Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Clin Pharmacokinet ; 62(12): 1755-1763, 2023 12.
Article in English | MEDLINE | ID: mdl-37882999

ABSTRACT

BACKGROUND: Ulotaront is a novel psychotropic agent with agonist activity at trace amine-associated receptor 1 (TAAR1) and 5-hydroxytryptamine type 1A (5-HT1A) receptors in phase III clinical development for the treatment of schizophrenia. OBJECTIVE: This study aimed to investigate the effect of paroxetine, a strong cytochrome P450 (CYP) 2D6 inhibitor, on ulotaront pharmacokinetics (PK) in healthy volunteers. METHODS: Subjects received a single oral dose of 25 mg ulotaront on Day 1 and an oral dose of 20 mg paroxetine once daily from Days 5 to 10 to achieve steady-state plasma paroxetine levels. On Day 11, subjects received another single oral dose of 25 mg ulotaront, with continued daily oral dosing of 20 mg paroxetine from Days 11 to 14. All 24 subjects were CYP2D6 normal metabolizers. RESULTS: Coadministration of paroxetine increased ulotaront maximum observed plasma concentration (Cmax) and area under the plasma concentration-time curve from time zero to infinity (AUC∞) by 31% and 72%, respectively, and decreased ulotaront apparent clearance (CL/F) by approximately 42%. While coadministration of paroxetine increased AUC∞ of active but minor metabolite SEP-363854 by 32%, it had no effect on SEP-363854 Cmax, or on SEP-363854 to the ulotaront AUC from time zero to the last quantifiable concentration (AUClast) ratio. Based on the acceptable adverse event profile of ulotaront across previous phase II studies, the increase in ulotaront exposure is unlikely to be clinically meaningful. CONCLUSIONS: Weak drug-drug interactions were observed between ulotaront and the strong CYP2D6 inhibitor paroxetine; however, dose adjustment as a precondition when ulotaront is coadministered with strong CYP2D6 inhibitors or administered to CYP2D6 poor metabolizers should not be necessary.


Subject(s)
Cytochrome P-450 CYP2D6 , Paroxetine , Humans , Cytochrome P-450 CYP2D6/metabolism , Paroxetine/adverse effects , Healthy Volunteers , Cytochrome P-450 CYP2D6 Inhibitors/pharmacokinetics , Drug Interactions , Enzyme Inhibitors , Area Under Curve
2.
Neurol Ther ; 10(2): 693-709, 2021 Dec.
Article in English | MEDLINE | ID: mdl-33991326

ABSTRACT

INTRODUCTION: In a pivotal study, apomorphine sublingual film (APL; KYNMOBI®) was an effective and generally well-tolerated on-demand treatment of "OFF" episodes in patients with Parkinson's disease (PD), approved across the dose range of 10-30 mg. Pharmacokinetics and comparative bioavailability of APL and two subcutaneous (SC) apomorphine formulations (SC-APO [APOKYN®] and SC-APO-GO [APO-go® PEN]) were evaluated in a randomized, three-way crossover, open-label study (NCT03292016). METHODS: Patients with PD and "OFF" episodes received an open-label randomized sequence of single doses of SC-APO and SC-APO-GO at the currently prescribed dose (2/3/4/5 mg) and APL doses with similar plasma exposure (15/20/25/30 mg) with ≥ 1-day washout between formulations. Plasma pharmacokinetics of apomorphine and apomorphine sulfate (major inactive metabolite) were measured 0-6 h postdose. RESULTS: Median time to maximum plasma concentration (tmax) of apomorphine was 0.63-0.75 h for APL and 0.25-0.38 h for SC-APO and SC-APO-GO. Geometric mean maximum plasma concentration (Cmax) of apomorphine was 4.31-11.2 ng/ml across APL doses and was generally lower compared with SC apomorphine formulations within dose groups. Area under the concentration-time curve from time 0 to infinity (AUC∞) was similar across apomorphine formulations within most dose groups. Relative bioavailability of APL was ~ 17% of SC apomorphine by AUC∞; SC-APO and SC-APO-GO had similar bioavailability (98% and 83% by AUC∞ and Cmax, respectively). Apomorphine sulfate exposure was ~ three-fold higher for APL versus SC-APO and SC-APO-GO by AUC∞ and Cmax. CONCLUSION: In patients with PD and "OFF" episodes, APL demonstrated lower Cmax and relative bioavailability but similar exposures (AUCs) versus SC apomorphine within the approved dose range. TRIAL REGISTRATION: ClinicalTrials.gov, NCT03292016.

