1
|
Bai J, Wen A, Li Z, Li X, Duan M. Population pharmacokinetics and dosing optimisation of imipenem in critically ill patients. Eur J Hosp Pharm 2024; 31:434-439. [PMID: 36948580 PMCID: PMC11347199 DOI: 10.1136/ejhpharm-2022-003403] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 02/06/2023] [Indexed: 03/24/2023] Open
Abstract
OBJECTIVE The objective of this study was to explore factors that affect the clearance of imipenem in critically ill patients and to provide a dosing regimen for such patients. METHODS A prospective open-label study enrolled 51 critically ill patients with sepsis. Patients were between the ages of 18 and 96. Blood samples were collected in duplicate before (0 hour) and at 0.5, 1, 1.5, 2, 3, 4, 6, and 8 hours after imipenem administration. The plasma imipenem concentration was determined by the high-performance liquid chromatography-ultraviolet detection (HPLC-UV) method. A population pharmacokinetic (PPK) model was developed using nonlinear mixed-effects modelling methods to identify covariates. Monte Carlo simulations were performed using the final PPK model to explore the effect of different dosing regimens on the probability of target attainment (PTA). RESULTS The imipenem concentration data were best described by a two-compartment model. Creatinine clearance (CrCl, mL/min) was a covariate that affected central clearance (CLc). Patients were divided into four subgroups based on different CrCl rates. Monte Carlo simulations were performed to assess the PTA differences between empirical dosing regimens (0.5 g every 6 hours (q6h), 0.5 g every 8 hours (q8h), 0.5 g every 12 hours (q12h), 1 g every 6 hours (q6h), 1 g every 8 hours (q8h), and 1 g every 12 hours (q12h)) and to determine the target achievement rate covariate. CONCLUSION This study identified covariates for CLc, and the proposed final model can be used to guide clinicians administering imipenem in this particular patient population.
Collapse
Affiliation(s)
- Jing Bai
- Department of Intensive Care Unit, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Aiping Wen
- Department of Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Zhe Li
- Department of Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Xingang Li
- Department of Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Meili Duan
- Department of Intensive Care Unit, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
2
|
Yang R, Ding Q, Ding J, Zhu L, Pei Q. Physiologically based pharmacokinetic modeling in obesity: applications and challenges. Expert Opin Drug Metab Toxicol 2024:1-12. [PMID: 39101366 DOI: 10.1080/17425255.2024.2388690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 07/11/2024] [Accepted: 08/01/2024] [Indexed: 08/06/2024]
Abstract
INTRODUCTION Rising global obesity rates pose a threat to people's health. Obesity causes a series of pathophysiologic changes, making the response of patients with obesity to drugs different from that of nonobese, thus affecting the treatment efficacy and even leading to adverse events. Therefore, understanding obesity's effects on pharmacokinetics is essential for the rational use of drugs in patients with obesity. AREAS COVERED Articles related to physiologically based pharmacokinetic (PBPK) modeling in patients with obesity from inception to October 2023 were searched in PubMed, Embase, Web of Science and the Cochrane Library. This review outlines PBPK modeling applications in exploring factors influencing obesity's effects on pharmacokinetics, guiding clinical drug development and evaluating and optimizing clinical use of drugs in patients with obesity. EXPERT OPINION Obesity-induced pathophysiologic alterations impact drug pharmacokinetics and drug-drug interactions (DDIs), altering drug exposure. However, there is a lack of universal body size indices or quantitative pharmacology models to predict the optimal for the patients with obesity. Therefore, dosage regimens for patients with obesity must consider individual physiological and biochemical information, and clinically individualize therapeutic drug monitoring for highly variable drugs to ensure effective drug dosing and avoid adverse effects.
Collapse
Affiliation(s)
- Ruwei Yang
- Department of Pharmacy, The Third XiangyHospital, Central South University, Changsha, Hunan, China
| | - Qin Ding
- Department of Pharmacy, The Third XiangyHospital, Central South University, Changsha, Hunan, China
| | - Junjie Ding
- Center for Tropical Medicine and Global Health, Oxford Medical School, Oxford, UK
| | - Liyong Zhu
- Department of Gastrointestinal Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qi Pei
- Department of Pharmacy, The Third XiangyHospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
3
|
Simeoli R, Lava SAG, Di Deo A, Roversi M, Cairoli S, Tambucci R, Rea F, Malamisura M, Angelino G, Biondi I, Simonetti A, De Angelis P, Dionisi Vici C, Rossi P, Pontrelli G, Della Pasqua O, Goffredo BM. Pharmacokinetic Evaluation of Oral Viscous Budesonide in Paediatric Patients with Eosinophilic Oesophagitis in Repaired Oesophageal Atresia. Pharmaceutics 2024; 16:872. [PMID: 39065569 PMCID: PMC11280286 DOI: 10.3390/pharmaceutics16070872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 06/24/2024] [Accepted: 06/27/2024] [Indexed: 07/28/2024] Open
Abstract
Eosinophilic oesophagitis is a long-term complication of oesophageal atresia (EA), an uncommon condition that affects approximately 1 in 3500 infants. An exploratory, open-label phase 2 clinical trial was conducted in paediatric eosinophilic oesophagitis after oesophageal atresia (EoE-EA) to assess the safety, pharmacokinetics, and efficacy of oral viscous budesonide (OVB). In total, eight patients were enrolled in the study and assigned to a twice-daily dosing regimen of either 0.8 or 1 mg OVB, depending on age and height, administered for 12 weeks. OVB was safe and effective in the treatment of EoE-EA. The current investigation focuses on the pharmacokinetics of budesonide and the impact of an oral viscous formulation on its absorption and bioavailability. Using a non-linear mixed effects approach, two distinct absorption profiles were identified, despite marked interindividual variability in drug concentrations. Budesonide exposure was higher than previously reported in children following oral inhalation. Even though no significant effect has been observed on serum cortisol levels, future studies should consider exploring different doses, schedules, and/or treatment durations, as there may be an opportunity to reduce the risk of cortisol suppression.
Collapse
Affiliation(s)
- Raffaele Simeoli
- Division of Metabolic Diseases, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (R.S.); (S.C.); (C.D.V.)
| | - Sebastiano A. G. Lava
- Clinical Pharmacology & Therapeutics Group, University College London, London WC1N 3JH, UK; (S.A.G.L.); (A.D.D.); (O.D.P.)
| | - Alessandro Di Deo
- Clinical Pharmacology & Therapeutics Group, University College London, London WC1N 3JH, UK; (S.A.G.L.); (A.D.D.); (O.D.P.)
| | - Marco Roversi
- PhD Program in Immunology, Molecular Medicine and Applied Biotechnology, University of Rome Tor Vergata, 00133 Rome, Italy;
| | - Sara Cairoli
- Division of Metabolic Diseases, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (R.S.); (S.C.); (C.D.V.)
| | - Renato Tambucci
- Digestive Endoscopy Unit, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (R.T.); (F.R.); (M.M.); (G.A.); (P.D.A.)
| | - Francesca Rea
- Digestive Endoscopy Unit, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (R.T.); (F.R.); (M.M.); (G.A.); (P.D.A.)
| | - Monica Malamisura
- Digestive Endoscopy Unit, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (R.T.); (F.R.); (M.M.); (G.A.); (P.D.A.)
| | - Giulia Angelino
- Digestive Endoscopy Unit, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (R.T.); (F.R.); (M.M.); (G.A.); (P.D.A.)
| | - Isabella Biondi
- Centre of Excellence for the Development and Implementation of Medicines, Vaccines, and Medical Devices for Pediatric Use, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (I.B.); (A.S.); (P.R.); (G.P.)
| | - Alessandra Simonetti
- Centre of Excellence for the Development and Implementation of Medicines, Vaccines, and Medical Devices for Pediatric Use, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (I.B.); (A.S.); (P.R.); (G.P.)
| | - Paola De Angelis
- Digestive Endoscopy Unit, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (R.T.); (F.R.); (M.M.); (G.A.); (P.D.A.)
| | - Carlo Dionisi Vici
- Division of Metabolic Diseases, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (R.S.); (S.C.); (C.D.V.)
| | - Paolo Rossi
- Centre of Excellence for the Development and Implementation of Medicines, Vaccines, and Medical Devices for Pediatric Use, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (I.B.); (A.S.); (P.R.); (G.P.)
| | - Giuseppe Pontrelli
- Centre of Excellence for the Development and Implementation of Medicines, Vaccines, and Medical Devices for Pediatric Use, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (I.B.); (A.S.); (P.R.); (G.P.)
| | - Oscar Della Pasqua
- Clinical Pharmacology & Therapeutics Group, University College London, London WC1N 3JH, UK; (S.A.G.L.); (A.D.D.); (O.D.P.)
| | - Bianca Maria Goffredo
- Division of Metabolic Diseases, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (R.S.); (S.C.); (C.D.V.)
| |
Collapse
|
4
|
D'Agate S, Ruiz Gabarre D, Della Pasqua O. Population pharmacokinetics and dose rationale for aciclovir in term and pre-term neonates with herpes. Pharmacol Res Perspect 2024; 12:e1193. [PMID: 38775304 PMCID: PMC11110484 DOI: 10.1002/prp2.1193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 01/23/2024] [Indexed: 05/25/2024] Open
Abstract
Aciclovir is considered the first-line treatment against Herpes simplex virus (HSV) infections in new-borns and infants. As renal excretion is the major route of elimination, in renally-impaired patients, aciclovir doses are adjusted according to the degree of impairment. However, limited attention has been given to the implications of immature renal function or dysfunction due to the viral disease itself. The aim of this investigation was to characterize the pharmacokinetics of aciclovir taking into account maturation and disease processes in the neonatal population. Pharmacokinetic data obtained from 2 previously published clinical trials (n = 28) were analyzed using a nonlinear mixed effects modeling approach. Post-menstrual age (PMA) and creatinine clearance (CLCR) were assessed as descriptors of maturation and renal function. Simulation scenarios were also implemented to illustrate the use of pharmacokinetic data to extrapolate efficacy from adults. Aciclovir pharmacokinetics was described by a one-compartment model with first-order elimination. Body weight and diagnosis (systemic infection) were statistically significant covariates on the volume of distribution, whereas body weight, CLCR and PMA had a significant effect on clearance. Median clearance varied from 0.2 to 1.0 L/h in subjects with PMA <34 or ≥34 weeks, respectively. Population estimate for volume of distribution was 1.93 L with systemic infection increasing this value by almost 3-fold (2.67 times higher). A suitable model parameterization was identified, which discriminates the effects of developmental growth, maturation, and organ function. Exposure to aciclovir was found to increase with decreasing PMA and renal function (CLCR), suggesting different dosing requirement for pre-term neonates.
Collapse
Affiliation(s)
- S. D'Agate
- Clinical Pharmacology & Therapeutics GroupUniversity College LondonLondonUK
| | - D. Ruiz Gabarre
- Clinical Pharmacology & Therapeutics GroupUniversity College LondonLondonUK
- Present address:
Institute for Regeneration and RepairUniversity of EdinburghEdinburghUK
| | - O. Della Pasqua
- Clinical Pharmacology & Therapeutics GroupUniversity College LondonLondonUK
| |
Collapse
|
5
|
Lava SAG, Laurence C, Di Deo A, Sekarski N, Burch M, Della Pasqua O. Dapagliflozin and Empagliflozin in Paediatric Indications: A Systematic Review. Paediatr Drugs 2024; 26:229-243. [PMID: 38635113 DOI: 10.1007/s40272-024-00623-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/27/2024] [Indexed: 04/19/2024]
Abstract
INTRODUCTION In adults, sodium-glucose cotransporter type 2 inhibitors have revolutionised the treatment of type 2 diabetes mellitus, heart failure, and chronic kidney disease. OBJECTIVE We aimed to review information on compassionate use, clinical pharmacology, efficacy, and safety of dapagliflozin and empagliflozin in children. METHODS We conducted a systematic review of published clinical trials, case reports, and observational studies in Medline, Excerpta Medica, and Web of Science databases from inception to September 2023. For the two randomised controlled trials on type 2 diabetes mellitus (T2DM), we implemented a meta-analysis on the primary outcome (mean difference in glycosylated haemoglobin [HbA1c] between intervention and placebo groups). Review Manager (RevMan), version 5.4.1, was used for this purpose. RESULTS Thirty-five articles (nine case reports, ten case series, one prospective non-controlled trial, four controlled randomised trials, two surveys, six pharmacokinetic studies, and three pharmacovigilance studies) were selected, in which 415 children were exposed to either dapagliflozin or empagliflozin: 189 diabetic patients (mean age 14.7 ± 2.9 years), 32 children with glycogen storage disease type Ib (GSD Ib), glucose-6-phosphatase catalytic subunit 3 (G6PC3) deficiency, or severe congenital neutropenia type 4 (8.5 ± 5.1 years), 47 children with kidney disease or heart failure (11.2 ± 6.1 years), 84 patients in pharmacokinetic studies (15.1 ± 2.3 years), and 63 patients in toxicological series. The effect of dapagliflozin and empagliflozin in T2DM was demonstrated by HbA1c reduction in two randomised trials among a total of 177 adolescents, with a mean HbA1c difference of -0.82% (95% confidence interval -1.34 to -0.29) as compared to placebo (no heterogeneity, I2 = 0%). Dosage ranged between 5 and 20 mg (mean 11.4 ± 3.7) once daily for dapagliflozin and between 5 and 25 mg (mean 15.4 ± 7.4) once daily for empagliflozin. Among the paediatric cases of GSD Ib, empagliflozin 0.1-1.3 mg/kg/day improved neutropenia, infections, and gastrointestinal health. Dapagliflozin (mean dosage 6.9 ± 5.2 mg once daily) was well-tolerated in children with chronic kidney disease and heart failure. Side effects were generally mild, the most frequent being hypoglycaemia in children with GSD Ib (33% of patients) or T2DM (14% of patients) on concomitant hypoglycaemic drugs. Diabetic ketoacidosis is rare in children. CONCLUSION Early evidence suggests that dapagliflozin and empagliflozin are well tolerated in children. A clinical pharmacology rationale currently exists only for adolescents with diabetes mellitus. PROSPERO REGISTRATION NUMBER CRD42023438162.
Collapse
Affiliation(s)
- Sebastiano A G Lava
- Pediatric Cardiology Unit, Department of Pediatrics, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Rue du Bugnon 46, 1011, Lausanne, Switzerland.
- Heart Failure and Transplantation, Department of Paediatric Cardiology, Great Ormond Street Hospital, London, UK.
- Clinical Pharmacology and Therapeutics Group, University College London, London, UK.
- Division of Clinical Pharmacology and Toxicology, Institute of Pharmacological Sciences of Southern Switzerland, Ente Ospedaliero Cantonale, Lugano, Switzerland.
| | - Craig Laurence
- Heart Failure and Transplantation, Department of Paediatric Cardiology, Great Ormond Street Hospital, London, UK
| | - Alessandro Di Deo
- Clinical Pharmacology and Therapeutics Group, University College London, London, UK
| | - Nicole Sekarski
- Pediatric Cardiology Unit, Department of Pediatrics, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Rue du Bugnon 46, 1011, Lausanne, Switzerland
| | - Michael Burch
- Heart Failure and Transplantation, Department of Paediatric Cardiology, Great Ormond Street Hospital, London, UK
| | - Oscar Della Pasqua
- Clinical Pharmacology and Therapeutics Group, University College London, London, UK
| |
Collapse
|
6
|
Chacko IA, Ramachandran G, Sudheesh MS. Unmet technological demands in orodispersible films for age-appropriate paediatric drug delivery. Drug Deliv Transl Res 2024; 14:841-857. [PMID: 37957474 DOI: 10.1007/s13346-023-01451-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/11/2023] [Indexed: 11/15/2023]
Abstract
Age-appropriateness of a formulation is the ability to deliver variable but accurate doses to the paediatric population in a safe and acceptable manner to improve medical adherence and reduce medication errors. Paediatric drug delivery is a challenging area of formulation research due to the existing gap in knowledge. This includes the unknown safety of excipients in the paediatric population, the need for an age-appropriate formulation, the lack of an effective taste-masking method and the lack of paediatric pharmacokinetic data and patient acceptability. It is equally important to establish methods for predicting the biopharmaceutical performance of a paediatric formulation as a function of age. Overcoming the challenges of existing technologies and providing custom-made solutions for the development of age-appropriate formulation is, therefore, a daunting task. Orodispersible films (ODF) are promising as age-appropriate formulations, an unmet need in paediatric drug delivery. New technological improvements in taste masking, improving solubility and rate of dissolution of insoluble drugs, the flexibility of dosing and extemporaneous preparation of these films in a hospital good manufacturing practises (GMP) setup using 3D printing can increase its acceptance among clinicians, patients and caregivers. The current review discusses the problems and possibilities in ODF technology to address the outstanding issues of age-appropriateness, which is the hallmark of patient acceptance and medical adherence in paediatrics.
