1
|
Yang Y, Qian Z, Wu C, Cheng Y, Yang B, Shao J, Zhao J, Zhu X, Jia X, Feng L. Differential absorption and metabolic characteristics of organic acid components in pudilan xiaoyan oral liquid between young rats and adult rats. JOURNAL OF ETHNOPHARMACOLOGY 2024; 334:118528. [PMID: 38972526 DOI: 10.1016/j.jep.2024.118528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 07/01/2024] [Accepted: 07/04/2024] [Indexed: 07/09/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Pudilan Xiaoyan Oral Liquid (PDL) is a proprietary Chinese medicinal preparation approved by the State for treating acute pharyngitis in both adults and children (Approval No. Z20030095). It is worth noting that children exhibit unique physiopathological characteristics compared to adults. However, the in vivo regulatory characteristics of PDL in treating acute pharyngitis in children remain incompletely understood. AIM OF THE STUDY The differential absorption and metabolism characteristics of the main pharmacological components in PDL in young and adult rats were investigated with a view to providing a reference for preclinical data of PDL in medication for children. MATERIALS AND METHODS This study utilized UPLC-Q-TOF-MS to investigate the pharmacodynamic material basis of PDL. The focus was on the gastrointestinal digestion and absorption characteristics of organic acid components in PDL (PDL-OAC), known as the primary pharmacodynamic components in this formulation. The research combined in vitro dynamic simulation and a Quadruple single-pass intestinal perfusion model to examine these characteristics. The permeability properties of PDL-OAC were evaluated using an artificial parallel membrane model. Additionally, an acute pharyngitis model was established to evaluate the histopathological condition of the pharynx in young rats using H&E staining. The levels of IL-1β, TNF-α, IL-6, and IL-10 in blood and pharyngeal tissue homogenates of young rats were quantified using ELISA kits. RESULTS A total of 91 components were identified in PDL, including 33 organic acids, 24 flavonoids, 14 alkaloids, 5 terpenoids and coumarins, 3 sugars, and 12 amino acids. The PDL-OAC exhibited a significant reduction in IL-1β, TNF-α, IL-6, and IL-10 levels in the pharyngeal tissues of young rats with acute pharyngitis. Results from dynamic simulation studies of gastrointestinal fluids revealed that the PDL-OAC (Specifically chlorogenic acid (CGA), gallic acid (GA), chicoric acid (CRA), and caffeic acid (CA)) were effectively stabilized in the gastrointestinal fluids of both children and adults in vitro. Young rats, characterized by thinner intestinal walls and higher permeability, efficiently absorbed the four organic acids across the entire intestinal segment. The absorption of CGA, GA, and CRA followed a concentration-dependent pattern, with CGA and GA absorption being influenced by exocytosis. CONCLUSION The efficacy of the PDL-OAC in treating acute pharyngitis was demonstrated in young rats. The absorption rate of these components was observed to be faster in young rats compared to adult rats, underscoring the need for dedicated studies on the drug's usage in children. This research provides valuable insights for the appropriate clinical use of PDL in pediatric patients.
Collapse
Affiliation(s)
- Yanjun Yang
- School of Traditional Chinese Pharmacy, Innovation Center for Industry-Education Integration of Pediatrics and Traditional Chinese Medicine, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 211198, PR China.
| | - Zhouyang Qian
- School of Traditional Chinese Pharmacy, Innovation Center for Industry-Education Integration of Pediatrics and Traditional Chinese Medicine, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 211198, PR China.
| | - Chenhui Wu
- School of Traditional Chinese Pharmacy, Innovation Center for Industry-Education Integration of Pediatrics and Traditional Chinese Medicine, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 211198, PR China.
| | - Yue Cheng
- School of Traditional Chinese Pharmacy, Innovation Center for Industry-Education Integration of Pediatrics and Traditional Chinese Medicine, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 211198, PR China.
| | - Bing Yang
- School of Traditional Chinese Pharmacy, Innovation Center for Industry-Education Integration of Pediatrics and Traditional Chinese Medicine, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 211198, PR China.
| | - Jianguo Shao
- Jiangsu Key Laboratory of Chinese Medicine and Characteristic Preparations for Paediatrics, Jumpcan Pharmaceutical Co., Ltd., Taixing, 225400, PR China.
| | - Jing Zhao
- Jiangsu Key Laboratory of Chinese Medicine and Characteristic Preparations for Paediatrics, Jumpcan Pharmaceutical Co., Ltd., Taixing, 225400, PR China.
| | - Xiangjun Zhu
- Jiangsu Health Development Research Center, National Health and Family Planning Commission Contraceptives Adverse Reaction Surveillance Center, Nanjing, 210036, PR China.
| | - Xiaobin Jia
- School of Traditional Chinese Pharmacy, Innovation Center for Industry-Education Integration of Pediatrics and Traditional Chinese Medicine, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 211198, PR China.
| | - Liang Feng
- School of Traditional Chinese Pharmacy, Innovation Center for Industry-Education Integration of Pediatrics and Traditional Chinese Medicine, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 211198, PR China.
| |
Collapse
|
2
|
Streekstra EJ, Keuper-Navis M, van den Heuvel JJMW, van den Broek P, Stommel MWJ, de Boode W, Botden S, Bervoets S, O’Gorman L, Greupink R, Russel FGM, van de Steeg E, de Wildt SN. Enteroids to Study Pediatric Intestinal Drug Transport. Mol Pharm 2024; 21:4983-4994. [PMID: 39279643 PMCID: PMC11462498 DOI: 10.1021/acs.molpharmaceut.4c00339] [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: 03/29/2024] [Revised: 08/30/2024] [Accepted: 08/30/2024] [Indexed: 09/18/2024]
Abstract
Intestinal maturational changes after birth affect the pharmacokinetics (PK) of drugs, having major implications for drug safety and efficacy. However, little is known about ontogeny-related PK patterns in the intestine. To explore the accuracy of human enteroid monolayers for studying drug transport in the pediatric intestine, we compared the drug transporter functionality and expression in enteroid monolayers and tissue from pediatrics and adults. Enteroid monolayers were cultured of 14 pediatric [median (range) age: 44 weeks (2 days-13 years)] and 5 adult donors, in which bidirectional drug transport experiments were performed. In parallel, we performed similar experiments with tissue explants in Ussing chamber using 11 pediatric [median (range) age: 54 weeks (15 weeks-10 years)] and 6 adult tissues. Enalaprilat, propranolol, talinolol, and rosuvastatin were used to test paracellular, transcellular, and transporter-mediated efflux by P-gp and breast cancer resistance protein (BCRP), respectively. In addition, we compared the expression patterns of ADME-related genes in pediatric and adult enteroid monolayers with tissues using RNA sequencing. Efflux transport by P-gp and BCRP was comparable between the enteroids and tissue. Efflux ratios (ERs) of talinolol and rosuvastatin by P-gp and BCRP, respectively, were higher in enteroid monolayers compared to Ussing chamber, likely caused by experimental differences in model setup and cellular layers present. Explorative statistics on the correlation with age showed trends of increasing ER with age for P-gp in enteroid monolayers; however, it was not significant. In the Ussing chamber setup, lower enalaprilat and propranolol transport was observed with age. Importantly, the RNA sequencing pathway analysis revealed that age-related variation in drug metabolism between neonates and adults was present in both enteroids and intestinal tissue. Age-related differences between 0 and 6 months old and adults were observed in tissue as well as in enteroid monolayers, although to a lesser extent. This study provides the first data for the further development of pediatric enteroids as an in vitro model to study age-related variation in drug transport. Overall, drug transport in enteroids was in line with data obtained from ex vivo tissue (using chamber) experiments. Additionally, pathway analysis showed similar PK-related differences between neonates and adults in both tissue and enteroid monolayers. Given the challenge to elucidate the effect of developmental changes in the pediatric age range in human tissue, intestinal enteroids derived from pediatric patients could provide a versatile experimental platform to study pediatric phenotypes.
Collapse
Affiliation(s)
- Eva J. Streekstra
- Division
of Pharmacology and Toxicology, Department of Pharmacy, Radboud University Medical Center, Nijmegen 6525GA, The Netherlands
- Department
of Metabolic Health Research, Netherlands
Organization for Applied Scientific Research (TNO), Leiden 2333BE, The Netherlands
| | - Marit Keuper-Navis
- Department
of Metabolic Health Research, Netherlands
Organization for Applied Scientific Research (TNO), Leiden 2333BE, The Netherlands
- Division
of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht 3584 CS, The Netherlands
| | - Jeroen J. M. W. van den Heuvel
- Division
of Pharmacology and Toxicology, Department of Pharmacy, Radboud University Medical Center, Nijmegen 6525GA, The Netherlands
| | - Petra van den Broek
- Division
of Pharmacology and Toxicology, Department of Pharmacy, Radboud University Medical Center, Nijmegen 6525GA, The Netherlands
| | - Martijn W. J. Stommel
- Department
of Surgery, Radboud University Medical Center, Nijmegen 6525GA, The Netherlands
| | - Willem de Boode
- Department
of Pediatrics, Division of Neonatology, Radboud University Medical Center, Amalia Children’s Hospital, Nijmegen 6525GA, The Netherlands
| | - Sanne Botden
- Department
of Surgery, Amalia Children’s Hospital, Radboud University Medical Center, Nijmegen 6525GA, The Netherlands
| | - Sander Bervoets
- Radboudumc
Technology Center for Bioinformatics, Department of Medical BioSciences, Radboud University Medical Center, Nijmegen 6525GA, The Netherlands
| | - Luke O’Gorman
- Radboudumc
Technology Center for Bioinformatics, Department of Medical BioSciences, Radboud University Medical Center, Nijmegen 6525GA, The Netherlands
| | - Rick Greupink
- Division
of Pharmacology and Toxicology, Department of Pharmacy, Radboud University Medical Center, Nijmegen 6525GA, The Netherlands
| | - Frans G. M. Russel
- Division
of Pharmacology and Toxicology, Department of Pharmacy, Radboud University Medical Center, Nijmegen 6525GA, The Netherlands
| | - Evita van de Steeg
- Department
of Metabolic Health Research, Netherlands
Organization for Applied Scientific Research (TNO), Leiden 2333BE, The Netherlands
| | - Saskia N. de Wildt
- Division
of Pharmacology and Toxicology, Department of Pharmacy, Radboud University Medical Center, Nijmegen 6525GA, The Netherlands
- Department
of Intensive Care, Radboud University Medical
Center, Nijmegen 6525GA, The Netherlands
- Department
of Neonatal and Pediatric Intensive Care, Erasmus MC Sophia Children’s Hospital, Rotterdam 3015 GD, The Netherlands
| |
Collapse
|
3
|
Streekstra EJ, Keuper-Navis M, van den Heuvel JJWM, van den Broek P, Greupink R, Stommel MWJ, de Boode WP, Botden SMBI, Russel FGM, van de Steeg E, de Wildt SN. The potential of enteroids derived from children and adults to study age-dependent differences in intestinal CYP3A4/5 metabolism. Eur J Pharm Sci 2024; 201:106868. [PMID: 39084538 DOI: 10.1016/j.ejps.2024.106868] [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] [Received: 03/29/2024] [Revised: 07/12/2024] [Accepted: 07/27/2024] [Indexed: 08/02/2024]
Abstract
Drug metabolism in the intestinal wall affects bioavailability of orally administered drugs and is influenced by age. Hence, it is important to fully understand the drug metabolizing capacity of the gut to predict systemic exposure. The aim of this study was to investigate the potential of enteroids as a tool to study CYP3A4/5 -mediated metabolism in both children and adults. Bioconversion of midazolam, a CYP3A4/5 model substrate, was studied using enteroid monolayers as well as tissue explants in the Ussing chamber, both derived from pediatric [median (range age): 54 weeks (2 days - 13 years), n = 21] and adult (n = 5) tissue. Caco-2 cellular monolayers were employed as controls. In addition, mRNA expression of CYP3A4 was determined in enteroid monolayers (n = 11), tissue (n = 23) and Caco-2 using RT-qPCR. Midazolam metabolism was successfully detected in all enteroid monolayers, as well as in all tissue explants studied in the Ussing chamber, whereas Caco-2 showed no significant metabolite formation. The extracted fraction of midazolam was similar between enteroid monolayers and tissue. The fraction of midazolam extracted increased with age in enteroid monolayers derived from 0 to 70 week old donors. No statistically significant correlation was observed in tissue likely due to high variability observed and the smaller donor numbers included in the study. At the level of gene expression, CYP3A4 increased with age in tissues (n = 32), while this was not reflected in enteroid monolayers (n = 16). Notably, asymmetric metabolite formation was observed in enteroids and tissue, with higher metabolite formation on the luminal side of the barrier. In summary, we demonstrated that enteroids can be used to measure CYP3A4/5 midazolam metabolism, which we show is similar as observed in fresh isolated tissue. This was the case both in children and adults, indicating the potential of enteroids to predict intestinal metabolism. This study provides promising data to further develop enteroids to study drug metabolism in vitro and potentially predict oral absorption for special populations as an alternative to using fresh tissue.