3.
Clin Transl Sci ; 14(4): 1464-1475, 2021 07.
Article in English | MEDLINE | ID: mdl-33650272

ABSTRACT

Apomorphine is an on-demand treatment of "OFF" episodes in patients with Parkinson's disease (PD). A joint parent-metabolite population pharmacokinetic (PK) model characterized apomorphine and apomorphine-sulfate following administration of apomorphine sublingual film (APL) and two formulations of subcutaneous apomorphine. Overall, 2485 samples from 87 healthy subjects and 71 patients with PD and "OFF" episodes were analyzed using nonlinear mixed-effects modeling. Apomorphine PK was adequately described by a two-compartment model with first-order transit absorption via both routes of administration and first-order metabolism to apomorphine-sulfate with one-compartment disposition and first-order elimination. Bioavailability of apomorphine sublingual film was ~ 18% relative to subcutaneous apomorphine. Among covariates tested, only body weight had a large effect on apomorphine exposure (maximum plasma concentration and area under the concentration-time curve [AUC0-∞ ]), with greater weight resulting in lower exposure. Model-predicted apomorphine exposure was similar between apomorphine sublingual film 30 mg and subcutaneous apomorphine 5 mg (median AUC0-24 , 66.7 ng•h/mL, geometric mean ratio of 0.99; 90% confidence interval [CI], 0.96-1.03) and was comparable between apomorphine sublingual film 35 mg and subcutaneous apomorphine 6 mg (median AUC0-24 , 75.4 and 80.0 ng•h/mL, respectively; geometric mean ratio of 0.94; 90% CI, 0.90-0.97) administered every 2 h for a maximum of 5 doses per day. In a typical patient with PD, predicted apomorphine exposure increased with increasing doses of apomorphine sublingual film; however, the increase was less than dose proportional. Similar apomorphine exposure was predicted in patients with mild renal impairment versus normal renal function. PK properties of apomorphine sublingual film support its administration for a wide range of patients with PD and "OFF" episodes, regardless of demographic and clinical characteristics.


Subject(s)
Antiparkinson Agents/pharmacokinetics , Apomorphine/pharmacokinetics , Models, Biological , Parkinson Disease/drug therapy , Administration, Sublingual , Adult , Aged , Aged, 80 and over , Antiparkinson Agents/administration & dosage , Apomorphine/administration & dosage , Area Under Curve , Biological Availability , Biological Variation, Population , Clinical Trials as Topic , Female , Healthy Volunteers , Humans , Injections, Subcutaneous , Male , Middle Aged , Oral Mucosal Absorption , Parkinson Disease/blood , Young Adult
4.
Clin Ther ; 38(1): 4-15, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26730454