Collapse
Affiliation(s)
- Indhu Annie Chacko
- Department of Pharmaceutics, Amrita School of Pharmacy, AIMS Health Sciences Campus, Amrita Vishwa Vidyapeetham, 682041, Ponekkara, Kochi, India
| | - Gayathri Ramachandran
- Department of Pharmaceutics, Amrita School of Pharmacy, AIMS Health Sciences Campus, Amrita Vishwa Vidyapeetham, 682041, Ponekkara, Kochi, India
| | - M S Sudheesh
- Department of Pharmaceutics, Amrita School of Pharmacy, AIMS Health Sciences Campus, Amrita Vishwa Vidyapeetham, 682041, Ponekkara, Kochi, India.
| |
Collapse
|
7
|
Wang G, Sun Q, Li X, Mei S, Li S, Li Z. A Cross-sectional Comparative Analysis of Eleven Population Pharmacokinetic Models for Docetaxel in Chinese Breast Cancer Patients. Curr Drug Metab 2024; 25:479-488. [PMID: 39161139 PMCID: PMC11826906 DOI: 10.2174/0113892002322494240816032948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 07/19/2024] [Accepted: 07/31/2024] [Indexed: 08/21/2024]
Abstract
OBJECTIVE Various population pharmacokinetic (PPK) models have been established to help determine the appropriate dosage of docetaxel, however, no clear consensus on optimal dosing has been achieved. The purpose of this study is to perform an external evaluation of published models in order to test their predictive performance, and to find an appropriate PPK model for Chinese breast cancer patients. METHODS A systematic literature search of docetaxel PPK models was performed using PubMed, Web of Science, China National Knowledge Infrastructure, and WanFang databases. The predictive performance of eleven identified models was evaluated using prediction-based and simulation-based diagnostics on an independent dataset (112 docetaxel concentrations from 56 breast cancer patients). The -2×log (likelihood) and Akaike information criterion were also calculated to evaluate model fit. RESULTS The median prediction error of eight of the eleven models was less than 10%. The model fitting results showed that the three-compartment model of Bruno et al. had the best prediction performance and that the three compartment model of Wang et al. had the best simulation effect. Furthermore, although the covariates that significantly affect PK parameters were different between them, seven models demonstrated that docetaxel PK parameters were influenced by liver function. CONCLUSIONS Three compartment PPK models may be predictive of optimal docetaxel dosage for Chinese breast cancer patients. However, for patients with impaired liver function, the choice of which model to use to predict the blood concentration of docetaxel still requires great care.
Collapse
Affiliation(s)
- Genzhu Wang
- Electric Power Teaching Hospital, Capital Medical University, Beijing, 100073, China
| | - Qiang Sun
- Electric Power Teaching Hospital, Capital Medical University, Beijing, 100073, China
| | - Xiaojing Li
- Electric Power Teaching Hospital, Capital Medical University, Beijing, 100073, China
| | - Shenghui Mei
- Beijing Tiantan Hospital,Capital Medical University, Beijing, 100070, China
| | - Shihui Li
- Electric Power Teaching Hospital, Capital Medical University, Beijing, 100073, China
| | - Zhongdong Li
- Electric Power Teaching Hospital, Capital Medical University, Beijing, 100073, China
| |
Collapse
|
8
|
Healy P, Verrest L, Felisi M, Ceci A, Della Pasqua O. Dose rationale for gabapentin and tramadol in pediatric patients with chronic pain. Pharmacol Res Perspect 2023; 11:e01138. [PMID: 37803937 PMCID: PMC10558965 DOI: 10.1002/prp2.1138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 07/14/2023] [Indexed: 10/08/2023] Open
Abstract
Despite off-label use, the efficacy and safety of gabapentin and tramadol in pediatric patients (3 months to <18 years old) diagnosed with chronic pain has not been characterized. However, generating evidence based on randomized clinical trials in this population has been extremely challenging. The current investigation illustrates the use of clinical trial simulations (CTSs) as a tool for optimizing doses and protocol design for a prospective investigation in pediatric patients with chronic pain. Pharmacokinetic (PK) modeling and CTSs were used to describe the PKs of gabapentin and tramadol in the target population. In the absence of biomarkers of analgesia, systemic exposure (AUC, Css) was used to guide dose selection under the assumption of a comparable exposure-response (PKPD) relationship for either compound between adults and children. Two weight bands were identified for gabapentin, with doses titrated from 5 to 63 mg/kg. This yields gabapentin exposures (AUC0-8 ) of approximately 35 mg/L*h (1200 mg/day adult dose equivalent). For tramadol, median steady state concentrations between 200 and 300 ng/mL were achieved after doses of 2-5 mg/kg, but concentrations showed high interindividual variability. Simulation scenarios showed that titration steps are required to explore therapeutically relevant dose ranges taking into account the safety profile of both drugs. Gabapentin can be used t.i.d. at doses between 7-63 and 5-45 mg/kg for patients receiving gabapentin weighing <15 and ≥15 kg, respectively, whereas a t.i.d. regimen with doses between 1 and 5 mg/kg can be used for tramadol in patients who are not fast metabolisers.
Collapse
Affiliation(s)
- Paul Healy
- Clinical Pharmacology & Therapeutics Group, School of PharmacyUniversity College LondonLondonUK
| | - Luka Verrest
- Clinical Pharmacology & Therapeutics Group, School of PharmacyUniversity College LondonLondonUK
| | | | - Adriana Ceci
- Fondazione per la Ricerca Farmacologica Gianni Benzi onlusValenzanoItaly
| | - Oscar Della Pasqua
- Clinical Pharmacology & Therapeutics Group, School of PharmacyUniversity College LondonLondonUK
| |
Collapse
|
9
|
Staubach P, Alvaro-Lozano M, Sekerel BE, Maurer M, Ben-Shoshan M, Porter M, Hua E, Ji Y, Burciu A, Savelieva M, Severin T, Drollmann A, Bienczak A. Ligelizumab in adolescents with chronic spontaneous urticaria: Results of a dedicated phase 2b randomized clinical trial supported with pharmacometric analysis. Pediatr Allergy Immunol 2023; 34:e13982. [PMID: 37492920 DOI: 10.1111/pai.13982] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 05/27/2023] [Accepted: 05/30/2023] [Indexed: 07/27/2023]
Abstract
BACKGROUND Chronic spontaneous urticaria (CSU), a long-lasting disease in children, impacts their quality of life. We report the results of a phase 2b dose-finding trial of ligelizumab (NCT03437278) and a high-affinity humanized monoclonal anti-IgE antibody, in adolescents with CSU, supported by modeling and simulation analyses, mitigating challenges in pediatric drug development. METHODS This multicenter, double-blind, placebo-controlled trial, randomized H1-antihistamine-refractory adolescent CSU patients (12-18 years) 2:1:1 to ligelizumab 24 mg, 120 mg, or placebo every 4 weeks for 24 weeks. Patients on placebo transitioned to ligelizumab 120 mg at week 12. Integrating data from the previous adult and present adolescent trial of ligelizumab, a nonlinear mixed-effects modeling described the longitudinal changes in ligelizumab pharmacokinetics, and its effect on weekly Urticaria Activity Score (UAS7). RESULTS Baseline UAS7 (mean ± SD) was 30.5 ± 7.3 (n = 24), 29.3 ± 7.7 (n = 13), and 32.5 ± 9.0 (n = 12) for patients (median age, 15 years) on ligelizumab 24 mg, 120 mg, and placebo, respectively. Change from baseline in UAS7 at week 12 with ligelizumab 24 mg, 120 mg, and placebo was -15.7 ± 10.9, -18.4 ± 12.3, and -13.0 ± 13.0, respectively. Ligelizumab was well-tolerated. The modeling analysis showed that body weight, but not age, affected ligelizumab's apparent clearance. No significant differences between adolescents and adults were detected on the model-estimated maximum effect and potency. CONCLUSIONS Ligelizumab exhibited efficacy and safety in adolescent CSU patients, consistent with that in adults. The PK and potency of ligelizumab were not impacted by age, and the same dose of ligelizumab can be used for treating adolescents and adults with CSU. Our study shows how modeling and simulation can complement pediatric drug development.
Collapse
Affiliation(s)
- Petra Staubach
- Department of Dermatology, University Medical Center, Mainz, Germany
| | - Montserrat Alvaro-Lozano
- Department of Pediatric Allergology and Clinical Immunology, Hospital Sant Joan de Déu, Esplugues (Barcelona), Institut de Recerca Sant Joan de Déu, Universitat de Barcelona, Barcelona, Spain
| | - Bulent Enis Sekerel
- Faculty of Medicine, Pediatric Allergy and Asthma Unit, Hacettepe University, Ankara, Turkey
| | - Marcus Maurer
- Institute of Allergology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Allergology and Immunology, Berlin, Germany
| | - Moshe Ben-Shoshan
- Division of Allergy and Clinical Immunology, Department of Paediatrics, Montreal Children's Hospital McGill University Health Centre, Montreal, Quebec, Canada
| | | | - Eva Hua
- Shanghai Novartis Trading Ltd., Shanghai, China
| | - Yan Ji
- Novartis Pharmaceuticals Corporation, East Hanover, New Jersey, USA
| | | | | | | | | | | |
Collapse
|
10
|
Lim A, Sharma P, Stepanov O, Reddy VP. Application of Modelling and Simulation Approaches to Predict Pharmacokinetics of Therapeutic Monoclonal Antibodies in Pediatric Population. Pharmaceutics 2023; 15:pharmaceutics15051552. [PMID: 37242793 DOI: 10.3390/pharmaceutics15051552] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/11/2023] [Accepted: 05/17/2023] [Indexed: 05/28/2023] Open
Abstract
Ethical regulations and limited paediatric participants are key challenges that contribute to a median delay of 6 years in paediatric mAb approval. To overcome these barriers, modelling and simulation methodologies have been adopted to design optimized paediatric clinical studies and reduce patient burden. The classical modelling approach in paediatric pharmacokinetic studies for regulatory submissions is to apply body weight-based or body surface area-based allometric scaling to adult PK parameters derived from a popPK model to inform the paediatric dosing regimen. However, this approach is limited in its ability to account for the rapidly changing physiology in paediatrics, especially in younger infants. To overcome this limitation, PBPK modelling, which accounts for the ontogeny of key physiological processes in paediatrics, is emerging as an alternative modelling strategy. While only a few mAb PBPK models have been published, PBPK modelling shows great promise demonstrating a similar prediction accuracy to popPK modelling in an Infliximab paediatric case study. To facilitate future PBPK studies, this review consolidated comprehensive data on the ontogeny of key physiological processes in paediatric mAb disposition. To conclude, this review discussed different use-cases for pop-PK and PBPK modelling and how they can complement each other to increase confidence in pharmacokinetic predictions.
Collapse
Affiliation(s)
- Andrew Lim
- Clinical Pharmacology and Pharmacometrics, Biopharmaceuticals R&D, AstraZeneca, Cambridge CB2 8PA, UK
- Faculty of Medicine, Imperial College London, London SW7 2AZ, UK
| | - Pradeep Sharma
- Clinical Pharmacology and Pharmacometrics, Biopharmaceuticals R&D, AstraZeneca, Cambridge CB2 8PA, UK
| | - Oleg Stepanov
- Clinical Pharmacology and Pharmacometrics, Biopharmaceuticals R&D, AstraZeneca, Cambridge CB2 8PA, UK
| | - Venkatesh Pilla Reddy
- Clinical Pharmacology and Pharmacometrics, Biopharmaceuticals R&D, AstraZeneca, Cambridge CB2 8PA, UK
| |
Collapse
|
11
|
Aurich B, Apele‐Freimane D, Banaschewski T, Chouchana L, Day S, Kaguelidou F, Kelly LE, Kindblom JM, Neubert A, Wong ICK. c4c: Paediatric pharmacovigilance: Methodological considerations in research and development of medicines for children - A c4c expert group white paper. Br J Clin Pharmacol 2022; 88:4997-5016. [PMID: 34699077 PMCID: PMC9788092 DOI: 10.1111/bcp.15119] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 10/12/2021] [Accepted: 10/15/2021] [Indexed: 12/30/2022] Open
Abstract
Children frequently respond differently to therapies compared to adults. Differences also exist between paediatric age groups for pharmacokinetics and pharmacodynamics in both efficacy and safety. Paediatric pharmacovigilance requires an understanding of the unique aspects of children with regard to, for example, drug response, growth and development, clinical presentation of adverse drug reactions (ADRs), how they can be detected and population-specific factors (e.g., more frequent use of off-label/unlicensed drugs). In recognition of these challenges, a group of experts has been formed in the context of the conect4children (c4c) project to support paediatric drug development. This expert group collaborated to develop methodological considerations for paediatric drug safety and pharmacovigilance throughout the life-cycle of medicinal products which are described in this article. These considerations include practical points to consider for the development of the paediatric section of the risk management plan (RMP), safety in paediatric protocol development, safety data collection and analysis. Furthermore, they describe the specific details of post-marketing pharmacovigilance in children using, for example, spontaneous reports, electronic health care records, registries and record-linkage, as well as the use of paediatric pharmacoepidemiology studies for risk characterisation. Next the details of the assessment of benefit-risk and challenges related to medicinal product formulation in the context of a Paediatric Investigation Plan (PIP) are presented. Finally, practical issues in paediatric signal detection and evaluation are included. This paper provides practical points to consider for paediatric pharmacovigilance throughout the life-cycle of medicinal products for RMPs, protocol development, safety data collection and analysis and PIPs.
