Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Clin Drug Investig ; 36(5): 341-56, 2016 May.
Article in English | MEDLINE | ID: mdl-27021968

ABSTRACT

Lisdexamfetamine dimesylate (LDX) is a long-acting d-amphetamine prodrug used to treat attention-deficit/hyperactivity disorder (ADHD) in children, adolescents and adults. LDX is hydrolysed in the blood to yield d-amphetamine, and the pharmacokinetic profile of d-amphetamine following oral administration of LDX has a lower maximum plasma concentration (Cmax), extended time to Cmax (Tmax) and lower inter- and intra-individual variability in exposure compared with the pharmacokinetic profile of an equivalent dose of immediate-release (IR) d-amphetamine. The therapeutic action of LDX extends to at least 13 h post-dose in children and 14 h post-dose in adults, longer than that reported for any other long-acting formulation. Drug-liking scores for LDX are lower than for an equivalent dose of IR d-amphetamine, which may result from the reduced euphorigenic potential associated with its pharmacokinetic profile. These pharmacokinetic and pharmacodynamic characteristics of LDX may be beneficial in the management of symptoms in children, adolescents and adults with ADHD.


Subject(s)
Amphetamine/administration & dosage , Central Nervous System Stimulants/administration & dosage , Drug Delivery Systems/methods , Lisdexamfetamine Dimesylate/administration & dosage , Prodrugs/administration & dosage , Administration, Oral , Adolescent , Adult , Amphetamine/blood , Amphetamine/chemistry , Attention Deficit Disorder with Hyperactivity/blood , Attention Deficit Disorder with Hyperactivity/drug therapy , Central Nervous System Stimulants/blood , Central Nervous System Stimulants/chemistry , Child , Humans , Lisdexamfetamine Dimesylate/blood , Lisdexamfetamine Dimesylate/chemistry , Male , Prodrugs/chemistry , Prodrugs/metabolism , Treatment Outcome
2.
Neuropsychiatr Dis Treat ; 10: 2275-80, 2014.
Article in English | MEDLINE | ID: mdl-25489246

ABSTRACT

Lisdexamfetamine dimesylate (LDX) is approved as a once-daily treatment for attention-deficit/hyperactivity disorder in children, adolescents, and adults in some countries. LDX is a prodrug comprising d-amphetamine covalently linked to l-lysine via a peptide bond. Following oral administration, LDX is rapidly taken up from the small intestine by active carrier-mediated transport, probably via peptide transporter 1. Enzymatic hydrolysis of the peptide bond to release d-amphetamine has previously been shown to occur in human red blood cells but not in several other tissues. Here, we report that LDX hydrolytic activity resides in human red blood cell lysate and cytosolic extract but not in the membrane fraction. Among several inhibitors tested, a protease inhibitor cocktail, bestatin, and ethylenediaminetetra-acetic acid each potently inhibited d-amphetamine production from LDX in cytosolic extract. These results suggest that an aminopeptidase is responsible for hydrolytic cleavage of the LDX peptide bond, although purified recombinant aminopeptidase B was not able to release d-amphetamine from LDX in vitro. The demonstration that aminopeptidase-like activity in red blood cell cytosol is responsible for the hydrolysis of LDX extends our understanding of the smooth and consistent systemic delivery of d-amphetamine by LDX and the long daily duration of efficacy of the drug in relieving the symptoms of attention-deficit/hyperactivity disorder.

3.
Neuropharmacology ; 87: 41-50, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24594478

ABSTRACT

Lisdexamfetamine dimesylate (LDX) is a novel pro-drug of d-amphetamine that is currently used for the treatment of attention-deficit/hyperactivity disorder in children aged ≥ 6 years and adults. LDX is enzymatically cleaved to form d-amphetamine following contact with red blood cells, which reduces the rate of appearance and magnitude of d-amphetamine concentration in the blood and hence the brain when compared with immediate-release d-amphetamine at equimolar doses. Thus, the increase of striatal dopamine efflux and subsequent increase of locomotor activity following d-amphetamine is less prominent and slower to attain maximal effect following an equimolar dose of LDX. Furthermore, unlike d-amphetamine, the pharmacodynamic effects of LDX are independent of the route of administration underlining the requirement to be hydrolyzed by contact with red blood cells. It is conceivable that these pharmacokinetic and pharmacodynamic differences may impact the psychostimulant properties of LDX in the clinic. This article reviews the preclinical pharmacokinetics, pharmacology, and toxicology of LDX. This article is part of the Special Issue entitled 'CNS Stimulants'.