ABSTRACT

PURPOSE: Characterization of dose-response relationships for psychotropic agents may be difficult to determine based on results of individual clinical studies, particularly those with a flexible dose design. The goal of this pharmacometric analysis was to characterize the dose-response profile for lurasidone in patients with bipolar depression. METHODS: The statistical modeling and simulation analyses reported here were derived from 2 randomized, 6-week, double-blind, placebo-controlled, flexible-dose studies (20-60 mg/d or 80-120 mg/d of lurasidone as monotherapy or 20-120 mg/d adjunct to lithium or valproate) in patients with bipolar depression. Pooled data included 5245 Montgomery-Åsberg Depression Rating Scale (MADRS) observations from 825 patients who had received lurasidone or placebo treatments, with or without lithium or valproate background medication. FINDINGS: The time course of placebo effect on the MADRS score was adequately described by an exponential asymptotic placebo model. A linear dose-response model best described the effect of lurasidone. The net improvement in MADRS score due to lurasidone treatment (the drug effect) was significant (P < 0.001), and a positive dose response was detected. Net drug effect after 6 weeks of treatment was estimated to be a 6.0-point decrease in MADRS score per 100 mg of lurasidone. Covariate effects (for age and lithium or valproate use) were significant only for placebo effect parameters; thus, no dose adjustment was necessary related to demographic covariates. IMPLICATIONS: This population dose-response modeling analysis indicates that higher doses of lurasidone are likely to produce greater therapeutic effects in patients with bipolar depression. The linear dose response was consistent for both lurasidone monotherapy and adjunctive therapy in patients with bipolar depression. ClinicalTrials.gov identifiers: NCT00868452, NCT00868699.


Subject(s)
Antipsychotic Agents/administration & dosage , Bipolar Disorder/drug therapy , Lurasidone Hydrochloride/administration & dosage , Models, Theoretical , Adult , Antimanic Agents/therapeutic use , Antipsychotic Agents/pharmacokinetics , Computer Simulation , Depressive Disorder , Dose-Response Relationship, Drug , Double-Blind Method , Drug Therapy, Combination , Female , Humans , Lithium Compounds/therapeutic use , Lurasidone Hydrochloride/pharmacokinetics , Male , Middle Aged , Placebo Effect , Psychiatric Status Rating Scales , Valproic Acid/therapeutic use
5.
Clin Ther ; 37(12): 2788-97, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26631428

ABSTRACT

PURPOSE: The aim of this study was to evaluate the pharmacokinetic (PK) profile and tolerability of lurasidone in children and adolescents with a range of psychiatric disorders. METHODS: This multicenter, open-label, single and multiple ascending-dose study of the PK profile of lurasidone (20, 40, 80, 120, and 160 mg/d) enrolled outpatients aged 6 to 17 years with a diagnosis of attention deficit/hyperactivity disorder, bipolar spectrum disorder, or other psychiatric disorder. Serial blood samples were collected for analysis of PK parameters, including Cmax, Tmax, and AUC0-24. FINDINGS: Exposure (Cmax and AUC0-24) to lurasidone and its active metabolites showed linear increases across the entire dose range. Slope estimates (95% CI) across the dose range studied was 0.90 ng · h/mL (0.74-1.06) for AUC0-24 and 0.70 ng/mL (0.52-0.87) for Cmax on day 10 or 12. Lurasidone exposure, after multiple-dose administration in this child and adolescent population, was similar to exposure observed at steady state in adults. The effects of dose on exposure to the 3 active metabolites of lurasidone were linear and similar after the administration of single and multiple doses. Adverse events were qualitatively similar to those reported in adults. Discontinuations due to adverse events were dose related, with doses <120 mg/d being better tolerated than higher doses, especially in younger children. IMPLICATIONS: In this child and adolescent population, exposure parameters for lurasidone and its active metabolites were dose proportional in the range of 20 to 160 mg/d after the administration of single and multiple doses. These results suggest that lurasidone doses <120 mg/d were better tolerated compared with higher doses, especially in younger children. ClinicalTrials.gov identifier: NCT01620060.