Collapse
Affiliation(s)
- Beate Aurich
- Department of PharmacologySaint‐Louis HospitalParisFrance
| | - Dina Apele‐Freimane
- Neonatal Intensive Care, Women and Child Health ClinicPauls Stradins Clinical University HospitalRigaLatvia
| | - Tobias Banaschewski
- Department of Child and Adolescent Psychiatry, Central Institute of Mental Health, Medical Faculty MannheimUniversity of HeidelbergMannheimGermany
| | - Laurent Chouchana
- Regional Center of Pharmacovigilance, Department of PharmacologyCochin Hospital, APHP, Centre, Université de ParisParisFrance
| | - Simon Day
- Clinical Trials Consulting and Training LimitedNorth MarstonUK
| | - Florentia Kaguelidou
- Department of Pediatric Pharmacology and Pharmacogenetics, APHP, Robert Debré Hospital; Center of Clinical Investigations, INSERM CIC1426Université de ParisParisFrance
| | - Lauren E. Kelly
- Department of Paediatrics and Child Health, Max Rady College of MedicineUniversity of ManitobaWinnipegCanada
| | - Jenny M. Kindblom
- Paediatric Clinical Research Center, Queen Silvia Children's HospitalSahlgrenska University HospitalGothenburgSweden
| | - Antje Neubert
- Department of Paediatrics and Adolescent MedicineUniversity Hospital Erlangen, Friedrich‐Alexander University Erlangen‐Nürnberg (FAU)ErlangenGermany
| | - Ian C. K. Wong
- Centre for Safe Medication Practice and Research, Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of MedicineThe University of Hong KongChina,Research Department of Practice and Policy, School of PharmacyUCLLondonUK
| |
Collapse
|
12
|
Gipson DS, Troost JP, Spino C, Attalla S, Tarnoff J, Massengill S, Lafayette R, Vega-Warner V, Adler S, Gipson P, Elliott M, Kaskel F, Fermin D, Moxey-Mims M, Fine RN, Brown EJ, Reidy K, Tuttle K, Gibson K, Lemley KV, Greenbaum LA, Atkinson MA, Hingorani S, Srivastava T, Sethna CB, Meyers K, Tran C, Dell KM, Wang CS, Yee JL, Sampson MG, Gbadegesin R, Lin JJ, Brady T, Rheault M, Trachtman H. Comparing Kidney Health Outcomes in Children, Adolescents, and Adults With Focal Segmental Glomerulosclerosis. JAMA Netw Open 2022; 5:e2228701. [PMID: 36006643 PMCID: PMC9412226 DOI: 10.1001/jamanetworkopen.2022.28701] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 07/11/2022] [Indexed: 11/14/2022] Open
Abstract
Importance Focal segmental glomerulosclerosis (FSGS) is a common cause of end-stage kidney disease (ESKD) across the lifespan. While 10% to 15% of children and 3% of adults who develop ESKD have FSGS, it remains uncertain whether the natural history differs in pediatric vs adult patients, and this uncertainty contributes to the exclusion of children and adolescents in clinical trials. Objective To examine whether there are differences in the kidney health outcomes among children, adolescents, and adults with FSGS. Design, Setting, and Participants This cohort study used pooled and parallel analyses, completed July 5, 2022, from 3 complimentary data sources: (1) Nephrotic Syndrome Rare Disease Clinical Research Network (NEPTUNE); (2) FSGS clinical trial (FSGS-CT); and (3) Kidney Research Network (KRN). NEPTUNE is a multicenter US/Canada cohort study; FSGS-CT is a multicenter US/Canada clinical trial; and KRN is a multicenter US electronic health record-based registry from academic and community nephrology practices. NEPTUNE included 166 patients with incident FSGS enrolled at first kidney biopsy; FSGS-CT included 132 patients with steroid-resistant FSGS randomized to cyclosporine vs dexamethasone with mycophenolate; and KRN included 184 patients with prevalent FSGS. Data were collected from November 2004 to October 2019 and analyzed from October 2020 to July 2022. Exposures Age: children (age <13 years) vs adolescents (13-17 years) vs adults (≥18 years). Covariates of interest included sex, disease duration, APOL1 genotype, urine protein-to-creatinine ratio, estimated glomerular filtration rate (eGFR), edema, serum albumin, and immunosuppressive therapy. Main Outcomes and Measures ESKD, composite outcome of ESKD or 40% decline in eGFR, and complete and/or partial remission of proteinuria. Results The study included 127 (26%) children, 102 (21%) adolescents, and 253 (52%) adults, including 215 (45%) female participants and 138 (29%) who identified as Black, 98 (20%) who identified as Hispanic, and 275 (57%) who identified as White. Overall, the median time to ESKD was 11.9 years (IQR, 5.2-19.1 years). There was no difference in ESKD risk among children vs adults (hazard ratio [HR], 0.67; 95% CI, 0.43-1.03) or adolescents vs adults (HR, 0.85; 95% CI, 0.52-1.36). The median time to the composite end point was 5.7 years (IQR 1.6-15.2 years), with hazard ratio estimates for children vs adults of 1.12 (95% CI, 0.83-1.52) and adolescents vs adults of 1.06 (95% CI, 0.75-1.50). Conclusions and Relevance In this study, the association of FSGS with kidney survival and functional outcomes was comparable at all ages.
Collapse
Affiliation(s)
- Debbie S. Gipson
- Division of Nephrology, Department of Pediatrics, University of Michigan, Ann Arbor
| | - Jonathan P. Troost
- Michigan Institute for Clinical and Health Research, University of Michigan, Ann Arbor
| | - Cathie Spino
- School of Public Health, Department of Biostatistics, University of Michigan, Ann Arbor
| | - Samara Attalla
- Division of Nephrology, Department of Pediatrics, University of Michigan, Ann Arbor
| | - Joshua Tarnoff
- NephCure Kidney International, King of Prussia, Pennsylvania
| | - Susan Massengill
- Division of Pediatric Nephrology, Department of Pediatrics, Levine Children’s Hospital, Atrium Health, Charlotte, North Carolina
| | - Richard Lafayette
- Department of Internal Medicine, Division of Nephrology, Stanford University, Palo Alto, California
| | - Virginia Vega-Warner
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor
| | - Sharon Adler
- Division of Nephrology and Hypertension, Los Angeles Biomedical Research Institute at Harbor–University of California, Torrance
| | - Patrick Gipson
- Division of Nephrology, Department of Pediatrics, University of Michigan, Ann Arbor
| | | | - Frederick Kaskel
- Division of Nephrology, Children’s Hospital at Montefiore; Albert Einstein College of Medicine, Bronx, New York
| | - Damian Fermin
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor
| | - Marva Moxey-Mims
- Division of Nephrology, Children’s National Hospital, Department of Pediatrics, The George Washington University School of Medicine, Washington, DC
| | - Richard N. Fine
- Renaissance School of Medicine at Stony Brook University, Stony Brook University Medical Center, Stony Brook, New York
| | - Elizabeth J. Brown
- Division of Nephrology, Department of Pediatrics, UT Southwestern Medical Center, Dallas, Texas
| | - Kimberly Reidy
- Division of Nephrology, Department of Pediatrics, Albert Einstein College of Medicine, Montefiore Medical Center, New York, New York
| | - Katherine Tuttle
- Providence Medical Research Center, Providence Health Care, Spokane, Washington
- Kidney Research Institute, Nephrology Division, and Institute for Translational Health Sciences, University of Washington, Seattle
| | - Keisha Gibson
- University of North Carolina Kidney Center at Chapel Hill
| | - Kevin V. Lemley
- Department of Pediatrics, USC Keck School of Medicine, Children’s Hospital Los Angeles, Los Angeles, California
| | - Larry A. Greenbaum
- Division of Pediatric Nephrology, Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, Atlanta, Georgia
| | - Meredith A. Atkinson
- Division of Pediatric Nephrology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Sangeeta Hingorani
- Department of Pediatrics, University of Washington and Division of Nephrology, Seattle Children’s, Seattle
| | - Tarak Srivastava
- Section of Nephrology, Children’s Mercy Hospital and University of Missouri at Kansas City
| | - Christine B. Sethna
- Pediatric Nephrology, Cohen Children’s Medical Center of New York, Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York
| | - Kevin Meyers
- Division of Nephrology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Cheryl Tran
- Children’s Center, Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, Minnesota
| | - Katherine M. Dell
- Center for Pediatric Nephrology, Cleveland Clinic Children’s, Cleveland, Ohio
| | - Chia-shi Wang
- Division of Pediatric Nephrology, Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, Atlanta, Georgia
| | - Jennifer Lai Yee
- Division of Nephrology, Department of Pediatrics, University of Michigan, Ann Arbor
| | - Matthew G. Sampson
- Division of Nephrology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
- Kidney Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | | | - J. J. Lin
- Pediatric Nephrology, Wake Forest Baptist Health, Winston Salem, North Carolina
| | - Tammy Brady
- Division of Pediatric Nephrology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Michelle Rheault
- Department of Pediatrics, Division of Nephrology, University of Minnesota, Minneapolis
| | - Howard Trachtman
- Division of Nephrology, Department of Pediatrics, University of Michigan, Ann Arbor
| |
Collapse
|
13
|
Wang Y, Travis J, Gajewski B. Bayesian adaptive design for pediatric clinical trials incorporating a community of prior beliefs. BMC Med Res Methodol 2022; 22:118. [PMID: 35448963 PMCID: PMC9027907 DOI: 10.1186/s12874-022-01569-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 03/09/2022] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Pediatric population presents several barriers for clinical trial design and analysis, including ethical constraints on the sample size and slow accrual rate. Bayesian adaptive design methods could be considered to address these challenges in pediatric clinical trials. METHODS We developed an innovative Bayesian adaptive design method and demonstrated the approach as a re-design of a published phase III pediatric trial. The innovative design used early success criteria based on skeptical prior and early futility criteria based on enthusiastic prior extrapolated from a historical adult trial, and the early and late stopping boundaries were calibrated to ensure a one-sided type I error of 2.5%. We also constructed several alternative designs which incorporated only one type of prior belief and the same stopping boundaries. To identify a preferred design, we compared operating characteristics including power, expected trial size and trial duration for all the candidate adaptive designs via simulation when performing an increasing number of equally spaced interim analyses. RESULTS When performing an increasing number of equally spaced interim analyses, the innovative Bayesian adaptive trial design incorporating both skeptical and enthusiastic priors at both interim and final analyses outperforms alternative designs which only consider one type of prior belief, because it allows more reduction in sample size and trial duration while still offering good trial design properties including controlled type I error rate and sufficient power. CONCLUSIONS Designing a Bayesian adaptive pediatric trial with both skeptical and enthusiastic priors can be an efficient and robust approach for early trial stopping, thus potentially saving time and money for trial conduction.
Collapse
Affiliation(s)
- Yu Wang
- Department of Biostatistics & Data Science, University of Kansas Medical Center, Robinson 5028, 3901 Rainbow Blvd., Kansas City, KS, 66160, USA.
| | - James Travis
- Division of Biometrics II, Office of Biostatistics, Office of Translational Sciences, Center of Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, 20993, USA
| | - Byron Gajewski
- Department of Biostatistics & Data Science, University of Kansas Medical Center, Robinson 5028, 3901 Rainbow Blvd., Kansas City, KS, 66160, USA
| |
Collapse
|
14
|
Babu M, Pavithran K. Therapeutic Drug Monitoring as a Tool for Therapy Optimization. Drug Metab Lett 2022; 15:DML-EPUB-122284. [PMID: 35382721 DOI: 10.2174/1872312815666220405122021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 01/21/2022] [Accepted: 02/09/2022] [Indexed: 11/22/2022]
Abstract
The use of pharmacotherapy for improving healthcare in society is increasing. A vast majority of patients have either received subtherapeutic treatment (which could result from low pharmacokinetic) or experienced adverse effects due to the toxic levels of the drug. The medicines used to treat chronic conditions, such as epilepsy; cardiovascular diseases; and oncological, neurological, and psychiatric disorders, require routine monitoring. New targeted therapies suggest an individualized treatment that can slowly move practitioners away from the concept of a one-size-fits-all-fixed-dosing approach. Therapeutic drug use can be monitored based on pharmacokinetic, pharmacodynamic, and pharmacometric methods. Based on the experiences of therapeutic drug monitoring of various agents across the globe, we can look ahead to the possible developments of therapeutic drug monitoring in India.
Collapse
Affiliation(s)
- Merin Babu
- Department of Medical Oncology, Amrita Institute of Medical Sciences and Research Centre Amrita Vishwa Vidyapeetham, Ponekkara P.O, Kochi, Kerala, India
| | - Keechilat Pavithran
- Department of Medical Oncology, Amrita Institute of Medical Sciences and Research Centre Amrita Vishwa Vidyapeetham, Ponekkara P.O, Kochi, Kerala, India
| |
Collapse
|
15
|
Prediction of lung exposure to anti-tubercular drugs using plasma pharmacokinetic data: implications for dose selection. Eur J Pharm Sci 2022; 173:106163. [DOI: 10.1016/j.ejps.2022.106163] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 12/28/2021] [Accepted: 03/02/2022] [Indexed: 01/08/2023]
|
16
|
Wang Y, Wang X, Harlin M, Larsen F, Panni M, Yildirim M, Madera J, Arias L, Forbes A, Mustafa N, Ruiz-White I, Raoufinia A. An alternative start regimen with aripiprazole once-monthly in patients with schizophrenia: population pharmacokinetic analysis of a single-day, two-injection start with gluteal and/or deltoid intramuscular injection. Curr Med Res Opin 2021; 37:1961-1972. [PMID: 34407720 DOI: 10.1080/03007995.2021.1965974] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
BACKGROUND The single-injection start regimen for aripiprazole once-monthly 400 mg (AOM 400) in patients with schizophrenia requires a single intramuscular injection in the gluteal or deltoid site and 14 days of concurrent oral therapy. A simplified, single-day regimen of two injections at separate gluteal and/or deltoid injection sites, together with a single 20-mg dose of oral aripiprazole on the 1st day, was assessed. PATIENTS AND METHODS A previously developed population-pharmacokinetic (popPK) model for characterizing aripiprazole PK following oral administration and gluteal intramuscular depot injection was expanded to include deltoid injection. Simulations were conducted to assess PK profiles following various (including two-injection) start regimens. Postmarketing data on patients who received higher-than-recommended AOM doses were used to assess overall safety/tolerability. RESULTS The two-injection start regimen with a single concurrent oral dose displayed a comparable PK profile to the single-injection start regimen with concurrent 14-day oral administration in simulations. The safety assessment indicated the two-injection start regimen was unlikely to be associated with safety concerns beyond those expected with a single-injection start regimen. CONCLUSION These data support use of the two-injection start regimen in clinical practice to reduce reliance on daily oral administration and optimize the therapeutic benefits of AOM 400 in patients with schizophrenia.
Collapse
Affiliation(s)
- Yanlin Wang
- Otsuka Pharmaceutical Development & Commercialization, Inc, Princeton, NJ, USA
| | - Xiaofeng Wang
- Otsuka Pharmaceutical Development & Commercialization, Inc, Princeton, NJ, USA
| | - Matt Harlin
- Otsuka Pharmaceutical Development & Commercialization, Inc, Princeton, NJ, USA
| | | | - Moeen Panni
- Otsuka Pharmaceutical Europe Ltd, Wexham, UK
| | | | - Jessica Madera
- Otsuka Pharmaceutical Development & Commercialization, Inc, Princeton, NJ, USA
| | | | - Andy Forbes
- Otsuka Pharmaceutical Development & Commercialization, Inc, Princeton, NJ, USA
| | - Nihal Mustafa
- Otsuka Pharmaceutical Development & Commercialization, Inc, Princeton, NJ, USA
| | - Inez Ruiz-White
- Otsuka Pharmaceutical Development & Commercialization, Inc, Princeton, NJ, USA
| | - Arash Raoufinia
- Otsuka Pharmaceutical Development & Commercialization, Inc, Princeton, NJ, USA
| |
Collapse
|
17
|
Yoneyama K, Schmitt C, Chang T, Dhalluin C, Nagami S, Petry C, Levy GG. A Model-Based Framework to Inform the Dose Selection and Study Design of Emicizumab for Pediatric Patients With Hemophilia A. J Clin Pharmacol 2021; 62:232-244. [PMID: 34545950 PMCID: PMC9298840 DOI: 10.1002/jcph.1968] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 09/10/2021] [Indexed: 12/13/2022]
Abstract
Emicizumab is a bispecific antibody mimicking the cofactor function of activated coagulation factor VIII to prevent bleeds in patients with hemophilia A. The dose selection for the first-in-child phase III study of emicizumab was addressed by pediatric pharmacokinetic prediction using an adult/adolescent population pharmacokinetic model developed in phase I-I/II studies. The model was modified to incorporate functions describing the age-dependent increase in body weight (BW) with or without clearance maturation to account for the differences in emicizumab pharmacokinetics between adults/adolescents and children. A minimal dose anticipated to achieve in children the same target efficacious exposure as for adults/adolescents was identified when considering BW and clearance maturation. It was the same BW-based dose as for adults/adolescents and was selected for the starting dose for the pediatric study. Whether considering clearance maturation or not in addition to BW led to uncertainty in the pediatric pharmacokinetic prediction and dose selection, which informed implementation of a dose-adapting scheme in the study design. Exposure matching to adults/adolescents was ultimately achieved in children with the starting dose, indicating that consideration of clearance maturation in addition to BW provided adequate pediatric pharmacokinetic predictions for emicizumab. This pharmacokinetic finding in conjunction with exposure-response information served as a basis for the efficacy demonstrated in children, avoiding a time-consuming process for exploring an optimal pediatric dose of emicizumab. This experience indicates that a model-based framework helped optimize the pediatric dose selection and study design, thereby streamlining the development process with extrapolation, of emicizumab for children.