Subject(s)
Central Nervous System Stimulants/pharmacology , Central Nervous System Stimulants/pharmacokinetics , Dextroamphetamine/pharmacology , Dextroamphetamine/pharmacokinetics , Prodrugs/pharmacology , Prodrugs/pharmacokinetics , Animals , Brain/drug effects , Brain/metabolism , Central Nervous System Stimulants/toxicity , Dextroamphetamine/toxicity , Humans , Lisdexamfetamine Dimesylate , Prodrugs/toxicity
4.
J Drug Assess ; 2(1): 17-20, 2013.
Article in English | MEDLINE | ID: mdl-27536433

ABSTRACT

OBJECTIVES: Lisdexamfetamine dimesylate (LDX), a long-acting pro-drug psychostimulant, requires conversion to d-amphetamine for therapeutic activity. Conversion of LDX to d-amphetamine occurs primarily in the blood, specifically red blood cells (RBCs). These in vitro studies examine potential conversion in blood-containing pathologically deformed RBCs. METHODS: Fresh blood samples from two human male donors with sickle cell disease and two healthy control donors were incubated for up to 4 h with LDX (1 µg/mL) at 37°C. LDX and d-amphetamine were measured by a validated liquid chromatographic mass spectrometric (LC/MS/MS) method. RESULTS: In incubations of blood from the two donors with sickle cell disease, LDX concentrations declined over time such that 14.1% and 15.3% of initial LDX remained after 4 h. Similarly, in incubations of blood from two healthy donors, LDX concentrations declined over time with 13.1% and 10.5% of initial LDX remaining. Half-life of LDX was 1.30 and 1.36 h for the donors with sickle cell disease and 1.15 and 1.13 h for the healthy donors. Concurrent with the decrease in LDX concentrations, the d-amphetamine concentrations rose in a similar fashion in samples from healthy controls and sickle cell donors. d-Amphetamine levels detected at 4 h with LC/MS/MS were 297.0 ng/mL and 324.3 ng/mL in the two healthy donors and 304.5 ng/mL and 286.6 ng/mL in the two sickle cell donors. CONCLUSIONS: While the current findings are derived from in vitro investigations on a small number of samples and the applicability of this in vitro experimental system to in vivo function has not been established, biotransformation of LDX and the resulting delivery of active d-amphetamine from LDX are likely to be similar in individuals with or without sickle cell disease.

5.
Ren Fail ; 34(3): 263-70, 2012.
Article in English | MEDLINE | ID: mdl-22250993

ABSTRACT

The effectiveness of phosphate binders can be assessed by evaluating urinary phosphorus excretion in healthy volunteers, which indicates the ability of the phosphate binder to reduce gastrointestinal phosphate absorption. Healthy volunteers were enrolled into one of five separate randomized trials; four were open label and one double blind. Following a screening period of ≤28 days, participants received differing tablets containing lanthanum carbonate [LC, 3000 mg/day of elemental lanthanum (in one study other doses were also used)]. Participants received a standardized phosphate diet and remained in the relevant study center throughout the duration of each treatment period. The end point in all studies was the reduction in urinary phosphorus excretion. Reductions in mean 24-h urinary phosphorus excretion in volunteers receiving a lanthanum dose of 3000 mg/day were between 236 and 468 mg/day over the five separate studies. These data in healthy volunteers can be used to estimate the amount of reduction of dietary phosphate absorption by LC. The reduction in 24-h urinary phosphorus excretion per tablet was compared with published data on other phosphate binders. Although there are limitations, evidence suggests that LC is a very effective phosphate binder in terms of binding per tablet.


Subject(s)
Lanthanum/administration & dosage , Phosphorus/urine , Administration, Oral , Adolescent , Adult , Cross-Over Studies , Dose-Response Relationship, Drug , Double-Blind Method , Female , Humans , Hyperphosphatemia/chemically induced , Hyperphosphatemia/urine , Kidney Failure, Chronic/diet therapy , Kidney Failure, Chronic/urine , Male , Phosphorus Compounds/metabolism , Phosphorus, Dietary/pharmacokinetics , Reference Values , Single-Blind Method , Young Adult
6.
Neuropsychiatr Dis Treat ; 6: 317-27, 2010 Jun 24.
Article in English | MEDLINE | ID: mdl-20628632