Collapse
Affiliation(s)
- Eva J Streekstra
- Division of Pharmacology and Toxicology, Department of Pharmacy, Radboud University Medical Center, Nijmegen, the Netherlands; Department of Metabolic Health Research, Netherlands Organization for Applied Scientific Research (TNO), Leiden, the Netherlands
| | - Marit Keuper-Navis
- Department of Metabolic Health Research, Netherlands Organization for Applied Scientific Research (TNO), Leiden, the Netherlands; Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, the Netherlands
| | - Jeroen J W M van den Heuvel
- Division of Pharmacology and Toxicology, Department of Pharmacy, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Petra van den Broek
- Division of Pharmacology and Toxicology, Department of Pharmacy, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Rick Greupink
- Division of Pharmacology and Toxicology, Department of Pharmacy, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Martijn W J Stommel
- Department of Surgery, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Willem P de Boode
- Department of Pediatrics, Division of Neonatology, Radboud University Medical Center, Amalia Children's Hospital, Nijmegen, the Netherlands
| | - Sanne M B I Botden
- Amalia Children's Hospital, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Frans G M Russel
- Division of Pharmacology and Toxicology, Department of Pharmacy, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Evita van de Steeg
- Department of Metabolic Health Research, Netherlands Organization for Applied Scientific Research (TNO), Leiden, the Netherlands
| | - Saskia N de Wildt
- Division of Pharmacology and Toxicology, Department of Pharmacy, Radboud University Medical Center, Nijmegen, the Netherlands; Department of Intensive Care, Radboud University Medical Center, Nijmegen, the Netherlands; Department of Neonatal and Pediatric Intensive Care, Erasmus MC Sophia Children's Hospital, Rotterdam, the Netherlands.
| |
Collapse
|
4
|
Meesters K, Balbas-Martinez V, Allegaert K, Downes KJ, Michelet R. Personalized Dosing of Medicines for Children: A Primer on Pediatric Pharmacometrics for Clinicians. Paediatr Drugs 2024; 26:365-379. [PMID: 38755515 DOI: 10.1007/s40272-024-00633-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/25/2024] [Indexed: 05/18/2024]
Abstract
The widespread use of drugs for unapproved purposes remains common in children, primarily attributable to practical, ethical, and financial constraints associated with pediatric drug research. Pharmacometrics, the scientific discipline that involves the application of mathematical models to understand and quantify drug effects, holds promise in advancing pediatric pharmacotherapy by expediting drug development, extending applications, and personalizing dosing. In this review, we delineate the principles of pharmacometrics, and explore its clinical applications and prospects. The fundamental aspect of any pharmacometric analysis lies in the selection of appropriate methods for quantifying pharmacokinetics and pharmacodynamics. Population pharmacokinetic modeling is a data-driven method ('top-down' approach) to approximate population-level pharmacokinetic parameters, while identifying factors contributing to inter-individual variability. Model-informed precision dosing is increasingly used to leverage population pharmacokinetic models and patient data, to formulate individualized dosing recommendations. Physiologically based pharmacokinetic models integrate physicochemical drug properties with biological parameters ('bottom-up approach'), and is particularly valuable in situations with limited clinical data, such as early drug development, assessing drug-drug interactions, or adapting dosing for patients with specific comorbidities. The effective implementation of these complex models hinges on strong collaboration between clinicians and pharmacometricians, given the pivotal role of data availability. Promising advancements aimed at improving data availability encompass innovative techniques such as opportunistic sampling, minimally invasive sampling approaches, microdialysis, and in vitro investigations. Additionally, ongoing research efforts to enhance measurement instruments for evaluating pharmacodynamics responses, including biomarkers and clinical scoring systems, are expected to significantly bolster our capacity to understand drug effects in children.
Collapse
Affiliation(s)
- Kevin Meesters
- Department of Pediatrics, University of British Columbia, 4480 Oak Street, Vancouver, BC, V6H 3V4, Canada.
- Vaccine Evaluation Center, BC Children's Hospital Research Institute, Vancouver, BC, Canada.
| | | | - Karel Allegaert
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
- Department of Hospital Pharmacy, Erasmus MC, Rotterdam, The Netherlands
| | - Kevin J Downes
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Division of Infectious Diseases, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Robin Michelet
- Department of Clinical Pharmacy and Biochemistry, Institute of Pharmacy, Freie Universität Berlin, Berlin, Germany
- qPharmetra LLC, Berlin, Germany
| |
Collapse
|
5
|
Caleffi‐Marchesini ER, Herling AA, Macente J, Bonan RH, de Freitas Lima P, Moreno R, Alexandre V, Charbe NB, Borghi‐Pangoni FB, Cristofoletti R, Diniz A. Adult and pediatric physiologically-based biopharmaceutics modeling to explain lamotrigine immediate release absorption process. CPT Pharmacometrics Syst Pharmacol 2024; 13:208-221. [PMID: 37916262 PMCID: PMC10864931 DOI: 10.1002/psp4.13071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 09/17/2023] [Accepted: 09/25/2023] [Indexed: 11/03/2023] Open
Abstract
Physiologically-based biopharmaceutics modeling (PBBM) has potential to accelerate the development of new drug and formulations. An important application of PBBM is for special populations such as pediatrics that have pharmacokinetics dependent on the maturation process. Lamotrigine (LTG) is a Biopharmaceutics Classification System (BCS) II drug and is widely prescribed. Therefore, the goal of this study was to assess the biopharmaceutics risk of the low-soluble drug LTG when the ontogeny on gastrointestinal tract (GIT) physiological parameters are considered. An oral physiologically-based pharmacokinetic model and a PBBM were developed and verified using GastroPlus™ software for both adults and children (2-12 years old, 12-52 kg). The biopharmaceutics properties and GIT physiological parameters were evaluated by sensitivity analysis. High doses were simulated assuming a worst case scenario, that is, the dose of 200 mg for adults and 5 mg/kg (up to the maximum of 200 mg) for 2-year-old children. Although several authors have suggested that ontogeny may have an effect on gastrointestinal fluid volume, our study found no evidence of interference between fluid and dose volumes with in vivo dissolution of LTG. The most impactful parameter was found to be the gastric transit time. Therefore, the hypothesis is developed to examine whether LTG exhibits characteristics of a BCS II classification in vitro while showing BCS I-like behavior in vivo. This hypothesis could act as a base for conducting novel studies on model-informed precision dosing, tailored to specific populations and clinical conditions. In addition, it could be instrumental in assessing the influence of various release profiles on in vivo performance for both adult and pediatric populations.
Collapse
Affiliation(s)
| | - Amanda Antunes Herling
- Pharmacokinetics and Biopharmaceutics LaboratoryState University of MaringáMaringáPRBrazil
| | - Julia Macente
- Pharmacokinetics and Biopharmaceutics LaboratoryState University of MaringáMaringáPRBrazil
| | | | | | - Rafaela Moreno
- Centro Universitário Barão de MauáRibeirão PretoSPBrazil
| | - Veriano Alexandre
- Hospital das Clínicas, Faculdade de MedicinaUniversidade de São PauloRibeirão PretoSPBrazil
| | - Nitin Bharat Charbe
- Center for Pharmacometrics & Systems PharmacologyUniversity of FloridaOrlandoFloridaUSA
| | | | - Rodrigo Cristofoletti
- Center for Pharmacometrics & Systems PharmacologyUniversity of FloridaOrlandoFloridaUSA
| | - Andréa Diniz
- Pharmacokinetics and Biopharmaceutics LaboratoryState University of MaringáMaringáPRBrazil
| |
Collapse
|
6
|
Zhang W, Zhang Q, Cao Z, Zheng L, Hu W. Physiologically Based Pharmacokinetic Modeling in Neonates: Current Status and Future Perspectives. Pharmaceutics 2023; 15:2765. [PMID: 38140105 PMCID: PMC10747965 DOI: 10.3390/pharmaceutics15122765] [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: 10/20/2023] [Revised: 12/07/2023] [Accepted: 12/09/2023] [Indexed: 12/24/2023] Open
Abstract
Rational drug use in special populations is a clinical problem that doctors and pharma-cists must consider seriously. Neonates are the most physiologically immature and vulnerable to drug dosing. There is a pronounced difference in the anatomical and physiological profiles be-tween neonates and older people, affecting the absorption, distribution, metabolism, and excretion of drugs in vivo, ultimately leading to changes in drug concentration. Thus, dose adjustments in neonates are necessary to achieve adequate therapeutic concentrations and avoid drug toxicity. Over the past few decades, modeling and simulation techniques, especially physiologically based pharmacokinetic (PBPK) modeling, have been increasingly used in pediatric drug development and clinical therapy. This rigorously designed and verified model can effectively compensate for the deficiencies of clinical trials in neonates, provide a valuable reference for clinical research design, and even replace some clinical trials to predict drug plasma concentrations in newborns. This review introduces previous findings regarding age-dependent physiological changes and pathological factors affecting neonatal pharmacokinetics, along with their research means. The application of PBPK modeling in neonatal pharmacokinetic studies of various medications is also reviewed. Based on this, we propose future perspectives on neonatal PBPK modeling and hope for its broader application.
Collapse
Affiliation(s)
| | | | | | - Liang Zheng
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; (W.Z.); (Q.Z.); (Z.C.)
| | - Wei Hu
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; (W.Z.); (Q.Z.); (Z.C.)
| |
Collapse
|
7
|
Islam N, Ullah Khan N, Razzaq A, Ullah Khan Z, Menaa F, Alfaifi MY, Elbehairi SEI, Iqbal H, Ni J. Self-emulsifying micelles as a drug nanocarrier system for itraconazole oral bioavailability enhancement; in vitro and in vivo assessment. Saudi Pharm J 2023; 31:101839. [PMID: 37965489 PMCID: PMC10641562 DOI: 10.1016/j.jsps.2023.101839] [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: 05/25/2023] [Accepted: 10/19/2023] [Indexed: 11/16/2023] Open
Abstract
Itraconazole (ITZ) is a renowned antifungal medication, however its therapeutic efficacy is limited by low solubility and oral bioavailability. The current research work attempted to augment the oral bioavailability of ITZ by incorporating into self-emulsifying micelles (SEMCs). To fabricate the SEMCs, various preparation techniques including physical mixture, melt-emulsification, solvent evaporation and kneading, were opted by using different weight ratio of drug and solubilizers i.e. Gelucire-50/13 or Gelucire-44/14 and characterized both in vitro and in vivo. The prepared SEMCs were found to be in the size range from 63.4 ± 5.2 to 284.2 ± 19.5 nm with surface charges ranging from -16 ± 1.2 to -27 ± 2.0 mV. The drug solubility was improved to a reasonable extent with all investigated formulations, however, SEMCs in group 6 prepared by kneading method (KMG6) using Gelucire-44/14: drug (10:1 presented 87.6 folds' increase (964.93 ± 2 μg/mL) compared to solubility of crystalline ITZ (11 ± 2 μg/mL) through kneading method. In addition, KMG6 SEMCs shows the fast drug release compared to other SEMCs. Further, KMG6 SEMCs also exhibited 5.12-fold higher relative intestinal serosal fluid absorption compared to crystalline ITZ. The pharmacokinetic parameters such Cmax, AUC and Tmax of KMG6 SEMCs significantly improved compared to crystalline ITZ. In conclusion, the manipulation of ITZ solubility, dissolution rate and absorption using SEMCs is a promising strategy for bioavailability enhancement.
Collapse
Affiliation(s)
- Nayyer Islam
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, GC University, Faisalabad, Pakistan
| | - Naveed Ullah Khan
- Department of Pharmacy, CECOS University of IT and Emerging Sciences, Peshawar 25000, Pakistan
| | - Anam Razzaq
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Zaheer Ullah Khan
- Department of Pharmacy, COMSATS University Islamabad, Abbottabad Campus, Pakistan
| | - Farid Menaa
- Departments of Oncology and Nanomedicine, California Innovations Corporation, San Diego, CA 92037, USA
| | - Mohammad Y. Alfaifi
- King Khalid University, Faculty of Science, Biology Department, Abha 9004, Saudi Arabia
| | | | - Haroon Iqbal
- Zhejiang Cancer Hospital, Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Jiang Ni
- Department of Pharmacy, Affiliated Hospital of Jiangnan University, Wuxi 214000, China
| |
Collapse
|
8
|
Ye J, Bi Y, Ting N. How to select the initial dose for a pediatric study? J Biopharm Stat 2023; 33:844-858. [PMID: 36476267 DOI: 10.1080/10543406.2022.2149770] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 11/16/2022] [Indexed: 12/13/2022]
Abstract
In typical clinical development programs, a new drug is first developed for the adult use. Drugs are often approved for adult use or in the process of obtaining approval in adults in the target indication before pediatric development is initiated. In designing the first pediatric clinical trial, one of the challenges is to select the initial dose to be tested. The ICH E11 R1 guidance advises that chronologic age alone may not always be the most appropriate categorical determinant to define developmental subgroups in pediatric studies. In this manuscript, the approaches to utilize available data in adults related to those factors beyond age to inform the starting dose selection in pediatric drug development are discussed. Practical considerations and approaches are provided for informing pediatric starting dose. Additional considerations to use pre-clinical information are provided in the case when adult information is limited or not available.
