Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters











Publication year range
1.
J Pediatr Pharmacol Ther ; 29(1): 49-52, 2024.
Article in English | MEDLINE | ID: mdl-38332965

ABSTRACT

OBJECTIVE: Sublingual buprenorphine has demonstrated efficacy for treatment of the neonatal opioid withdrawal syndrome (NOWS), but the current formulation used in clinical practice contains 30% ethanol. Ethanol as a pharmacologically active excipient ideally should be removed from neonatal formulations. The objective of this study was to determine the relative bioavailability of a novel ethanol-free -formulation (CHF6563) compared with the commonly used ethanolic solution in a phase I, open-label, 2-period, -single-dose, crossover study in healthy adults. METHODS: Eighteen adult opioid-naïve volunteers were administered one of the formulations in a randomized crossover treatment. After a 10-day washout period, subjects received the other formulation. Serial blood samples were drawn for pharmacokinetic analysis over 48 hours. RESULTS: The geometric mean ratio (90% CIs) of the ethanol-free buprenorphine solution AUC0-last was 0.80 (0.65-0.99) and Cmax was 0.81 (0.66-0.99) compared with reference ethanolic formulation. The -ethanol-free formulation had a greater degree of intersubject variability than the ethanol-containing -reference formulation (coefficient of variation of 59% vs 31.5%, respectively, for AUC0-last). CONCLUSIONS: In an adult population, a novel ethanol-free formulation of buprenorphine containing widely used excipients demonstrated a slight decrease in bioavailability when compared with an ethanolic solution. These results will inform those seeking to develop ethanol-free pediatric drug formulations.

2.
Clin Ther ; 43(11): 1934-1947.e4, 2021 11.
Article in English | MEDLINE | ID: mdl-34600734

ABSTRACT

PURPOSE: A number of single-inhaler, fixed-dose, triple combinations are available for the management of chronic obstructive pulmonary disease and/or asthma. One of these is the extrafine formulation beclomethasone dipropionate, formoterol fumarate, glycopyrronium bromide (BDP/FF/GB). Given that differences in ethnicity can result in differences in systemic exposure, we evaluated the relative pharmacokinetic (PK) profiles of BDP/FF/GB in Japanese vs Caucasian healthy volunteers to assess the need for dose adjustment. METHODS: This randomized, double-blind, single-dose, 4-way crossover study recruited healthy men and women 20 to 55 years of age; for each Japanese person a Caucasian was enrolled who matched in terms of sex, age, and weight. Study treatments included BDP/FF/GB 200/12/25 and 400/12/25 µg (therapeutic), 800/48/100 µg (supratherapeutic), and placebo. PK blood samples were taken up to 24 hours for evaluation of BDP, beclomethasone 17-monopropionate (B17MP, an active metabolite of BDP), and formoterol and up to 48 h for GB. The primary objective was to characterize the PK profiles of BDP, FF, and GB after administration of a single dose of BDP/FF/GB in Caucasian and Japanese healthy volunteers in terms of the AUC0-t and Cmax of B17MP, formoterol, and GB. FINDINGS: Of the 32 recruited participants (16 Japanese and 16 Caucasian ), 30 completed the study. A clear plasma exposure dose-response relationship was found for all 4 molecules. B17MP Cmax geometric mean ratios for Japanese vs Caucasian participants for the 3 study treatments ranged from 1.17 to 1.26, and AUC0-t ratios ranged from 1.16 to 1.22; thus, the findings were comparable between the ethnicities. Formoterol exposure was higher in Japanese than Caucasian participants (Cmax, 1.22-1.53; AUC0-t, 1.23-1.40). The GB Cmax with BDP/FF/GB 400/12/25 µg (1.09) and AUC0-t values for all three doses (0.98-1.17) were comparable in the 2 populations, but Cmax with 200/12/25 and 800/48/100 µg were higher in Japanese participants (1.32 and 1.42, respectively). Pharmacodynamic (cortisol, potassium, glucose, blood pressure, heart rate, and QT interval with the Fridericia correction) and safety profile results were similar in the 2 ethnicities, with most patients not experiencing any adverse events. IMPLICATIONS: Exposure to BDP/FF/GB pressurized metered dose inhaler at therapeutic and supratherapeutic doses was associated with higher plasma levels in Japanese versus Caucasian healthy volunteers. These PK differences did not translate into meaningful differences in the safety or pharmacodynamic parameters assessed in this study and were consistent with the results of other long-term (52-week) published studies. Dose adjustments in Japanese people are not deemed necessary. CLINICALTRIALS. GOV IDENTIFIER: NCT03859414.