ABSTRACT

These studies investigated the absorption and metabolic conversion of lisdexamfetamine dimesylate (LDX), a prodrug stimulant that requires conversion to d-amphetamine for activity. Oral absorption of LDX was assessed in rat portal and jugular blood, and perfusion of LDX into isolated intestinal segments of anesthetized rats was used to assess regional absorption. Carrier-mediated transport of LDX was investigated in Caco-2 cells and Chinese hamster ovary (CHO) cells expressing human peptide transporter-1 (PEPT1). LDX metabolism was studied in rat and human tissue homogenates and human blood fractions. LDX was approximately10-fold higher in portal blood versus systemic blood. LDX and d-amphetamine were detected in blood following perfusion of the rat small intestine but not the colon. Transport of LDX in Caco-2 cells had permeability apparently similar to cephalexin and was reduced with concurrent PEPT1 inhibitor. Affinity for PEPT1 was also demonstrated in PEPT1-transfected CHO cells. LDX metabolism occurred primarily in whole blood (rat and human), only with red blood cells. Slow hydrolysis in liver and kidney homogenates was probably due to residual blood. The carrier-mediated absorption of intact LDX, likely by the high-capacity PEPT1 transporter, and subsequent metabolism to d-amphetamine in a high-capacity system in blood (ie, red blood cells) may contribute to the consistent, reproducible pharmacokinetic profile of LDX.

7.
Clin Drug Investig ; 28(12): 745-55, 2008.
Article in English | MEDLINE | ID: mdl-18991468

ABSTRACT

BACKGROUND AND OBJECTIVES: Attention-deficit/hyperactivity disorder (ADHD) in children often persists into adulthood and is potentially associated with significant social and occupational impairments. It is important to understand the effects of pharmacological treatments of ADHD in adults. This study aimed to assess the absorption, metabolism and elimination of lisdexamfetamine dimesylate in normal, healthy adult subjects following a single oral dose. A secondary objective was to assess the safety and tolerability of treatment. METHODS: In an open-label, single-centre study, six healthy adult volunteers aged 22-52 years received a single oral 70 mg dose of (14)C-radiolabelled lisdexamfetamine dimesylate in solution following a 10-hour fast. Blood samples drawn pre-dose and at time points up to 120 hours post-dose were used for plasma pharmacokinetic analysis of the active d-amphetamine and the intact parent compound lisdexamfetamine dimesylate. Recovery of radioactivity was determined by liquid scintillation counting of blood samples (whole blood and plasma), urine samples and faecal samples collected pre-dose and at designated time points up to 120 hours post-dose. Urine samples were also analysed for the presence of amphetamine-derived metabolites. Safety was assessed by adverse event reporting, changes in physical findings, vital sign measurements, ECG measurements, and clinical laboratory test results. RESULTS: For intact lisdexamfetamine dimesylate, the median time to reach maximum plasma drug concentration (t(max)) was 1.00 hour, and the mean maximum plasma drug concentration (C(max)) was 58.2 +/- 28.1 ng/mL. Intact lisdexamfetamine dimesylate exhibited modest systemic exposure (area under the drug concentration-time curve from time 0 to infinity [AUC(infinity)] 67.04 +/- 18.94 ng . h/mL), and rapid elimination (mean apparent terminal elimination half-life [t((1/2)beta)] 0.47 hours). For d-amphetamine, the median t(max) was 3.00 hours, and the mean C(max) was 80.3 +/- 11.8 ng/mL. The AUC(infinity) of d-amphetamine was 1342 +/- 216.9 ng . h/mL, and elimination occurred as a first-order process. The t((1/2)beta) of d-amphetamine was 10.39 hours. Peaks consistent with amphetamine and hippuric acid were identified in urine samples by high-performance liquid chromatography radioactive profiling. Relative to dose administered, 41.5% was recovered in urine as d-amphetamine, 24.8% as hippuric acid and 2.2% as intact lisdexamfetamine dimesylate. Less than 0.3% of the administered dose was recovered in the faeces. During the 0- to 48-hour urine samples, no unexpected adverse events or clinically significant laboratory, ECG or physical examination findings related to the study medication were observed. CONCLUSIONS: Following a single 70 mg oral dose, lisdexamfetamine dimesylate was quickly absorbed, extensively metabolized to d-amphetamine and its derivatives, and rapidly eliminated. Systemic exposure to d-amphetamine was approximately 20-fold higher than systemic exposure to intact lisdexamfetamine dimesylate in healthy adults. Lisdexamfetamine dimesylate, administered as a single 70 mg dose, was generally well tolerated in this study.


