Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 19 de 19
1.
Clin Pharmacol Ther ; 115(2): 248-255, 2024 02.
Article En | MEDLINE | ID: mdl-38082506

Anthrax Immune Globulin Intravenous (AIGIV [ANTHRASIL]), was developed for the treatment of toxemia associated with inhalational anthrax. It is a plasma product collected from individuals vaccinated with anthrax vaccine and contains antitoxin IgG antibodies against Bacillus anthracis protective antigen. A pharmacokinetic (PK) and exposure-response model was constructed to assess the PKs of AIGIV in anthrax-free and anthrax-exposed rabbits, non-human primates and anthrax-free humans, as well as the relationship between AIGIV exposure and survival from anthrax, based on available preclinical/clinical studies. The potential effect of anthrax on the PKs of AIGIV was evaluated and estimates of survival odds following administration of AIGIV protective doses with and without antibiotic co-treatment were established. As the developed PK model can simulate exposure of AIGIV in any species for any dosing scenario, the relationship between the predicted area under the concentration curve of AIGIV in humans and the probability of survival observed in preclinical studies was explored. Based on the simulation results, the intravenous administration of 420 U (units of potency as measured by validated Toxin Neutralization Assay) of AIGIV is expected to result in a > 80% probability of survival in more than 90% of the human population. Additional simulations suggest that exposure levels were similar in healthy and obese humans, and exposure in pediatrics is expected to be up to approximately seven-fold higher than in healthy adults, allowing for doses in pediatric populations that ranged from one to seven vials. Overall, the optimal human dose was justified based on the PK/pharmacodynamic (PD) properties of AIGIV in animals and model-based translation of PK/PD to predict human exposure and efficacy.


Anthrax Vaccines , Anthrax , Antitoxins , Respiratory Tract Infections , Adult , Animals , Humans , Rabbits , Child , Anthrax/drug therapy , Anthrax/prevention & control , Immunoglobulin G , Antitoxins/pharmacology , Antitoxins/therapeutic use , Anthrax Vaccines/therapeutic use , Anti-Bacterial Agents , Antigens, Bacterial/therapeutic use
2.
Pharm Res ; 40(7): 1657-1672, 2023 Jul.
Article En | MEDLINE | ID: mdl-36418671

PURPOSE: Long-acting formulations of the potent antiretroviral prodrug tenofovir alafenamide (TAF) hold potential as biomedical HIV prevention modalities. Here, we present a rigorous comparison of three animal models, C57BL/6 J mice, beagle dogs, and merino sheep for evaluating TAF implant pharmacokinetics (PKs). METHODS: Implants delivering TAF over a wide range of controlled release rates were tested in vitro and in mice and dogs. Our existing PK model, supported by an intravenous (IV) dosing dog study, was adapted to analyze mechanistic aspects underlying implant TAF delivery. RESULTS: TAF in vitro release in the 0.13 to 9.8 mg d-1 range with zero order kinetics were attained. Implants with equivalent fabrication parameters released TAF in mice and sheep at rates that were not statistically different, but were 3 times higher in dogs. When two implants were placed in the same subcutaneous pocket, a two-week creep to Cmax was observed in dogs for systemic drug and metabolite concentrations, but not in mice. Co-modeling IV and TAF implant PK data in dogs led to an apparent TAF bioavailability of 9.6 in the single implant groups (compared to the IV group), but only 1.5 when two implants were placed in the same subcutaneous pocket. CONCLUSIONS: Based on the current results, we recommend using mice and sheep, with macaques as a complementary species, for preclinical TAF implant evaluation with the caveat that our observations may be specific to the implant technology used here. Our report provides fundamental, translatable insights into multispecies TAF delivery via long-acting implants.


Anti-HIV Agents , HIV Infections , Pre-Exposure Prophylaxis , Animals , Mice , Dogs , Sheep , Tenofovir , HIV Infections/drug therapy , HIV Infections/prevention & control , Pre-Exposure Prophylaxis/methods , Mice, Inbred C57BL , Adenine , Alanine
3.
Blood ; 141(10): 1147-1158, 2023 03 09.
Article En | MEDLINE | ID: mdl-36108308