Collapse
Affiliation(s)
| | | | - Tiffany Chang
- Genentech, Inc., South San Francisco, California, USA
| | | | | | | | - Gallia G Levy
- Genentech, Inc., South San Francisco, California, USA
| |
Collapse
|
18
|
Model-Informed Repurposing of Medicines for SARS-CoV-2: Extrapolation of Antiviral Activity and Dose Rationale for Paediatric Patients. Pharmaceutics 2021; 13:pharmaceutics13081299. [PMID: 34452260 PMCID: PMC8399437 DOI: 10.3390/pharmaceutics13081299] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 08/03/2021] [Accepted: 08/04/2021] [Indexed: 12/17/2022] Open
Abstract
Repurposing of remdesivir and other drugs with potential antiviral activity has been the basis of numerous clinical trials aimed at SARS-CoV-2 infection in adults. However, expeditiously designed trials without careful consideration of dose rationale have often resulted in treatment failure and toxicity in the target patient population, which includes not only adults but also children. Here we show how paediatric regimens can be identified using pharmacokinetic-pharmacodynamic (PKPD) principles to establish the target exposure and evaluate the implications of dose selection for early and late intervention. Using in vitro data describing the antiviral activity and published pharmacokinetic data for the agents of interest, we apply a model-based approach to assess the exposure range required for adequate viral clearance and eradication. Pharmacokinetic parameter estimates were subsequently used with clinical trial simulations to characterise the probability target attainment (PTA) associated with enhanced antiviral activity in the lungs. Our analysis shows that neither remdesivir, nor anti-malarial drugs can achieve the desirable target exposure range based on a mg/kg dosing regimen, due to a limited safety margin and high concentrations needed to ensure the required PTA. To date, there has been limited focus on suitable interventions for children affected by COVID-19. Most clinical trials have defined doses selection criteria empirically, without thorough evaluation of the PTA. The current results illustrate how model-based approaches can be used for the integration of clinical and nonclinical data, providing a robust framework for assessing the probability of pharmacological success and consequently the dose rationale for antiviral drugs for the treatment of SARS-CoV-2 infection in children.
Collapse
|
19
|
Wang K, Jiang K, Wei X, Li Y, Wang T, Song Y. Physiologically Based Pharmacokinetic Models Are Effective Support for Pediatric Drug Development. AAPS PharmSciTech 2021; 22:208. [PMID: 34312742 PMCID: PMC8312709 DOI: 10.1208/s12249-021-02076-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 06/16/2021] [Indexed: 12/30/2022] Open
Abstract
Pediatric drug development faces many difficulties. Traditionally, pediatric drug doses are simply calculated linearly based on the body weight, age, and body surface area of adults. Due to the ontogeny of children, this simple linear scaling may lead to drug overdose in pediatric patients. The physiologically based pharmacokinetic (PBPK) model, as a mathematical model, contributes to the research and development of pediatric drugs. An example of a PBPK model guiding drug dose selection in pediatrics has emerged and has been approved by the relevant regulatory agencies. In this review, we discuss the principle of the PBPK model, emphasize the necessity of establishing a pediatric PBPK model, introduce the absorption, distribution, metabolism, and excretion of the pediatric PBPK model, and understand the various applications and related prospects of the pediatric PBPK model.
Collapse
|
20
|
Nguyen D, Shaik JS, Tai G, Tiffany C, Perry C, Dumont E, Gardiner D, Barth A, Singh R, Hossain M. Comparison between physiologically based pharmacokinetic and population pharmacokinetic modelling to select paediatric doses of gepotidacin in plague. Br J Clin Pharmacol 2021; 88:416-428. [PMID: 34289143 PMCID: PMC9293063 DOI: 10.1111/bcp.14996] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 05/03/2021] [Accepted: 05/09/2021] [Indexed: 12/31/2022] Open
Abstract
Aims To develop physiologically based pharmacokinetic (PBPK) and population pharmacokinetic (PopPK) models to predict effective doses of gepotidacin in paediatrics for the treatment of pneumonic plague (Yersinia pestis). Methods A gepotidacin PBPK model was constructed using a population‐based absorption, distribution, metabolism and excretion simulator, Simcyp®, with physicochemical and in vitro data, optimized with clinical data from a dose‐escalation intravenous (IV) study and a human mass balance study. A PopPK model was developed with pooled PK data from phase 1 studies with IV gepotidacin in healthy adults. Results For both the PopPK and PBPK models, body weight was found to be a key covariate affecting gepotidacin clearance. With PBPK, ~90% of the predicted PK for paediatrics fell between the 5th and 95th percentiles of adult values except for subjects weighing ≤5 kg. PopPK‐simulated paediatric means for Cmax and AUC(0‐τ) were similar to adult exposures across various weight brackets. The proposed dosing regimens were weight‐based for subjects ≤40 kg and fixed‐dose for subjects >40 kg. Comparison of observed and predicted exposures in adults indicated that both PBPK and PopPK models achieved similar AUC and Cmax for a given dose, but the Cmax predictions with PopPK were slightly higher than with PBPK. The two models differed on dose predictions in children <3 months old. The PopPK model may be suboptimal for low age groups due to the absence of maturation characterization of drug‐metabolizing enzymes involved with clearance in adults. Conclusions Both PBPK and PopPK approaches can reasonably predict gepotidacin exposures in children.
Collapse
Affiliation(s)
- Dung Nguyen
- GlaxoSmithKline, Collegeville, PA, United States
| | | | - Guoying Tai
- GlaxoSmithKline, Collegeville, PA, United States
| | | | | | | | | | - Aline Barth
- GlaxoSmithKline, Collegeville, PA, United States
| | | | | |
Collapse
|
21
|
Oosterholt SP, Della Pasqua O. Population pharmacokinetics of raxibacumab in healthy adult subjects. Br J Clin Pharmacol 2021; 87:4718-4725. [PMID: 33973655 DOI: 10.1111/bcp.14894] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 03/29/2021] [Accepted: 04/20/2021] [Indexed: 01/24/2023] Open
Abstract
AIMS Raxibacumab is a fully humanized monoclonal antibody that blocks the interaction of Bacillus anthracis toxins, thereby protecting target cells from its effects. Raxibacumab is approved in the USA for the treatment of adults and children with inhalational anthrax in combination with antibiotics, and for prophylaxis of inhalational anthrax. The aim of this investigation was to characterise the population pharmacokinetics and assess the effect of baseline demographic covariates on the disposition of raxibacumab. METHODS The data used for this analysis were obtained from 3 clinical trials and include 2229 blood samples from 322 healthy subjects who were randomised to receive a 40 mg/kg intravenous dose of raxibacumab over a period of 2.25 hours. Population pharmacokinetic modelling was performed using a nonlinear mixed effects approach. Secondary parameters of interest were the area under the curve, maximum concentration and the time of serum raxibacumab concentrations greater than or equimolar to the highest serum protective antigen concentrations observed for at least 28 days in any monkey challenged with B. anthracis that died. RESULTS Raxibacumab exposure in healthy subjects was described by a 2-compartment model. Interindividual variability was estimated for all model parameters, whilst residual variability was described by a proportional and additive error model. Weight was the only influential covariate with significant effect on disposition parameters. CONCLUSIONS A dose of 40 mg/kg provided comparable exposure across the overall healthy subject population. Interindividual variability in raxibacumab vs. time profiles could partially be accounted for by differences in body weight.
Collapse
Affiliation(s)
- Sean P Oosterholt
- Clinical Pharmacology & Therapeutics Group, University College London, London, UK
| | - Oscar Della Pasqua
- Clinical Pharmacology & Therapeutics Group, University College London, London, UK.,Clinical Pharmacology Modelling and Simulation, GlaxoSmithKline, Brentford, UK
| |
Collapse
|
22
|
Oosterholt SP, Della Pasqua O. Extrapolation and dosing recommendations for raxibacumab in children from birth to age <18 years. Br J Clin Pharmacol 2021; 87:4709-4717. [PMID: 33974281 DOI: 10.1111/bcp.14893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 03/29/2021] [Accepted: 03/31/2021] [Indexed: 11/27/2022] Open
Abstract
AIMS The US Food and Drug Administration's Animal Rule allows for the approval of drugs when human efficacy studies are not ethical. While the therapeutic doses of raxibacumab, a monoclonal antibody for the prophylaxis and treatment of inhalational anthrax, have been based on pharmacokinetic data from adult subjects, its disposition in children has not been investigated in clinical trials. Here we evaluate the effect of demographic covariates and maturation processes on the pharmacokinetics of raxibacumab and explore opportunities for the optimisation of paediatric doses. METHODS A population pharmacokinetic model was used as basis for the extrapolation of raxibacumab disposition from adults to children. Different extrapolation scenarios, including weight-banded dosing regimens, were considered to assess the effect of growth and maturation on the pharmacokinetic parameters of interest. Area under the concentration-time curve, maximum plasma concentration and the time of serum raxibacumab concentrations greater than or equimolar to the highest serum protective antigen concentrations observed for at least 28 days in any monkey challenged with Bacillus anthracis that died were derived and compared with the currently approved US doses. RESULTS Based on practical considerations, a weight-banded dosing regimen consisting of 4 dose levels (75 mg/kg for individuals ≤1.5 kg, 55 mg/kg for individuals <10 kg, 45 mg/kg for individuals <50 kg, 40 mg/kg for all individuals >50 kg) was required to optimise target exposure across the paediatric population. CONCLUSIONS Age-related maturation processes may affect raxibacumab clearance in very young patients. The proposed dosing regimens take into account effects of body weight and maturation processes on the elimination of raxibacumab.
Collapse
Affiliation(s)
- Sean P Oosterholt
- Clinical Pharmacology & Therapeutics Group, University College London, UK
| | - Oscar Della Pasqua
- Clinical Pharmacology & Therapeutics Group, University College London, UK.,Clinical Pharmacology Modelling and Simulation, GlaxoSmithKline, Brentford, UK
| |
Collapse
|
23
|
Michelet R, Ursino M, Boulet S, Franck S, Casilag F, Baldry M, Rolff J, van Dyk M, Wicha SG, Sirard JC, Comets E, Zohar S, Kloft C. The Use of Translational Modelling and Simulation to Develop Immunomodulatory Therapy as an Adjunct to Antibiotic Treatment in the Context of Pneumonia. Pharmaceutics 2021; 13:601. [PMID: 33922017 PMCID: PMC8143524 DOI: 10.3390/pharmaceutics13050601] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/16/2021] [Accepted: 04/20/2021] [Indexed: 11/16/2022] Open
Abstract
The treatment of respiratory tract infections is threatened by the emergence of bacterial resistance. Immunomodulatory drugs, which enhance airway innate immune defenses, may improve therapeutic outcome. In this concept paper, we aim to highlight the utility of pharmacometrics and Bayesian inference in the development of immunomodulatory therapeutic agents as an adjunct to antibiotics in the context of pneumonia. For this, two case studies of translational modelling and simulation frameworks are introduced for these types of drugs up to clinical use. First, we evaluate the pharmacokinetic/pharmacodynamic relationship of an experimental combination of amoxicillin and a TLR4 agonist, monophosphoryl lipid A, by developing a pharmacometric model accounting for interaction and potential translation to humans. Capitalizing on this knowledge and associating clinical trial extrapolation and statistical modelling approaches, we then investigate the TLR5 agonist flagellin. The resulting workflow combines expert and prior knowledge on the compound with the in vitro and in vivo data generated during exploratory studies in order to construct high-dimensional models considering the pharmacokinetics and pharmacodynamics of the compound. This workflow can be used to refine preclinical experiments, estimate the best doses for human studies, and create an adaptive knowledge-based design for the next phases of clinical development.
Collapse
Affiliation(s)
- Robin Michelet
- Department of Clinical Pharmacy & Biochemistry, Institute of Pharmacy, Freie Universitaet Berlin, 12169 Berlin, Germany; (S.F.); (C.K.)
| | - Moreno Ursino
- Unit of Clinical Epidemiology, Assistance Publique-Hôpitaux de Paris, CHU Robert Debré, Université de Paris, Sorbonne Paris-Cité, Inserm U1123 and CIC-EC 1426, F-75019 Paris, France;
- INSERM, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, F-75006 Paris, France; (S.B.); (S.Z.)
| | - Sandrine Boulet
- INSERM, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, F-75006 Paris, France; (S.B.); (S.Z.)
- HeKA, Inria, F-75006 Paris, France
| | - Sebastian Franck
- Department of Clinical Pharmacy & Biochemistry, Institute of Pharmacy, Freie Universitaet Berlin, 12169 Berlin, Germany; (S.F.); (C.K.)
- Department of Clinical Pharmacy, Institute of Pharmacy, University of Hamburg, 20146 Hamburg, Germany;
| | - Fiordiligie Casilag
- CNRS, Inserm, CHU Lille, Institute Pasteur de Lille, U1019-UMR9017-CIIL-Centre for Infection and Immunity of Lille, Université de Lille, F-59000 Lille, France; (F.C.); (M.B.); (J.-C.S.)
| | - Mara Baldry
- CNRS, Inserm, CHU Lille, Institute Pasteur de Lille, U1019-UMR9017-CIIL-Centre for Infection and Immunity of Lille, Université de Lille, F-59000 Lille, France; (F.C.); (M.B.); (J.-C.S.)
| | - Jens Rolff
- Department of Evolutionary Biology, Institute of Biology, Freie Universitaet Berlin, 14195 Berlin, Germany;
| | - Madelé van Dyk
- Flinders Centre for Innovation in Cancer, College of Medicine and Public Health, Flinders University, Adelaide 5042, Australia;
| | - Sebastian G. Wicha
- Department of Clinical Pharmacy, Institute of Pharmacy, University of Hamburg, 20146 Hamburg, Germany;
| | - Jean-Claude Sirard
- CNRS, Inserm, CHU Lille, Institute Pasteur de Lille, U1019-UMR9017-CIIL-Centre for Infection and Immunity of Lille, Université de Lille, F-59000 Lille, France; (F.C.); (M.B.); (J.-C.S.)
| | - Emmanuelle Comets
- INSERM, University Rennes-1, CIC 1414, F-35000 Rennes, France;
- INSERM, IAME, Université de Paris, F-75006 Paris, France
| | - Sarah Zohar
- INSERM, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, F-75006 Paris, France; (S.B.); (S.Z.)
- HeKA, Inria, F-75006 Paris, France
| | - Charlotte Kloft
- Department of Clinical Pharmacy & Biochemistry, Institute of Pharmacy, Freie Universitaet Berlin, 12169 Berlin, Germany; (S.F.); (C.K.)
| |
Collapse
|
24
|
Orito Y, Kakara M, Okada A, Nagai N. Model-based approach to sampling optimization in studies of antibacterial drugs for infants and young children. Clin Transl Sci 2021; 14:1543-1553. [PMID: 33742784 PMCID: PMC8301546 DOI: 10.1111/cts.13018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 02/02/2021] [Accepted: 02/17/2021] [Indexed: 11/28/2022] Open
Abstract
Clinical trials for pediatric indications and new pediatric drugs face challenges, including the limited blood volume due to the patients’ small bodies. In Japan, the Evaluation Committee on Unapproved or Off‐labeled Drugs with High Medical Needs has discussed the necessity of pediatric indications against the background of a lack of Japanese pediatric data. The limited treatment options regarding antibiotics for pediatric patients are associated with the emergence of antibiotic‐resistant bacteria. Regulatory guidelines promote the use of model‐based drug development to reduce practical and ethical constraints for pediatric patients. Sampling optimization is one of the key study designs for pediatric drug development. In this simulation study, we evaluated the precision of the empirical Bayes estimates of pharmacokinetic (PK) parameters based on the sampling times optimized by published pediatric population PK models. We selected three previous PK studies of cefepime and ciprofloxacin in infants and young children as paradigms. The number of sampling times was reduced from original full sampling times to two to four sampling times based on the Fisher information matrix. We observed that the precision of empirical Bayes estimates of the key PK parameters and the predicted efficacy based on the reduced sampling times were generally comparable to those based on the original full sampling times. The model‐based approach to sampling optimization provided a maximization of PK information with a minimum burden on infants and young children for the future development of pediatric drugs.