Subject(s)
Dextroamphetamine/pharmacokinetics , Adult , Dextroamphetamine/adverse effects , Female , Humans , Lisdexamfetamine Dimesylate , Male , Middle Aged , Tissue Distribution
8.
Clin Pharmacokinet ; 47(9): 553-63, 2008.
Article in English | MEDLINE | ID: mdl-18698878

ABSTRACT

Lanthanum carbonate is considered to be the most potent of a new generation of noncalcium phosphate binders used to treat hyperphosphataemia in chronic kidney disease (CKD), a condition associated with progressive bone and cardiovascular pathology and a markedly elevated risk of death. Its phosphate-binding action involves ionic binding and precipitation of insoluble complexes within the lumen of the intestine, thereby preventing absorption of dietary phosphate. While pharmacokinetics have little relevance to the efficacy of lanthanum carbonate, they are of fundamental importance when it comes to evaluating safety. When administered as lanthanum carbonate, the oral bioavailability of lanthanum is low (approximately 0.001%). The small absorbed fraction is excreted predominantly in bile, with less than 2% being eliminated by the kidneys. Predictably, therefore, plasma exposure and pharmacokinetics have been shown to be similar in healthy human volunteers and CKD stage 5 patients. With almost complete plasma protein binding, free lanthanum concentrations in patients at steady state are <3 pg/mL. These properties greatly reduce systemic exposure, tissue deposition and the potential for adverse effects. While lanthanum has a variety of calcium-like actions in vitro, there is little or no evidence that these occur in vivo. This paradox is explained by the very low concentrations of circulating free lanthanum ions, which are many orders of magnitude lower than reported effect concentrations in vitro. Safety pharmacology and toxicology evaluations have failed to reveal any significant calcium-like actions in vivo, despite inclusion of high intravenous doses in some cases.Lanthanum carbonate has a low propensity to cause systemic drug interactions due to its poor absorption. However, the higher concentrations present in the gastrointestinal tract can form chelates with some drugs, such as fluoroquinolones, and reduce their absorption. The improved understanding of the pharmacokinetics of lanthanum that has emerged in recent years has helped to explain why the myriad of calcium-like effects described in vitro for lanthanum have little if any relevance in vivo. The pharmacokinetic investigations of lanthanum carbonate formed an important part of the stringent premarketing safety assessment process and have been influential in reassuring both regulators and physicians that the agent can be used safely and effectively in this vulnerable dialysis population.


Subject(s)
Hyperphosphatemia/drug therapy , Lanthanum/pharmacokinetics , Phosphates/metabolism , Animals , Biological Availability , Clinical Trials as Topic , Humans , Hyperphosphatemia/etiology , Kidney Failure, Chronic/complications , Kidney Failure, Chronic/drug therapy , Lanthanum/pharmacology , Tissue Distribution
9.
Clin Pharmacokinet ; 47(8): 543-52, 2008.
Article in English | MEDLINE | ID: mdl-18611063

ABSTRACT

OBJECTIVE: Lanthanum carbonate (Fosrenol) is a non-calcium phosphate binder that controls hyperphosphataemia without increasing calcium intake above guideline targets. The biological fate and bone load of lanthanum were modelled with the aid of a four-compartment kinetic model, analogous to that of calcium. METHODS: The model used data from healthy subjects who received intravenous lanthanum chloride or oral lanthanum carbonate, and bone lanthanum concentration data collected from dialysis patients during three long-term trials (up to 5 years). RESULTS: Infusion of lanthanum chloride or ingestion of lanthanum carbonate led to a rapid rise in plasma lanthanum concentrations, followed by an exponential decrease. Comparison of oral and intravenous exposure confirmed that lanthanum is very poorly absorbed. On a typical intake of lanthanum (3000 mg/day as lanthanum carbonate), the rate of absorption was calculated as 2.2 microg/h, with a urinary excretion rate constant of 0.004-0.01 h(-1). The faecal content of endogenous lanthanum was estimated to be 8- to 20-fold greater than that of urine, compared with a ratio of only about 1 for calcium. The model predicts that upon multiple dosing, plasma lanthanum concentrations rise rapidly to a near plateau and then increase by about 3% per year. However, this small change is obscured by the variability of the study data, which show that a plateau is rapidly attained by 2 weeks and is thereafter maintained for at least 2 years. The initial deposition rate of lanthanum in bone was 1 microg/g/year and, after 10 years of lanthanum carbonate treatment, the model predicts a 7-fold increase in total bone lanthanum (from 10 mg to 69 mg [from 1 microg/g wet weight to 6.6 microg/g wet weight]), with lanthanum cleared after cessation of treatment at 13% per year. The model indicates that lanthanum flow from bone surface to bone interior is much lower than that of calcium. CONCLUSION: Bone is the major reservoir for metals, but bone lanthanum concentrations are predicted to remain low after long-term treatment because of very poor intestinal absorption.