Factor VIII (FVIII) circulates in a noncovalent complex with von Willebrand Factor (VWF), the latter determining FVIII half-life. The VWF-binding aptamer rondaptivon pegol (BT200) increases plasma levels of VWF/FVIII in healthy volunteers. This trial assessed its safety, pharmacokinetics, and pharmacodynamics in hemophilia A. Nineteen adult patients (ages 20-62 years, 4 women) with hemophilia A (8 mild, 2 moderate, and 9 severe) received subcutaneous injections of rondaptivon pegol. After an initial fixed dose of 3 mg on days 0 and 4, patients received weekly doses of 2 to 9 mg until day 28. Severe hemophilia A patients underwent sparse-sampling population pharmacokinetics individual profiling after the final dose of rondaptivon pegol. Adverse events, pharmacokinetics, and pharmacodynamics were assessed. FVIII activity and VWF levels were measured. All patients tolerated rondaptivon pegol well. The geometric mean half-life of rondaptivon pegol was 5.4 days and rondaptivon pegol significantly increased VWF levels. In severe hemophilia A, 6 doses of rondaptivon pegol increased the half-lives of 5 different FVIII products from a median of 10.4 hours to 31.1 hours (range, 20.8-56.0 hours). Median FVIII increased from 22% to 48% in mild hemophilia A and from 3% to 7.5% in moderate hemophilia A. Rondaptivon pegol is a first-in-class prohemostatic molecule that extended the half-life of substituted FVIII approximately 3-fold and increased endogenous FVIII levels approximately 2-fold in hemophilia patients. This trial was registered at www.clinicaltrials.gov as #NCT04677803.


Hemophilia A , Hemostatics , Adult , Humans , Female , Young Adult , Middle Aged , von Willebrand Factor/therapeutic use , Hemophilia A/drug therapy , Factor VIII , Hemostatics/therapeutic use , Half-Life
4.
Sci Rep ; 12(1): 8224, 2022 05 17.
Article En | MEDLINE | ID: mdl-35581262

Global efforts aimed at preventing human immunodeficiency virus type one (HIV-1) infection in vulnerable populations appear to be stalling, limiting our ability to control the epidemic. Long-acting, controlled drug administration from subdermal implants holds significant potential by reducing the compliance burden associated with frequent dosing. We, and others, are exploring the development of complementary subdermal implant technologies delivering the potent prodrug, tenofovir alafenamide (TAF). The current report addresses knowledge gaps in the preclinical pharmacology of long-acting, subdermal TAF delivery using several mouse models. Systemic drug disposition during TAF implant dosing was explained by a multi-compartment pharmacokinetic (PK) model. Imaging mass spectrometry was employed to characterize the spatial distribution of TAF and its principal five metabolites in local tissues surrounding the implant. Humanized mouse studies determined the effective TAF dose for preventing vaginal and rectal HIV-1 acquisition. Our results represent an important step in the development of a safe and effective TAF implant for HIV-1 prevention.


Anti-HIV Agents , HIV Infections , Adenine , Alanine/therapeutic use , Animals , Female , HIV Infections/drug therapy , HIV Infections/prevention & control , Mice , Tenofovir/analogs & derivatives , Tenofovir/therapeutic use
5.
Clin Pharmacol Ther ; 112(1): 171-180, 2022 07.
Article En | MEDLINE | ID: mdl-35467014

Botulism antitoxin heptavalent (A, B, C, D, E, F, and G - Equine; BAT) product is a sterile solution of F(ab')2 and F(ab')2 -related antibody fragments prepared from plasma obtained from horses that have been immunized with a specific serotype of botulinum toxoid and toxin. BAT product is indicated for the treatment of symptomatic botulism following documented or suspected exposure to botulinum neurotoxin serotypes A to G in adults and pediatric patients. Pharmacokinetic and exposure-response models were used to explore the relationship between BAT product exposure and the probability of survival, and the occurrence of relevant moderate clinical signs observed during the preclinical development of BAT product to justify the clinical dose. The predicted probability of survival in humans for all serotypes of botulinum neurotoxin was more than 95.9% following intravenous administration of one vial of BAT product. Furthermore, this BAT product dose is expected to result in significant protection against clinical signs in human adults for all botulinum neurotoxin serotypes. Our exposure response model indicates that we have sufficient antitoxin levels to give full protection at various theoretical exposure levels and, based on neutralization capacity/potency of one dose of BAT product, it is expected to exceed the amount of circulating botulinum neurotoxin.


Botulinum Antitoxin , Botulinum Toxins , Botulism , Animals , Botulinum Antitoxin/therapeutic use , Botulinum Toxins/adverse effects , Botulism/drug therapy , Botulism/prevention & control , Horses , Humans
6.
Haematologica ; 107(9): 2121-2132, 2022 09 01.
Article En | MEDLINE | ID: mdl-34818873