Subject(s)
Antipsychotic Agents/adverse effects , Antipsychotic Agents/pharmacokinetics , Attention Deficit Disorder with Hyperactivity/drug therapy , Bipolar Disorder/drug therapy , Lurasidone Hydrochloride/adverse effects , Lurasidone Hydrochloride/pharmacokinetics , Adolescent , Antipsychotic Agents/blood , Antipsychotic Agents/therapeutic use , Child , Humans , Lurasidone Hydrochloride/blood , Lurasidone Hydrochloride/therapeutic use
6.
Drug Metabol Drug Interact ; 29(3): 191-202, 2014.
Article in English | MEDLINE | ID: mdl-24825095

ABSTRACT

BACKGROUND: To evaluate potential drug-drug interactions with the atypical antipsychotic lurasidone. METHODS: Seven phase I studies were conducted to investigate the effects of repeated dosing of ketoconazole, diltiazem, rifampin, or lithium on the pharmacokinetics (PK) of single oral doses of lurasidone, or the effects of repeated dosing of lurasidone on the PK of digoxin, midazolam, or the oral contraceptive norgestimate/ethinyl estradiol. Two 6-week, phase III studies included evaluation of the potential for interaction between lurasidone and lithium or valproate. Maximum serum or plasma concentration (Cmax) and area under the concentration-time curve (AUC) were calculated. RESULTS: Concomitant ketoconazole administration resulted in a 6.8-fold increase in lurasidone Cmax and a 9.3-fold increase in lurasidone AUC; concomitant diltiazem administration resulted in 2.1- and 2.2-fold increases, respectively. Rifampin decreased lurasidone Cmax and AUC (one-seventh and one-fifth of lurasidone alone, respectively). Steady-state dosing with lurasidone increased Cmax and AUC0-24 (AUC from time 0 to 24 h postdose) of digoxin by 9% and 13%, respectively, and of midazolam by 21% and 44%, respectively. There were no significant interactions between lurasidone and lithium, valproate, ethinyl estradiol, or norelgestromin (the major active metabolite of norgestimate). CONCLUSIONS: Lurasidone PK is altered by strong cytochrome P450 (CYP) 3A4 inhibitors or inducers, and coadministration is contraindicated; whereas moderate CYP3A4 inhibitors have less effect, and lurasidone dosage restrictions are recommended. No dose adjustment for lurasidone is needed when administered with lithium or valproate. Dose adjustment is not required for lithium, valproate, digoxin (a P-glycoprotein substrate), or midazolam or oral contraceptives (CYP3A4 substrates) when coadministered with lurasidone.


Subject(s)
Antipsychotic Agents/pharmacokinetics , Isoindoles/pharmacokinetics , Thiazoles/pharmacokinetics , Antipsychotic Agents/adverse effects , Area Under Curve , Clinical Trials, Phase I as Topic , Cytochrome P-450 CYP3A/pharmacology , Cytochrome P-450 CYP3A Inducers/pharmacology , Cytochrome P-450 CYP3A Inhibitors/pharmacology , Diltiazem/pharmacology , Drug Interactions , Drug Therapy, Combination , Humans , Isoindoles/adverse effects , Ketoconazole/pharmacology , Lithium Compounds/pharmacology , Lurasidone Hydrochloride , Rifampin/pharmacology , Thiazoles/adverse effects
7.
Hum Psychopharmacol ; 28(5): 495-505, 2013 Sep.
Article in English | MEDLINE | ID: mdl-24014143