Subject(s)
Glycopyrrolate , Pulmonary Disease, Chronic Obstructive , Administration, Inhalation , Beclomethasone/therapeutic use , Bronchodilator Agents/therapeutic use , Cross-Over Studies , Double-Blind Method , Drug Combinations , Ethnicity , Female , Formoterol Fumarate/therapeutic use , Humans , Japan , Male , Metered Dose Inhalers , Nebulizers and Vaporizers , Pulmonary Disease, Chronic Obstructive/drug therapy
3.
Adv Ther ; 38(10): 5317-5332, 2021 10.
Article in English | MEDLINE | ID: mdl-34515977

ABSTRACT

INTRODUCTION: Tacrolimus, the cornerstone of transplantation immunosuppression, is a narrow therapeutic index drug with a low and highly variable bioavailability. Therapeutic drug monitoring based on trough level assessment is mandatory in order to target a personalised exposure and avoid both rejection and toxicity. Population pharmacokinetic (POPPK) models might be a useful tool for improving early attainment of target range by guiding initial doses until steady state is reached and trough levels can be reliably used as surrogate marker of exposure. Here we present the first POPPK for predicting the initial doses of the once-daily prolonged release tacrolimus Envarsus (LCPT) in adult kidney recipients. METHODS: The model was developed exploiting the data from a recent pharmacokinetic randomised clinical study, in which 69 de novo kidney recipients, 33 of whom treated with LCPT, underwent an intensive blood sampling strategy for tacrolimus including four complete pharmacokinetic profiles. RESULTS: The complex and prolonged absorption of LCPT is well described by the three-phase model that incorporates body weight and CYP3A5 genotype as significant covariates accounting for a great proportion of the inter-patient variability: in particular, CYP3A5*1/*3 expressors had a 66% higher LCPT clearance. We have then generated by simulation a personalised dosing strategy based on the model that could improve the early attainment of therapeutic trough levels by almost doubling the proportion of patients within target range (69.3% compared to 36.1% with the standard body weight-based approach) on post-transplantation day 4 and significantly reduce the proportion of overexposed patients at risk of toxicity. CONCLUSIONS: A POPPK model was successfully developed for LCPT in de novo kidney recipients. The model could guide a personalised dosing strategy early after transplantation. For the model to be translated into clinical practice, its beneficial impact of earlier attainment of therapeutic trough levels should be demonstrated on hard clinical outcomes in further studies.


Subject(s)
Kidney Transplantation , Tacrolimus , Adult , Drug Administration Schedule , Drug Monitoring , Graft Rejection , Humans , Immunosuppressive Agents
4.
Int J Pharm ; 606: 120893, 2021 Sep 05.
Article in English | MEDLINE | ID: mdl-34274456