Von Willebrand factor (VWF) and factor VIII (FVIII) circulate in a noncovalent complex in blood and promote primary hemostasis and clotting, respectively. A new VWF A1-domain binding aptamer, BT200, demonstrated good subcutaneous bioavailability and a long half-life in non-human primates. This first-in-human, randomized, placebo-controlled, doubleblind trial tested the hypothesis that BT200 is well tolerated and has favorable pharmacokinetic and pharmacodynamic effects in 112 volunteers. Participants received one of the following: a single ascending dose of BT200 (0.18-48 mg) subcutaneously, an intravenous dose, BT200 with concomitant desmopressin or multiple doses. Pharmacokinetics were characterized, and the pharmacodynamic effects were measured by VWF levels, FVIII clotting activity, ristocetin-induced aggregation, platelet function under high shear rates, and thrombin generation. The mean half-lives ranged from 7-12 days and subcutaneous bioavailability increased dose-dependently exceeding 55% for doses of 6-48 mg. By blocking free A1 domains, BT200 dose-dependently decreased ristocetin-induced aggregation, and prolonged collagen-adenosine diphosphate and shear-induced platelet plug formation times. However, BT200 also increased VWF antigen and FVIII levels 4-fold (P<0.001), without increasing VWF propeptide levels, indicating decreased VWF/FVIII clearance. This, in turn, increased thrombin generation and accelerated clotting. Desmopressin-induced VWF/FVIII release had additive effects on a background of BT200. Tolerability and safety were generally good, but exaggerated pharmacology was seen at saturating doses. This trial identified a novel mechanism of action for BT200: BT200 dose-dependently increases VWF/FVIII by prolonging half-life at doses well below those which inhibit VWF-mediated platelet function. This novel property can be exploited therapeutically to enhance hemostasis in congenital bleeding disorders.


von Willebrand Diseases , von Willebrand Factor , Deamino Arginine Vasopressin , Factor VIII , Humans , Ristocetin/pharmacology , Thrombin , von Willebrand Factor/metabolism
7.
Pharmacol Res Perspect ; 8(2): e00567, 2020 04.
Article En | MEDLINE | ID: mdl-32155317

Eptinezumab is a humanized mAb that targets calcitonin gene-related peptide and is under regulatory review for the prevention of episodic and chronic migraine (EM, CM). It is important to determine whether exposures achieved with intravenous (IV) administration of eptinezumab achieve desired pharmacologic effects. Population pharmacokinetics, including dose- and exposure-response analyses, were performed using patient-level data from the eptinezumab clinical trial program with IV doses ranging from 10 to 1000 mg in pharmacokinetic analyses or 10 to 300 mg in phase 2/3 clinical studies in patients with EM or CM. Exposure-response analysis explored the relationship between eptinezumab exposure metrics and efficacy parameters including monthly migraine days. The pharmacokinetic profile of eptinezumab was characterized by rapid attainment of maximum plasma concentration (ie, end of IV administration) and a terminal half-life of 27 days. Covariate analysis found that patient characteristics had no clinically significant effects on pharmacokinetic parameters and were insufficient to influence dosing. Dose- and exposure-response analyses found exposure with single doses ≥100 mg was associated with greater efficacy compared with doses ≤30 mg and a plateau of effect between 100 and 300 mg. A saturable inhibitory Emax model found the exposure over 12 weeks produced by single-dose eptinezumab 100 and 300 mg exceeded the exposure estimates required to achieve 90% of the maximal efficacy (EC90 ). This pharmacokinetic analysis of eptinezumab supports dosing every 12 weeks with no adjustment for patient characteristics, including exposures associated with 100- or 300-mg doses producing optimal efficacy effects. The similar efficacy profiles support 100 mg as the lowest effective dose of eptinezumab.


Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/pharmacokinetics , Models, Biological , Adolescent , Adult , Aged , Calcitonin Gene-Related Peptide/antagonists & inhibitors , Chronic Disease , Dose-Response Relationship, Drug , Female , Humans , Male , Middle Aged , Migraine Disorders/drug therapy , Migraine Disorders/metabolism , Young Adult
8.
Clin Pharmacol Ther ; 104(4): 655-663, 2018 10.
Article En | MEDLINE | ID: mdl-29737533

Aberrant activation of the classical complement pathway is the common underlying pathophysiology of orphan diseases such as bullous pemphigoid, antibody-mediated rejection of organ transplants, cold agglutinin disease, and warm autoimmune hemolytic anemia. Therapeutic options for these complement-mediated disorders are limited and sutimlimab, a humanized monoclonal antibody directed against complement factor C1s, may be potentially useful for inhibition of the classical complement pathway. A phase I, first-in-human, double-blind, randomized, placebo-controlled, dose-escalation trial of single and multiple doses of sutimlimab or placebo was conducted in 64 volunteers to evaluate safety, tolerability, pharmacokinetic, and pharmacodynamic profiles. Single and multiple infusions of sutimlimab were well tolerated without any safety concerns. sutimlimab exhibited a steep concentration-effect relationship with a Hill coefficient of 2.4, and an IC90 of 15.5 µg/mL. This study establishes the foundation for using sutimlimab as a highly selective inhibitor of the classical complement pathway in different diseases.