ABSTRACT

OBJECTIVE: This study aimed to investigate the effect of prandial status and caloric and fat composition of meals on the pharmacokinetics of lurasidone. METHODS: Two randomized, open-label, crossover studies were conducted in clinically stable adults with schizophrenia or schizoaffective disorder. Study 1 (n = 16) evaluated the effect of fasting and three meal types (100 kcal/medium fat, 200 kcal/medium fat, and 800-1000 kcal/high fat), and Study 2 (n = 26) evaluated the effect of fasting and five meal types (350 kcal/high fat, 500 kcal/low fat, 500 kcal/high fat, 800-1000 kcal/low fat, and 800-1000 kcal/high fat) on the bioavailability of lurasidone. Subjects received lurasidone 120 mg once daily. Maximum serum concentration (Cmax ) and area under the serum concentration-time curve over the dosing interval (AUC0-tau ) were determined on Day 5 for each meal type. RESULTS: In Study 1, the geometric mean Cmax in the fasted state was 56.7 ng/mL compared with 123.0 ng/mL for the 800- to 1000-kcal meal; mean AUC0-tau was 360.0 versus 752.4 ng·h/mL (both p < 0.001). Lurasidone exposure following meals containing 100 and 200 kcal was substantially lower than with meals containing 800-1000 kcal. In Study 2, the geometric mean Cmax was 52.9 ng/mL in the fasted state, 161 ng/mL for the 350-kcal/high-fat meal, 135 ng/mL for the 500-kcal/high-fat meal, and 131 ng/mL for the 800- to 1000-kcal/high-fat meal; mean AUC0-tau was 390, 743, 727, and 769 ng·h/mL, respectively. For all comparisons, the 90% confidence interval of the fed to fasted ratios indicated nonequivalence. Lurasidone exposure was similar following meals containing 350-1000 kcal and was independent of fat content. CONCLUSION: Lurasidone should be administered with food-at least 350 kcal-to ensure maximum exposure.


Subject(s)
Food-Drug Interactions/physiology , Isoindoles/administration & dosage , Isoindoles/blood , Schizophrenia/blood , Schizophrenia/drug therapy , Thiazoles/administration & dosage , Thiazoles/blood , Adult , Cross-Over Studies , Female , Humans , Lurasidone Hydrochloride , Male , Middle Aged
8.
HIV Clin Trials ; 9(2): 73-82, 2008.
Article in English | MEDLINE | ID: mdl-18474492

ABSTRACT

PURPOSE: To evaluate the safety and antiviral activity of once-daily (qd) enfuvirtide (ENF) compared with twice daily (bid) ENF. METHOD: T20-401 was a phase 2, open-label, randomized, 48-week pilot study comparing ENF 180 mg qd versus ENF 90 mg bid, added to an optimized background (OB) regimen. Patients were randomized 1:1 to receive ENF 180 mg qd given as two 90-mg injections (n = 30) or one 90-mg injection bid (n = 31), plus OB. The primary efficacy endpoint was the proportion of patients achieving a HIV-1 RNA viral load <400 copies/mL. Adherence, pharmacokinetics, safety, and tolerability parameters, including injection site reactions (ISRs), were compared between treatment arms. RESULTS: At Week 48, 23.3% of patients on once daily versus 22.6% on twice daily (p = .969) reached <400 copies/mL and 13.3% and 22.6% (p = .323), respectively, reached <50 copies/mL. The proportion reporting 1 adverse event or ISRs was comparable between arms, despite an increased incidence of grade 4 erythema (13% vs. 0%), induration (33% vs. 12%), and ecchymosis (7% vs. 0%) on twice daily versus once daily. ENF adherence (95% of prescribed doses) was higher on once daily (80.0%) versus twice daily (58.1%). CONCLUSION: The antiviral efficacy, safety, and tolerability of 180 mg ENF qd appeared comparable with that of 90 mg ENF bid at Week 48, although the study was not powered to demonstrate the noninferiority of once daily versus twice daily.


Subject(s)
HIV Envelope Protein gp41/administration & dosage , HIV Envelope Protein gp41/adverse effects , HIV Fusion Inhibitors/administration & dosage , HIV Fusion Inhibitors/adverse effects , HIV Infections/drug therapy , Peptide Fragments/administration & dosage , Peptide Fragments/adverse effects , Adult , Enfuvirtide , Female , HIV Envelope Protein gp41/pharmacokinetics , HIV Fusion Inhibitors/pharmacokinetics , HIV Infections/virology , HIV-1/drug effects , Humans , Male , Middle Aged , Patient Compliance , Peptide Fragments/pharmacokinetics , Pilot Projects , RNA, Viral/blood , Treatment Outcome , Viral Load
9.
J Acquir Immune Defic Syndr ; 47(3): 342-5, 2008 Mar 01.
Article in English | MEDLINE | ID: mdl-18091608