Collapse
|
25
|
D'Agate S, Musuamba FT, Jacqz-Aigrain E, Della Pasqua O. Simplified Dosing Regimens for Gentamicin in Neonatal Sepsis. Front Pharmacol 2021; 12:624662. [PMID: 33762945 PMCID: PMC7982486 DOI: 10.3389/fphar.2021.624662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 01/04/2021] [Indexed: 11/30/2022] Open
Abstract
Background: The effectiveness of antibiotics for the treatment of severe bacterial infections in newborns in resource-limited settings has been determined by empirical evidence. However, such an approach does not warrant optimal exposure to antibiotic agents, which are known to show different disposition characteristics in this population. Here we evaluate the rationale for a simplified regimen of gentamicin taking into account the effect of body size and organ maturation on pharmacokinetics. The analysis is supported by efficacy data from a series of clinical trials in this population. Methods: A previously published pharmacokinetic model was used to simulate gentamicin concentration vs. time profiles in a virtual cohort of neonates. Model predictive performance was assessed by supplementary external validation procedures using therapeutic drug monitoring data collected in neonates and young infants with or without sepsis. Subsequently, clinical trial simulations were performed to characterize the exposure to intra-muscular gentamicin after a q.d. regimen. The selection of a simplified regimen was based on peak and trough drug levels during the course of treatment. Results: In contrast to current World Health Organization guidelines, which recommend gentamicin doses between 5 and 7.5 mg/kg, our analysis shows that gentamicin can be used as a fixed dose regimen according to three weight-bands: 10 mg for patients with body weight <2.5 kg, 16 mg for patients with body weight between 2.5 and 4 kg, and 30 mg for those with body weight >4 kg. Conclusion: The choice of the dose of an antibiotic must be supported by a strong scientific rationale, taking into account the differences in drug disposition in the target patient population. Our analysis reveals that a simplified regimen is feasible and could be used in resource-limited settings for the treatment of sepsis in neonates and young infants with sepsis aged 0–59 days.
Collapse
Affiliation(s)
- S D'Agate
- Clinical Pharmacology and Therapeutics Group, University College London, London, United Kingdom
| | - F Tshinanu Musuamba
- Clinical Pharmacology and Therapeutics Group, University College London, London, United Kingdom
| | - E Jacqz-Aigrain
- Department of Paediatric Pharmacology and Pharmacogenetics, Centre Hospitalier Universitaire, Hôpital Robert Debré, Paris, France
| | - O Della Pasqua
- Clinical Pharmacology and Therapeutics Group, University College London, London, United Kingdom
| |
Collapse
|
26
|
Zempsky WT, Bhagat PK, Siddiqui K. Practical Challenges-Use of Paracetamol in Children and Youth Who are Overweight or Obese: A Narrative Review. Paediatr Drugs 2020; 22:525-534. [PMID: 32918268 PMCID: PMC7529628 DOI: 10.1007/s40272-020-00417-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Worldwide, > 380 million children and adolescents are overweight or obese, including 41 million children aged < 5 years. Obesity can change the pharmacokinetic properties of drugs by altering their distribution, metabolism, and elimination. Thus, children who are overweight or obese are at increased risk for receiving inappropriate doses of commonly used drugs, which can result in treatment failure, adverse events, and/or drug toxicity. This review analyzes available data on paracetamol dosing for pain and fever in children and adolescents who are overweight or obese to identify gaps and challenges in optimal dosing strategies. Literature searches using Medline, Embase, and ClinicalTrials.gov were conducted to identify English-language articles reporting paracetamol pharmacokinetics, dosing practices, and guidelines in children and adolescents who are overweight or obese. Of 24 relevant studies identified, 20 were specific to overweight/obese individuals and 15 were specific to children and/or adolescents. Data on paracetamol pharmacokinetics in children and adolescents who are overweight or obese are lacking, and there is no high-quality evidence to guide paracetamol prescribing practices in these patients. Adult data have been extrapolated to pediatric populations; however, extrapolation does not address differences in paracetamol metabolism in adults versus children; the efficacy and safety effects of such differences are unknown. Given the growing worldwide prevalence of obesity in children and adolescents and the likelihood that paracetamol use in this population will increase accordingly, obesity-specific pediatric dosing guidelines for paracetamol are urgently needed. High-quality research is necessary to inform such guidelines.
Collapse
Affiliation(s)
- William T Zempsky
- Department of Pediatrics, Connecticut Children's Medical Center, University of Connecticut, 282 Washington St, Hartford, CT, 06106, USA.
| | - Preeti K Bhagat
- Consumer Healthcare R & D, GlaxoSmithKline Consumer Healthcare, Singapore, Singapore
| | - Kamran Siddiqui
- Consumer Healthcare R & D, GlaxoSmithKline Consumer Healthcare, Singapore, Singapore
| |
Collapse
|
27
|
D'Agate S, Musuamba FT, Della Pasqua O. Dose Rationale for Amoxicillin in Neonatal Sepsis When Referral Is Not Possible. Front Pharmacol 2020; 11:521933. [PMID: 33117151 PMCID: PMC7549385 DOI: 10.3389/fphar.2020.521933] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 08/24/2020] [Indexed: 01/17/2023] Open
Abstract
Background Despite the widespread use of amoxicillin in young children, efforts to establish the feasibility of simplified dosing regimens in resource-limited settings have relied upon empirical evidence of efficacy. Given the antibacterial profile of beta-lactams, understanding of the determinants of pharmacokinetic variability may provide a more robust guidance for the selection of a suitable regimen. Here we propose a simplified dosing regimen based on pharmacokinetic-pharmacodynamic principles, taking into account the impact of growth, renal maturation and disease processes on the systemic exposure to amoxicillin. Materials and Methods A meta-analytical modeling approach was applied to allow the adaptation of an existing pharmacokinetic model for amoxicillin in critically ill adults. Model parameterization was based on allometric concepts, including a maturation function. Clinical trial simulations were then performed to characterize exposure, as defined by secondary pharmacokinetic parameters (AUC, Cmax, Cmin) and T>MIC. The maximization of the T>MIC was used as criterion for the purpose of this analysis and results compared to current WHO guidelines. Results A two-compartment model with first order absorption and elimination was found to best describe the pharmacokinetics of amoxicillin in the target population. In addition to the changes in clearance and volume distribution associated with demographic covariates, our results show that sepsis alters drug distribution, leading to lower amoxicillin levels and longer half-life as compared to non-systemic disease conditions. In contrast to the current WHO guidelines, our analysis reveals that amoxicillin can be used as a fixed dose regimen including two weight bands: 125 mg b.i.d. for patients with body weight < 4.0 kg and 250 mg b.i.d. for patients with body weight ≥ 4.0 kg. Conclusions In addition to the effect of developmental growth and renal maturation, sepsis also alters drug disposition. The use of a model-based approach enabled the integration of these factors when defining the dose rationale for amoxicillin. A simplified weight-banded dosing regimen should be considered for neonates and young infants with sepsis when referral is not possible.
Collapse
Affiliation(s)
- Salvatore D'Agate
- Clinical Pharmacology and Therapeutics Group, University College London, London, United Kingdom
| | - Flora Tshinanu Musuamba
- Clinical Pharmacology and Therapeutics Group, University College London, London, United Kingdom
| | - Oscar Della Pasqua
- Clinical Pharmacology and Therapeutics Group, University College London, London, United Kingdom
| |
Collapse
|
28
|
Li Z, Bai J, Wen A, Shen S, Duan M, Li X. Pharmacokinetic and Pharmacodynamic Analysis of Critically Ill Patients Undergoing Continuous Renal Replacement Therapy With Imipenem. Clin Ther 2020; 42:1564-1577.e8. [PMID: 32741646 DOI: 10.1016/j.clinthera.2020.06.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/12/2020] [Accepted: 06/21/2020] [Indexed: 01/30/2023]
Abstract
PURPOSE This study explores factors that affect behavior in critically ill patients receiving continuous renal replacement therapy (CRRT) with imipenem and provides dosing regimens for these patients. METHODS A prospective, open-label study was conducted in a clinical setting. Both blood and effluent samples were collected pairwise at the scheduled time points. Plasma and effluent imipenem concentrations were determined by HPLC-UV. A population pharmacokinetic model was developed using a nonlinear mixed-effects modeling method. The final model was evaluated by a bootstrap and visual predictive check. A population pharmacokinetic and pharmacodynamic analysis using Monte Carlo simulations was performed to explore the effects of empirically used dosing regimens (0.5 g q6h, 0.5 g q8h, 0.5 g q12h, 1 g q6h, 1 g q8h, and 1 g q12h) on the probability of target attainment. FINDINGS Thirty patients were included in the population model analysis. Imipenem concentration data were best described by a 3-compartment model (central, peripheral, and dialysis compartments). The clearance of the dialysis compartment (CLd) was used to characterize drug elimination from the dialyzer. Creatinine clearance (CrCl) was the covariate that influenced the central clearance (CLc), and the effects of dialysate flow (Qd) was significant for CLd. Model validation revealed that the final model had qualified stability and acceptable predictive properties. A pharmacokinetic and pharmacodynamic analysis was conducted by Monte Carlo simulation, and patients were categorized into 12 subgroups based on different CrCl values (<30, 31-60, 61-90, and >90 mL/min) and Qd values (300, 500, and 1000 mL/h). Under the same MIC value and administration regimen, probability of target attainment values decreased with an increase of CrCl and Qd. IMPLICATIONS CrCl and Qd had significant effects on CLc and CLd, respectively. The proposed final model may be used to guide practitioners in imipenem dosing in this specific patient population.
Collapse
Affiliation(s)
- Zhe Li
- Department of Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Jing Bai
- Department of Intensive Care Unit, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Aiping Wen
- Department of Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Su Shen
- Department of Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Meili Duan
- Department of Intensive Care Unit, Beijing Friendship Hospital, Capital Medical University, Beijing, China.
| | - Xingang Li
- Department of Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
29
|
Sassen SDT, Zwaan CM, van der Sluis IM, Mathôt RAA. Pharmacokinetics and population pharmacokinetics in pediatric oncology. Pediatr Blood Cancer 2020; 67:e28132. [PMID: 31876123 DOI: 10.1002/pbc.28132] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 11/19/2019] [Accepted: 11/24/2019] [Indexed: 12/28/2022]
Abstract
Pharmacokinetic research has become increasingly important in pediatric oncology as it can have direct clinical implications and is a crucial component in individualized medicine. Population pharmacokinetics has become a popular method especially in children, due to the potential for sparse sampling, flexible sampling times, computing of heterogeneous data, and identification of variability sources. However, population pharmacokinetic reports can be complex and difficult to interpret. The aim of this article is to provide a basic explanation of population pharmacokinetics, using clinical examples from the field of pediatric oncology, to facilitate the translation of pharmacokinetic research into the daily clinic.
Collapse
Affiliation(s)
- Sebastiaan D T Sassen
- Department of Pediatric Oncology, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - C Michel Zwaan
- Department of Pediatric Oncology, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands.,Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | | | - Ron A A Mathôt
- Department of Hospital Pharmacy, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| |
Collapse
|
30
|
Borella E, Oosterholt S, Magni P, Della Pasqua O. Use of prior knowledge and extrapolation in paediatric drug development: A case study with deferasirox. Eur J Pharm Sci 2019; 136:104931. [PMID: 31108206 DOI: 10.1016/j.ejps.2019.05.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 03/09/2019] [Accepted: 05/13/2019] [Indexed: 01/19/2023]
Abstract
The characterisation of pharmacokinetics, pharmacodynamics and dose-exposure-response relationships requires data arising from well-designed study protocols and a relatively large sample from the target patient population. Such a prerequisite is unrealistic for paediatric rare diseases, where the patient population is often vulnerable and very small. In such cases, different sources of data and knowledge need to be considered to ensure trial designs are truly informative and oncoming data can be analysed efficiently. Here, we use clinical trial simulations to assess the contribution of historical data for (1) the analysis of sparse samples from a limited number of children and (2) the optimisation of study design when an increase in the number of subjects is not feasible. The evaluation of the pharmacokinetics of deferasirox in paediatric patients affected by haemoglobinopathies was used as case study. Our investigation shows that the incorporation of prior knowledge increases parameter precision and probability of successful convergence from only 12% with no priors to 56% and 75% for weakly and highly informative priors, respectively. In addition, results suggest that even when only one sample is collected per subject, as implemented in the original trial and in many other examples in clinical research, there is a 60% probability of biased parameter estimates (>25%). In conjunction with adult prior information and optimisation techniques, the probability of bias could be limited to <20% by increasing the number of samples/subject from 1 to 3. The methodology described here can be easily applied to other studies in small populations.
Collapse
Affiliation(s)
- Elisa Borella
- Dipart. Ingegneria Industriale e dell'Informazione, Università degli Studi di Pavia, Pavia, Italy
| | - Sean Oosterholt
- Clinical Pharmacology & Therapeutics Group, University College London, London, UK
| | - Paolo Magni
- Dipart. Ingegneria Industriale e dell'Informazione, Università degli Studi di Pavia, Pavia, Italy
| | - Oscar Della Pasqua
- Clinical Pharmacology & Therapeutics Group, University College London, London, UK; Clinical Pharmacology Modelling & Simulation, GlaxoSmithKline, Uxbridge, UK.
| |
Collapse
|
31
|
van Dijkman SC, De Cock PAJG, Smets K, Decaluwe W, Smits A, Allegaert K, Vande Walle J, De Paepe P, Della Pasqua O. Dose rationale and pharmacokinetics of dexmedetomidine in mechanically ventilated new-borns: impact of design optimisation. Eur J Clin Pharmacol 2019; 75:1393-1404. [PMID: 31312867 DOI: 10.1007/s00228-019-02708-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Accepted: 06/11/2019] [Indexed: 12/11/2022]
Abstract
PURPOSE There is a need for alternative analgosedatives such as dexmedetomidine in neonates. Given the ethical and practical difficulties, protocol design for clinical trials in neonates should be carefully considered before implementation. Our objective was to identify a protocol design suitable for subsequent evaluation of the dosing requirements for dexmedetomidine in mechanically ventilated neonates. METHODS A published paediatric pharmacokinetic model was used to derive the dosing regimen for dexmedetomidine in a first-in-neonate study. Optimality criteria were applied to optimise the blood sampling schedule. The impact of sampling schedule optimisation on model parameter estimation was assessed by simulation and re-estimation procedures for different simulation scenarios. The optimised schedule was then implemented in a neonatal pilot study. RESULTS Parameter estimates were more precise and similarly accurate in the optimised scenarios, as compared to empirical sampling (normalised root mean square error: 1673.1% vs. 13,229.4% and relative error: 46.4% vs. 9.1%). Most importantly, protocol deviations from the optimal design still allowed reasonable parameter estimation. Data analysis from the pilot group (n = 6) confirmed the adequacy of the optimised trial protocol. Dexmedetomidine pharmacokinetics in term neonates was scaled using allometry and maturation, but results showed a 20% higher clearance in this population compared to initial estimates obtained by extrapolation from a slightly older paediatric population. Clearance for a typical neonate, with a post-menstrual age (PMA) of 40 weeks and weight 3.4 kg, was 2.92 L/h. Extension of the study with 11 additional subjects showed a further increased clearance in pre-term subjects with lower PMA. CONCLUSIONS The use of optimal design in conjunction with simulation scenarios improved the accuracy and precision of the estimates of the parameters of interest, taking into account protocol deviations, which are often unavoidable in this event-prone population.