Antibodies, Monoclonal, Humanized/administration & dosage , Complement C1s/antagonists & inhibitors , Complement Inactivating Agents/administration & dosage , Complement Pathway, Classical/drug effects , Adult , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/pharmacokinetics , Austria , Complement C1s/immunology , Complement Inactivating Agents/adverse effects , Complement Inactivating Agents/pharmacokinetics , Dose-Response Relationship, Drug , Double-Blind Method , Drug Administration Schedule , Female , Healthy Volunteers , Humans , Infusions, Intravenous , Male , Middle Aged , Treatment Outcome , Young Adult
9.
Antimicrob Agents Chemother ; 59(7): 3913-9, 2015 Jul.
Article En | MEDLINE | ID: mdl-25896688

Oral or topical daily administration of antiretroviral (ARV) drugs to HIV-1-negative individuals in vulnerable populations is a promising strategy for HIV-1 prevention. Adherence to the dosing regimen has emerged as a critical factor determining efficacy outcomes of clinical trials. Because adherence to therapy is inversely related to the dosing period, sustained release or long-acting ARV formulations hold significant promise for increasing the effectiveness of HIV-1 preexposure prophylaxis (PrEP) by reducing dosing frequency. A novel, subdermal implant delivering the potent prodrug tenofovir alafenamide (TAF) with controlled, sustained, zero-order (linear) release characteristics is described. A candidate device delivering TAF at 0.92 mg day(-1) in vitro was evaluated in beagle dogs over 40 days for pharmacokinetics and preliminary safety. No adverse events related to treatment with the test article were noted during the course of the study, and no significant, unusual abnormalities were observed. The implant maintained a low systemic exposure to TAF (median, 0.85 ng ml(-1); interquartile range [IQR], 0.60 to 1.50 ng ml(-1)) and tenofovir (TFV; median, 15.0 ng ml(-1); IQR, 8.8 to 23.3 ng ml(-1)), the product of in vivo TAF hydrolysis. High concentrations (median, 512 fmol/10(6) cells over the first 35 days) of the pharmacologically active metabolite, TFV diphosphate, were observed in peripheral blood mononuclear cells at levels over 30 times higher than those associated with HIV-1 PrEP efficacy in humans. Our report on the first sustained-release nucleoside reverse transcriptase inhibitor (NRTI) for systemic delivery demonstrates a successful proof of principle and holds significant promise as a candidate for HIV-1 prophylaxis in vulnerable populations.


Adenine/analogs & derivatives , Anti-HIV Agents/pharmacokinetics , Anti-HIV Agents/therapeutic use , HIV Infections/prevention & control , Reverse Transcriptase Inhibitors/pharmacokinetics , Reverse Transcriptase Inhibitors/therapeutic use , Adenine/administration & dosage , Adenine/pharmacokinetics , Adenine/therapeutic use , Alanine , Animals , Anti-HIV Agents/administration & dosage , Dogs , Drug Implants , Equipment Design , HIV-1/drug effects , Male , Monocytes/metabolism , Prodrugs , Reverse Transcriptase Inhibitors/administration & dosage , Tenofovir/analogs & derivatives
10.
Antimicrob Agents Chemother ; 59(1): 59-66, 2015 Jan.
Article En | MEDLINE | ID: mdl-25313219

Mother-to-child transmission (MTCT) of HIV-1 remains a global health problem. The World Health Organization (WHO) recommendations advise the administration of a once-daily, oral, prophylactic regimen of the nonnucleoside reverse transcriptase inhibitor nevirapine (NVP) from birth until 4 to 6 weeks of age for infants born to HIV-infected mothers in regions without access to safe and nutritionally adequate alternatives to breast milk. A critical factor driving the successful implementation of the WHO guidelines involves sustaining high adherence to the frequent dosing. With these challenges in mind, we have developed the first injectable, sustained-release NVP formulations with the goal of providing, for 6 weeks or longer, preventative plasma drug levels from a single subcutaneous administration at birth. The long-acting NVP consists of large (>50 µm), monodisperse NVP particles coated with biocompatible polymers that control the drug release kinetics. Two lead formulations exhibiting burst-free, sustained-release kinetics for up to 75 days in vitro were developed. Subsequent in vivo studies in rats demonstrated no toxicity related to the formulations. Rat plasma NVP concentrations were above the analytical assay's limit of quantification for up to 28 days. Pharmacokinetic analysis of the rat plasma NVP concentration-time data allowed absorption rate constants to be calculated. These data then were used to simulate infant NVP exposure from a single injected dose (<200 mg) of our long-acting formulations, demonstrating preliminary feasibility of the technology to maintain safe, preventative NVP plasma levels (0.2 to 3.0 µg ml(-1)) for 6 weeks or longer.


Acquired Immunodeficiency Syndrome/drug therapy , Acquired Immunodeficiency Syndrome/prevention & control , Infectious Disease Transmission, Vertical/prevention & control , Nevirapine/pharmacokinetics , Nevirapine/therapeutic use , Animals , Breast Feeding , Delayed-Action Preparations/therapeutic use , Humans , Infant , Infant, Newborn , Nevirapine/adverse effects , Nevirapine/blood , Rats , Rats, Sprague-Dawley
11.
Bone ; 49(2): 250-6, 2011 Aug.
Article En | MEDLINE | ID: mdl-21458605