Subject(s)
Bone and Bones/metabolism , Lanthanum/pharmacokinetics , Models, Biological , Administration, Oral , Calcium/pharmacokinetics , Drug Administration Schedule , Humans , Injections, Intravenous , Lanthanum/administration & dosage , Phosphates/metabolism , Randomized Controlled Trials as Topic , Renal Dialysis , Time Factors
10.
Pharmacotherapy ; 27(9): 1253-62, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17723079

ABSTRACT

STUDY OBJECTIVE: To evaluate the single- and multiple-dose pharmacokinetics of an oral extended-release formulation of guanfacine in children and adolescents with a diagnosis of attention-deficit-hyperactivity disorder (ADHD). DESIGN: Phase I-II, open-label, dose-escalation study. SETTING: Clinical study center. PATIENTS: Fourteen children (aged 6-12 yrs) and 14 adolescents (aged 13-17 yrs) with ADHD. INTERVENTION: All patients received guanfacine as a single 2-mg dose on day 1. They received a daily dose of 2 mg on days 9-15, 3 mg on days 16-22, and 4 mg on days 23-29. MEASUREMENTS AND MAIN RESULTS: Blood samples, vital signs, and electrocardiograms (ECGs) were obtained before dosing on day 1 and at intervals over 24 hours, with repeat measurements on days 14 and 28. Guanfacine demonstrated linear pharmacokinetics. Mean plasma concentrations, peak exposure (C(max)), and total or 24-hour exposure (area under the concentration-time curve [AUC](0-infinity) or AUC(0-24), respectively) were as follows in children and adolescents, respectively: after a single 2-mg dose, AUC(0-infinity) was 65.2 +/- 23.9 ng x hour/ml and 47.3 +/- 13.7 ng x hour/ml and C(max) was 2.55 +/- 1.03 ng x ml and 1.69 +/- 0.43 ng/ml after multiple 2-mg doses, AUC(0-24) was 70.0 +/- 28.3 ng x hour/ml and 48.2 +/- 16.1 ng x hour/ml and C(max) was 4.39 +/- 1.66 ng/ml and 2.86 +/- 0.77 ng/ml; and after multiple 4-mg doses, AUC(0-24) was 162 +/- 116 ng x hour/ml and 117 +/- 28.4 ng x hour/ml and C(max) was 10.1 +/- 7.09 ng/ml and 7.01 +/- 1.53 ng/ml. After a single 2-mg dose, half-life was 14.4 +/- 2.39 hours in children and 17.9 +/- 5.77 hours in adolescents. The most frequent treatment-emergent adverse events were somnolence, insomnia, headache, blurred vision, and altered mood. Most were mild to moderate in severity, with the highest frequency associated with the 4-mg doses. Blood pressure, pulse, and ECG reading.hour/ml s were all within normal limits. CONCLUSION: Guanfacine extended-release formulation demonstrated linear pharmacokinetics. Plasma concentrations and concentration-related pharmacokinetic parameters were higher in children than in adolescents. These differences are likely due to heavier body weights in adolescents and young male subjects. No serious adverse events were reported.


Subject(s)
Adrenergic alpha-Agonists/pharmacokinetics , Attention Deficit Disorder with Hyperactivity/drug therapy , Guanfacine/pharmacokinetics , Adolescent , Adrenergic alpha-Agonists/administration & dosage , Adrenergic alpha-Agonists/adverse effects , Age Factors , Area Under Curve , Blood Pressure/drug effects , Body Weight , Child , Delayed-Action Preparations , Dose-Response Relationship, Drug , Electrocardiography , Female , Guanfacine/administration & dosage , Guanfacine/adverse effects , Half-Life , Heart Rate/drug effects , Humans , Male , Sex Factors , Tissue Distribution
11.
Clin Ther ; 29(4): 617-25, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17617285