ABSTRACT

OBJECTIVE: The aim of this study was to examine the influence of kidney disease and hemodialysis on the pharmacokinetics ofenfuvirtide. DESIGN: An open-label, multicenter, parallel group study of HIV-1-infected patients with varying degrees of kidney dysfunction. METHODS: A 90-mg dose of enfuvirtide was administered by subcutaneous injection to 3 groups of patients: group A, patients with normal kidney function; group B, patients with chronic kidney disease; and group C, patients with end-stage renal disease (ESRD) requiring dialysis. Patients with ESRD requiring dialysis received the 90-mg dose of enfuvirtide on 2 separate occasions; a dialysis day and a nondialysis day. After each dose, a full 48-hour pharmacokinetic profile was collected and pharmacokinetic parameters were estimated using model-independent techniques. RESULTS: Enfuvirtide area under the curve (AUCinfinity) and maximum observed enfuvirtide plasma concentration (Cmax) for patients with normal kidney function (group A) was 49.6 microg h/mL and 3.79 microg/mL, respectively. Patients with chronic kidney disease (group B) had higher AUCinfinity (80.3 microg h/mL) and Cmax (5.72 microg/mL), which was similar to patients with ESRD (group C) on both nondialysis days (AUCinfinity 71.1 microg h/mL; Cmax 5.34 microg/mL) and dialysis days (AUCinfinity 66.9 microg h/mL; Cmax 6.31 microg/mL). An average of< 13% of enfuvirtide was removed during the dialysis procedure. The incidence of adverse events was comparable for all study groups. CONCLUSION: Enfuvirtide exposure observed in patients with ESRD requiring dialysis or chronic kidney disease was slightly higher than in patients with normal kidney function and similar to historical Cmax and AUC values from studies in patients with normal kidney function. Thus, enfuvirtide does not require dosage adjustment in patients with impaired kidney function.


Subject(s)
HIV Envelope Protein gp41/pharmacokinetics , HIV Infections/drug therapy , HIV-1/drug effects , Kidney Failure, Chronic/physiopathology , Peptide Fragments/pharmacokinetics , Area Under Curve , Dose-Response Relationship, Drug , Enfuvirtide , Female , HIV Envelope Protein gp41/therapeutic use , HIV Fusion Inhibitors/pharmacokinetics , HIV Fusion Inhibitors/therapeutic use , HIV Infections/metabolism , Humans , Kidney/metabolism , Kidney/physiopathology , Kidney Failure, Chronic/therapy , Male , Metabolic Clearance Rate , Peptide Fragments/therapeutic use , Renal Dialysis
10.
Br J Clin Pharmacol ; 63(5): 527-33, 2007 May.
Article in English | MEDLINE | ID: mdl-17324249

ABSTRACT

AIMS: To evaluate the pharmacokinetics of R667, a novel emphysema agent, in patients with moderate to severe emphysema. METHODS: Multiple-dose pharmacokinetics of R667 and its metabolites in emphysematous patients were studied in a multicentre, randomized, single-blind, and placebo-controlled trial. Four groups of 10 patients per group received placebo, 0.2, 0.5, or 1 mg R667 once a day for 14-16 days. On day 14 (+/-1), blood samples were taken at predose and 1, 2, 3, 4, 6, 8, 10, 12, 16 and 24 h after dosing. RESULTS: Pharmacokinetic analysis of the data indicated that the mean steady-state C(max) and AUC(0,tau) of R667 appeared to be dose proportional over the dose range of 0.2-1 mg when administered to emphysematous patients. Mean metabolite to R667 ratios for C(max) or AUC(0,tau) were, in general, similar across the dose range of 0.2-1 mg. CONCLUSIONS: The pharmacokinetics of R667 and its metabolites appeared to be similar for patients with emphysema and healthy volunteers. Multiple-dose administration of 0.2-1 mg of R667 for up to 16 days was well tolerated in patients with emphysema.