Collapse
Affiliation(s)
- Jingjing Ye
- Global Statistics and Data Science (GSDS), Fulton, MD, USA
| | - Youwei Bi
- Division of Pharmacometrics, Office of Translational Sciences (OTS), Center for Drug Evaluation and Research (CDER), US Food and Drug Administration (FDA), Silver Spring, MD, USA
| | - Naitee Ting
- Biostatistics and Data Science, Boehringer Ingelheim Pharmaceuticals, Inc, Ridgefield, CT, USA
| |
Collapse
|
9
|
Developmental Pharmacokinetics of Antibiotics Used in Neonatal ICU: Focus on Preterm Infants. Biomedicines 2023; 11:biomedicines11030940. [PMID: 36979919 PMCID: PMC10046592 DOI: 10.3390/biomedicines11030940] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/06/2023] [Accepted: 03/07/2023] [Indexed: 03/22/2023] Open
Abstract
Neonatal Infections are among the most common reasons for admission to the intensive care unit. Neonatal sepsis (NS) significantly contributes to mortality rates. Empiric antibiotic therapy of NS recommended by current international guidelines includes benzylpenicillin, ampicillin/amoxicillin, and aminoglycosides (gentamicin). The rise of antibacterial resistance precipitates the growth of the use of antibiotics of the Watch (second, third, and fourth generations of cephalosporines, carbapenems, macrolides, glycopeptides, rifamycins, fluoroquinolones) and Reserve groups (fifth generation of cephalosporines, oxazolidinones, lipoglycopeptides, fosfomycin), which are associated with a less clinical experience and higher risks of toxic reactions. A proper dosing regimen is essential for effective and safe antibiotic therapy, but its choice in neonates is complicated with high variability in the maturation of organ systems affecting drug absorption, distribution, metabolism, and excretion. Changes in antibiotic pharmacokinetic parameters result in altered efficacy and safety. Population pharmacokinetics can help to prognosis outcomes of antibiotic therapy, but it should be considered that the neonatal population is heterogeneous, and this heterogeneity is mainly determined by gestational and postnatal age. Preterm neonates are common in clinical practice, and due to the different physiology compared to the full terms, constitute a specific neonatal subpopulation. The objective of this review is to summarize the evidence about the developmental changes (specific for preterm and full-term infants, separately) of pharmacokinetic parameters of antibiotics used in neonatal intensive care units.
Collapse
|
10
|
Sun S, Wang H. Clocking Epilepsies: A Chronomodulated Strategy-Based Therapy for Rhythmic Seizures. Int J Mol Sci 2023; 24:4223. [PMID: 36835631 PMCID: PMC9962262 DOI: 10.3390/ijms24044223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 02/08/2023] [Accepted: 02/14/2023] [Indexed: 02/22/2023] Open
Abstract
Epilepsy is a neurological disorder characterized by hypersynchronous recurrent neuronal activities and seizures, as well as loss of muscular control and sometimes awareness. Clinically, seizures have been reported to display daily variations. Conversely, circadian misalignment and circadian clock gene variants contribute to epileptic pathogenesis. Elucidation of the genetic bases of epilepsy is of great importance because the genetic variability of the patients affects the efficacies of antiepileptic drugs (AEDs). For this narrative review, we compiled 661 epilepsy-related genes from the PHGKB and OMIM databases and classified them into 3 groups: driver genes, passenger genes, and undetermined genes. We discuss the potential roles of some epilepsy driver genes based on GO and KEGG analyses, the circadian rhythmicity of human and animal epilepsies, and the mutual effects between epilepsy and sleep. We review the advantages and challenges of rodents and zebrafish as animal models for epileptic studies. Finally, we posit chronomodulated strategy-based chronotherapy for rhythmic epilepsies, integrating several lines of investigation for unraveling circadian mechanisms underpinning epileptogenesis, chronopharmacokinetic and chronopharmacodynamic examinations of AEDs, as well as mathematical/computational modeling to help develop time-of-day-specific AED dosing schedules for rhythmic epilepsy patients.
Collapse
Affiliation(s)
- Sha Sun
- Center for Circadian Clocks, Soochow University, Suzhou 215123, China
- School of Biology and Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou 215123, China
| | - Han Wang
- Center for Circadian Clocks, Soochow University, Suzhou 215123, China
- School of Biology and Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou 215123, China
| |
Collapse
|
11
|
Meesters K, Alemayehu T, Benou S, Buonsenso D, Decloedt EH, Pillay-Fuentes Lorente V, Downes KJ, Allegaert K. Pharmacokinetics of Antimicrobials in Children with Emphasis on Challenges Faced by Low and Middle Income Countries, a Clinical Review. Antibiotics (Basel) 2022; 12:17. [PMID: 36671218 PMCID: PMC9854442 DOI: 10.3390/antibiotics12010017] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/16/2022] [Accepted: 12/18/2022] [Indexed: 12/25/2022] Open
Abstract
Effective antimicrobial exposure is essential to treat infections and prevent antimicrobial resistance, both being major public health problems in low and middle income countries (LMIC). Delivery of drug concentrations to the target site is governed by dose and pharmacokinetic processes (absorption, distribution, metabolism and excretion). However, specific data on the pharmacokinetics of antimicrobials in children living in LMIC settings are scarce. Additionally, there are significant logistical constraints to therapeutic drug monitoring that further emphasize the importance of understanding pharmacokinetics and dosing in LMIC. Both malnutrition and diarrheal disease reduce the extent of enteral absorption. Multiple antiretrovirals and antimycobacterial agents, commonly used by children in low resource settings, have potential interactions with other antimicrobials. Hypoalbuminemia, which may be the result of malnutrition, nephrotic syndrome or liver failure, increases the unbound concentrations of protein bound drugs that may therefore be eliminated faster. Kidney function develops rapidly during the first years of life and different inflammatory processes commonly augment renal clearance in febrile children, potentially resulting in subtherapeutic drug concentrations if doses are not adapted. Using a narrative review approach, we outline the effects of growth, maturation and comorbidities on maturational and disease specific effects on pharmacokinetics in children in LMIC.
Collapse
Affiliation(s)
- Kevin Meesters
- Department of Pediatrics, BC Children’s Hospital and The University of British Columbia, 4500 Oak Street, Vancouver, BC V6H 3N1, Canada
| | - Tinsae Alemayehu
- Division of Pediatric Infectious Diseases, Department of Pediatrics and Child Health, St. Paul’s Hospital Millennium Medical College, Addis Ababa P.O. Box 1271, Ethiopia
- Division of Infectious Diseases and Travel Medicine, American Medical Center, Addis Ababa P.O. Box 62706, Ethiopia
| | - Sofia Benou
- Department of Pediatrics, General University Hospital of Patras, Medical School, University of Patras, 26504 Rion, Greece
| | - Danilo Buonsenso
- Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del Sacro Cuore, Largo A. Gemelli 8, 00168 Rome, Italy
- Centro di Salute Globale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Eric H. Decloedt
- Division of Clinical Pharmacology, Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town 7500, South Africa
| | - Veshni Pillay-Fuentes Lorente
- Division of Clinical Pharmacology, Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town 7500, South Africa
| | - Kevin J. Downes
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA 19104, USA
- Division of Infectious Diseases, The Children’s Hospital of Philadelphia, 3401 Civic Center Blvd, Philadelphia, PA 19104, USA
| | - Karel Allegaert
- Department of Development and Regeneration, KU Leuven, Herestraat 49, B-3000 Leuven, Belgium
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Herestraat 49, B-3000 Leuven, Belgium
- Department of Clinical Pharmacy, Erasmus Medical Center, Doctor Molewaterplein 40, 3015 GD Rotterdam, The Netherlands
| |
Collapse
|
12
|
Smits A, Annaert P, Cavallaro G, De Cock PAJG, de Wildt SN, Kindblom JM, Lagler FB, Moreno C, Pokorna P, Schreuder MF, Standing JF, Turner MA, Vitiello B, Zhao W, Weingberg AM, Willmann R, van den Anker J, Allegaert K. Current knowledge, challenges and innovations in developmental pharmacology: A combined conect4children Expert Group and European Society for Developmental, Perinatal and Paediatric Pharmacology White Paper. Br J Clin Pharmacol 2022; 88:4965-4984. [PMID: 34180088 PMCID: PMC9787161 DOI: 10.1111/bcp.14958] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 05/22/2021] [Accepted: 05/30/2021] [Indexed: 12/30/2022] Open
Abstract
Developmental pharmacology describes the impact of maturation on drug disposition (pharmacokinetics, PK) and drug effects (pharmacodynamics, PD) throughout the paediatric age range. This paper, written by a multidisciplinary group of experts, summarizes current knowledge, and provides suggestions to pharmaceutical companies, regulatory agencies and academicians on how to incorporate the latest knowledge regarding developmental pharmacology and innovative techniques into neonatal and paediatric drug development. Biological aspects of drug absorption, distribution, metabolism and excretion throughout development are summarized. Although this area made enormous progress during the last two decades, remaining knowledge gaps were identified. Minimal risk and burden designs allow for optimally informative but minimally invasive PK sampling, while concomitant profiling of drug metabolites may provide additional insight in the unique PK behaviour in children. Furthermore, developmental PD needs to be considered during drug development, which is illustrated by disease- and/or target organ-specific examples. Identifying and testing PD targets and effects in special populations, and application of age- and/or population-specific assessment tools are discussed. Drug development plans also need to incorporate innovative techniques such as preclinical models to study therapeutic strategies, and shift from sequential enrolment of subgroups, to more rational designs. To stimulate appropriate research plans, illustrations of specific PK/PD-related as well as drug safety-related challenges during drug development are provided. The suggestions made in this joint paper of the Innovative Medicines Initiative conect4children Expert group on Developmental Pharmacology and the European Society for Developmental, Perinatal and Paediatric Pharmacology, should facilitate all those involved in drug development.
Collapse
Affiliation(s)
- Anne Smits
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium.,Neonatal intensive Care unit, University Hospitals Leuven, Leuven, Belgium
| | - Pieter Annaert
- Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Giacomo Cavallaro
- Neonatal intensive care unit, Fondazione IRCCS Ca' Grande Ospedale Maggiore Policlinico, Milan, Italy
| | - Pieter A J G De Cock
- Department of Pediatric Intensive Care, Ghent University Hospital, Ghent, Belgium.,Heymans Institute of Pharmacology, Ghent University, Ghent, Belgium.,Department of Pharmacy, Ghent University Hospital, Ghent, Belgium
| | - Saskia N de Wildt
- Intensive Care and Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, the Netherlands.,Department of Pharmacology and Toxicology, Radboud Institute Health Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Jenny M Kindblom
- Pediatric Clinical Research Center, Queen Silvia Children's Hospital, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Florian B Lagler
- Institute for Inherited Metabolic Diseases and Department of Pediatrics, Paracelsus Medical University, Clinical Research Center Salzburg, Salzburg, Austria
| | - Carmen Moreno
- Institute of Psychiatry and Mental Health, Child and Adolescent Psychiatry Department, Hospital General Universitario Gregorio Marañón, School of Medicine, Universidad Complutense, IiSGM, CIBERSAM, Madrid, Spain
| | - Paula Pokorna
- Intensive Care and Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, the Netherlands.,Department of Pharmacology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic.,Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic.,Department of Physiology and Pharmacology, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Michiel F Schreuder
- Department of Pediatric Nephrology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Amalia Children's Hospital, Nijmegen, the Netherlands
| | - Joseph F Standing
- UCL Great Ormond Street Institute of Child Health, London, UK.,Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK.,Institute for Infection and Immunity, St George's, University of London, London, UK
| | - Mark A Turner
- Department of Women's and Children's Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool Health Partners, Liverpool, UK
| | - Benedetto Vitiello
- Division of Child and Adolescent Neuropsychiatry, Department of Public Health and Pediatrics, University of Torino, Torino, Italy
| | - Wei Zhao
- Department of Clinical Pharmacy, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, China.,Department of Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China.,Clinical Research Centre, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | | | | | - John van den Anker
- Intensive Care and Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, the Netherlands.,Paediatric Pharmacology and Pharmacometrics, University Children's Hospital Basel (UKBB), University of Basel, Basel, Switzerland.,Division of Clinical Pharmacology, Children's National Hospital, Washington, DC, USA
| | - Karel Allegaert
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium.,Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium.,Department of Hospital Pharmacy, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
13
|
Oral drug delivery strategies for development of poorly water soluble drugs in paediatric patient population. Adv Drug Deliv Rev 2022; 190:114507. [PMID: 36049580 DOI: 10.1016/j.addr.2022.114507] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 06/14/2022] [Accepted: 08/17/2022] [Indexed: 01/24/2023]
Abstract
Selecting the appropriate formulation and solubility-enabling technology for poorly water soluble drugs is an essential element in the development of formulations for paediatric patients. Different methodologies and structured strategies are available to select a suitable approach and guide formulation scientists for development of adult formulations. However, there is paucity of available literature for selection of technology and overcoming the challenges in paediatric formulation development. The need for flexible dosing, and the limited knowledge of the safety of many formulation excipients in paediatric subjects, impose significant constraints and in some instances require adaptation of the approaches taken to formulating these drugs for the adult population. Selection of the best drug delivery system for paediatrics requires an efficient, systematic approach that considers a drug's physical and chemical properties and the targeted patient population's requirements. This review is a step towards development of a strategy for the design of solubility enhancing paediatric formulations of highly insoluble drugs. The aim of this review is to provide an overview of different approaches and strategies to consider in order to assist development of paediatric formulation for poorly water-soluble drugs with the provision of examples of some marketed products. In addition, it provides recommendations to overcome the range of challenges posed by these strategies and adaptations of the adult approach/product presentation required to enable paediatric drug development and administration.
Collapse
|
14
|
Patient-specific in vitro drug release testing coupled with in silico PBPK modeling to forecast the in vivo performance of oral extended-release levodopa formulations in Parkinson's disease patients. Eur J Pharm Biopharm 2022; 180:101-118. [PMID: 36150616 DOI: 10.1016/j.ejpb.2022.09.015] [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: 02/22/2022] [Revised: 08/24/2022] [Accepted: 09/15/2022] [Indexed: 11/24/2022]
Abstract
Biorelevant in vitro release models are valuable analytical tools for oral drug development but often tailored to gastrointestinal conditions in 'average' healthy adults. However, predicting in vivo performance in individual patients whose gastrointestinal conditions do not match those of healthy adults would be of great value for optimizing oral drug therapy for such patients. This study focused on establishing patient-specific in vitro and in silico models to predict the in vivo performance of levodopa extended-release products in Parkinsońs disease patients. Current knowledge on gastrointestinal conditions in these patients was incorporated into model development. Relevant in vivo pharmacokinetic data and patient-specific in vitro release data from a novel in vitro test setup were integrated into patient-specific physiologically-based pharmacokinetic models. AUC, cmax and tmax of the computed plasma profiles were calculated using PK-Sim®. For the products studied, levodopa plasma concentration-time profiles modeled using this novel approach compared far better with published average plasma profiles in Parkinsońs disease patients than those derived from in vitro release data obtained from the 'average' healthy adult setup. Although further work is needed, results of this study highlight the importance of addressing patient-specific gastrointestinal conditions when aiming to predict drug release in such specific patient groups.