Collapse
Affiliation(s)
- Sven C van Dijkman
- Division of Pharmacology, Leiden Academic Centre for Drug Research, Leiden, The Netherlands
| | - Pieter A J G De Cock
- Heymans Institute of Pharmacology, Ghent University, Ghent, Belgium
- Department of Pharmacy, Ghent University Hospital, Ghent, Belgium
| | - Koenraad Smets
- Department of Neonatology, Ghent University Hospital, Ghent, Belgium
| | - Wim Decaluwe
- Department of Neonatology, AZ Sint Jan Brugge-Oostende AV, Bruges, Belgium
| | - Anne Smits
- Neonatal Intensive Care Unit, University Hospital Leuven, Leuven, Belgium
| | - Karel Allegaert
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Johan Vande Walle
- Department of Paediatric Nephrology, Ghent University Hospital, Ghent, Belgium
| | - Peter De Paepe
- Heymans Institute of Pharmacology, Ghent University, Ghent, Belgium
| | - Oscar Della Pasqua
- Clinical Pharmacology and Therapeutics, University College London, BMA House, Tavistock Square, London, WC1H 9JP, UK.
- Clinical Pharmacology Modelling & Simulation, GlaxoSmithKline, Uxbridge, UK.
| |
Collapse
|
32
|
Kaguelidou F, Le Roux E, Mangiarini L, Lundin R, de Leeuw TG, Della Pasqua O, Felisi M, Bonifazi D, Tibboel D, Ceci A, de Wildt SN, Alberti C. Non-inferiority double-blind randomised controlled trial comparing gabapentin versus tramadol for the treatment of chronic neuropathic or mixed pain in children and adolescents: the GABA-1 trial-a study protocol. BMJ Open 2019; 9:e023296. [PMID: 30787078 PMCID: PMC6398794 DOI: 10.1136/bmjopen-2018-023296] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 12/16/2018] [Accepted: 12/19/2018] [Indexed: 01/24/2023] Open
Abstract
INTRODUCTION Gabapentin is currently used 'off-label' in children and adolescents with chronic neuropathic pain, and reliable evidence of its effects and optimal dosing are lacking. OBJECTIVES The GABA-1 trial aims to compare the efficacy and safety of gabapentin liquid formulation relative to tramadol and to explore the pharmacokinetics of both drugs in the treatment of chronic, neuropathic or mixed pain in the paediatric population. METHODS AND ANALYSIS The trial is a multicentre, double-blind, double-dummy, randomised, active-controlled, non-inferiority trial. Participants aged from 3 months to <18 years of age with moderate to severe (≥4/10 in age-appropriate pain scales) chronic neuropathic or mixed pain will be recruited in 14 clinical sites in eight European countries. A total of 94 subjects will be randomised to receive gabapentin and tramadol placebo or tramadol and gabapentin placebo throughout 16-19 weeks (including 3 weeks of titration [optimisation period], 12 weeks of treatment at a stable dose [maintenance period] and 1-4 weeks of tapering [discontinuation period]). The primary objective is to assess the efficacy of gabapentin relative to tramadol for the treatment of moderate to severe chronic neuropathic or mixed pain by comparing the difference in average pain scores (assessed by age-appropriate pain scales) between intervention arms after 15 weeks of treatment. Secondary objectives include the assessment of the safety, quality of life and global satisfaction with treatment and the description of the pharmacokinetic-pharmacodynamic relationship of gabapentin liquid formulation and tramadol oral drops to validate the recommended paediatric doses. Only rescue pain medication by paracetamol and/or ibuprofen is allowed during the trial. ETHICS AND DISSEMINATION Ethic approval was obtained in the eight participating countries. Results will be submitted for publication in a peer-reviewed journal and presented at one or more scientific conferences. TRIAL REGISTRATION NUMBERS 2014-004851-30 and NCT02722603. TRIAL STATUS Ongoing research study, currently recruiting.
Collapse
Affiliation(s)
- Florentia Kaguelidou
- Department of Paediatric Pharmacology and Pharmacogenetics, AP-HP, Hôpital Robert Debré, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Paris, France
- Inserm, CIC 1426, F-75019, Paris, France
| | - Enora Le Roux
- AP-HP, Hôpital Robert Debré, Unité d’Epidémiologie Clinique, Paris, France
- UMR-1123 ECEVE, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | | | | | - Thomas G de Leeuw
- Department of Anaesthesia, Erasmus MC Sophia Children’s Hospital & Center for Pain Medicine Rotterdam, Rotterdam, The Netherlands
| | - Oscar Della Pasqua
- Clinical Pharmacology and Therapeutics Group, University College London, London, UK
| | - Mariagrazia Felisi
- PHARM – Pharmaceutical Research Management SRL, Lodi, Italy
- CVBF–Consorzio per le Valutazioni Biologiche e Farmacologiche, Pavia, Italy
| | - Donato Bonifazi
- PHARM – Pharmaceutical Research Management SRL, Lodi, Italy
- CVBF–Consorzio per le Valutazioni Biologiche e Farmacologiche, Pavia, Italy
| | - Dick Tibboel
- Intensive Care and Department of Pediatric Surgery, Erasmus MC Sophia Children’s Hospital, Rotterdam, The Netherlands
| | - Adriana Ceci
- Fondazione per la Ricerca Farmacologica Gianni Benzi onlus, Valenzano, Italy
| | - Saskia N de Wildt
- Intensive Care and Department of Pediatric Surgery, Erasmus MC Sophia Children’s Hospital, Rotterdam, The Netherlands
- Department of Pharmacology and Toxicology, Radboud Universiteit Faculteit der Rechtsgeleerdheid, Nijmegen, The Netherlands
| | - Corinne Alberti
- Université Paris Diderot, Sorbonne Paris Cité, Paris, France
- AP-HP, Hôpital Robert Debré, Unité d’Epidémiologie Clinique, Paris, France
- Inserm, U1123 and CICEC 1426, F-75019, Paris, France
| |
Collapse
|
33
|
Kaddi CD, Niesner B, Baek R, Jasper P, Pappas J, Tolsma J, Li J, van Rijn Z, Tao M, Ortemann‐Renon C, Easton R, Tan S, Puga AC, Schuchman EH, Barrett JS, Azer K. Quantitative Systems Pharmacology Modeling of Acid Sphingomyelinase Deficiency and the Enzyme Replacement Therapy Olipudase Alfa Is an Innovative Tool for Linking Pathophysiology and Pharmacology. CPT Pharmacometrics Syst Pharmacol 2018; 7:442-452. [PMID: 29920993 PMCID: PMC6063739 DOI: 10.1002/psp4.12304] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 03/27/2018] [Accepted: 04/10/2018] [Indexed: 12/12/2022] Open
Abstract
Acid sphingomyelinase deficiency (ASMD) is a rare lysosomal storage disorder with heterogeneous clinical manifestations, including hepatosplenomegaly and infiltrative pulmonary disease, and is associated with significant morbidity and mortality. Olipudase alfa (recombinant human acid sphingomyelinase) is an enzyme replacement therapy under development for the non-neurological manifestations of ASMD. We present a quantitative systems pharmacology (QSP) model supporting the clinical development of olipudase alfa. The model is multiscale and mechanistic, linking the enzymatic deficiency driving the disease to molecular-level, cellular-level, and organ-level effects. Model development was informed by natural history, and preclinical and clinical studies. By considering patient-specific pharmacokinetic (PK) profiles and indicators of disease severity, the model describes pharmacodynamic (PD) and clinical end points for individual patients. The ASMD QSP model provides a platform for quantitatively assessing systemic pharmacological effects in adult and pediatric patients, and explaining variability within and across these patient populations, thereby supporting the extrapolation of treatment response from adults to pediatrics.
Collapse
Affiliation(s)
| | - Bradley Niesner
- Translational Informatics, TMED, Sanofi, BridgewaterNew JerseyUSA
| | - Rena Baek
- Sanofi Genzyme, CambridgeMassachusettsUSA
| | | | | | | | - Jing Li
- Translational Informatics, TMED, Sanofi, BridgewaterNew JerseyUSA
| | - Zachary van Rijn
- Translational Informatics, TMED, Sanofi, BridgewaterNew JerseyUSA
| | - Mengdi Tao
- Translational Informatics, TMED, Sanofi, BridgewaterNew JerseyUSA
| | | | - Rachael Easton
- Translational Informatics, TMED, Sanofi, BridgewaterNew JerseyUSA
| | - Sharon Tan
- Sanofi Genzyme, CambridgeMassachusettsUSA
| | | | - Edward H. Schuchman
- Genetics & Genomic Sciences, Icahn School of Medicine at Mount SinaiNew YorkNYUSA
| | | | - Karim Azer
- Translational Informatics, TMED, Sanofi, BridgewaterNew JerseyUSA
| |
Collapse
|
34
|
Chitty KM, Chan B, Pulanco CL, Luu S, Egunsola O, Buckley NA. Discontinuities and disruptions in drug dosage guidelines for the paediatric population. Br J Clin Pharmacol 2018; 84:1029-1037. [PMID: 29411410 DOI: 10.1111/bcp.13511] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 12/21/2017] [Accepted: 12/23/2017] [Indexed: 12/30/2022] Open
Abstract
AIMS This study investigates paediatric drug dosage guidelines with the aim of investigating their agreement with body surface area (BSA) scaling principles. METHODS A total of 454 drug dosage guidelines listed in the AMH-CDC 2015 were examined. Data extracted included the administration, frequency and dose per age bracket from 0 to 18 years. Drug treatments were categorized as follows: (1) The same dose recommendation in milligrams per kilogram (mg kg-1 ) for all age/weights; (2) Change in the mg kg-1 dosing according to age/weight; (3) Change in dose in mg according to age/weight; (4) Change from mg kg-1 dosing to a dose in mg according to age/weight; (5) The same recommendation for all age/weight groups in mg; or (6) BSA dosing. Example drugs were selected to illustrate dose progression across ages. RESULTS Most drug treatments (63%) have the same mg kg-1 dose for all age/weight groups, 14% are dosed in mg kg-1 across all ages with dose changes according to age/weight, 13% were dosed in mg across all ages with dose changes, 10% switched from mg kg-1 to a set dose in mg, 4.2% have the same dose in mg for all age and weight groups and 2.2% are dosed according to BSA. CONCLUSIONS Paediatric dosage guidelines are based on weight-based formulas, available dosing formulations and prior patterns of use. Substantial variation from doses predicted by BSA scaling are common, as are large shifts in recommended doses at age thresholds. Further research is required to determine if better outcomes could be achieved by adopting biologically based scaling of paediatric doses.
Collapse
Affiliation(s)
- Kate M Chitty
- Discipline of Pharmacology, Sydney Medical School, Translational Australian Clinical Toxicology Program, The University of Sydney, NSW, Australia, 2006
| | - Bosco Chan
- Discipline of Pharmacology, Sydney Medical School, Translational Australian Clinical Toxicology Program, The University of Sydney, NSW, Australia, 2006
| | - Camille L Pulanco
- Discipline of Pharmacology, Sydney Medical School, Translational Australian Clinical Toxicology Program, The University of Sydney, NSW, Australia, 2006
| | - Sonya Luu
- Discipline of Pharmacology, Sydney Medical School, Translational Australian Clinical Toxicology Program, The University of Sydney, NSW, Australia, 2006
| | - Oluwaseun Egunsola
- Discipline of Pharmacology, Sydney Medical School, Translational Australian Clinical Toxicology Program, The University of Sydney, NSW, Australia, 2006
| | - Nicholas A Buckley
- Discipline of Pharmacology, Sydney Medical School, Translational Australian Clinical Toxicology Program, The University of Sydney, NSW, Australia, 2006
| |
Collapse
|
35
|
Model Informed Pediatric Development Applied to Bilastine: Ontogenic PK Model Development, Dose Selection for First Time in Children and PK Study Design. Pharm Res 2017; 34:2720-2734. [PMID: 28971281 DOI: 10.1007/s11095-017-2248-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 08/21/2017] [Indexed: 12/21/2022]
Abstract
PURPOSE Bilastine is an H1 antagonist whose pharmacokinetics (PK) and pharmacodynamics (PD) have been resolved in adults with a therapeutic oral dose of 20 mg/day. Bilastine has favorable characteristics for use in pediatrics but the PK/PD and the optimal dose in children had yet to be clinically explored. The purpose is to: (1) Develop an ontogenic predictive model of bilastine PK linked to the PD in adults by integrating current knowledge; (2) Use the model to design a PK study in children; (3) Confirm the selected dose and the study design through the evaluation of model predictability in the first recruited children; (4) Consider for inclusion the group of younger children (< 6 years). METHODS A semi-mechanistic approach was applied to predict bilastine PK in children assuming the same PD as described in adults. The model was used to simulate the time evolution of plasma levels and wheal and flare effects after several doses and design an adaptive PK trial in children that was then confirmed using data from the first recruits by comparing observations with model predictions. RESULTS PK/PD simulations supported the selection of 10 mg/day in 2 to <12 year olds. Results from the first interim analysis confirmed the model predictions and design hence trial continuation. CONCLUSION The model successfully predicted bilastine PK in pediatrics and optimally assisted the selection of the dose and sampling scheme for the trial in children. The selected dose was considered suitable for younger children and the forthcoming safety study in children aged 2 to <12 years.
Collapse
|
36
|
Quantitative determination of dobutamine in newborn pig plasma samples by HPLC–MS/MS. J Pharm Biomed Anal 2017; 145:178-185. [DOI: 10.1016/j.jpba.2017.06.050] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 06/16/2017] [Accepted: 06/20/2017] [Indexed: 01/17/2023]
|
37
|
Chong LSH, Sautenet B, Tong A, Hanson CS, Samuel S, Zappitelli M, Dart A, Furth S, Eddy AA, Groothoff J, Webb NJA, Yap HK, Bockenhauer D, Sinha A, Alexander SI, Goldstein SL, Gipson DS, Raman G, Craig JC. Range and Heterogeneity of Outcomes in Randomized Trials of Pediatric Chronic Kidney Disease. J Pediatr 2017; 186:110-117.e11. [PMID: 28449820 DOI: 10.1016/j.jpeds.2017.03.034] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 02/07/2017] [Accepted: 03/10/2017] [Indexed: 01/16/2023]
Abstract
OBJECTIVE To determine the range and heterogeneity of outcomes reported in randomized controlled trials of interventions for children with chronic kidney disease (CKD). STUDY DESIGN The Cochrane Kidney and Transplant Specialized Register was searched to March 2016. Randomized trials involving children across all stages of CKD were selected. All outcome domains and measurements were extracted from included trials. The frequency and characteristics of the outcome domains and measures were evaluated. RESULTS From 205 trials included, 6158 different measurements of 100 different outcome domains were reported, with a median of 22 domains per trial (IQR 13-41). Overall, 52 domains (52%) were surrogate, 38 (38%) were clinical, and 10 (10%) were patient-reported. The 5 most commonly reported domains were blood pressure (76 [37%] trials), relapse/remission (70 [34%]), kidney function (66 [32%]), infection (61 [30%]), and height/pubertal development (51 [25%]). Mortality (14%), cardiovascular disease (4%), and quality of life (1%) were reported infrequently. The 2 most frequently reported outcomes, blood pressure and relapse/remission, had 56 and 81 different outcome measures, respectively. CONCLUSIONS The outcomes reported in clinical trials involving children with CKD are extremely heterogeneous and are most often surrogate outcomes, rather than clinical and patient-centered outcomes such as cardiovascular disease and quality of life. Efforts to ensure consistent reporting of outcomes that are important to patients and clinicians will improve the value of trials to guide clinical decision-making. In our study, non-English articles were excluded.