Hypophosphatasia (HPP) features rickets or osteomalacia from tissue-nonspecific alkaline phosphatase (TNSALP) deficiency due to deactivating mutations within the ALPL gene. Enzyme replacement therapy with a bone-targeted, recombinant TNSALP (sALP-FcD(10), renamed ENB-0040) prevents manifestations of HPP when initiated at birth in TNSALP knockout (Akp2(-/-)) mice. Here, we evaluated the dose-response relationship of ENB-0040 to various phenotypic traits of Akp2(-/-) mice receiving daily subcutaneous (SC) injections of ENB-0040 from birth at 0.5, 2.0, or 8.2mg/kg for 43days. Radiographs, µCT, and histomorphometric analyses documented better bone mineralization with increasing doses of ENB-0040. We found a clear, positive correlation between ENB-0040 dose and prevention of mineralization defects of the feet, rib cage, lower limbs, and jaw bones. According to a dose-response model, the ED(80) (the dose that prevents bone defects in 80% of mice) was 3.2, 2.8 and 2.9mg/kg/day for these sites, respectively. Long bones seemed to respond to lower daily doses of ENB-0040. There was also a positive relationship between ENB-0040 dose and survival. Median survival, body weight, and bone length all improved with increasing doses of ENB-0040. Urinary PP(i) concentrations remained elevated in all treatment groups, indicating that while this parameter is a good biochemical marker for diagnosing HPP in patients, it may not be a good follow up marker for evaluating response to treatment when administering bone-targeted TNSALP to mice. These dose-response relationships strongly support the pharmacological efficacy of ENB-0040 for HPP, and provide the experimental basis for the therapeutic range of ENB-0040 chosen for clinical trials.


Alkaline Phosphatase/metabolism , Hypophosphatasia/drug therapy , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Alkaline Phosphatase/genetics , Animals , Enzyme Replacement Therapy/methods , Mice , Mice, Knockout , Osteomalacia/drug therapy , Recombinant Proteins/genetics
12.
Mol Genet Metab ; 100(3): 221-8, 2010 Jul.
Article En | MEDLINE | ID: mdl-20382058

UNLABELLED: Glycerol phenylbutyrate (glyceryl tri (4-phenylbutyrate)) (GPB) is being studied as an alternative to sodium phenylbutyrate (NaPBA) for the treatment of urea cycle disorders (UCDs). This phase 2 study explored the hypothesis that GPB offers similar safety and ammonia control as NaPBA, which is currently approved as adjunctive therapy in the chronic management of UCDs, and examined correlates of 24-h blood ammonia. METHODS: An open-label, fixed sequence switch-over study was conducted in adult UCD patients taking maintenance NaPBA. Blood ammonia and blood and urine metabolites were compared after 7 days (steady state) of TID dosing on either drug, both dosed to deliver the same amount of phenylbutyric acid (PBA). RESULTS: Ten subjects completed the study. Adverse events were comparable for the two drugs; 2 subjects experienced hyperammonemic events on NaPBA while none occurred on GPB. Ammonia values on GPB were approximately 30% lower than on NaPBA (time-normalized AUC=26.2 vs. 38.4 micromol/L; Cmax=56.3 vs. 79.1 micromol/L; not statistically significant), and GPB achieved non-inferiority to NaPBA with respect to ammonia (time-normalized AUC) by post hoc analysis. Systemic exposure (AUC(0-24)) to PBA on GPB was 27% lower than on NaPBA (540 vs. 739 microgh/mL), whereas exposure to phenylacetic acid (PAA) (575 vs. 596 microg h/mL) and phenylacetylglutamine (PAGN) (1098 vs. 1133 microg h/mL) were similar. Urinary PAGN excretion accounted for approximately 54% of PBA administered for both NaPBA and GPB; other metabolites accounted for <1%. Intact GPB was generally undetectable in blood and urine. Blood ammonia correlated strongly and inversely with urinary PAGN (r=-0.82; p<0.0001) but weakly or not at all with blood metabolite levels. CONCLUSIONS: Safety and ammonia control with GPB appear at least equal to NaPBA. Urinary PAGN, which is stoichiometrically related to nitrogen scavenging, may be a useful biomarker for both dose selection and adjustment for optimal control of venous ammonia.


Ammonia/blood , Phenylbutyrates/therapeutic use , Urea Cycle Disorders, Inborn/drug therapy , Urea Cycle Disorders, Inborn/metabolism , Adult , Aged , Cross-Over Studies , Female , Glutamine/analogs & derivatives , Glutamine/blood , Glycerol/analogs & derivatives , Glycerol/blood , Glycerol/pharmacokinetics , Glycerol/therapeutic use , Humans , Male , Middle Aged , Phenylacetates/blood , Phenylbutyrates/blood , Phenylbutyrates/pharmacokinetics , Urea Cycle Disorders, Inborn/blood , Young Adult
13.
J Clin Pharmacol ; 50(1): 36-49, 2010 Jan.
Article En | MEDLINE | ID: mdl-19773525