Subject(s)
Pulmonary Emphysema/blood , Pyrazoles/blood , Aged , Aged, 80 and over , Body Mass Index , Dose-Response Relationship, Drug , Female , Forced Expiratory Volume , Humans , Male , Middle Aged , Pulmonary Emphysema/physiopathology , Pyrazoles/administration & dosage , Pyrazoles/adverse effects , Single-Blind Method , Vital Capacity
11.
Pharmacotherapy ; 26(12): 1679-86, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17125431

ABSTRACT

STUDY OBJECTIVES: To compare the relative bioavailability of enfuvirtide, a human immunodeficiency virus type 1 (HIV-1) fusion inhibitor, injected with the Biojector 2000 (B2000) needle-free device versus a 27-gauge half-inch needle-syringe; and to assess safety, tolerability, and patient preference for the two devices. DESIGN: Open-label, randomized, two-period crossover bioequivalence evaluation. SETTING: Clinical research center. PATIENTS: Twenty-seven adults with HIV-1 viral loads below 1000 copies/ml. INTERVENTION: Each patient received enfuvirtide 90 mg subcutaneously with the B2000 and with the needle-syringe, with a 1-week washout between treatments. MEASUREMENTS AND MAIN RESULTS: Twenty-six and 27 patients were included in the bioequivalence and safety analyses, respectively. Plasma enfuvirtide concentrations were measured at baseline and at several intervals after each injection. The B2000:needle-syringe ratios of maximum concentration (C(max)), area under the concentration-time curve from time zero extrapolated to infinity (AUC(0-infinity)), and AUC from time zero to tau (dosing interval) (AUC(0-tau)) served as criteria for bioequivalence determination. The two drug delivery systems were considered bioequivalent if the 90% confidence intervals (CIs) for the ratios were within 0.8-1.25. Safety and tolerability were evaluated based on documentation of adverse events, graded laboratory toxicities, and local injection-site reactions. Patient surveys provided feedback on device preference. Ratios of C(max), AUC(0-infinity), and AUC(0-tau) were 0.95 (90% CI 0.84-1.09), 0.99 (90% CI 0.93-1.05), and 0.99 (90% CI 0.93-1.05), respectively. The frequency of injection-site reactions was low, and severity was generally mild for both devices. Survey results showed 18 patients (69%) had a positive overall impression of the B2000 and 14 (54%) felt safer injecting with this device. Overall, 17 patients (65%) preferred the B2000 over the needle-syringe. CONCLUSION: Bioavailability of enfuvirtide with the B2000 and needle-syringe was equivalent based on C(max), AUC(0-tau), and AUC(0-infinity). Safety profiles and injection-site reactions were comparable between the devices, but patients preferred the B2000. Delivery of enfuvirtide with the B2000 is a feasible alternative to standard needle administration and warrants further evaluation.


Subject(s)
HIV Envelope Protein gp41/therapeutic use , HIV Fusion Inhibitors/pharmacokinetics , HIV Fusion Inhibitors/therapeutic use , HIV Infections/drug therapy , Peptide Fragments/pharmacokinetics , Peptide Fragments/therapeutic use , Area Under Curve , Cross-Over Studies , Enfuvirtide , Female , HIV Envelope Protein gp41/administration & dosage , HIV Fusion Inhibitors/administration & dosage , Humans , Injections, Jet , Injections, Subcutaneous , Male , Middle Aged , Patient Satisfaction , Peptide Fragments/administration & dosage , Therapeutic Equivalency
12.
AIDS ; 20(3): 397-404, 2006 Feb 14.
Article in English | MEDLINE | ID: mdl-16439873