Collapse
Affiliation(s)
- Lauren S H Chong
- Sydney School of Public Health, The University of Sydney, Sydney, Australia; Centre for Kidney Research, The Children's Hospital at Westmead, Sydney, Australia
| | - Benedicte Sautenet
- Sydney School of Public Health, The University of Sydney, Sydney, Australia; Centre for Kidney Research, The Children's Hospital at Westmead, Sydney, Australia; University Francois Rabelais, Tours, France; Department of Nephrology and Clinical Immunology, Tours Hospital, Tours, France; INSERM (U1153), Paris, France
| | - Allison Tong
- Sydney School of Public Health, The University of Sydney, Sydney, Australia; Centre for Kidney Research, The Children's Hospital at Westmead, Sydney, Australia.
| | - Camilla S Hanson
- Sydney School of Public Health, The University of Sydney, Sydney, Australia; Centre for Kidney Research, The Children's Hospital at Westmead, Sydney, Australia
| | - Susan Samuel
- Department of Pediatrics, Section of Nephrology, University of Calgary, Calgary, Canada
| | - Michael Zappitelli
- Department of Pediatrics, Division of Pediatric Nephrology, Montreal Children's Hospital, McGill University Health Centre, McGill University, Montreal, Canada
| | - Allison Dart
- Department of Pediatrics and Child Health, The Children's Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, Canada
| | - Susan Furth
- Departments of Pediatrics and Epidemiology, Perelman School of Medicine, Division of Nephrology, The Children's Hospital of Philadelphia, Philadelphia, PA
| | - Allison A Eddy
- Department of Pediatrics, University of British Columbia and the British Columbia Children's Hospital, Vancouver, Canada
| | - Jaap Groothoff
- Department of Pediatric Nephrology, Emma Children's Hospital, Academic Medical Center, Amsterdam, The Netherlands
| | - Nicholas J A Webb
- Department of Pediatric Nephrology and National Institute for Health Research/Wellcome Trust Clinical Research Facility, University of Manchester, Manchester Academic Health Science Centre, Royal Manchester Children's Hospital, Manchester, United Kingdom
| | - Hui-Kim Yap
- Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Detlef Bockenhauer
- University College London Centre for Nephrology, Great Ormond Street Hospital for Children National Health Service Foundation Trust, London, United Kingdom
| | - Aditi Sinha
- Division of Nephrology, Department of Pediatrics, All India Institute of Medical Sciences, India
| | - Stephen I Alexander
- Centre for Kidney Research, The Children's Hospital at Westmead, Sydney, Australia
| | - Stuart L Goldstein
- Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Debbie S Gipson
- Department of Pediatrics, School of Medicine, University of Michigan, Ann Arbor, MI
| | - Gayathri Raman
- Sydney School of Public Health, The University of Sydney, Sydney, Australia
| | - Jonathan C Craig
- Sydney School of Public Health, The University of Sydney, Sydney, Australia; Centre for Kidney Research, The Children's Hospital at Westmead, Sydney, Australia
| |
Collapse
|
38
|
Vermeulen E, van den Anker JN, Della Pasqua O, Hoppu K, van der Lee JH, Global Research in Paediatrics (GRiP). How to optimise drug study design: pharmacokinetics and pharmacodynamics studies introduced to paediatricians. J Pharm Pharmacol 2017; 69:439-447. [PMID: 27671925 PMCID: PMC6084327 DOI: 10.1111/jphp.12637] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 08/10/2016] [Indexed: 02/06/2023]
Abstract
OBJECTIVES In children, there is often lack of sufficient information concerning the pharmacokinetics (PK) and pharmacodynamics (PD) of a study drug to support dose selection and effective evaluation of efficacy in a randomised clinical trial (RCT). Therefore, one should consider the relevance of relatively small PKPD studies, which can provide the appropriate data to optimise the design of an RCT. METHODS Based on the experience of experts collaborating in the EU-funded Global Research in Paediatrics consortium, we aimed to inform clinician-scientists working with children on the design of investigator-initiated PKPD studies. KEY FINDINGS The importance of the identification of an optimal dose for the paediatric population is explained, followed by the differences and similarities of dose-ranging and efficacy studies. The input of clinical pharmacologists with modelling expertise is essential for an efficient dose-finding study. CONCLUSIONS The emergence of new laboratory techniques and statistical tools allows for the collection and analysis of sparse and unbalanced data, enabling the implementation of (observational) PKPD studies in the paediatric clinic. Understanding of the principles and methods discussed in this study is essential to improve the quality of paediatric PKPD investigations, and to prevent the conduct of paediatric RCTs that fail because of inadequate dosing.
Collapse
Affiliation(s)
- Eric Vermeulen
- Pediatric Clinical Research OfficeEmma Children's HospitalAcademic Medical CenterAmsterdamThe Netherlands
| | - John N. van den Anker
- Division of Pediatric Clinical PharmacologyChildren's National Health SystemWashingtonDCUSA
- Division of Paediatric Pharmacology and PharmacometricsUniversity of Basel Children's HospitalBaselSwitzerland
- Intensive Care and Department of Pediatric SurgeryErasmus Medical CenterSophia Children's HospitalRotterdamThe Netherlands
| | - Oscar Della Pasqua
- Clinical Pharmacology Modelling & SimulationGlaxoSmithKlineStockley ParkUK
- Clinical Pharmacology & TherapeuticsUniversity College LondonLondonUK
| | - Kalle Hoppu
- Poison Information CentreHelsinki University Central HospitalHelsinkiFinland
| | - Johanna H. van der Lee
- Pediatric Clinical Research OfficeEmma Children's HospitalAcademic Medical CenterAmsterdamThe Netherlands
| | | |
Collapse
|
39
|
Piana C, Danhof M, Della Pasqua O. Impact of disease, drug and patient adherence on the effectiveness of antiviral therapy in pediatric HIV. Expert Opin Drug Metab Toxicol 2017; 13:497-511. [PMID: 28043170 DOI: 10.1080/17425255.2017.1277203] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Maintaining effective antiretroviral treatment for life is a major problem in both resource-limited and resource-rich countries. Despite the progress observed in paediatric antiretroviral therapy, approximately 12% of children still experience treatment failure due to drug resistance, inadequate dosing and poor adherence. We explore the current status of antiretroviral therapy in children with focus on the interaction between disease, drug pharmacokinetics and patient behavior, all of which are strongly interconnected and determine treatment outcome. Areas covered: An overview is provided of the viral characteristics and available drug combinations aimed at the prevention of resistance. In this context, the role of patient adherence is scrutinized. A detailed assessment of factors affecting adherence is presented together with the main strategies to enhance treatment response in children. Expert opinion: Using modeling and simulation, a framework for characterizing the forgiveness of non-adherence for specific antiretroviral drugs in children is proposed in which information on pharmacokinetics, pharmacokinetic-pharmacodynamic relationships and viral dynamics is integrated. This approach represents an opportunity for the simplification of dosing regimens taking into account the interaction between these factors. Based on clinical trial simulation scenarios, we envisage the possibility of assessing the impact of variable adherence to antiretroviral drug combinations in HIV-infected children.
Collapse
Affiliation(s)
- Chiara Piana
- a Division of Pharmacology, Leiden Academic Centre for Drug Research , Leiden University , Leiden , The Netherlands
| | - Meindert Danhof
- a Division of Pharmacology, Leiden Academic Centre for Drug Research , Leiden University , Leiden , The Netherlands
| | - Oscar Della Pasqua
- b Clinical Pharmacology Modelling & Simulation , GlaxoSmithKline , Uxbridge , United Kingdom.,c Clinical Pharmacology & Therapeutics Group , University College London , London , United Kingdom
| |
Collapse
|
40
|
Bellanti F, Del Vecchio GC, Putti MC, Maggio A, Filosa A, Cosmi C, Mangiarini L, Spino M, Connelly J, Ceci A, Della Pasqua O. Population pharmacokinetics and dosing recommendations for the use of deferiprone in children younger than 6 years. Br J Clin Pharmacol 2016; 83:593-602. [PMID: 27641003 DOI: 10.1111/bcp.13134] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Revised: 08/26/2016] [Accepted: 09/11/2016] [Indexed: 01/19/2023] Open
Abstract
AIMS Despite long clinical experience with deferiprone, there is limited information on its pharmacokinetics in children aged <6 years. Here we assess the impact of developmental growth on the pharmacokinetics of deferiprone in this population using a population approach. Based on pharmacokinetic bridging concepts, we also evaluate whether the recommended doses yield appropriate systemic exposure in this group of patients. METHODS Data from a study in which 18 paediatric patients were enrolled were available for the purposes of this analysis. Patients were randomised to three deferiprone dose levels (8.3, 16.7 and 33.3 mg kg-1 ). Blood samples were collected according to an optimised sampling scheme in which each patient contributed to a maximum of five samples. A population pharmacokinetic model was developed using NONMEM v.7.2. Model selection criteria were based on graphical and statistical summaries. RESULTS A one-compartment model with first-order absorption and first-order elimination best described the pharmacokinetics of deferiprone. Drug disposition parameters were affected by body weight, with both clearance and volume increasing allometrically with size. Simulation scenarios show that comparable systemic exposure (AUC) is achieved in children and adults after similar dose levels in mg kg-1 , with median (5-95th quantiles) AUC values, respectively, of 340.6 (223.2-520.0) μmol l-1 h and 318.5 (200.4-499.0) μmol l-1 h at 75 mg kg-1 day-1 , and 453.7 (297.3-693.0) μmol l-1 h and 424.2 (266.9-664.0) μmol l-1 h at 100 mg kg-1 day-1 given as three times daily (t.i.d.) doses. CONCLUSIONS Based on the current findings, a dosing regimen of 25 mg kg-1 t.i.d. is recommended in children aged <6 years, with the possibility of titration up to 33.3 mg kg-1 t.i.d.
Collapse
Affiliation(s)
| | - Giovanni C Del Vecchio
- Paediatric Hematology Unit, Azienda Ospedaliero-Universitaria Consorziale Policlinico di Bari, Italy
| | | | - Aurelio Maggio
- Azienda Ospedaliera Ospedali Riuniti Villa Sofia - Cervello, Palermo, Italy
| | - Aldo Filosa
- Azienda Ospedaliera Antonio Cardarelli, Naples, Italy
| | - Carlo Cosmi
- Clinica Pediatrica Università di Sassari - ASL1, Sassari, Italy
| | - Laura Mangiarini
- Consorzio per Valutazioni Biologiche e Farmacologiche, Pavia, Italy
| | | | | | - Adriana Ceci
- Consorzio per Valutazioni Biologiche e Farmacologiche, Pavia, Italy
| | - Oscar Della Pasqua
- Leiden Academic Centre for Drug Research, Leiden, The Netherlands.,Clinical Pharmacology & Therapeutics, University College London, UK
| | | |
Collapse
|
41
|
Jorga K, Chavanne C, Frey N, Lave T, Lukacova V, Parrott N, Peck R, Reigner B. Bottom-up Meets Top-down: Complementary Physiologically Based Pharmacokinetic and Population Pharmacokinetic Modeling for Regulatory Approval of a Dosing Algorithm of Valganciclovir in Very Young Children. Clin Pharmacol Ther 2016; 100:761-769. [PMID: 27530217 DOI: 10.1002/cpt.449] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 08/01/2016] [Accepted: 08/10/2016] [Indexed: 01/28/2023]
Abstract
Population pharmacokinetic (PopPK) and physiologically based pharmacokinetic (PBPK) models are frequently used to support pediatric drug development. Both methods have strengths and limitations and we used them complementarily to support the regulatory approval of a dosing algorithm for valganciclovir (VGCV) in children <4 months old. An existing pediatric PBPK model was extended to neonates and showed that potential physiological differences compared with older children are minor. The PopPK model was used to simulate ganciclovir (GCV) exposures in children with population typical combinations of body size and renal function and to assess the effectiveness of an alternative dosing algorithm suggested by the US Food and Drug Administration. PBPK and PopPK confirmed that the proposed VGCV dosing algorithm achieves similar GCV exposures in children of all ages and that the alternative dosing algorithm leads to underexposure in a substantial fraction of patients. Our approach raised the confidence in the VGCV dosing algorithm for children <4 months old and supported the regulatory approval.
Collapse
Affiliation(s)
- K Jorga
- KarinJorga Life Science Consulting GmbH, Basel, Switzerland
| | - C Chavanne
- Pharma Research & Development, Clinical Pharmacology, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - N Frey
- Pharma Research & Development, Clinical Pharmacology, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - T Lave
- Pharma Research & Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - V Lukacova
- SimulationsPlus, Inc., Lancaster, California, USA
| | - N Parrott
- Pharma Research & Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - R Peck
- Pharma Research & Development, Clinical Pharmacology, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - B Reigner
- Pharma Research & Development, Clinical Pharmacology, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| |
Collapse
|
42
|
Li X, Wu Y, Sun S, Zhao Z, Wang Q. Population Pharmacokinetics of Vancomycin in Postoperative Neurosurgical Patients and the Application in Dosing Recommendation. J Pharm Sci 2016; 105:3425-3431. [PMID: 27671237 DOI: 10.1016/j.xphs.2016.08.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 08/14/2016] [Accepted: 08/18/2016] [Indexed: 11/25/2022]
Abstract
Our previous study indicates that cerebrospinal fluid (CSF) albumin level is a determinant of CSF vancomycin concentration for postoperative neurosurgical patients. We aimed to develop an improved vancomycin population pharmacokinetic model with incorporation of more covariates, and to provide dosing guidance for clinicians. Vancomycin was administered intravenously to 20 patients with external ventricular drains after neurosurgical operation. Blood and CSF were collected and vancomycin concentrations were measured by HPLC. A separate CSF compartment was considered, and was linked to the central compartment by a first-order process (QCSF). The clearance of the CSF compartment (ClCSF) was used to characterize vancomycin elimination from CSF through external ventricular drain. Nonlinear mixed-effects modeling approach was used to develop the model. The CSF albumin level (mg/dL) was the covariate influencing QCSF: QCSF=0.0049+0.000021×(CSF albumin-279). The effect of body weight (BW, kg) was significant on central volume (VC): VC=27.84+0.96×(BW-69). All parameters were estimated with an acceptable precision (relative standard error: RSE% < 30.26). The performance of the final model was acceptable with our previous dataset. A simple to use dosage regimen table was created to guide clinicians with vancomycin dosing. This model incorporates variables of both CSF albumin and BW, which offers improvements to the previous pharmacokinetics model.
Collapse
Affiliation(s)
- Xingang Li
- Department of Pharmacy, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, China; Precision Medicine Research Center for Neurological Disorders, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, China
| | - Yuanxing Wu
- Respiratory and Critical Care Medicine, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Shusen Sun
- College of Pharmacy, Western New England University, Springfield, Massachusetts 01119
| | - Zhigang Zhao
- Department of Pharmacy, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, China; Precision Medicine Research Center for Neurological Disorders, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, China
| | - Qiang Wang
- Intensive Care Unit, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, China.
| |
Collapse
|
43
|
Brussee JM, Calvier EAM, Krekels EHJ, Välitalo PAJ, Tibboel D, Allegaert K, Knibbe CAJ. Children in clinical trials: towards evidence-based pediatric pharmacotherapy using pharmacokinetic-pharmacodynamic modeling. Expert Rev Clin Pharmacol 2016; 9:1235-44. [PMID: 27269200 DOI: 10.1080/17512433.2016.1198256] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
INTRODUCTION In pediatric pharmacotherapy, many drugs are still used off-label, and their efficacy and safety is not well characterized. Different efficacy and safety profiles in children of varying ages may be anticipated, due to developmental changes occurring across pediatric life. AREAS COVERED Beside pharmacokinetic (PK) studies, pharmacodynamic (PD) studies are urgently needed. Validated PKPD models can be used to derive optimal dosing regimens for children of different ages, which can be evaluated in a prospective study before implementation in clinical practice. Strategies should be developed to ensure that formularies update their drug dosing guidelines regularly according to the most recent advances in research, allowing for clinicians to integrate these guidelines in daily practice. Expert commentary: We anticipate a trend towards a systems-level approach in pediatric modeling to optimally use the information gained in pediatric trials. For this approach, properly designed clinical PKPD studies will remain the backbone of pediatric research.