Teduglutide is a GLP-2 analog currently evaluated for the treatment of short bowel syndrome, Crohn's disease, and other gastrointestinal disorders. The population pharmacokinetics (PK) of teduglutide were assessed following daily subcutaneous (SC) administrations of 2.5 to 80 mg doses in a total of 256 patients. A 1-compartment model with a site-specific rate constant of absorption in the abdomen, arm, and thigh was used to assess the PK of teduglutide. Apparent clearance (CL/F) of teduglutide in male participants was approximately 18% higher than that observed in female participants (12.4 vs 10.5 L/h, respectively). Body weight was detected as a significant covariate explaining the volume of distribution of teduglutide. The elimination half-life (t((1/2))) of teduglutide was also influenced by the body weight of participants. For a male patient weighing 50 and 90 kg, t((1/2)) of teduglutide was 0.897 and 2.99 hours, respectively. Renal and hepatic function of patients did not affect the PK of teduglutide. As a result, no dose adjustment was deemed necessary in patients with altered renal or liver function. The population PK model will help to support adequate drug labeling following SC administrations in patients and determine whether an individualized dosage is required.


Crohn Disease/drug therapy , Dose-Response Relationship, Drug , Gastrointestinal Agents/pharmacokinetics , Peptides/pharmacokinetics , Short Bowel Syndrome/drug therapy , Adolescent , Adult , Aged , Clinical Trials as Topic , Female , Humans , Injections, Subcutaneous , Kidney Diseases/metabolism , Liver Diseases/metabolism , Male , Middle Aged , Models, Statistical , Peptides/administration & dosage , Sex Characteristics
14.
Ther Drug Monit ; 31(4): 502-10, 2009 Aug.
Article En | MEDLINE | ID: mdl-19494795

Capsaicin, a pungent compound in chili peppers, is a highly selective agonist for the transient receptor potential vanilloid 1 receptor expressed in nociceptive sensory nerves. A high-concentration (640 microg/cm2) capsaicin patch, designated NGX-4010, is in clinical evaluation for the management of peripheral neuropathic pain. To determine systemic capsaicin exposure after single 60- or 90-minute NGX-4010 applications, plasma samples were collected from 173 patients with postherpetic neuralgia (PHN), painful human immunodeficiency virus-associated neuropathy (HIV-AN), and painful diabetic neuropathy (PDN). The percentages of patients with quantifiable levels of capsaicin at any time point were 31% for PHN (30 of 96), 7% for HIV-AN (3 of 44), and 3% for PDN (1 of 33). The maximum plasma concentration observed in any patient was 17.8 ng/mL. Due to the limited number of quantifiable levels, a population analysis was performed to characterize the pharmacokinetics (PK) of capsaicin. Plasma concentrations were fitted adequately using a 1-compartment model with first-order absorption and linear elimination. Capsaicin levels declined very rapidly, with a mean population elimination half-life of 1.64 hours. Mean area under the curve and C max values after a 60-minute application were 7.42 ng x h/mL and 1.86 ng/mL, respectively. Only a few correlations between calculated PK parameters and patient characteristics were observed. Duration and area of application of the patch were detected as significant covariates explaining the PK of capsaicin. Ninety-minute applications of NGX-4010 resulted in capsaicin area under the curve and Cmax values approximately 1.78- and 2.15-fold higher than those observed in patients treated for 60 minutes. Treatment on the feet (patients with HIV-AN and PDN) produced far lower systemic exposure than treatment on the trunk (patients with PHN). Finally, larger treatment areas were associated with statistically higher Vc/F values. The low systemic exposure and very rapid elimination half-life of capsaicin after NGX-4010 administration are unlikely to result in systemic effects and support the overall safety profile of this investigational cutaneous patch.


Capsaicin/pharmacokinetics , Pain/metabolism , Peripheral Nervous System Diseases/metabolism , Administration, Cutaneous , Administration, Topical , Capsaicin/administration & dosage , Capsaicin/pharmacology , Capsicum , Dosage Forms , Dose-Response Relationship, Drug , Female , HIV Infections , HIV-1 , Half-Life , Humans , Kinetics , Male , Pain Measurement , Treatment Outcome
15.
J Clin Pharmacol ; 48(11): 1289-99, 2008 Nov.
Article En | MEDLINE | ID: mdl-18974283