ABSTRACT

BACKGROUND: Guanfacine is an alpha(2)-adrenoreceptor agonist used to treat children and adults with attention-deficit/hyperactivity disorder. An extended-release formulation of guanfacine is currently under development. OBJECTIVE: The objective of this study was to assess the single-dose pharmacokinetic properties and dose proportionality of guanfacine extended-release (GXR) tablets after oral administration in healthy adults. METHODS: This was a Phase I, randomized, open-label, single-dose, crossover trial of GXR 1-, 2-, and 4-mg tablets in healthy adults. In the lead-in period (period 1), subjects received a single GXR 1-mg tablet, and then were randomized to receive single GXR 2- or 4-mg tablets during 4 separate weekly visits. Vital signs were monitored, blood samples were obtained, and subjects underwent electrocardiography (ECG) before dose administration and at regular intervals over 96 hours. The pharmacokinetic parameters of CmaX, AUC(0-t), and AUC(0-infinity) were determined after each dose of GXR in all subjects. Summary statistics for the concentration-time data were analyzed to assess between-dose linearity. An analysis-of-variance model was constructed to test the concentration-time data for dose proportionality. Tolerability was assessed at each visit through the analysis of standard serology tests; urinalysis/drug screen reports; and physical examination, including height and weight measurements; vital-sign data; and ECG findings. RESULTS: The total study enrollment was 52 subjects, including 28 men (53.8%) and 24 (46.2%) women. The subjects had a mean (SD) age of 32.9 (10.3) years (range, 18-54 years) and a mean (SD) body weight of 73.4 (15.7) kg (range, 49.6-120.0 kg). Forty (76.9%) subjects were Hispanic, 7 (13.5%) were white, and 5 (9.6%) were black. Three subjects were discontinued by the study investigators because of noncompliance with study procedures or use of concomitant medications. Forty-nine subjects completed the study. Mean (SD) values for guanfacine plasma concentrations with GXR 1, 2, and 4 mg, respectively, were 0.98 (0.26), 1.57 (0.51), and 3.58 (1.39) ng/mL for C(max); 29.3 (8.84), 54.5 (17.7), and 119.1 (42.3) ng/mL . h(-1) for AUC(0-t); and 32.4 (8.78), 58.0 (18.9), and 124.1 (45.1) ng/mL . h(-1) for AUC(0-infinity) . Mean (SD) t((1/2)) values were 17.5 (3.8), 16.6 (3.8), and 16.7 (4.90) hours for GXR 1, 2, and 4 mg, respectively. The geometric mean ratios for C(max), AUC(0-t), and AUC(0-infinity) were proportional to dose between GXR 1 and 2 mg, 1 and 4 mg, and 2 and 4 mg, except for the increase in C(max) between GXR 1 and 2 mg. All treatment-emergent adverse events (AEs) were assessed as mild or moderate and resolved without treatment with the exception of headache in 3 subjects and 1 case of lower back discomfort, which resolved with therapy. Left rib pain was reported in 1 subject, but it is unknown if it had resolved, since the subject was lost to followup. No subjects withdrew from participation or were discontinued by the study investigators as a result of AEs. The most common treatment-emergent AE, somnolence, occurred in 33 (63.5%) of 52 subjects. All mean vital-sign measurements and mean ECG parameters remained within normal limits after dosing and no marked changes from baseline measurements were noted. Mean values for all test results of hematology and serum chemistry panels were within the reference range at completion of the study, with no significant changes from baseline. CONCLUSIONS: In these 49 healthy adult subjects, the single-dose pharmacokinetic properties of GXR 1-, 2-, and 4-mg tablets appeared to be statistically linear; that is, increases in mean C(max), AUC(0-t), and AUC(0-infinity) of guanfacine were proportional to dose, with the exception of the increase in mean C(max) between GXR 1 and 2 mg. All doses appeared to be well tolerated, with no serious AEs or withdrawal or discontinuation from study participation due to AEs reported.


Subject(s)
Adrenergic alpha-2 Receptor Agonists , Guanfacine/administration & dosage , Guanfacine/pharmacokinetics , Adolescent , Adult , Analysis of Variance , Area Under Curve , Attention Deficit Disorder with Hyperactivity/drug therapy , Cross-Over Studies , Delayed-Action Preparations , Drug Administration Schedule , Female , Guanfacine/adverse effects , Humans , Male , Middle Aged , Tablets
12.
Toxicol Lett ; 171(1-2): 69-77, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17570622