Collapse
Affiliation(s)
- Janneke M Brussee
- a Division of Pharmacology, Leiden Academic Centre for Drug Research , Leiden University , Leiden , The Netherlands
| | - Elisa A M Calvier
- a Division of Pharmacology, Leiden Academic Centre for Drug Research , Leiden University , Leiden , The Netherlands
| | - Elke H J Krekels
- a Division of Pharmacology, Leiden Academic Centre for Drug Research , Leiden University , Leiden , The Netherlands
| | - Pyry A J Välitalo
- a Division of Pharmacology, Leiden Academic Centre for Drug Research , Leiden University , Leiden , The Netherlands
| | - Dick Tibboel
- b Intensive Care and Department of Surgery , Erasmus MC-Sophia Children's Hospital , Rotterdam , The Netherlands
| | - Karel Allegaert
- b Intensive Care and Department of Surgery , Erasmus MC-Sophia Children's Hospital , Rotterdam , The Netherlands.,c Department of Development and Regeneration , KU Leuven , Leuven , Belgium
| | - Catherijne A J Knibbe
- a Division of Pharmacology, Leiden Academic Centre for Drug Research , Leiden University , Leiden , The Netherlands.,d Department of Clinical Pharmacy , St. Antonius Hospital , Nieuwegein , The Netherlands
| |
Collapse
|
44
|
Shawahna R. Pediatric Biopharmaceutical Classification System: Using Age-Appropriate Initial Gastric Volume. AAPS J 2016; 18:728-36. [PMID: 26935428 PMCID: PMC5256604 DOI: 10.1208/s12248-016-9885-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 02/01/2016] [Indexed: 01/22/2023] Open
Abstract
Development of optimized pediatric formulations for oral administration can be challenging, time consuming, and financially intensive process. Since its inception, the biopharmaceutical classification system (BCS) has facilitated the development of oral drug formulations destined for adults. At least theoretically, the BCS principles are applied also to pediatrics. A comprehensive age-appropriate BCS has not been fully developed. The objective of this work was to provisionally classify oral drugs listed on the latest World Health Organization's Essential Medicines List for Children into an age-appropriate BCS. A total of 38 orally administered drugs were included in this classification. Dose numbers were calculated using age-appropriate initial gastric volume for neonates, 6-month-old infants, and children aging 1 year through adulthood. Using age-appropriate initial gastric volume and British National Formulary age-specific dosing recommendations in the calculation of dose numbers, the solubility classes shifted from low to high in pediatric subpopulations of 12 years and older for amoxicillin, 5 years, 12 years and older for cephalexin, 9 years and older for chloramphenicol, 3-4 years, 9-11 and 15 years and older for diazepam, 18 years and older (adult) for doxycycline and erythromycin, 8 years and older for phenobarbital, 10 years and older for prednisolone, and 15 years and older for trimethoprim. Pediatric biopharmaceutics are not fully understood where several knowledge gaps have been recently emphasized. The current biowaiver criteria are not suitable for safe application in all pediatric populations.
Collapse
Affiliation(s)
- Ramzi Shawahna
- Department of Physiology, Pharmacology and Toxicology, Faculty of Medicine & Health Sciences, An-Najah National University, New Campus, Building: 19, Office: 1340, P.O. Box 7, Nablus, Palestine.
- An-Najah BioSciences Unit, Center for Poison Control, Chemical and Biological Analyses, An-Najah National University, Nablus, Palestine.
| |
Collapse
|
45
|
Bellanti F, Di Iorio VL, Danhof M, Della Pasqua O. Sampling Optimization in Pharmacokinetic Bridging Studies: Example of the Use of Deferiprone in Children With β-Thalassemia. J Clin Pharmacol 2016; 56:1094-103. [PMID: 26785826 DOI: 10.1002/jcph.708] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2015] [Accepted: 01/13/2016] [Indexed: 01/19/2023]
Abstract
Despite wide clinical experience with deferiprone, the optimum dosage in children younger than 6 years remains to be established. This analysis aimed to optimize the design of a prospective clinical study for the evaluation of deferiprone pharmacokinetics in children. A 1-compartment model with first-order oral absorption was used for the purposes of the analysis. Different sampling schemes were evaluated under the assumption of a constrained population size. A sampling scheme with 5 samples per subject was found to be sufficient to ensure accurate characterization of the pharmacokinetics of deferiprone. Whereas the accuracy of parameters estimates was high, precision was slightly reduced because of the small sample size (CV% >30% for Vd/F and KA). Mean AUC ± SD was found to be 33.4 ± 19.2 and 35.6 ± 20.2 mg · h/mL, and mean Cmax ± SD was found to be 10.2 ± 6.1 and 10.9 ± 6.7 mg/L based on sparse and frequent sampling, respectively. The results showed that typical frequent sampling schemes and sample sizes do not warrant accurate model and parameter identifiability. Expectation of the determinant (ED) optimality and simulation-based optimization concepts can be used to support pharmacokinetic bridging studies. Of importance is the accurate estimation of the magnitude of the covariate effects, as they partly determine the dose recommendation for the population of interest.
Collapse
Affiliation(s)
- Francesco Bellanti
- Division of Pharmacology, Leiden Academic Centre for Drug Research, London, UK
| | | | - Meindert Danhof
- Division of Pharmacology, Leiden Academic Centre for Drug Research, London, UK
| | - Oscar Della Pasqua
- Division of Pharmacology, Leiden Academic Centre for Drug Research, London, UK.,Clinical Pharmacology Modelling & Simulation, GlaxoSmithKline, Stockley Park, UK.,Clinical Pharmacology & Therapeutics, University College London, London, UK
| |
Collapse
|
46
|
Rodieux F, Wilbaux M, van den Anker JN, Pfister M. Effect of Kidney Function on Drug Kinetics and Dosing in Neonates, Infants, and Children. Clin Pharmacokinet 2015; 54:1183-204. [PMID: 26138291 PMCID: PMC4661214 DOI: 10.1007/s40262-015-0298-7] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Neonates, infants, and children differ from adults in many aspects, not just in age, weight, and body composition. Growth, maturation and environmental factors affect drug kinetics, response and dosing in pediatric patients. Almost 80% of drugs have not been studied in children, and dosing of these drugs is derived from adult doses by adjusting for body weight/size. As developmental and maturational changes are complex processes, such simplified methods may result in subtherapeutic effects or adverse events. Kidney function is impaired during the first 2 years of life as a result of normal growth and development. Reduced kidney function during childhood has an impact not only on renal clearance but also on absorption, distribution, metabolism and nonrenal clearance of drugs. 'Omics'-based technologies, such as proteomics and metabolomics, can be leveraged to uncover novel markers for kidney function during normal development, acute kidney injury, and chronic diseases. Pharmacometric modeling and simulation can be applied to simplify the design of pediatric investigations, characterize the effects of kidney function on drug exposure and response, and fine-tune dosing in pediatric patients, especially in those with impaired kidney function. One case study of amikacin dosing in neonates with reduced kidney function is presented. Collaborative efforts between clinicians and scientists in academia, industry, and regulatory agencies are required to evaluate new renal biomarkers, collect and share prospective pharmacokinetic, genetic and clinical data, build integrated pharmacometric models for key drugs, optimize and standardize dosing strategies, develop bedside decision tools, and enhance labels of drugs utilized in neonates, infants, and children.
Collapse
Affiliation(s)
- Frederique Rodieux
- Department of Pediatric Clinical Pharmacology, Pediatric Pharmacology and Pharmacometrics Research Center, University Children's Hospital (UKBB), University of Basel, Spitalstrasse 33, CH-4056, Basel, Switzerland.
| | - Melanie Wilbaux
- Department of Pediatric Clinical Pharmacology, Pediatric Pharmacology and Pharmacometrics Research Center, University Children's Hospital (UKBB), University of Basel, Spitalstrasse 33, CH-4056, Basel, Switzerland
| | - Johannes N van den Anker
- Department of Pediatric Clinical Pharmacology, Pediatric Pharmacology and Pharmacometrics Research Center, University Children's Hospital (UKBB), University of Basel, Spitalstrasse 33, CH-4056, Basel, Switzerland.
- Division of Pediatric Clinical Pharmacology, Children's National Health System, Washington, DC, USA.
- Intensive Care, Erasmus Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands.
| | - Marc Pfister
- Department of Pediatric Clinical Pharmacology, Pediatric Pharmacology and Pharmacometrics Research Center, University Children's Hospital (UKBB), University of Basel, Spitalstrasse 33, CH-4056, Basel, Switzerland
- Quantitative Solutions LP, Menlo Park, CA, USA
| |
Collapse
|
47
|
Zhao W, Kaguelidou F, Biran V, Zhang D, Allegaert K, Capparelli EV, Holford N, Kimura T, Lo YL, Peris JE, Thomson A, van den Anker JN, Fakhoury M, Jacqz-Aigrain E. External Evaluation of Population Pharmacokinetic Models of Vancomycin in Neonates: The transferability of published models to different clinical settings. Br J Clin Pharmacol 2015; 75:1068-80. [PMID: 23148919 DOI: 10.1111/j.1365-2125.2012.04406.x] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Vancomycin is one of the most evaluated antibiotics in neonates using modeling and simulation approaches. However no clear consensus on optimal dosing has been achieved. The objective of the present study was to perform an external evaluation of published models, in order to test their predictive performances in an independent dataset and to identify the possible study-related factors influencing the transferability of pharmacokinetic models to different clinical settings. METHOD Published neonatal vancomycin pharmacokinetic models were screened from the literature. The predictive performance of 6 models was evaluated using an independent dataset (112 concentrations from 78 neonates). The evaluation procedures used simulation-based diagnostics (visual predictive check [VPC] and normalized prediction distribution errors [NPDE]). RESULTS Differences in predictive performances of models for vancomycin pharmacokinetics in neonates were found. The mean of NPDE for 6 evaluated models were 1.35, -0.22, -0.36, 0.24, 0.66, 0.48, respectively. These differences were explained, at least partly, by taking into account the method used to measure serum creatinine concentrations. The adult conversion factor of 1.3 (enzymatic to Jaffé) was tested with an improvement in the VPC and NPDE, but it still need to be evaluated and validated in neonates. Differences were also identified between analytical methods for vancomycin. CONCLUSION The importance of analytical techniques for serum creatinine concentrations and vancomycin as a predictor of vancomycin concentrations in neonates has been confirmed. Dosage individualisation of vancomycin in neonates should consider not only patients' characteristics and clinical conditions, but also the methods used to measure serum creatinine and vancomycin.
Collapse
Affiliation(s)
- Wei Zhao
- Department of Pediatric Pharmacology and Pharmacogenetics, Hôpital Robert Debré, AP-HP, Paris, France; Clinical Investigation Center CIC9202, AP-HP-INSERM, Paris, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Baverel PG, Jain M, Stelmach I, She D, Agoram B, Sandbach S, Piper E, Kuna P. Pharmacokinetics of tralokinumab in adolescents with asthma: implications for future dosing. Br J Clin Pharmacol 2015; 80:1337-49. [PMID: 26182954 DOI: 10.1111/bcp.12725] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Revised: 07/10/2015] [Accepted: 07/13/2015] [Indexed: 12/31/2022] Open
Abstract
AIMS Tralokinumab, an investigational human immunoglobulin G4 monoclonal antibody, potently and specifically neutralizes interleukin-13, a central mediator of asthma. Tralokinumab has shown improvements in clinical endpoints in adults with uncontrolled asthma. The present study explored the pharmacokinetics (PK) and safety of a single tralokinumab dose, and utilized a population PK modelling and simulation approach to evaluate the optimal dosing strategy for adolescents. METHODS Adolescent subjects with asthma, using daily controller medication, received a single subcutaneous dose of tralokinumab 300 mg. Safety, immunogenicity and PK data were collected during a 57-day follow-up. A population PK model was developed using data from the present study and prior studies in adults. Simulations were performed to evaluate dose adjustment requirements for adolescents. RESULTS Twenty adolescents (12-17 years) were enrolled; all completed the study. No clinically relevant safety findings or antidrug antibodies were detected. PK parameters were similar to those observed in adults. PK modelling showed that body weight was a minor predictor of tralokinumab PK; after incorporating body weight into the PK model, a 15% (nonparametric 95% confidence interval 5%, 26%) lower clearance was found in adolescents compared with adults [173 (151, 209) vs. 204 (191, 229) ml day(-1)]. Simulations showed no therapeutically relevant differences in exposures between adolescent and adult populations, and similar PK profiles for weight-based (4 mg kg(-1)) and fixed (300 mg) fortnightly subcutaneous doses of tralokinumab. CONCLUSION Single-dose administration of tralokinumab 300 mg in adolescents was well tolerated, with a PK profile similar to that in adults. Exposure predictions suggest that dose adjustment is not required for adolescents.
Collapse
Affiliation(s)
- Paul G Baverel
- Clinical Pharmacology, Drug Metabolism and Pharmacokinetics, MedImmune, Cambridge, UK
| | - Meena Jain
- Clinical Development, MedImmune, Cambridge, UK
| | - Iwona Stelmach
- Department of Paediatrics and Allergy, Medical University of Łódź, Poland
| | - Dewei She
- Biostatistics, MedImmune, Gaithersburg, MD, USA
| | - Balaji Agoram
- Clinical Pharmacology, Drug Metabolism and Pharmacokinetics, MedImmune, Cambridge, UK
| | | | | | - Piotr Kuna
- Department of Internal Medicine, Asthma and Allergy, Barlicki University Hospital, Medical University of Łódź, Poland
| |
Collapse
|
49
|
Piana C, Danhof M, Della Pasqua O. Influence of covariate distribution on the predictive performance of pharmacokinetic models in paediatric research. Br J Clin Pharmacol 2015; 78:145-57. [PMID: 24433411 DOI: 10.1111/bcp.12322] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Accepted: 11/06/2013] [Indexed: 01/03/2023] Open
Abstract
AIMS The accuracy of model-based predictions often reported in paediatric research has not been thoroughly characterized. The aim of this exercise is therefore to evaluate the role of covariate distributions when a pharmacokinetic model is used for simulation purposes. METHODS Plasma concentrations of a hypothetical drug were simulated in a paediatric population using a pharmacokinetic model in which body weight was correlated with clearance and volume of distribution. Two subgroups of children were then selected from the overall population according to a typical study design, in which pre-specified body weight ranges (10-15 kg and 30-40 kg) were used as inclusion criteria. The simulated data sets were then analyzed using non-linear mixed effects modelling. Model performance was assessed by comparing the accuracy of AUC predictions obtained for each subgroup, based on the model derived from the overall population and by extrapolation of the model parameters across subgroups. RESULTS Our findings show that systemic exposure as well as pharmacokinetic parameters cannot be accurately predicted from the pharmacokinetic model obtained from a population with a different covariate range from the one explored during model building. Predictions were accurate only when a model was used for prediction in a subgroup of the initial population. CONCLUSIONS In contrast to current practice, the use of pharmacokinetic modelling in children should be limited to interpolations within the range of values observed during model building. Furthermore, the covariate point estimate must be kept in the model even when predictions refer to a subset different from the original population.
Collapse
Affiliation(s)
- Chiara Piana
- LACDR, Division of Pharmacology, Leiden University, Leiden, The Netherlands
| | | | | |
Collapse
|
50
|
Broglio KR, Sandalic L, Albertson T, Berry SM. Bayesian dose escalation in oncology with sharing of information between patient populations. Contemp Clin Trials 2015; 44:56-63. [DOI: 10.1016/j.cct.2015.07.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 07/01/2015] [Accepted: 07/03/2015] [Indexed: 10/23/2022]
|