Teduglutide, a glucagon-like peptide-2 (GLP-2) analog, is currently being evaluated for the treatment of short-bowel syndrome, Crohn's disease, and other gastrointestinal disorders. The pharmacokinetics, safety, and tolerability of teduglutide in healthy subjects (N = 64) were assessed following daily subcutaneous administrations for 8 days in a double-blinded, randomized, placebo-controlled, ascending-dose study. Teduglutide treatments were administered as a 50-mg/mL (10, 15, 20, 25, 30, 50, and 80 mg) or 20-mg/mL (20 mg) formulation. Blood samples were collected on days 1 and 8, and plasma concentrations of teduglutide were measured using a liquid chromatography/tandem mass spectrometry method. Mean systemic exposures to teduglutide were very similar on days 1 and 8, suggesting minimal, if any, accumulation following once-daily repeated administrations. The apparent clearance of teduglutide following administration of the 50-mg/mL formulation was constant over the dose range, with mean values in male and female subjects of 0.155 and 0.159 L/h/kg, respectively. Peak plasma concentrations and total exposure of teduglutide after subcutaneous injection of a 20-mg/mL formulation (1.0 mL) were approximately 15% and 78% higher than those observed with the 50-mg/mL formulation (0.4 mL), respectively. Teduglutide treatments were safe and well tolerated. All but 1 adverse event was assessed as mild or moderate in severity. No relationship between teduglutide treatments and frequency of adverse events was observed, with the exception of injection site pain, which increased as a function of dose and injected volume. Results from the current study will assist in the dose selection in future efficacy studies.


Glucagon-Like Peptide 2/chemistry , Peptides/administration & dosage , Peptides/pharmacokinetics , Administration, Cutaneous , Adult , Cohort Studies , Double-Blind Method , Female , Humans , Male , Middle Aged , Peptides/adverse effects , Peptides/chemistry , Young Adult
16.
Chem Res Toxicol ; 18(3): 475-85, 2005 Mar.
Article En | MEDLINE | ID: mdl-15777087

The objectives of this study were to (i) develop quantitative structure-property relationships (QSPRs) for blood:air partition coefficients (Pb:a), tissue:air partition coefficients (Pt:a), and hepatic clearance (CLh) and (ii) conduct interspecies extrapolations of the pharmacokinetics of low molecular weight volatile organic chemicals (VOCs) by incorporating the above QSPRs within a physiologically based pharmacokinetic (PBPK) modeling framework. Pb:a and Pt:a were predicted using the following algorithm: FnlxPo:a+FwxPw:a+f(b)xFpxPp:a, where Fnl=content of neutral lipid equivalents in biological matrix, Fw=content of water equivalents in biological matrix, Fp=protein content of blood and tissues, Po:a=vegetable oil:air partition coefficient, Pw:a=water:air partition coefficient, f(b)=fraction of total protein involved in the partitioning process, and Pp:a=protein:air partition coefficient. CLh was estimated as follows: Qlx[(CLintxC(P4502E1)xVl)/(Ql+CLintxC(P4502E1)xVl)], where CLint=intrinsic clearance normalized for P450 2E1 content, Ql=blood flow to the liver, C(P4502E1)=hepatic concentration of P450 2E1 in the species of interest, and Vl=volume of liver. QSPRs relating molecular fragments of 46 VOCs and parameters required for estimating Pb:a, Pt:a, and CLh (namely, Po:a, Pw:a, Pp:a, and CLint) were established using a group contribution method (f(i)xCi, where f=frequency of occurrence of the group i in a given molecule and Ci=contribution of the group i to Po:a, Pw:a, Pp:a, or CLint). Values of group contributions were determined by multilinear regression of experimental data. The species specific parameters required for solving the above algorithms were obtained from the literature. These algorithms, once incorporated into a multispecies PBPK modeling framework, enabled extrapolation of the kinetics of chemicals across species. The inhalation pharmacokinetics of dichloromethane and toluene as well as two de novo compounds (1,2,4-trimethyl benzene and ethyl benzene) were extrapolated from rat to human, using the present modeling methodology. This study has demonstrated that it is possible to extrapolate the pharmacokinetic behavior of chemicals from rats to humans on the basis of QSPRs and species specific physiological information.


Air Pollutants/pharmacology , Inhalation Exposure , Organic Chemicals/pharmacokinetics , Quantitative Structure-Activity Relationship , Administration, Inhalation , Air Pollutants/chemistry , Air Pollutants/classification , Animals , Humans , Organic Chemicals/chemistry , Organic Chemicals/classification , Rats , Species Specificity
17.
Toxicol Mech Methods ; 15(5): 361-6, 2005.
Article En | MEDLINE | ID: mdl-20021057

Chronic exposure to volatile organic chemicals (VOCs) in the environment leads to steady-state conditions. The establishment of quantitative relationships between steady-state blood concentrations and molecular structures of VOCs can be potentially useful. The objective of this study was therefore to investigate the relationship between the steady-state arterial blood concentration (Ca(ss)) in the rat and the molecular structures of 19 VOCs belonging to multiple chemical families (alkanes, haloalkanes, haloalkenes, and aromatics). The overall approach consisted of developing quantitative relationships between molecular fragments (CH(3), CH(2), CH, C, C horizontal lineC, H, Cl, benzene ring, and H in benzene ring structure) in alkanes, haloalkanes, haloethylenes, and aromatic hydrocarbons, as well as their Ca(ss) (associated with 1 mu mu mol/L inhalation exposure) according to an additive fragment model. This modeling approach implies that each fragment in the structure of a chemical has an additive and constant contribution to its Ca(ss). A multilinear regression was performed using a commercially available statistical software package, and the results obtained were essentially the contributions associated with each of the nine structural fragments toward Ca(ss) in the rat continuously exposed to 1 mu mu mol/L VOC in the air. The resulting model estimated adequately the Ca(ss) of VOCs initially used in the calibration (estimated/experimental ratio: 1.04 +/- 0.30, mean +/- standard deviation [SD]). This molecular structure vs. Ca(ss) relationship was then evaluated using an external dataset on Ca(ss) for three aliphatic hydrocarbons (octane, 2-methyl octane, and 1-nonene; 100 ppm exposures). The ratio of predicted to experimental Ca(ss) for these chemicals ranged from 0.6 to 1.2. The results of this study suggest that steady-state blood concentrations of inhaled VOCs can be predicted using structure-activity type models.