ABSTRACT

Orally inhaled products (OIPs) are gaining increased attention, as pulmonary delivery is a preferred route for the treatment of various diseases. Yet, the field of inhalation biopharmaceutics is still in development phase. For a successful correlation between various in vitro data obtained during formulation characterization and in vivo performance, it is necessary to understand the impact of parameters such as solubility and dissolution of drugs. In this work, we used in vitro-in silico feedback-feedforward approach to gain a better insight into the biopharmaceutics behavior of inhaled Salbutamol Sulphate (SS) and Budesonide (BUD). The thorough characterization of the in vitro test media and the impact of different in vitro fluid components such as lipids and protein on the solubility of aforementioned drugs was studied. These results were subsequently used as an input into the developed in silico models to investigate potential PK parameter changes in vivo. Results revealed that media comprising lipids and albumin were the most biorelevant and impacted the solubility of BUD the most. On the contrary, no notable impact was seen in case of SS. The use of simple media such as phosphate buffer saline (PBS) might be sufficient to use in solubility studies of the highly soluble and permeable drugs. However, its use for the poorly soluble drugs is limited due to the greater potential for interactions within in vivo environment. The use of in silico tools showed that the model response varies, depending on the used media. Therefore, this work highlights the relevance of carefully selecting the media composition when investigating solubility and dissolution behavior, especially in the early phases of drug development and of poorly soluble drugs.


Subject(s)
Models, Biological , Pharmaceutical Preparations , Administration, Inhalation , Administration, Oral , Computer Simulation , Intestinal Absorption , Lung , Solubility
5.
CPT Pharmacometrics Syst Pharmacol ; 8(7): 460-468, 2019 07.
Article in English | MEDLINE | ID: mdl-31077576

ABSTRACT

Concentration-QTcF data obtained from two phase I studies in healthy volunteers treated with a novel phosphodiesterase-4 inhibitor currently under development for the treatment of chronic obstructive pulmonary disease were analyzed by means of mixed-effects modeling. A simple linear mixed-effects model and a more complex model that included oscillatory functions were employed and compared. The slope of the concentration-QTcF relationship was not significantly greater than 0 in both approaches, and the simulations showed that the upper limit of the 90% confidence interval around the mean ΔΔQTcF is not expected to exceed 10 ms within the range of clinically relevant concentrations. An additional simulation study confirmed the robustness of the simple linear mixed-effects model for the analysis of concentration-QT data and supported the modeling of data obtained from studies with different designs (parallel and crossover).


Subject(s)
Heart Rate/drug effects , Phosphodiesterase 4 Inhibitors/administration & dosage , Sulfonamides/administration & dosage , para-Aminobenzoates/administration & dosage , Clinical Trials, Phase I as Topic , Female , Healthy Volunteers , Humans , Male , Models, Theoretical , Phosphodiesterase 4 Inhibitors/pharmacology , Randomized Controlled Trials as Topic , Sulfonamides/pharmacology , para-Aminobenzoates/pharmacology
6.
PLoS One ; 12(9): e0184822, 2017.
Article in English | MEDLINE | ID: mdl-28922421

ABSTRACT

The development of novel therapeutics to prevent cognitive decline of Alzheimer's disease (AD) is facing paramount difficulties since the translational efficacy of rodent models did not resulted in better clinical results. Currently approved treatments, including the acetylcholinesterase inhibitor donepezil (DON) and the N-methyl-D-aspartate antagonist memantine (MEM) provide marginal therapeutic benefits to AD patients. There is an urgent need to develop a predictive animal model that is phylogenetically proximal to humans to achieve better translation. The non-human primate grey mouse lemur (Microcebus murinus) is increasingly used in aging research, but there is no published results related to the impact of known pharmacological treatments on age-related cognitive impairment observed in this primate. In the present study we investigated the effects of DON and MEM on sleep-deprivation (SD)-induced memory impairment in young and aged male mouse lemurs. In particular, spatial memory impairment was evaluated using a circular platform task after 8 h of total SD. Acute single doses of DON or MEM (0.1 and 1mg/kg) or vehicle were administered intraperitoneally 3 h before the cognitive task during the SD procedure. Results indicated that both doses of DON were able to prevent the SD-induced deficits in retrieval of spatial memory as compared to vehicle-treated animals, both in young and aged animals Likewise, MEM show a similar profile at 1 mg/kg but not at 0.1mg/kg. Taken together, these results indicate that two widely used drugs for mitigating cognitive deficits in AD were partially effective in sleep deprived mouse lemurs, which further support the translational potential of this animal model. Our findings demonstrate the utility of this primate model for further testing cognitive enhancing drugs in development for AD or other neuropsychiatric conditions.