Collapse
|
15
|
Zhao Y, Lin S, Fang R, Shi Y, Wu W, Zhang W, Chen H. Mechanism of Enhanced Oral Absorption of a Nano-Drug Delivery System Loaded with Trimethyl Chitosan Derivatives. Int J Nanomedicine 2022; 17:3313-3324. [PMID: 35937081 PMCID: PMC9346306 DOI: 10.2147/ijn.s358832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 07/18/2022] [Indexed: 11/28/2022] Open
Abstract
Introduction In the previous study, nanoparticles coated with trimethyl chitosan (TMC) derivatives (PPTT-NPs) could promote the oral bioavailability of panax notoginseng saponins (PNS). Herein, we chose PPTT-NPs as a model drug to study the property and mechanism of intestinal absorption in vitro and in vivo. Methods The stability of PPTT-NPs was evaluated using simulated gastric fluid and simulated intestinal fluid. The uptake and transport of PPTT-NPs were investigated in Caco-2 and Caco-2&HT29 co-culture cells. The biosafety, intestinal permeability, adhesion, and absorption mechanism of PPTT-NPs were investigated using SD rats in vivo. The live imaging and biodistribution of PPTT-NPs were observed by IVIS. Furthermore, the effects on CYP3A4 of PPTT-NPs were investigated using testosterone as the probe substrate. Results The results of the stability assay showed that PPTT-NPs had a strong tolerance to the pH and digestive enzymes in the gastrointestinal environment. In vitro cell experiments showed that the uptake of drugs exhibited a time-dependent. When the ratio of TMC-VB12 and TMC-Cys was 1:3, the uptake capacity of PPTT-NPs was the highest. PPTT-NPs could enhance the paracellular transport of drugs by reversibly opening a tight junction. Animal experiments demonstrated that PPTT-NPs have good biological safety. PPTT-NPs had good adhesion and permeability to small intestinal mucosa. Meanwhile, PPTT-NPs needed energy and various protein to participate in the uptake of drugs. The live imaging of NPs illustrated that PPTT-NPs could prolong the residence time in the intestine. Moreover, TMC-VB12 and TMC-Cys could reduce the metabolism of drugs by inhibiting CYP3A4 to a certain extent. Conclusion The results show that TMC-VB12 and TMC-Cys are effective in the transport of PPTT-NPs. PPTT-NPs can increase the intestinal adhesion of drugs and exert high permeation by intestinal enterocytes which demonstrate significant and efficient potential for oral delivery of the BCS III drugs.
Collapse
Affiliation(s)
- Ying Zhao
- College of Pharmacy, Guilin Medical University, Guilin, 541199, People’s Republic of China
| | - Shiyuan Lin
- College of Pharmacy, Guilin Medical University, Guilin, 541199, People’s Republic of China
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, People’s Republic of China
| | - Ruiyue Fang
- College of Pharmacy, Guilin Medical University, Guilin, 541199, People’s Republic of China
| | - Yaling Shi
- College of Pharmacy, Guilin Medical University, Guilin, 541199, People’s Republic of China
| | - Wei Wu
- College of Pharmacy, Guilin Medical University, Guilin, 541199, People’s Republic of China
| | - Wei Zhang
- College of Pharmacy, Guilin Medical University, Guilin, 541199, People’s Republic of China
| | - Hui Chen
- College of Pharmacy, Guilin Medical University, Guilin, 541199, People’s Republic of China
- Correspondence: Hui Chen; Wei Zhang, College of Pharmacy, Guilin Medical University, No. 1 Zhiyuan Road, Guilin, 541199, People’s Republic of China, Email ;
| |
Collapse
|
16
|
Hamimed M, Leblond P, Dumont A, Gattacceca F, Tresch-Bruneel E, Probst A, Chastagner P, Pagnier A, De Carli E, Entz-Werlé N, Grill J, Aerts I, Frappaz D, Bertozzi-Salamon AI, Solas C, André N, Ciccolini J. Impact of pharmacogenetics on variability in exposure to oral vinorelbine among pediatric patients: a model-based population pharmacokinetic analysis. Cancer Chemother Pharmacol 2022; 90:29-44. [PMID: 35751658 DOI: 10.1007/s00280-022-04446-y] [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: 01/31/2022] [Accepted: 06/04/2022] [Indexed: 11/02/2022]
Abstract
PURPOSE Better understanding of pharmacokinetics of oral vinorelbine (VNR) in children would help predicting drug exposure and, beyond, clinical outcome. Here, we have characterized the population pharmacokinetics of oral VNR and studied the factors likely to explain the variability observed in VNR exposure among young patients. DESIGN/METHODS We collected blood samples from 36 patients (mean age 11.6 years) of the OVIMA multicentric phase II study in children with recurrent/progressive low-grade glioma. Patients received 60 mg/m2 of oral VNR on days 1, 8, and 15 during the first 28-day treatment cycle and 80 mg/m2, unless contraindicated, from cycle 2-12. Population pharmacokinetic analysis was performed using nonlinear mixed-effects modeling within the Monolix® software. Fifty SNPs of pharmacokinetic-related genes were genotyped. The influence of demographic, biological, and pharmacogenetic covariates on pharmacokinetic parameters was investigated using a stepwise multivariate procedure. RESULTS A three-compartment model, with a delayed double zero-order absorption and a first-order elimination, best described VNR pharmacokinetics in children. Typical population estimates for the apparent central volume of distribution (Vc/F) and elimination rate constant were 803 L and 0.60 h-1, respectively. Following covariate analysis, BSA, leukocytes count, and drug transport ABCB1-rs2032582 SNP showed a dramatic impact on Vc/F. Conversely, age and sex had no significant effect on VNR pharmacokinetics. CONCLUSION Beyond canonical BSA and leukocytes, ABCB1-rs2032582 polymorphism showed a meaningful impact on VNR systemic exposure. Simulations showed that the identified covariates could have an impact on both efficacy and toxicity outcomes. Thus, a personalized dosing strategy, using those covariates, could help to optimize the efficacy/toxicity balance of VNR in children.
Collapse
Affiliation(s)
- Mourad Hamimed
- SMARTc Unit, Cancer Research Center of Marseille, Inserm U1068-CNRS UMR 7258, Aix-Marseille University U105, 27 Boulevard Jean Moulin, 13385, Marseille, France. .,Inria-Inserm COMPO Team, Centre Inria Sophia Antipolis - Méditerranée, Inserm U1068-CNRS UMR 7258, Aix-Marseille University U105, Marseille, France.
| | - Pierre Leblond
- Institute of Pediatric Hematology and Oncology IHOPe, Léon Bérard Cancer Center, Lyon, France.,Department of Pediatric Oncology, Oscar Lambret Cancer Center, Lille, France
| | - Aurélie Dumont
- Unité d'Oncologie Moléculaire Humaine, Oscar Lambret Cancer Center, Lille, France
| | - Florence Gattacceca
- SMARTc Unit, Cancer Research Center of Marseille, Inserm U1068-CNRS UMR 7258, Aix-Marseille University U105, 27 Boulevard Jean Moulin, 13385, Marseille, France.,Inria-Inserm COMPO Team, Centre Inria Sophia Antipolis - Méditerranée, Inserm U1068-CNRS UMR 7258, Aix-Marseille University U105, Marseille, France
| | | | - Alicia Probst
- Département de la Recherche Clinique et Innovation, Oscar Lambret Cancer Center, Lille, France
| | - Pascal Chastagner
- Service d'Hémato-Oncologie Pédiatrique, Nancy University Hospital, Nancy, France
| | - Anne Pagnier
- Service d'Hémato-Oncologie Pédiatrique, Grenoble University Hospital, Grenoble, France
| | - Emilie De Carli
- Service d'Hémato-Oncologie Pédiatrique, Angers University Hospital, Angers, France
| | - Natacha Entz-Werlé
- Pédiatrie Onco-Hématologie Université de Strasbourg, CHRU Hautepierre, UMR CNRS 7021, Strasbourg, France
| | - Jacques Grill
- Département de Cancérologie de l'Enfant et de l'Adolescent et UMR CNRS 8203 Université Paris Saclay, Gustave Roussy, Villejuif, France
| | - Isabelle Aerts
- SIREDO Centre (Care, Innovation and Research in Paediatric, Adolescent and Young Adult Oncology), Institut Curie-Oncology Center, Paris, France
| | - Didier Frappaz
- Institute of Pediatric Hematology and Oncology IHOPe, Léon Bérard Cancer Center, Lyon, France
| | | | - Caroline Solas
- Unité des Virus Émergents (UVE), Aix-Marseille Univ-IRD 190-Inserm 1207, Marseille, France.,Clinical Pharmacokinetics and Toxicology Laboratory, La Timone University Hospital of Marseille, APHM, Marseille, France
| | - Nicolas André
- Department of Pediatric Oncology, La Timone University Hospital of Marseille, APHM, Marseille, France
| | - Joseph Ciccolini
- SMARTc Unit, Cancer Research Center of Marseille, Inserm U1068-CNRS UMR 7258, Aix-Marseille University U105, 27 Boulevard Jean Moulin, 13385, Marseille, France.,Inria-Inserm COMPO Team, Centre Inria Sophia Antipolis - Méditerranée, Inserm U1068-CNRS UMR 7258, Aix-Marseille University U105, Marseille, France.,Clinical Pharmacokinetics and Toxicology Laboratory, La Timone University Hospital of Marseille, APHM, Marseille, France
| |
Collapse
|
17
|
Sengupta S, Bhattacharya P, Nag DS, Sahay N. Search for the ideal route of premedication in children.. far from over? Indian J Anaesth 2022; 66:S188-S192. [PMID: 35874489 PMCID: PMC9298932 DOI: 10.4103/ija.ija_415_22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 05/22/2022] [Accepted: 05/23/2022] [Indexed: 11/04/2022] Open
Affiliation(s)
- Saikat Sengupta
- Senior Consultant, Apollo Multispeciality Hospitals, Kolkata, West Bengal
| | - Prithwis Bhattacharya
- Professor, Department of Anaesthesiology, Pacific Medical College and Hospital, Udaipur, Rajasthan
| | - Deb S. Nag
- Senior Consultant, Anaesthesiology, Tata Main Hospital, Jamshedpur, Jharkhand
| | - Nishant Sahay
- Additional Professor, All India Institute of Medical Sciences, Patna, Bihar, India
| |
Collapse
|
18
|
Wollmer E, Ungell AL, Nicolas JM, Klein S. Review of paediatric gastrointestinal physiology relevant to the absorption of orally administered medicines. Adv Drug Deliv Rev 2022; 181:114084. [PMID: 34929252 DOI: 10.1016/j.addr.2021.114084] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 11/13/2021] [Accepted: 12/13/2021] [Indexed: 12/11/2022]
Abstract
Despite much progress in regulations to improve paediatric drug development, there remains a significant need to develop better medications for children. For the design of oral dosage forms, a detailed understanding of the specific gastrointestinal (GI) conditions in children of different age categories and how they differ from GI conditions in adults is essential. Several review articles have been published addressing the ontogeny of GI characteristics, including luminal conditions in the GI tract of children. However, the data reported in most of these reviews are of limited quality because (1) information was cited from very old publications and sometimes low quality sources, (2) data gaps in the original data were filled with textbook knowledge, (3) data obtained on healthy and sick children were mixed, (4) average data obtained on groups of patients were mixed with data obtained on individual patients, and (5) results obtained using investigative techniques that may have altered the outcome of the respective studies were considered. Consequently, many of these reviews draw conclusions that may be incorrect. The aim of the present review was to provide a comprehensive and updated overview of the available original data on the ontogeny of GI luminal conditions relevant to oral drug absorption in the paediatric population. To this end, the PubMed and Web of Science metadatabases were searched for appropriate studies that examined age-related conditions in the oral cavity, esophagus, stomach, small intestine, and colon. Maturation was observed for several GI parameters, and corresponding data sets were identified for each paediatric age group. However, it also became clear that the ontogeny of several GI traits in the paediatric population is not yet known. The review article provides a robust and valuable data set for the development of paediatric in vitro and in silico biopharmaceutical tools to support the development of age-appropriate dosage forms. In addition, it provides important information on existing data gaps and should provide impetus for further systematic and well-designed in vivo studies on GI physiology in children of specific age groups in order to close existing knowledge gaps and to sustainably improve oral drug therapy in children.