ABSTRACT

Lanthanum carbonate is a non-calcium-based oral phosphate binder for the control of hyperphosphataemia in patients with chronic kidney disease Stage 5. As part of its pre-clinical safety evaluation, studies were conducted in rats to determine the extent of absorption and routes of excretion. Following oral gavage of a single 1500 mg/kg dose, the peak plasma lanthanum concentration was 1.04+/-0.31 ng/mL, 8 h post-dose. Lanthanum was almost completely bound to plasma proteins (>99.7%). Within 24h of administration of a single oral dose, 97.8+/-2.84% of the lanthanum was recovered in the faeces of rats. Comparing plasma exposure after oral and intravenous administration of lanthanum yielded an absolute oral bioavailability of 0.0007%. Following intravenous administration of lanthanum chloride (0.3 mg/kg), 74.1+/-5.82% of the dose (96.9+/-0.50% of recovered lanthanum) was excreted in faeces in 42 days, and in bile-duct cannulated rats, 10.0+/-2.46% of the dose (85.6+/-2.97% of recovered lanthanum) was excreted in bile in 5 days. Renal excretion was negligible, with <2% of the intravenous dose recovered in urine. These studies demonstrate that lanthanum undergoes extremely low intestinal absorption and that absorbed drug is predominantly excreted in the bile.


Subject(s)
Bile/metabolism , Lanthanum/pharmacokinetics , Administration, Oral , Animals , Area Under Curve , Biological Availability , Dogs , Drug Evaluation, Preclinical/methods , Feces/chemistry , Female , Glycoproteins/metabolism , Humans , Injections, Intravenous , Intubation, Gastrointestinal , Lanthanum/administration & dosage , Lanthanum/blood , Lanthanum/pharmacology , Male , Metabolic Clearance Rate/drug effects , Mice , Protein Binding , Rabbits , Rats , Rats, Sprague-Dawley , Serum Albumin/metabolism , Sex Factors , Sodium Chloride/administration & dosage
13.
Curr Med Res Opin ; 23(5): 1067-75, 2007 May.
Article in English | MEDLINE | ID: mdl-17519073

ABSTRACT

OBJECTIVE: To compare the single-dose pharmacokinetics of triple-bead mixed amphetamine salts (MAS), an oral, once-daily, enhanced extended-release amphetamine formulation, with MAS extended release (MAS XR) (Adderall XR) + MAS immediate release (MAS IR) administered 8 h later. METHODS: This was a phase I, randomized, open-label, single-dose, single-center, two-period, crossover study in healthy adult volunteers designed to evaluate the bioavailability of triple-bead MAS over the course of a full day. Subjects were randomized to triple-bead MAS 37.5 mg or MAS XR 25 mg + MAS IR 12.5 mg administered 8 h later (MAS XR + MAS IR). The reference treatment was designed to mimic the clinical practice of providing extended coverage by supplementing a morning dose of MAS XR with a dose of MAS IR 8 h later in order to increase the duration of action. Plasma was assayed for d-amphetamine and l-amphetamine. Treatment-emergent adverse events (TEAEs), vital signs, electrocardiograms (ECGs), and laboratory data were also collected for safety evaluation. RESULTS: Exposure to d- and l-amphetamine was equivalent between triple-bead MAS and MAS XR + MAS IR based on maximum plasma concentration (Cmax) and area under the plasma concentration-time curve from time 0 to infinity (AUC(0-infinity)). For Cmax, least-squares mean ratios comparing triple-bead MAS with MAS XR + MAS IR were 101.0% and 90.9% for d-amphetamine and l-amphetamine, respectively, and for AUC(0-infinity) were 104.4% and 95.3% for d-amphetamine and l-amphetamine, respectively. Median time to maximum observed plasma concentration (Tmax) values for d-amphetamine and l-amphetamine were 8.0 h for triple-bead MAS and 10.0 h for MAS XR + MAS IR. There were no clinically meaningful differences between the study formulations for TEAEs or laboratory values. One subject experienced an ECG abnormality (asymptomatic premature ventricular contractions) leading to early termination from the study. CONCLUSIONS: In healthy adults, the exposure observed with triple-bead MAS 37.5 mg was bioequivalent to MAS XR 25 mg supplemented by MAS IR 12.5 mg administered 8 h later. These data demonstrate that a single morning dose of triple-bead MAS provides equivalent plasma concentrations to those observed with a dose-augmentation strategy of MAS XR in the morning followed by MAS IR in the afternoon, while minimizing peak-to-trough fluctuations. Triple-bead MAS was also generally well-tolerated in this study.