Subject(s)
Aging/drug effects , Indans/pharmacology , Memantine/pharmacology , Memory Disorders/drug therapy , Piperidines/pharmacology , Sleep Deprivation/drug therapy , Spatial Memory/drug effects , Alzheimer Disease/complications , Alzheimer Disease/drug therapy , Alzheimer Disease/physiopathology , Animals , Cheirogaleidae , Disease Models, Animal , Donepezil , Male , Memory Disorders/etiology , Memory Disorders/physiopathology , Sleep Deprivation/complications , Sleep Deprivation/physiopathology
7.
Neuropharmacology ; 85: 206-14, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24878242

ABSTRACT

Memory loss is one of the key features of cognitive impairment in either aging, Mild Cognitive Impairment (MCI) or dementia. Pharmacological treatments for memory loss are today focused on addressing symptomatology. One of these approved compounds is memantine, a partial NMDA receptor antagonist that has proved its beneficial effects in cognition. The Octodon degus (O. degus) has been recently proposed as a potential model relevant for neurodegenerative diseases. However, there are no previous studies investigating the effect of pharmacological treatments for age-related cognitive impairment in this rodent. In this work we aimed to evaluate the effect of memantine on sleep deprivation (SD)-induced memory impairment in young and old O. degus. Young and old animals were trained in different behavioral paradigms validated for memory evaluation, and randomly assigned to a control (CTL, n=14) or an SD (n=14) condition, and treated with vehicle or memantine (10-mg/Kg i.p.) before the SD started. We demonstrate that SD impairs memory in both young and old animals, although the effect in the old group was significantly more severe (P<0.05). Memantine pretreatment was able to prevent the cognitive impairment caused by SD in both age groups, while it had no negative effect on CTL animals. The positive effect of memantine in counteracting the negative effect of SD on the retrieval process even in the aged O. degus further supports the translational potential of both the challenge and the species, and will enable a better understanding of the behavioral features of memantine effects, especially related with reference and working memories.


Subject(s)
Aging/drug effects , Memantine/pharmacology , Memory Disorders/prevention & control , Memory, Short-Term/drug effects , Nootropic Agents/pharmacology , Sleep Deprivation/complications , Aging/physiology , Animals , Female , Maze Learning/drug effects , Maze Learning/physiology , Memantine/blood , Memory Disorders/etiology , Memory Disorders/physiopathology , Memory, Short-Term/physiology , Neuropsychological Tests , Nootropic Agents/blood , Octodon , Random Allocation , Recognition, Psychology/drug effects , Recognition, Psychology/physiology , Sleep Deprivation/physiopathology
8.
Br J Clin Pharmacol ; 74(3): 525-35, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22300419

ABSTRACT

AIM: The objective of this investigation was to assess the performance of an allometric model as the basis for interpolating drug exposure in the context of pharmacokinetic bridging across paediatric subpopulations. METHODS: Midazolam was selected as a paradigm compound. Two nonlinear mixed effects models were developed to describe midazolam pharmacokinetics in infants, toddlers and adults (model 1) and in children and adolescents (model 2). Subsequently, systemic drug exposure, expressed in terms of the area under the concentration vs. time curve (AUC), in children and adolescents was interpolated based on pharmacokinetic parameter distributions obtained from the model describing infants, toddlers and adults (model 1). Results were compared with the values obtained from modelling of the data in the corresponding population (model 2). RESULTS: The two pharmacokinetic models accurately described midazolam exposure in the population on which they were built. However, the model based on data from infants, toddlers and adults failed to predict the exposure observed in children and adolescents: the mean difference between the predicted and estimated AUC(0-180) was of -17.8%, with a range of -6.8 to -38.4%.The discrepancy between estimated and interpolated exposure increased proportionally with body weight. CONCLUSIONS: The current results indicate that irrespective of whether extrapolation or interpolation methods are to be applied during paediatric drug development, model predictions beyond the range of the data used for parameter estimation may be biased. For accurate inter- or extrapolation to different populations, the assumption of identical parameter-covariate correlations across age groups may not be taken for granted.