ABSTRACT

OBJECTIVE: To investigate the pharmacokinetics, safety/tolerability and antiviral activity of enfuvirtide administered once-daily (QD) versus twice-daily (BID). DESIGN: An open-label, randomized, multiple dose, two-period crossover study comparing 180 mg enfuvirtide, two injections QD versus 90 mg enfuvirtide, two injections, BID. METHODS: Steady-state intensive pharmacokinetic samples were obtained on days 7 and 14. RESULTS: Thirty-seven subjects received at least one dose of enfuvirtide. Thirty-three subjects completed both dosing periods. The regimens were bioequivalent based on the ratio of geometric mean area under the curve (AUC)0-tau [112 +/- 6.2 microg x h/ml QD; 115 +/- 6.4 microg x h/ml 2 x BID; QD/BID 0.98; 90% confidence interval (CI) 0.89,1.07]. The maximum observed plasma concentration within a dosing interval (Cmax) was 49% higher for QD (9.5 +/- 2.7 microg/ml) versus BID (6.3 +/- 1.7 microg/ml) and the pre-dose plasma concentration (Ctrough) was 57% lower for QD (1.6 +/- 1.1 microg/ml) versus BID (3.8 +/- 1.3 microg/ml). The LSM decrease in viral load from baseline to day 7 was 1.0 +/- 0.14 log10 (n = 18) for QD and 1.4 +/- 0.2 log10 (n = 17) for BID (LSM difference 0.385; P = 0.07). Linear regression analysis suggested that decline in viral load up to day 7 was associated with Ctrough but not Cmax or AUC. There were no significant differences in adverse events between the two dosing regimens. CONCLUSIONS: Administration of enfuvirtide 180 mg QD results in bioequivalence compared with 90 mg BID based on AUC with a similar short-term safety profile, but a trend towards a weaker antiretroviral effect. Larger and longer-term studies are needed to determine if 180 mg once daily is an effective dosing alternative for enfuvirtide.


Subject(s)
HIV Envelope Protein gp41/administration & dosage , HIV Fusion Inhibitors/administration & dosage , HIV Infections/drug therapy , Peptide Fragments/administration & dosage , Adult , Area Under Curve , Cross-Over Studies , Dose-Response Relationship, Drug , Enfuvirtide , Female , HIV Envelope Protein gp41/adverse effects , HIV Fusion Inhibitors/adverse effects , HIV Fusion Inhibitors/pharmacokinetics , Humans , Male , Peptide Fragments/adverse effects , Peptide Fragments/pharmacokinetics
13.
Pharm Res ; 20(5): 749-56, 2003 May.
Article in English | MEDLINE | ID: mdl-12751630

ABSTRACT

PURPOSE: The purpose of this work was to determine the jejunal permeability of cyclosporin A (CsA) in humans and whether formulation variables modulate the effects of P-glycoprotein (P-gp) on the permeability of CsA in Caco-2 cells. METHODS: A solution containing CsA, phenylalanine, propranolol, polyethyleneglycol (PEG) 400, and PEG 4000 was perfused through a 10-cm jejunal segment in 12 subjects. Caco-2 transport studies were performed using previously reported methodology. RESULTS: The mean Peff (+/- SD) of CsA in humans was 1.65 (0.53). The mean permeabilities for phenylalanine, propranolol, and PEG 400 were 4.54 (2.39), 2.90 (1.28), and 0.83 (0.51) x 10(-4) cm/s, respectively. The presence of surfactants significantly decreased the permeabilities of CsA in both directions in Caco-2 cells. CONCLUSIONS: The results suggest that the effects of surfactants via micellar solubilization and inhibition of P-gp efflux on CsA transport in Caco-2 cells are significant. CsA can rightly be classified as a low solubility-high permeability Class II BCS drug and its highly variable absorption from Sandimmune oral formulations is the result of poor dissolution characteristics.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Cyclosporine/pharmacokinetics , Intestinal Absorption/physiology , Jejunum/metabolism , Surface-Active Agents/pharmacokinetics , Adult , Caco-2 Cells , Humans , Intestinal Absorption/drug effects , Jejunum/drug effects , Surface-Active Agents/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...