Collapse
|
19
|
Hamimed M, Gattacceca F, André N, Tresch-Bruneel E, Probst A, Chastagner P, Pagnier A, De Carli E, Entz-Werlé N, Grill J, Aerts I, Frappaz D, Bertozzi-Salamon AI, Solas C, Leblond P. Pharmacokinetics of oral vinorelbine in French children with recurrent or progressive primary low-grade glioma. Br J Clin Pharmacol 2021; 88:2096-2117. [PMID: 34709655 DOI: 10.1111/bcp.15131] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 09/29/2021] [Accepted: 10/21/2021] [Indexed: 11/30/2022] Open
Abstract
AIM There is a crucial need for pharmacokinetic (PK) data of oral vinorelbine (VNR) in pediatric population. The aim of this work was to assess the PK profile of orally administered VNR in children with recurrent/progressive primary low-grade glioma (LGG). METHODS A multicentric, open-label, single-arm intervention phase II study was conducted. Patients, aged between 6 and 18 years, with histologically confirmed recurrent or progressive primary LGG or non-documented typical optic pathway tumors, were included. PK parameters were estimated by non-compartmental analysis using Phoenix WinNonlin® software (version 8.0, Certara, Inc.). The Influence of demographic and biological covariates on VNR PK parameters was investigated using a multivariate linear regression analysis. RESULTS PK analysis included 36 patients with a median age (range) of 11 (6-17) years. Estimates of apparent oral clearance (CL/F), apparent volume of distribution (V/F), half-life (t1/2 ) and their between-subject variability (CV%) at 60 mg.m-2 dose level, were 472 L.h-1 (51.8%), 7002 L (57.9%) and 10 h (21.0%), respectively. Negligible accumulation of VNR between C1 and C2 was observed. CL/F and V/F were found to increase with body surface area (BSA) (p = 0.004). Lower area under the concentration-time curve (AUC) levels were observed among children in comparison to adults. CONCLUSION Higher doses may be necessary for children with LGG. BSA showed a significant impact on VNR systemic exposure. We believe that our findings will serve as a basis for further studies to better characterize the concentration-response relationships of VNR among pediatric patients.
Collapse
Affiliation(s)
- Mourad Hamimed
- SMARTc Unit, Cancer Research Center of Marseille, Inserm U1068 - CNRS UMR 7258 - Aix-Marseille University U105, Marseille, France.,Inria - Inserm COMPO team, Centre Inria Sophia Antipolis - Méditerranée, Inserm U1068 - CNRS UMR 7258 - Aix-Marseille University U105, Marseille, France
| | - Florence Gattacceca
- SMARTc Unit, Cancer Research Center of Marseille, Inserm U1068 - CNRS UMR 7258 - Aix-Marseille University U105, Marseille, France.,Inria - Inserm COMPO team, Centre Inria Sophia Antipolis - Méditerranée, Inserm U1068 - CNRS UMR 7258 - Aix-Marseille University U105, Marseille, France
| | - Nicolas André
- SMARTc Unit, Cancer Research Center of Marseille, Inserm U1068 - CNRS UMR 7258 - Aix-Marseille University U105, Marseille, France.,Department of Pediatric Oncology, La Timone University Hospital of Marseille, APHM, Marseille, France
| | | | - Alicia Probst
- Département de la Recherche Clinique et Innovation,Oscar Lambret Cancer Center, Lille, France
| | - Pascal Chastagner
- Service d'hémato-oncologie pédiatrique, Nancy University Hospital, Nancy, France
| | - Anne Pagnier
- Service d'hémato-oncologie pédiatrique, Grenoble University Hospital, Grenoble, France
| | - Emilie De Carli
- Service d'hémato-oncologie pédiatrique, Angers University Hospital, Angers, France
| | - Natacha Entz-Werlé
- Pédiatrie Onco-Hématologie Université de Strasbourg, CHRU Hautepierre- - UMR CNRS 7021, Strasbourg, France
| | - Jacques Grill
- Département de Cancérologie de l'Enfant et de l'Adolescent et UMR CNRS 8203 Université Paris Saclay, Gustave Roussy, Villejuif, France
| | - Isabelle Aerts
- SIREDO Centre (Care, innovation and research in paediatric, adolescent and young adult oncology), Institut Curie- Oncology Center, Paris, France
| | - Didier Frappaz
- Institute of Pediatric Hematology and Oncology IHOPe, Léon Bérard Cancer Center, Lyon, France
| | | | - Caroline Solas
- Unité des Virus Émergents (UVE), Aix-Marseille Univ-IRD 190-Inserm 1207, Marseille, France.,Clinical Pharmacokinetics and Toxicology Laboratory, La Timone University Hospital of Marseille, APHM, Marseille, France
| | - Pierre Leblond
- Institute of Pediatric Hematology and Oncology IHOPe, Léon Bérard Cancer Center, Lyon, France.,Department of Pediatric Oncology, Oscar Lambret Cancer Center, Lille, France
| |
Collapse
|
20
|
Quantification of Fluid Volume and Distribution in the Paediatric Colon via Magnetic Resonance Imaging. Pharmaceutics 2021; 13:pharmaceutics13101729. [PMID: 34684022 PMCID: PMC8540766 DOI: 10.3390/pharmaceutics13101729] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/08/2021] [Accepted: 10/14/2021] [Indexed: 12/16/2022] Open
Abstract
Previous studies have used magnetic resonance imaging (MRI) to quantify the fluid in the stomach and small intestine of children, and the stomach, small intestine and colon of adults. This is the first study to quantify fluid volumes and distribution using MRI in the paediatric colon. MRI datasets from 28 fasted (aged 0-15 years) and 18 fluid-fed (aged 10-16 years) paediatric participants were acquired during routine clinical care. A series of 2D- and 3D-based software protocols were used to measure colonic fluid volume and localisation. The paediatric colon contained a mean volume of 22.5 mL ± 41.3 mL fluid, (range 0-167.5 mL, median volume 0.80 mL) in 15.5 ± 17.5 discreet fluid pockets (median 12). The proportion of the fluid pockets larger than 1 mL was 9.6%, which contributed to 94.5% of the total fluid volume observed. No correlation was detected between all-ages and colonic fluid volume, nor was a difference in colonic fluid volumes observed based on sex, fed state or age group based on ICH-classifications. This study quantified fluid volumes within the paediatric colon, and these data will aid and accelerate the development of biorelevant tools to progress paediatric drug development for colon-targeting formulations.
Collapse
|
21
|
Dyavar SR, Kumar S, Gautam N, Podany AT, Winchester LC, Weinhold JA, Mykris TM, Nallasamy P, Alnouti Y, Fletcher CV. Intramuscular and subcutaneous administration of antiretroviral drugs, compared with oral, enhances delivery to lymphoid tissues in BALB/c mice. J Antimicrob Chemother 2021; 76:2651-2658. [PMID: 34312680 DOI: 10.1093/jac/dkab228] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 06/01/2021] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Multiple tissue reservoirs are established soon after HIV infection, and some tissues may also be pharmacological sanctuaries. Parenteral administration of antiretroviral (ARV) drugs for treatment and prevention of HIV infection is an active area of drug development. The influence of route of administration on ARV tissue pharmacokinetics is not known. OBJECTIVES To investigate ARV pharmacokinetics in lymphatic and select non-lymphatic tissues (e.g. brain and testes) after intramuscular and subcutaneous administration compared with oral in BALB/c mice. METHODS Tissue concentrations of cobicistat, efavirenz, elvitegravir, maraviroc, rilpivirine, tenofovir alafenamide and tenofovir disoproxil fumarate were determined. The tissue penetration ratio (TPR) was the primary measure for comparison; a change in TPR arises from factors affecting tissue distribution controlling for changes in systemic bioavailability. RESULTS Intramuscular and subcutaneous delivery increased TPRs in the lymph node and spleen for 27 of 28 (96%) drug administration events. Decreased TPRs, however, were found in some tissues such as the brain and testes. CONCLUSIONS These results demonstrate a change in route of drug administration from oral to intramuscular or subcutaneous can change tissue uptake. This has implications for HIV pharmacotherapy. For example, HIV persists in lymphoid tissues despite long-term oral ARV therapy, and low ARV concentrations have been found in lymphoid tissues. The improved ARV lymphatic tissue bioavailability with intramuscular and subcutaneous administration allows future studies to investigate these routes of drug administration as a therapeutic manoeuvre to limit viral persistence and eliminate viral sanctuaries in the lymphatic tissues, which is a prerequisite for eradication of HIV.
Collapse
Affiliation(s)
- Shetty Ravi Dyavar
- Antiviral Pharmacology Laboratory, Center for Drug Discovery, University of Nebraska Medical Center (UNMC), Omaha, NE, USA
| | - Sushil Kumar
- Department of Pharmaceutical Sciences, College of Pharmacy, UNMC, Omaha, NE, USA
| | - Nagsen Gautam
- Department of Pharmaceutical Sciences, College of Pharmacy, UNMC, Omaha, NE, USA
| | - Anthony T Podany
- Antiviral Pharmacology Laboratory, Center for Drug Discovery, University of Nebraska Medical Center (UNMC), Omaha, NE, USA
| | - Lee C Winchester
- Antiviral Pharmacology Laboratory, Center for Drug Discovery, University of Nebraska Medical Center (UNMC), Omaha, NE, USA
| | - Jonathan A Weinhold
- Antiviral Pharmacology Laboratory, Center for Drug Discovery, University of Nebraska Medical Center (UNMC), Omaha, NE, USA
| | - Timothy M Mykris
- Antiviral Pharmacology Laboratory, Center for Drug Discovery, University of Nebraska Medical Center (UNMC), Omaha, NE, USA
| | - Palanisamy Nallasamy
- Department of Biochemistry and Molecular Biology, College of Medicine, UNMC, Omaha, NE, 68198, USA
| | - Yazen Alnouti
- Department of Pharmaceutical Sciences, College of Pharmacy, UNMC, Omaha, NE, USA
| | - Courtney V Fletcher
- Antiviral Pharmacology Laboratory, Center for Drug Discovery, University of Nebraska Medical Center (UNMC), Omaha, NE, USA
| |
Collapse
|
22
|
Walsh J, Schaufelberger D, Iurian S, Klein S, Batchelor H, Turner R, Gizurarson S, Boltri L, Alessandrini E, Tuleu C. Path towards efficient paediatric formulation development based on partnering with clinical pharmacologists and clinicians, a conect4children expert group white paper. Br J Clin Pharmacol 2021; 88:5034-5051. [PMID: 34265091 DOI: 10.1111/bcp.14989] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 06/10/2021] [Accepted: 07/07/2021] [Indexed: 12/18/2022] Open
Abstract
Improved global access to novel age-appropriate formulations for paediatric subsets, either of new chemical entities or existing drugs, is a priority to ensure that medicines meet the needs of these patients. However, despite regulatory incentives, the introduction to the market of paediatric formulations still lags behind adult products. This is mainly caused by additional complexities associated with the development of acceptable age-appropriate paediatric medicines. This position paper recommends the use of a paediatric Quality Target Product Profile as an efficient tool to facilitate early planning and decision making across all teams involved in paediatric formulation development during the children-centric formulation design for new chemical entities, or to repurpose/reformulate off-patent drugs. Essential key attributes of a paediatric formulation are suggested and described. Moreover, greater collaboration between formulation experts and clinical colleagues, including healthcare professionals, is advocated to lead to safe and effective, age-appropriate medicinal products. Acceptability testing should be a secondary endpoint in paediatric clinical trials to ensure postmarketing adherence is not compromised by a lack of acceptability. Not knowing the indications and the related age groups and potential dosing regimens early enough is still a major hurdle for efficient paediatric formulation development; however, the proposed paediatric Quality Target Product Profile could be a valuable collaborative tool for planning and decision making to expedite paediatric product development, particularly for those with limited experience in developing a paediatric product.
Collapse
Affiliation(s)
- Jennifer Walsh
- BioCity Nottingham, Jenny Walsh Consulting Ltd, Nottingham, UK
| | - Daniel Schaufelberger
- School of Medicine, All Children's Hospital, Schaufelberger Consulting LLC and Johns Hopkins University, St. Petersburg, FL, USA
| | - Sonia Iurian
- Department of Pharmaceutical Technology and Biopharmacy, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Sandra Klein
- Department of Pharmacy, Institute of Biopharmaceutics and Pharmaceutical Technology, University of Greifswald, Greifswald, Germany
| | - Hannah Batchelor
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Roy Turner
- Idorsia Pharmaceuticals Ltd, Allschwil, Switzerland
| | - Sveinbjörn Gizurarson
- Faculty of Pharmaceutical Sciences, University of Iceland, Reykjavik, Iceland; and Pharmacy Department, Kamuzu University of Health Sciences, Blantyre, Malawi
| | - Luigi Boltri
- Pharmaceutical Development, Adare Pharmaceuticals, Milan, Italy
| | | | | |
Collapse
|
23
|
Sharma S, Prasad B. Meta-Analysis of Food Effect on Oral Absorption of Efflux Transporter Substrate Drugs: Does Delayed Gastric Emptying Influence Drug Transport Kinetics? Pharmaceutics 2021; 13:1035. [PMID: 34371727 PMCID: PMC8309017 DOI: 10.3390/pharmaceutics13071035] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/02/2021] [Accepted: 07/04/2021] [Indexed: 01/07/2023] Open
Abstract
The oral route of drug administration is the most convenient method of drug delivery, but it is associated with variable bioavailability. Food is one of the major factors that affect oral drug absorption by influencing drug properties (e.g., solubility and dissolution rate) and physiological factors (e.g., metabolism and transport across the gastrointestinal tract). The aim of this work was to investigate the effect of food on the high-affinity intestinal efflux transporter substrate drugs. We hypothesized that transport efficiency is higher in the fed state as compared to the fasted state because of the lower intestinal lumen drug concentration due to prolonged gastric emptying time. A systematic analysis of reported clinical food-effect (FE) studies on 311 drugs was performed and the association of the efflux transport efficiency was investigated on the FE magnitude, i.e., changes in maximal plasma concentration and area under the plasma concentration-time profile curve for both solubility and permeability-limited drugs. In total, 124 and 88 drugs showed positive and negative FE, respectively, whereas 99 showed no FE. As expected, the solubility-limited drugs showed positive FE, but interestingly, drugs with a high potential for efflux transport, were associated with negative FE. Moreover, a high-fat diet was associated with a higher magnitude of negative FE for high-affinity efflux transporter substrates as compared to a low-fat diet. To account for changes in drug absorption after food intake, the prolonged gastric emptying time should be considered in the physiologically based pharmacokinetic (PBPK) modeling of orally absorbed efflux transporter substrate drugs.