Subject(s)
Amphetamines/pharmacokinetics , Attention Deficit Disorder with Hyperactivity/drug therapy , Administration, Oral , Adult , Amphetamines/administration & dosage , Amphetamines/adverse effects , Biological Availability , Central Nervous System Agents/administration & dosage , Central Nervous System Agents/adverse effects , Central Nervous System Agents/pharmacokinetics , Cross-Over Studies , Delayed-Action Preparations/administration & dosage , Delayed-Action Preparations/adverse effects , Delayed-Action Preparations/pharmacokinetics , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Combinations , Drug Therapy, Combination , Female , Humans , Male , Middle Aged , Placebos , Salts/administration & dosage , Salts/adverse effects , Salts/pharmacokinetics , Therapeutic Equivalency
14.
J Clin Pharmacol ; 46(7): 738-46, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16809799

ABSTRACT

Lanthanum carbonate [La2(CO3)3] is a noncalcium, non-aluminum phosphate binder indicated for hyperphosphatemia treatment in end-stage renal disease. A randomized, open-label, parallel-group, phase I study was conducted to determine absolute bioavailability and investigate excretory routes for systemic lanthanum in healthy subjects. Twenty-four male subjects were randomized to a single lanthanum chloride (LaCl3) intravenous infusion (120 microg elemental lanthanum over a 4-hour period), a single 1-g oral dose [chewable La2(CO3)3 tablets; 4 x 250 mg elemental lanthanum], or no treatment (control). Serial blood, urine, and fecal samples were collected for 7 days postdosing. The absolute bioavailability of lanthanum [administered as La2(CO3)3] was extremely low (0.00127% +/- 0.00080%), with individual values in the range of 0.00015% to 0.00224%. Renal clearance was negligible following oral administration (1.36 +/- 1.43 mL/min). Intravenous administration confirmed low renal clearance (0.95 +/- 0.60 mL/min), just 1.7% of total plasma clearance. Fecal lanthanum excretion was not quantifiable after intravenous administration owing to high and variable background fecal lanthanum and constraints on the size of the intravenous dose. These findings demonstrate that lanthanum absorption from the intestinal tract into the systemic circulation is extremely low and that absorbed drug is cleared predominantly by nonrenal mechanisms.


Subject(s)
Lanthanum/pharmacokinetics , Administration, Oral , Adult , Biological Availability , Humans , Infusions, Intravenous , Intestinal Absorption , Lanthanum/administration & dosage , Male , Reference Values
15.
Med Sci Monit ; 9(8): PI99-104, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12942041

ABSTRACT

BACKGROUND: Marimastat is a potent inhibitor of matrix metalloproteinases and in preclinical studies enhances the anti-tumor activity of certain chemotherapeutics. We performed a phase I clinical evaluation of the combination of oral marimastat and intravenous paclitaxel, to determine if these drugs could be co-administered safely, and to determine whether marimastat alters paclitaxel pharmacokinetics. MATERIAL/METHODS: Marimastat was administered twice daily and paclitaxel as a three hour infusion every three weeks. Doses of both marimastat and paclitaxel were escalated in cohorts of patients up to maximal doses of 10 mg for marimastat and 175 mg/m2 for paclitaxel. Paclitaxel plasma pharmacokinetic parameters were assessed in the absence (cycle 1) and presence (cycle 2) of marimastat. Trough marimastat plasma levels were evaluated during cycle 2. RESULTS: A total of 19 patients were treated at three different dose levels. There were no dose-limiting toxicities during the first cycle of therapy, resulting in dose escalation up to the planned maximal dose for each drug. Neutropenia was the most common significant toxicity at the highest dose level, with grade 3 or higher neutropenia occurring in 38% of patients. There were no complete or partial responses. Pharmacokinetic analyses indicate that marimastat does not alter paclitaxel clearance. At the 10 mg dose, the mean trough marimastat level was 14.8 Kg/L. CONCLUSIONS: Marimastat and paclitaxel can be co-administered safely at doses equivalent to those recommended for single-agent administration. Additional studies are necessary to determine whether this combination is more effective in controlling tumor progression than paclitaxel alone.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Antineoplastic Combined Chemotherapy Protocols , Enzyme Inhibitors/therapeutic use , Hydroxamic Acids/therapeutic use , Neoplasms/drug therapy , Paclitaxel/therapeutic use , Adult , Aged , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/pharmacokinetics , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacokinetics , Female , Humans , Hydroxamic Acids/chemistry , Hydroxamic Acids/pharmacokinetics , Male , Middle Aged , Molecular Structure , Paclitaxel/chemistry , Paclitaxel/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...