Subject(s)
Hypnotics and Sedatives/pharmacokinetics , Midazolam/pharmacokinetics , Models, Biological , Adolescent , Adult , Age Factors , Area Under Curve , Body Weight , Child , Child, Preschool , Double-Blind Method , Female , Humans , Infant , Male , Pilot Projects , Young Adult
9.
Br J Clin Pharmacol ; 74(2): 346-53, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22256787

ABSTRACT

AIMS: To assess whether an adaptive design in early clinical trials based on the paradigm of variable dosing and controlled exposure can provide better dosing recommendations compared with the standard fixed dose approach. METHODS: In a clinical trial simulation setting, a paediatric study was simulated using a pharmacokinetic model previously developed for abacavir. Plasma concentrations following the current recommended dose (8 mg kg⁻¹) were taken at standard sampling times, exposures (AUC) were calculated and doses individually adapted to reach the target exposure (i.e. effective exposure in adults). A second round of simulations followed with the adapted doses, and the resulting concentrations were fitted again with the same model. Exposure distributions in both conditions (i.e. fixed dose and controlled exposure) were compared with the target exposure. RESULTS: The AUC distribution after the current dose resulted in a median exposure of 6.43 mg h l⁻¹ (90th percentile 3.13-10.67 mg h l⁻¹). A total of 61 of 128 subjects showed AUC values either too low or to high compared with the target exposure. After dose adjustment, the median exposure was 6.94 mg h l⁻¹ (5.57-8.25 mg h l⁻¹), and only 14 subjects deviated from the target range. CONCLUSIONS: Adaptive randomization can be used to optimize dosing regimens in early paediatric clinical trials. The randomization of patients to target exposure rather than dose increases the probability of demonstrating efficacy (i.e. study power) compared with dose-controlled trials. Furthermore, it contributes to further understanding of the role of dose on the total heterogeneity in clinical response.


Subject(s)
Aging/metabolism , Dideoxynucleosides/pharmacokinetics , Randomized Controlled Trials as Topic/methods , Research Design , Reverse Transcriptase Inhibitors/pharmacokinetics , Adolescent , Age Factors , Area Under Curve , Child , Child, Preschool , Computer Simulation , Dideoxynucleosides/administration & dosage , Dideoxynucleosides/adverse effects , Dose-Response Relationship, Drug , Drug Dosage Calculations , Humans , Medication Errors/prevention & control , Models, Biological , Patient Safety , Reverse Transcriptase Inhibitors/administration & dosage , Reverse Transcriptase Inhibitors/adverse effects , Uncertainty
10.
Br J Clin Pharmacol ; 73(4): 641-50, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21988586