Collapse
Affiliation(s)
- Sheena Sharma
- Department of Pharmaceutical Sciences, Washington State University, 412 E Spokane Falls Blvd, Spokane, WA 99202, USA
| | - Bhagwat Prasad
- Department of Pharmaceutical Sciences, Washington State University, 412 E Spokane Falls Blvd, Spokane, WA 99202, USA
| |
Collapse
|
24
|
van den Anker J, Allegaert K. Considerations for Drug Dosing in Premature Infants. J Clin Pharmacol 2021; 61 Suppl 1:S141-S151. [PMID: 34185893 DOI: 10.1002/jcph.1884] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 04/21/2021] [Indexed: 12/13/2022]
Abstract
In premature infants, effective and safe drug therapy depends on optimal dose selection and requires a thorough understanding of the underlying disease(s) of these fragile infants as well as the pharmacokinetics and pharmacodynamics of the drugs selected to treat their diseases. Differences in gestational and postnatal age or weight are the major determinants of the observed variability in drug disposition and effect in these infants. This article presents an outline on how to translate the results of a population pharmacokinetic/pharmacodynamic study into rational dosing regimens, and how physiologically based pharmacokinetic modeling, electronic health records, and the abundantly available data of vital functions of premature infants during their stay in the neonatal intensive care unit for evaluation of their pharmacotherapy can be used to tailor the most safe and effective dose in these infants.
Collapse
Affiliation(s)
- John van den Anker
- Division of Clinical Pharmacology, Children's National Hospital, Washington, DC, USA.,Division of Paediatric Pharmacology and Pharmacometrics, University Children's Hospital Basel, University of Basel, Basel, Switzerland.,Intensive Care and Department of Pediatric Surgery, Erasmus Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Karel Allegaert
- Department of Hospital Pharmacy, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands.,Department of Development and Regeneration, KU Leuven, Leuven, Belgium.,Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| |
Collapse
|
25
|
Liu XI, van den Anker JN, Burckart GJ, Dallmann A. Evaluation of Physiologically Based Pharmacokinetic Models to Predict the Absorption of BCS Class I Drugs in Different Pediatric Age Groups. J Clin Pharmacol 2021; 61 Suppl 1:S94-S107. [PMID: 34185902 DOI: 10.1002/jcph.1845] [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: 01/03/2021] [Accepted: 02/17/2021] [Indexed: 11/06/2022]
Abstract
Age-related changes in many parameters affecting drug absorption remain poorly characterized. The objective of this study was to apply physiologically based pharmacokinetic (PBPK) models in pediatric patients to investigate the absorption and pharmacokinetics of 4 drugs belonging to the Biopharmaceutics Classification System (BCS) class I administered as oral liquid formulations. Pediatric PBPK models built with PK-Sim/MoBi were used to predict the pharmacokinetics of acetaminophen, emtricitabine, theophylline, and zolpidem in different pediatric populations. The model performance for predicting drug absorption and pharmacokinetics was assessed by comparing the predicted absorption profile with the deconvoluted dose fraction absorbed over time and predicted with observed plasma concentration-time profiles. Sensitivity analyses were performed to analyze the effects of changes in relevant input parameters on the model output. Overall, most pharmacokinetic parameters were predicted within a 2-fold error range. The absorption profiles were generally reasonably predicted, but relatively large differences were observed for acetaminophen. Sensitivity analyses showed that the predicted absorption profile was most sensitive to changes in the gastric emptying time (GET) and the specific intestinal permeability. The drug's solubility played only a minor role. These findings confirm that gastric emptying time, more than intestinal permeability or solubility, is a key factor affecting BCS class I drug absorption in children. As gastric emptying time is prolonged in the fed state, a better understanding of the interplay between food intake and gastric emptying time in children is needed, especially in the very young in whom the (semi)fed condition is the prevailing prandial state, and hence prolonged gastric emptying time seems more plausible than the fasting state.
Collapse
Affiliation(s)
- Xiaomei I Liu
- Division of Clinical Pharmacology, Children's National Hospital, Washington, District of Columbia, USA
| | - John N van den Anker
- Division of Clinical Pharmacology, Children's National Hospital, Washington, District of Columbia, USA.,Division of Pediatric Pharmacology and Pharmacometrics, University of Basel Children's Hospital, Basel, Switzerland
| | - Gilbert J Burckart
- Office of Clinical Pharmacology, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - André Dallmann
- Pharmacometrics/Modeling & Simulation, Research & Development, Pharmaceuticals, Bayer AG, Leverkusen, Germany
| |
Collapse
|
26
|
Bi Y, Liu J, Li F, Yu J, Bhattaram A, Bewernitz M, Li RJ, Ahn J, Earp J, Ma L, Zhuang L, Yang Y, Zhang X, Zhu H, Wang Y. Model-Informed Drug Development in Pediatric Dose Selection. J Clin Pharmacol 2021; 61 Suppl 1:S60-S69. [PMID: 34185906 DOI: 10.1002/jcph.1848] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 02/18/2021] [Indexed: 01/12/2023]
Abstract
Model-informed drug development (MIDD) has been a powerful and efficient tool applied widely in pediatric drug development due to its ability to integrate and leverage existing knowledge from different sources to narrow knowledge gaps. The dose selection is the most common MIDD application in regulatory submission related to pediatric drug development. This article aims to give an overview of the 3 broad categories of use of MIDD in pediatric dose selection: leveraging from adults to pediatric patients, leveraging from animals to pediatric patients, and integrating mechanism in infants and neonates. Population pharmacokinetic analyses with allometric scaling can reasonably predict the clearance in pediatric patients aged >5 years. A mechanistic-based approach, such as physiologically based pharmacokinetic accounting for ontogeny, or an allometric model with age-dependent exponent, can be applied to select the dose in pediatric patients aged ≤2 years. The exposure-response relationship from adults or from other drugs in the same class may be useful in aiding the pediatric dose selection and benefit-risk assessment. Increasing application and understanding of use of MIDD have contributed greatly to several policy developments in the pediatric field. With the increasing efforts of MIDD under the Prescription Drug User Fee Act VI, bigger impacts of MIDD approaches in pediatric dose selection can be expected. Due to the complexity of model-based analyses, early engagement between drug developers and regulatory agencies to discuss MIDD issues is highly encouraged, as it is expected to increase the efficiency and reduce the uncertainty.
Collapse
Affiliation(s)
- Youwei Bi
- Office of Clinical Pharmacology, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Jiang Liu
- Office of Clinical Pharmacology, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Fang Li
- Office of Clinical Pharmacology, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Jingyu Yu
- Office of Clinical Pharmacology, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Atul Bhattaram
- Office of Clinical Pharmacology, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Michael Bewernitz
- Office of Clinical Pharmacology, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Ruo-Jing Li
- Office of Clinical Pharmacology, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Jihye Ahn
- Office of Clinical Pharmacology, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Justin Earp
- Office of Clinical Pharmacology, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Lian Ma
- Office of Clinical Pharmacology, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Luning Zhuang
- Office of Clinical Pharmacology, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Yuching Yang
- Office of Clinical Pharmacology, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Xinyuan Zhang
- Office of Clinical Pharmacology, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Hao Zhu
- Office of Clinical Pharmacology, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Yaning Wang
- Office of Clinical Pharmacology, Food and Drug Administration, Silver Spring, Maryland, USA
| |
Collapse
|
27
|
Palmeirim MS, Specht S, Scandale I, Gander-Meisterernst I, Chabicovsky M, Keiser J. Preclinical and Clinical Characteristics of the Trichuricidal Drug Oxantel Pamoate and Clinical Development Plans: A Review. Drugs 2021; 81:907-921. [PMID: 33929716 PMCID: PMC8144136 DOI: 10.1007/s40265-021-01505-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/17/2021] [Indexed: 12/31/2022]
Abstract
Soil-transmitted helminths (Ascaris lumbricoides, hookworm and Trichuris trichiura) infect about one-fifth of the world's population. The currently available drugs are all highly efficacious against A. lumbricoides. However, they are only moderately efficacious against hookworm and poorly efficacious against T. trichiura. Oxantel, a tetrahydropyrimidine derivative discovered in the 1970s, has recently been brought back to our attention given its high efficacy against T. trichiura infections (estimated 76% cure rate and 85% egg reduction rate at a 20 mg/kg dose). This review summarizes the current knowledge on oxantel pamoate and its use against T. trichiura infections in humans. Oxantel pamoate acts locally in the human gastrointestinal tract and binds to the parasite's nicotinic acetylcholine receptor (nAChR), leading to a spastic paralysis of the worm and subsequent expulsion. The drug is metabolically stable, shows low permeability and low systemic bioavailability after oral use. Oxantel pamoate was found to be safe in humans, with only a few mild adverse events reported. Several clinical trials have investigated the efficacy of this drug against T. trichiura and suggest that oxantel pamoate is more efficacious against T. trichiura than the currently recommended drugs, which makes it a strong asset to the depleted drug armamentarium and could help delay or even prevent the development of resistance to existing drugs. We highlight existing data to support the use of oxantel pamoate against T. trichiura infections.
Collapse
Affiliation(s)
- Marta S Palmeirim
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, P.O. Box, 4002, Basel, Switzerland
- University of Basel, P.O. Box, 4003, Basel, Switzerland
| | - Sabine Specht
- Drugs for Neglected Diseases Initiative, 15 Chemin Louis-Dunant, 1202, Geneva, Switzerland
| | - Ivan Scandale
- Drugs for Neglected Diseases Initiative, 15 Chemin Louis-Dunant, 1202, Geneva, Switzerland
| | | | - Monika Chabicovsky
- MC Toxicology Consulting GmbH, Siebensterngasse 31/8, 1070, Vienna, Austria
| | - Jennifer Keiser
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, P.O. Box, 4002, Basel, Switzerland.
- University of Basel, P.O. Box, 4003, Basel, Switzerland.
| |
Collapse
|
28
|
Gleeson JP, Fein KC, Whitehead KA. Oral delivery of peptide therapeutics in infants: Challenges and opportunities. Adv Drug Deliv Rev 2021; 173:112-124. [PMID: 33774115 PMCID: PMC8178217 DOI: 10.1016/j.addr.2021.03.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 03/09/2021] [Accepted: 03/18/2021] [Indexed: 12/14/2022]
Abstract
The vast majority of drugs are not designed or developed for pediatric and infant populations. Peptide drugs, which have become increasingly relevant in the past several decades, are no exception. Unfortunately, nearly all of the 60+ approved peptide drugs are formulated for injection, a particularly unfriendly mode of administration for infants. Although three peptide drugs were recently approved for oral formulations, this major advance in peptide drug delivery is available only for adults. In this review, we consider the current challenges and opportunities for the oral formulation of peptide therapeutics, specifically for infant populations. We describe the strategies that enable oral protein delivery and the potential impact of infant physiology on those strategies. We also detail the limited but encouraging progress towards 1) adapting conventional drug development and delivery approaches to infants and 2) designing novel infant-centric formulations. Together, these efforts underscore the feasibility of oral peptide delivery in infants and provide motivation to increase attention paid to this underserved area of drug delivery and formulation.
Collapse
Affiliation(s)
- John P Gleeson
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, United States
| | - Katherine C Fein
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, United States
| | - Kathryn A Whitehead
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, United States; Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, United States.
| |
Collapse
|
29
|
Ara-Montojo MF, Bustamante J, Sainz T, Pérez S, Jiménez-Moreno B, Ruiz-Carrascoso G, Rodríguez-Molino P, Villota J, García-López-Hortenano M, Mellado-Peña MJ. Intestinal giardiasis in children: Five years' experience in a reference unit. Travel Med Infect Dis 2021; 42:102082. [PMID: 34020030 DOI: 10.1016/j.tmaid.2021.102082] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 04/13/2021] [Accepted: 04/23/2021] [Indexed: 11/18/2022]
Abstract
BACKGROUND Giardiasis is highly prevalent in children and is often mildly symptomatic. First-line treatment is metronidazole, but treatment failure is not uncommon. We describe a paediatric series, to identify risk factors for treatment failure and to analyse the safety and effectiveness of other treatment strategies. METHODS Retrospective observational study, including children diagnosed with giardiasis from 2014 to 2019. Diagnosis was based on direct visualisation by microscopy after concentration using an alcohol-based fixative, antigen detection and/or DNA detection by polymerase chain reaction in stool. Treatment failure was considered when GI was detected 4 weeks after treatment. RESULTS A total of 120 patients were included, 71.6% internationally adopted, median age 4.2 (2.3-7.3) years. Only 50% presented with symptoms, mainly diarrhoea (35%) and abdominal pain (14.1%); co-parasitism was frequent (45%). First-line treatment failure after a standard dose of metronidazole was 20%, lowering to 8.3% when a higher dose was administered (p < 0.001). Quinacrine was administered in 10 patients, with 100% effectiveness. Children <2 years were at higher risk of treatment failure (OR 3.49; 95% CI 1.06-11.53; p = 0.040). CONCLUSIONS In children with giardiasis, treatment failure is frequent, especially before 2 years of age. Quinacrine can be considered as a second-line treatment. After treatment, eradication should be confirmed.