18.
Toxicol Appl Pharmacol ; 189(3): 221-32, 2003 Jun 15.
Article En | MEDLINE | ID: mdl-12791307

The objective of present study was to develop quantitative structure-property relationships (QSPRs) for the chemical-specific input parameters of rat physiologically based pharmacokinetic (PBPK) models (i.e., blood:air partition coefficient (P(b)), liver:air partition coefficient (P(l)), muscle:air partition coefficient (P(m)), fat:air partition coefficient (P(f)), and hepatic clearance (CL(h))), for simulating the inhalation pharmacokinetics of volatile organic chemicals (VOCs). The literature data on P(b), P(l), P(f), and P(m) for 46 low-molecular-weight VOCs as well as CL(h) for 25 such VOCs primarily metabolized by CYP2E1 (alkanes, haloalkanes, haloethylenes, and aromatic hydrocarbons) were analysed to develop QSPRs. The QSPRs developed in this study were essentially multilinear additive models, which imply that each fragment in the molecular structure has an additive and constant contribution to partition coefficients and hepatic clearance. Most of the values in the calibration set could be reproduced adequately with the QSPR approach, which involved the calculation of the sum of the frequency of occurrence of fragments (CH(3), CH(2), CH, C, C=C, H, Cl, Br, F, benzene ring, and H in benzene ring structure) times the fragment-specific contributions determined in this study. The QSPRs for P(b), P(l), P(m), P(f), and CL(h) were then included within a PBPK model, which only required the specification of the frequency of occurrence of fragments in a molecule along with exposure concentration and duration as input for conducting pharmacokinetic simulations. This QSPR-PBPK model framework facilitated the prediction of the inhalation pharmacokinetics of four VOCs present in the calibration dataset (toluene, dichloromethane, trichloroethylene, and 1,1,1-trichloroethane) and four VOCs that were not part of the calibration set (1,2,4-trimethyl benzene, ethyl benzene, 1,3-dichloropropene, and 2,2-dichloro-1,1,1-trifluoroethane) but that could be described using the molecular fragments investigated in the present study. The QSPRs developed in this study should be potentially useful for providing a first-cut evaluation of the inhalation pharmacokinetics of VOCs prior to experimentation, as long as the number and nature of the fragments do not exceed the ones in the calibration dataset used in this study.


Models, Biological , Organic Chemicals/chemistry , Organic Chemicals/pharmacokinetics , Quantitative Structure-Activity Relationship , Animals , Models, Chemical , Rats , Volatilization
19.
Environ Health Perspect ; 110 Suppl 6: 989-94, 2002 Dec.
Article En | MEDLINE | ID: mdl-12634130

The available data on binary interactions are yet to be considered within the context of mixture risk assessment because of our inability to predict the effect of a third or a fourth chemical in the mixture on the interacting binary pairs. Physiologically based pharmacokinetic (PBPK) models represent a potentially useful framework for predicting the consequences of interactions in mixtures of increasing complexity. This article highlights the conceptual basis and validity of PBPK models for extrapolating the occurrence and magnitude of interactions from binary to more complex chemical mixtures. The methodology involves the development of PBPK models for all mixture components and interconnecting them at the level of the tissue where the interaction is occurring. Once all component models are interconnected at the binary level, the PBPK framework simulates the kinetics of all mixture components, accounting for the interactions occurring at various levels in more complex mixtures. This aspect was validated by comparing the simulations of a binary interaction-based PBPK model with experimental data on the inhalation kinetics of m-xylene, toluene, ethyl benzene, dichloromethane, and benzene in mixtures of varying composition and complexity. The ability to predict the kinetics of chemicals in complex mixtures by accounting for binary interactions alone within a PBPK model is a significant step toward the development of interaction-based risk assessment for chemical mixtures.


Environmental Pollutants/adverse effects , Environmental Pollutants/pharmacokinetics , Models, Theoretical , Pharmacokinetics , Xenobiotics/adverse effects , Xenobiotics/pharmacokinetics , Animals , Drug Interactions , Forecasting , Humans , Reproducibility of Results , Risk Assessment/methods
...