ABSTRACT

WHAT IS ALREADY KNOWN ABOUT THIS SUBJECT: Abacavir is used to treat HIV infection in both adults and children. The recommended paediatric dose is 8 mg kg(-1) twice daily up to a maximum of 300 mg twice daily. Weight was identified as the central covariate influencing pharmacokinetics of abacavir in children. WHAT THIS STUDY ADDS: A population pharmacokinetic model was developed to describe both once and twice daily pharmacokinetic profiles of abacavir in infants and toddlers. Standard dosage regimen is associated with large interindividual variability in abacavir concentrations. A maximum a posteriori probability Bayesian estimator of AUC(0-) (t) based on three time points (0, 1 or 2, and 3 h) is proposed to support area under the concentration-time curve (AUC) targeted individualized therapy in infants and toddlers. AIMS: To develop a population pharmacokinetic model for abacavir in HIV-infected infants and toddlers, which will be used to describe both once and twice daily pharmacokinetic profiles, identify covariates that explain variability and propose optimal time points to optimize the area under the concentration-time curve (AUC) targeted dosage and individualize therapy. METHODS: The pharmacokinetics of abacavir was described with plasma concentrations from 23 patients using nonlinear mixed-effects modelling (NONMEM) software. A two-compartment model with first-order absorption and elimination was developed. The final model was validated using bootstrap, visual predictive check and normalized prediction distribution errors. The Bayesian estimator was validated using the cross-validation and simulation-estimation method. RESULTS: The typical population pharmacokinetic parameters and relative standard errors (RSE) were apparent systemic clearance (CL) 13.4 () h−1 (RSE 6.3%), apparent central volume of distribution 4.94 () (RSE 28.7%), apparent peripheral volume of distribution 8.12 () (RSE14.2%), apparent intercompartment clearance 1.25 () h−1 (RSE 16.9%) and absorption rate constant 0.758 h−1 (RSE 5.8%). The covariate analysis identified weight as the individual factor influencing the apparent oral clearance: CL = 13.4 × (weight/12)1.14. The maximum a posteriori probability Bayesian estimator, based on three concentrations measured at 0, 1 or 2, and 3 h after drug intake allowed predicting individual AUC0­t. CONCLUSIONS: The population pharmacokinetic model developed for abacavir in HIV-infected infants and toddlers accurately described both once and twice daily pharmacokinetic profiles. The maximum a posteriori probability Bayesian estimator of AUC(0-) (t) was developed from the final model and can be used routinely to optimize individual dosing.


Subject(s)
Anti-HIV Agents/pharmacokinetics , Dideoxynucleosides/pharmacokinetics , HIV Infections/metabolism , Area Under Curve , Bayes Theorem , Body Weight , Child, Preschool , Dose-Response Relationship, Drug , Drug Dosage Calculations , Female , HIV Infections/drug therapy , Humans , Infant , Male , Models, Biological
11.
Br J Clin Pharmacol ; 72(3): 454-64, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21501213

ABSTRACT

AIMS: To assess the predictive value of a model-based approach for dose selection across paediatric populations in early clinical drug development. METHODS: Abacavir was selected as a paradigm compound using data across a wide age range. Abacavir pharmacokinetics (PK) in children were analysed separately from infants and toddlers. Two independent models were obtained, and systemic exposure (AUC) was then simulated across populations based on the estimates from each model. Drug exposures in infants and toddlers were predicted using pharmacokinetic parameter distributions obtained from children, and the other way around. RESULTS: The pharmacokinetic models (a two-compartment PK model for infants and toddlers and a one compartment PK model for children) accurately described the exposure in the population from which they were built. However, neither model predicted exposure in a different population: in infants, the median AUC (95%(-) CI) was estimated at 7.03 (6.72, 7.48) µg ml(-1) h, whilst it was predicted at 5.75 (4.82, 6.26) µg ml(-1) h; in children, the estimated median AUC was 6.96 (5.85, 7.91) µg ml(-1) h, whilst the predicted value was 6.45 (5.80, 7.01) µg ml(-1) h. CONCLUSIONS: These findings suggest that the assumption of an identical (linear or nonlinear) correlation between pharmacokinetic parameters and demographic factors may not hold true across age groups. Whilst the use of modelling enables accurate characterization of pharmacokinetic properties, extrapolations based on such parameter estimates may have limited value due to differences in the impact of developmental growth across populations.