Collapse
Affiliation(s)
- M F Ara-Montojo
- Paediatric Tropical and Infectious Diseases, Department of Paediatrics, La Paz University Hospital, Madrid, Spain
| | - J Bustamante
- Paediatric Tropical and Infectious Diseases, Department of Paediatrics, La Paz University Hospital, Madrid, Spain; La Paz Research Institute (IdiPAZ), Madrid, Spain
| | - T Sainz
- Paediatric Tropical and Infectious Diseases, Department of Paediatrics, La Paz University Hospital, Madrid, Spain; La Paz Research Institute (IdiPAZ), Madrid, Spain; Translational Research Network in Paediatric Infectious Diseases (RITIP, Red de Investigación Translacional en Infectología Pediátrica), Spain.
| | - S Pérez
- Paediatric Tropical and Infectious Diseases, Department of Paediatrics, La Paz University Hospital, Madrid, Spain
| | | | - G Ruiz-Carrascoso
- Department of Microbiology, La Paz University Hospital, Madrid, Spain
| | - P Rodríguez-Molino
- Paediatric Tropical and Infectious Diseases, Department of Paediatrics, La Paz University Hospital, Madrid, Spain
| | - J Villota
- Paediatric Tropical and Infectious Diseases, Department of Paediatrics, La Paz University Hospital, Madrid, Spain; La Paz Research Institute (IdiPAZ), Madrid, Spain
| | - M García-López-Hortenano
- Paediatric Tropical and Infectious Diseases, Department of Paediatrics, La Paz University Hospital, Madrid, Spain; La Paz Research Institute (IdiPAZ), Madrid, Spain; Translational Research Network in Paediatric Infectious Diseases (RITIP, Red de Investigación Translacional en Infectología Pediátrica), Spain
| | - M J Mellado-Peña
- Paediatric Tropical and Infectious Diseases, Department of Paediatrics, La Paz University Hospital, Madrid, Spain; La Paz Research Institute (IdiPAZ), Madrid, Spain; Translational Research Network in Paediatric Infectious Diseases (RITIP, Red de Investigación Translacional en Infectología Pediátrica), Spain
| |
Collapse
|
30
|
Impact of gastrointestinal tract variability on oral drug absorption and pharmacokinetics: An UNGAP review. Eur J Pharm Sci 2021; 162:105812. [PMID: 33753215 DOI: 10.1016/j.ejps.2021.105812] [Citation(s) in RCA: 140] [Impact Index Per Article: 46.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 02/19/2021] [Accepted: 03/16/2021] [Indexed: 12/17/2022]
Abstract
The absorption of oral drugs is frequently plagued by significant variability with potentially serious therapeutic consequences. The source of variability can be traced back to interindividual variability in physiology, differences in special populations (age- and disease-dependent), drug and formulation properties, or food-drug interactions. Clinical evidence for the impact of some of these factors on drug pharmacokinetic variability is mounting: e.g. gastric pH and emptying time, small intestinal fluid properties, differences in pediatrics and the elderly, and surgical changes in gastrointestinal anatomy. However, the link of colonic factors variability (transit time, fluid composition, microbiome), sex differences (male vs. female) and gut-related diseases (chronic constipation, anorexia and cachexia) to drug absorption variability has not been firmly established yet. At the same time, a way to decrease oral drug pharmacokinetic variability is provided by the pharmaceutical industry: clinical evidence suggests that formulation approaches employed during drug development can decrease the variability in oral exposure. This review outlines the main drivers of oral drug exposure variability and potential approaches to overcome them, while highlighting existing knowledge gaps and guiding future studies in this area.
Collapse
|
31
|
Chapron BD, Chapron A, Leeder JS. Recent advances in the ontogeny of drug disposition. Br J Clin Pharmacol 2021; 88:4267-4284. [PMID: 33733546 DOI: 10.1111/bcp.14821] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 02/12/2021] [Accepted: 02/22/2021] [Indexed: 12/11/2022] Open
Abstract
Developmental changes that occur throughout childhood have long been known to impact drug disposition. However, pharmacokinetic studies in the paediatric population have historically been limited due to ethical concerns arising from incorporating children into clinical trials. As such, much of the early work in the field of developmental pharmacology was reliant on difficult-to-interpret in vitro and in vivo animal studies. Over the last 2 decades, our understanding of the mechanistic processes underlying age-related changes in drug disposition has advanced considerably. Progress has largely been driven by technological advances in mass spectrometry-based methods for quantifying proteins implicated in drug disposition, and in silico tools that leverage these data to predict age-related changes in pharmacokinetics. This review summarizes our current understanding of the impact of childhood development on drug disposition, particularly focusing on research of the past 20 years, but also highlighting select examples of earlier foundational research. Equally important to the studies reviewed herein are the areas that we cannot currently describe due to the lack of research evidence; these gaps provide a map of drug disposition pathways for which developmental trends still need to be characterized.
Collapse
Affiliation(s)
- Brian D Chapron
- Division of Clinical Pharmacology, Toxicology and Therapeutic Innovation, Department of Pediatrics, Children's Mercy Hospital, Kansas City, MO, USA
| | - Alenka Chapron
- Division of Clinical Pharmacology, Toxicology and Therapeutic Innovation, Department of Pediatrics, Children's Mercy Hospital, Kansas City, MO, USA
| | - J Steven Leeder
- Division of Clinical Pharmacology, Toxicology and Therapeutic Innovation, Department of Pediatrics, Children's Mercy Hospital, Kansas City, MO, USA.,Schools of Medicine and Pharmacy, University of Missouri-Kansas City, MO, USA
| |
Collapse
|
32
|
Kacirova I, Grundmann M, Brozmanova H. Umbilical Cord, Maternal Milk, and Breastfed Infant Levetiracetam Concentrations Monitoring at Delivery and during Early Postpartum Period. Pharmaceutics 2021; 13:398. [PMID: 33802733 PMCID: PMC8002441 DOI: 10.3390/pharmaceutics13030398] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 03/12/2021] [Accepted: 03/14/2021] [Indexed: 11/22/2022] Open
Abstract
(1) To obtain objective information about levetiracetam transplacental passage and its transport into colostrum, mature milk, and breastfed infants, we analyzed data from women treated for epilepsy between October 2006 and January 2021; (2) in this cohort study, maternal, umbilical cord, milk, and infant serum concentrations were measured at delivery, 2-4 days postpartum (colostrum) and 7-31 days postpartum (mature milk). Paired umbilical cord serum, maternal serum, breastfed infant serum, and milk concentrations were used to assess the ratios of umbilical cord/maternal serum, milk/maternal serum, and infant/maternal serum concentrations. The influence of combined treatment with enzyme-inducing antiseizure medication carbamazepine was assessed; (3) the umbilical cord/maternal serum concentration ratio ranged between 0.75 and 1.78 (mean 1.10 ± 0.33), paired maternal and umbilical cord serum concentrations were not significantly different, and a highly significant correlation was found between both concentrations. The mean milk/maternal serum concentration ratio was 1.14 ± 0.27 (2-4 days postpartum) and 1.04 ± 0.24 (7-31 days postpartum) while the mean infant/maternal serum concentration ratio was markedly lower (0.19 ± 0.13 and 0.14 ± 0.05, respectively); (4) levetiracetam was found in the umbilical cord at a concentration similar to those in maternal serum. All of the breastfed infant serum concentrations were below the reference range used for the general epileptic population.
Collapse
Affiliation(s)
- Ivana Kacirova
- Department of Clinical Pharmacology, Faculty of Medicine, University of Ostrava, 70300 Ostrava, Czech Republic; (I.K.); (H.B.)
- Department of Clinical Pharmacology, Department of Laboratory Medicine, University Hospital Ostrava, 70852 Ostrava, Czech Republic
| | - Milan Grundmann
- Department of Clinical Pharmacology, Faculty of Medicine, University of Ostrava, 70300 Ostrava, Czech Republic; (I.K.); (H.B.)
| | - Hana Brozmanova
- Department of Clinical Pharmacology, Faculty of Medicine, University of Ostrava, 70300 Ostrava, Czech Republic; (I.K.); (H.B.)
- Department of Clinical Pharmacology, Department of Laboratory Medicine, University Hospital Ostrava, 70852 Ostrava, Czech Republic
| |
Collapse
|
33
|
Nabbout R, Chemaly N, Chiron C, Kuchenbuch M. Safety considerations selecting antiseizure medications for the treatment of individuals with Dravet syndrome. Expert Opin Drug Saf 2021; 20:561-576. [PMID: 33645379 DOI: 10.1080/14740338.2021.1890025] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Introduction: Management of individuals with Dravet Syndrome has evolved significantly over the past 10 years. Progress has been made in understanding the pathophysiology, the long-term outcome and possible consequences of inappropriate therapies, new drugs have been approved by the regulatory authorities and patients and families expressed their needs beyond seizures' control.Areas covered: The authors aimed at providing an overview of the main antiseizure medications used in Dravet syndrome with a particular focus on safety considerations. As the highly active phase of seizures takes place before the age of 5 years, the characteristics of antiseizure medications in infancy and childhood have also been considered due to their impact on antiseizure medication safety.Expert opinion: Recent treatments, evaluated via randomized clinical trials, are promising in terms of efficacy and safety in individuals with DS. However, the balance between expected benefits and risks taken must be accurately assessed on an individual basis. There is a lack of data to understand the needs of patients and families, a major point particularly in this population, where the evaluation of efficacy and safety beyond seizures is difficult due to cognitive delay and behavioral disorders and where this evaluation is coming almost exclusively from caregivers.
Collapse
Affiliation(s)
- Rima Nabbout
- Reference Centre for Rare Epilepsies, Department of Pediatric Neurology, Necker Enfants Malades University Hospital, Université De Paris, Paris, France.,Institut National De La Santé Et De La Recherche Médicale (INSERM), UMR 1163, Institut Imagine, Université De Paris, Paris, France
| | - N Chemaly
- Reference Centre for Rare Epilepsies, Department of Pediatric Neurology, Necker Enfants Malades University Hospital, Université De Paris, Paris, France.,Institut National De La Santé Et De La Recherche Médicale (INSERM), UMR 1163, Institut Imagine, Université De Paris, Paris, France
| | - C Chiron
- Reference Centre for Rare Epilepsies, Department of Pediatric Neurology, Necker Enfants Malades University Hospital, Université De Paris, Paris, France.,INSERM U1141, Paris, France & Neurospin, CEA, Gif/Yvette, France
| | - M Kuchenbuch
- Reference Centre for Rare Epilepsies, Department of Pediatric Neurology, Necker Enfants Malades University Hospital, Université De Paris, Paris, France.,Institut National De La Santé Et De La Recherche Médicale (INSERM), UMR 1163, Institut Imagine, Université De Paris, Paris, France
| |
Collapse
|
34
|
Spencer CE, Flint LE, Duckett CJ, Cole LM, Cross N, Smith DP, Clench MR. Role of MALDI-MSI in combination with 3D tissue models for early stage efficacy and safety testing of drugs and toxicants. Expert Rev Proteomics 2020; 17:827-841. [PMID: 33440126 PMCID: PMC8396712 DOI: 10.1080/14789450.2021.1876568] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 01/12/2021] [Indexed: 12/13/2022]
Abstract
Introduction: Three-dimensional (3D) cell cultures have become increasingly important materials to investigate biological processes and drug efficacy and toxicity. The ability of 3D cultures to mimic the physiology of primary tissues and organs in the human body enables further insight into cellular behavior and is hence highly desirable in early-stage drug development. Analyzing the spatial distribution of drug compounds and endogenous molecules provides an insight into the efficacy of a drug whilst simultaneously giving information on biological responses. Areas Covered: In this review we will examine the main 3D cell culture systems employed and applications, which describe their integration with mass spectrometry imaging (MSI). Expert Opinion: MSI is a powerful technique that can map a vast range of molecules simultaneously in tissues without the addition of labels that can provide insights into the efficacy and safety of a new drug. The combination of MSI and 3D cell cultures has emerged as a promising tool in early-stage drug analysis. However, the most common administration route for pharmaceutical drugs is via oral delivery. The use of MSI in combination with models of the GI tract is an area that has been little explored to date, the reasons for this are discussed.
Collapse
Affiliation(s)
- Chloe E Spencer
- Centre for Mass Spectrometry Imaging, Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield, UK
| | - Lucy E Flint
- Centre for Mass Spectrometry Imaging, Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield, UK
| | - Catherine J Duckett
- Centre for Mass Spectrometry Imaging, Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield, UK
| | - Laura M Cole
- Centre for Mass Spectrometry Imaging, Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield, UK
| | - Neil Cross
- Centre for Mass Spectrometry Imaging, Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield, UK
| | - David P Smith
- Centre for Mass Spectrometry Imaging, Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield, UK
| | - Malcolm R Clench
- Centre for Mass Spectrometry Imaging, Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield, UK
| |
Collapse
|
35
|
Marsh MC, Watson JR, Holton C, Hill N, Saldivar L, Janse S, Haberman C. Relationship Between Clinical Factors and Duration of IV Antibiotic Treatment in Neonatal UTI. Hosp Pediatr 2020; 10:743-749. [PMID: 32817062 DOI: 10.1542/hpeds.2019-0325] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
OBJECTIVES To describe practice patterns of intravenous (IV) antibiotic treatment duration in term neonates ≤28 days old with a urinary tract infection (UTI). METHODS We performed a retrospective chart review of term neonates ≤28 days old hospitalized for UTI at 2 academic pediatric hospitals from 2012 to 2018. Neonates who were admitted to the PICU or with known preexisting renal and/or urologic anomalies or concomitant bacteremia were excluded. We examined clinical features, complications, and duration of IV antibiotic therapy. Univariate and multivariate analyses of long duration of IV antibiotics (>48 hours) were performed by using logistic regression. RESULTS Of 310 neonates identified by diagnostic codes and chart review, 112 met criteria for inclusion. The median IV antibiotic duration was 49 hours (51% received IV antibiotics for >48 hours), and the median total antibiotic duration was 10 days. No demographic features or laboratory values correlated with IV antibiotic duration apart from age <7 days. The odds of long IV antibiotic duration increased if the neonate had a secondary diagnosis extending hospitalization (adjusted odds ratio [aOR] = 3.2; P = .002; 95% confidence interval [CI], 1.2-8.7), subspecialty consult (aOR = 4.79; P < .001; 95% CI, 1.87-12.3), or an abnormal renal ultrasound (aOR = 2.26; P = .02; 95% CI, 1.01-5.08). Only 1 neonate experienced treatment failure. CONCLUSIONS Our study revealed the recent trend toward shorter IV antibiotic courses for healthy term neonates with UTI is inclusive of infants ≤28 days at these 2 sites. Few factors associated with neonates' initial clinical presentation appear to influence the length of IV antibiotic treatment.
