß deposition in mouse brains after intragastric administration of HBME was also observed. Combined with the in vitro and in vivo results, HBME was proposed to be a drug candidate against AD, its working mechanism appeared to be mediated by various effects of protecting mitochondrial damages.


Alzheimer Disease/drug therapy , Mitochondria/metabolism , Neuroprotective Agents/therapeutic use , 3-Hydroxybutyric Acid/pharmacokinetics , 3-Hydroxybutyric Acid/pharmacology , 3-Hydroxybutyric Acid/therapeutic use , Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/metabolism , Animals , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Apoptosis/drug effects , Atrophy , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Blood-Brain Barrier/physiopathology , Caspase 3/genetics , Caspase 3/metabolism , Disease Models, Animal , Down-Regulation/drug effects , Electron Transport/drug effects , Glucose/pharmacology , Hydroxybutyrates/pharmacokinetics , Hydroxybutyrates/pharmacology , Learning/drug effects , Magnetic Resonance Imaging , Membrane Potential, Mitochondrial/drug effects , Memory, Short-Term/drug effects , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Neuroprotective Agents/pharmacokinetics , Neuroprotective Agents/pharmacology , PC12 Cells , Prohibitins , Rats , Transcription, Genetic/drug effects
...