Subject(s)
Anti-HIV Agents/pharmacokinetics , Dideoxynucleosides/pharmacokinetics , Drug Evaluation/methods , Models, Biological , Age Factors , Area Under Curve , Child , Child, Preschool , Data Interpretation, Statistical , Drug Approval/methods , Female , Humans , Infant , Male
13.
Clin Pharmacokinet ; 49(4): 269-75, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20214410

ABSTRACT

For propofol clearance, allometric scaling has been applied successfully for extrapolations between species (rats and humans) and within the human bodyweight range (children and adults). In this analysis, the human bodyweight range is explored to determine for which range an allometric model with a fixed or estimated exponent can be used to predict propofol clearance, without correction for maturation. The predictive value of the allometric equation, clearance (CL) is equal to 0.071 x bodyweight in kg0.78, which was developed from rats, children and adults, and the predictive value of a fixed exponent allometric model derived from the basal metabolic rate, CL is equal to CL standardized to a 70 kg adult x (bodyweight in kg standardized to a 70 kg adult)0.75, were evaluated across five independent patient groups including (i) 25 (pre)term neonates with a postmenstrual age of 27-43 weeks; (ii) 22 postoperative infants aged 4-18 months; (iii) 12 toddlers aged 1-3 years; (iv) 14 adolescents aged 10-20 years; and (v) 26 critically ill adults sedated long term. The median percentage error of the predictions was calculated using the equation %error = (CL(allometric) - CL(i))/CL(i) x 100, where CL(allometric) is the predicted propofol clearance from the allometric equations for each individual and CL(i) is the individual-predicted (post hoc) propofol clearance value derived from published population pharmacokinetic models. In neonates, the allometric model developed from rats, children and adults, and the fixed-exponent allometric model, systematically overpredicted individual propofol clearance, with median percentage errors of 288% and 216%, respectively, whereas in infants, both models systematically underpredicted individual propofol clearance, with median percentage errors of -43% and -55%, respectively. In toddlers, adolescents and adults, both models performed reasonably well, with median percentage errors of -12% and -32%, respectively, in toddlers, 16% and -14%, respectively, in adolescents, and 12% and -18%, respectively, in adults. Both allometric models based on bodyweight alone may be of use to predict propofol clearance in individuals older than 2 years. Approaches that also incorporate maturation are required to predict clearance under the age of 2 years.


Subject(s)
Anesthetics, Intravenous/pharmacokinetics , Models, Biological , Propofol/pharmacokinetics , Adolescent , Adult , Age Factors , Aged , Aged, 80 and over , Algorithms , Animals , Body Weight , Child , Child, Preschool , Humans , Infant , Infant, Newborn , Infant, Premature , Metabolic Clearance Rate , Middle Aged , Rats , Species Specificity
14.
Expert Opin Pharmacother ; 8(12): 1787-99, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17696784

ABSTRACT

The area of pharmacogenetics (PGt) is evolving rapidly. However, ongoing efforts in this field are not aligned with the requirements for the inclusion of clinically relevant findings into the label, especially with reference to paediatric indications. Clinical research in children poses unique issues from a practical and technical perspective, but many challenges can be overcome by applying advanced study design and data analysis methods. When investigating the role of PGt factors on treatment effect, all features that influence drug response must be taken into account. Yet, PGt often has a privileged status in research protocols, with PGt factors evaluated independently from other determinants of response, instead of being regarded as other demographic or clinical covariates (e.g., age, renal function). At present, guidelines to incorporate PGt findings into label statements are lacking in part because this is a new and incompletely understood area. This situation is no longer acceptable. To achieve the potential that PGt can offer to drug development and ultimately to drug prescription, academia, industry and regulatory agencies need to pool resources on the revision of study design and data analysis requisites, bringing in model-based methodologies to enable accurate interpretation of results and provide appropriate labelling recommendations.


Subject(s)
Drug Design , Drug Labeling , Pharmacogenetics , Age Factors , Child , Clinical Trials as Topic/methods , Dose-Response Relationship, Drug , Humans , Pharmaceutical Preparations/administration & dosage , Polymorphism, Genetic , Research Design
SELECTION OF CITATIONS
SEARCH DETAIL