Collapse
Affiliation(s)
| | | | - Cara Holton
- Children's Mercy Hospital, Kansas City, Missouri
| | - Nina Hill
- College of Medicine, The Ohio State University, Columbus, Ohio; and
| | - Laura Saldivar
- College of Medicine, The Ohio State University, Columbus, Ohio; and
| | - Sarah Janse
- Nationwide Children's Hospital, Columbus, Ohio.,College of Medicine, The Ohio State University, Columbus, Ohio; and
| | - Cara Haberman
- Brenner Children's Hospital, Wake Forest Baptist Health, Winston Salem, North Carolina
| |
Collapse
|
36
|
Xie F, Van Bocxlaer J, Vermeulen A. Physiologically based pharmacokinetic modelling of lisinopril in children: A case story of angiotensin converting enzyme inhibitors. Br J Clin Pharmacol 2020; 87:1203-1214. [PMID: 32693432 DOI: 10.1111/bcp.14492] [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: 01/27/2020] [Revised: 07/09/2020] [Accepted: 07/12/2020] [Indexed: 11/26/2022] Open
Abstract
AIMS Lisinopril is an angiotensin converting enzyme inhibitor to treat hypertension. It shows complex pharmacokinetics (PK), and its PK behaviour in paediatric populations is not well characterized. The aim of this study was to develop a physiologically based PK (PBPK) model for lisinopril to describe the drug's PK in children. METHODS The PBPK model development was performed in a step-wise manner. An adult model was initially developed to characterize lisinopril's disposition and absorption and verified using literature data. Subsequently, the adult PBPK model was extrapolated to the paediatric population (0.5-18 years old) by accounting for age-dependent physiological and anatomical changes. Model performance was evaluated by comparing the PK profiles and drug exposures of observed vs predicted data. RESULTS The disposition of lisinopril was well described by a minimal PBPK model-an effective strategy to capture the biphasic elimination of the drug. The absorption of lisinopril was described by the intestinal peptide transporter-mediated uptake. The adult model adequately described the literature data with predictions within a twofold range of clinical observations. Good model predictivity was also observed in children older than 6 years of age. The model overpredicted the drug exposure in children under 6 years, probably due to not incorporating the actual, unknown ontogeny of the intestinal peptide transporter. CONCLUSIONS The PBPK model predicted the PK of lisinopril in adults and children above 6 years of age well. Model refinement in children under 6 years warrants future informative ontogeny data of the intestinal peptide transporter.
Collapse
Affiliation(s)
- Feifan Xie
- Division of Biopharmaceutics and Pharmacokinetics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China.,Laboratory of Medical Biochemistry and Clinical Analysis, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
| | - Jan Van Bocxlaer
- Laboratory of Medical Biochemistry and Clinical Analysis, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
| | - An Vermeulen
- Laboratory of Medical Biochemistry and Clinical Analysis, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
| |
Collapse
|
37
|
Bhatt-Mehta V, Hammoud H, Amidon GL. A proposed pediatric biopharmaceutical classification system for medications for chronic diseases in children. Eur J Pharm Sci 2020; 152:105437. [PMID: 32598914 DOI: 10.1016/j.ejps.2020.105437] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 06/15/2020] [Accepted: 06/25/2020] [Indexed: 01/04/2023]
Abstract
Age-appropriate pediatric formulations for oral administration can be challenging to formulate. Development of such formulations is often time consuming, labor-intensive and costly. The Biopharmaceutical Classification System (BCS), developed more than two decades ago, is used to develop suitable oral drug formulations for adult use. In theory, some of the same principles could be applied to formulate pediatric oral liquid dosage forms. However, the present BCS system was developed using adult gastrointestinal physiologic factors. Direct extrapolation of this method to develop pediatric oral dosage forms is inappropriate due to differences in adult and pediatric gastrointestinal physiologic differences during development. To date age-appropriate BCS to guide pediatric oral liquid formulation development has not been developed for various pediatric subpopulations. The objective of this study was to provisionally classify oral liquid formulations of extemporaneously prepared drugs at our institution into an age-appropriate BCS class after elimination of any duplicate listing when matched with the most current World Health Organization's Essential Medicines List for Children available at the time of this study and other published studies that may have reported BCS classification of drugs used as extemporaneous oral liquid formulations in children to treat chronic or rare diseases. A total of 96 orally administered extemporaneously compounded liquid formulations were included in this classification. Dose numbers were calculated using age-appropriate initial gastric volume for neonates, 6-month-old infants, and children up to 6 years of age. Using age-appropriate initial gastric volumes and pediatric and neonatal Lexicomp® age-specific maximal dosing recommendations for calculation of dose numbers, the solubility classes shifted for 62.5% of the drugs studied. A significant number of currently used extemporaneously compounded oral liquid formulations for age groups of children included in this study may not provide formulations with predictable safety and efficacy. Factors used in development of adult BCS cannot be applied directly to pediatric subpopulations.
Collapse
Affiliation(s)
- Varsha Bhatt-Mehta
- Department of Clinical Pharmacy, College of Pharmacy, University of Michigan, Ann Arbor, MI, USA; Department of Pediatrics and Communicable Diseases, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA.
| | - Hannah Hammoud
- Department of Clinical Pharmacy, College of Pharmacy, University of Michigan, Ann Arbor, MI, USA
| | - Gordon L Amidon
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
38
|
delMoral-Sanchez JM, Gonzalez-Alvarez I, Gonzalez-Alvarez M, Navarro-Ruiz A, Bermejo M. Availability of Authorizations from EMA and FDA for Age-Appropriate Medicines Contained in the WHO Essential Medicines List for Children 2019. Pharmaceutics 2020; 12:pharmaceutics12040316. [PMID: 32244848 PMCID: PMC7238124 DOI: 10.3390/pharmaceutics12040316] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 03/21/2020] [Accepted: 03/26/2020] [Indexed: 11/16/2022] Open
Abstract
Lack of age-appropriate commercially drug products availability is a common problem in pediatric therapeutics; this population needs improved and safer drug delivery. In addition, biopharmaceutic aspects, dosage requirements, and swallowing abilities demand pediatric forms different to adult formulations. The objective of this study was to evaluate the authorization availability from United States Food and Drug Administration (FDA) and European Medicines Agency (EMA) of oral essential medicines for children and analyze its age-appropriateness for oral administration in children. All oral drugs from 7th List of Essential Medicines for Children by World Health Organization (WHO) were selected. Availability of commercial drug products was collected from OrangeBook, Spanish drug product catalogue, British electronic Medicines Compendium, and the International Vademecum. Tablets, effervescent tablets, and capsules were considered as not age-appropriate forms. Liquid forms, powder for oral suspension, mini tablets, granules, and soluble films were considered as age-appropriate forms due to their flexibility. More than 80% of the studied drugs possess a commercial authorization in oral forms in both EMA and FDA. Nevertheless, around 50% of these formulations are not age-appropriate for most pediatric groups. This study shows the lack of age-appropriate medicines for children. More efforts are needed to improve development and approval of pediatric medicines.
Collapse
Affiliation(s)
- Jose-Manuel delMoral-Sanchez
- Department of Pharmacokinetics and Pharmaceutical Technology, Miguel Hernandez University, 03550 San Juan de Alicante, Spain; (J.-M.d.-S.); (M.G.-A.); (M.B.)
- Institute of Molecular and Cellular Biology of Miguel Hernandez University, 03202 Elche, Spain
| | - Isabel Gonzalez-Alvarez
- Department of Pharmacokinetics and Pharmaceutical Technology, Miguel Hernandez University, 03550 San Juan de Alicante, Spain; (J.-M.d.-S.); (M.G.-A.); (M.B.)
- Correspondence: ; Tel.: +34-965919217
| | - Marta Gonzalez-Alvarez
- Department of Pharmacokinetics and Pharmaceutical Technology, Miguel Hernandez University, 03550 San Juan de Alicante, Spain; (J.-M.d.-S.); (M.G.-A.); (M.B.)
| | | | - Marival Bermejo
- Department of Pharmacokinetics and Pharmaceutical Technology, Miguel Hernandez University, 03550 San Juan de Alicante, Spain; (J.-M.d.-S.); (M.G.-A.); (M.B.)
| |
Collapse
|
39
|
Germovsek E, Barker CIS, Sharland M, Standing JF. Pharmacokinetic-Pharmacodynamic Modeling in Pediatric Drug Development, and the Importance of Standardized Scaling of Clearance. Clin Pharmacokinet 2020; 58:39-52. [PMID: 29675639 PMCID: PMC6325987 DOI: 10.1007/s40262-018-0659-0] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Pharmacokinetic/pharmacodynamic (PKPD) modeling is important in the design and conduct of clinical pharmacology research in children. During drug development, PKPD modeling and simulation should underpin rational trial design and facilitate extrapolation to investigate efficacy and safety. The application of PKPD modeling to optimize dosing recommendations and therapeutic drug monitoring is also increasing, and PKPD model-based dose individualization will become a core feature of personalized medicine. Following extensive progress on pediatric PK modeling, a greater emphasis now needs to be placed on PD modeling to understand age-related changes in drug effects. This paper discusses the principles of PKPD modeling in the context of pediatric drug development, summarizing how important PK parameters, such as clearance (CL), are scaled with size and age, and highlights a standardized method for CL scaling in children. One standard scaling method would facilitate comparison of PK parameters across multiple studies, thus increasing the utility of existing PK models and facilitating optimal design of new studies.
Collapse
Affiliation(s)
- Eva Germovsek
- Infection, Inflammation and Rheumatology Section, UCL Great Ormond Street Institute of Child Heath, University College London, London, UK. .,Pharmacometrics Research Group, Department of Pharmaceutical Biosciences, Uppsala University, PO Box 591, 751 24, Uppsala, Sweden.
| | - Charlotte I S Barker
- Infection, Inflammation and Rheumatology Section, UCL Great Ormond Street Institute of Child Heath, University College London, London, UK.,Paediatric Infectious Diseases Research Group, Institute for Infection and Immunity, St George's, University of London, London, UK.,St George's University Hospitals NHS Foundation Trust, London, UK
| | - Mike Sharland
- Paediatric Infectious Diseases Research Group, Institute for Infection and Immunity, St George's, University of London, London, UK.,St George's University Hospitals NHS Foundation Trust, London, UK
| | - Joseph F Standing
- Infection, Inflammation and Rheumatology Section, UCL Great Ormond Street Institute of Child Heath, University College London, London, UK.,Paediatric Infectious Diseases Research Group, Institute for Infection and Immunity, St George's, University of London, London, UK
| |
Collapse
|
40
|
Toward precision medicine in pediatric population using cytochrome P450 phenotyping approaches and physiologically based pharmacokinetic modeling. Pediatr Res 2020; 87:441-449. [PMID: 31600772 DOI: 10.1038/s41390-019-0609-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 09/04/2019] [Accepted: 09/22/2019] [Indexed: 01/18/2023]
Abstract
The activity of drug-metabolizing enzymes (DME) shows high inter- and intra-individual variability. Genetic polymorphisms, exposure to drugs, and environmental toxins are known to significantly alter DME expression. In addition, the activity of these enzymes is highly age-dependent due to maturation processes that occur during development. Currently, there is a vast choice of phenotyping methods in adults using exogenous probes to characterize the activity of these enzymes. However, this can hardly be applied to children since it requires the intake of non-therapeutic xenobiotics. In addition, sampling may be challenging in the pediatric population for a variety of reasons: limited volume (e.g., blood), inappropriate sampling methods for age (e.g., urine), and metric requiring invasive or multiple blood samples. This review covers the main existing methods that can be used in the pediatric population to determine DME activity, with a particular focus on cytochrome P450 enzymes. Less invasive tools are described, including phenotyping using endogenous probes. Finally, the potential of pediatric model-informed precision dosing using physiologically based pharmacokinetic modeling is discussed.
Collapse
|
41
|
Classification of WHO Essential Oral Medicines for Children Applying a Provisional Pediatric Biopharmaceutics Classification System. Pharmaceutics 2019; 11:pharmaceutics11110567. [PMID: 31683740 PMCID: PMC6920833 DOI: 10.3390/pharmaceutics11110567] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 10/21/2019] [Accepted: 10/30/2019] [Indexed: 12/23/2022] Open
Abstract
The objective was using the Essential Medicines List for children by the World Health Organization (WHO) to create a pediatric biopharmaceutics classification system (pBCS) of the oral drugs included in the Essential Medicines List by the World Health Organization and to compare our results with the BCS for adults (aBCS). Several methods to estimate the oral drug dose in different pediatric groups were used to calculate dose number (Do) and solubility (high/low). The estimation of the gastrointestinal water volume was adapted to each pediatric group. Provisional permeability classification was done by comparison of each drug lipophilicity versus metoprolol as the model drug of high permeability. As a result, 24.5% of the included drugs moved from the favorable to unfavorable class (i.e., from high to low solubility). Observed changes point out potential differences in product performance in pediatrics compared to adults, due to changes in the limiting factors for absorption. BCS Class Changes 1 to 2 or 3 to 4 are indicative of drugs that could be more sensitive to the choice of appropriate excipient in the development process. Validating a pBCS for each age group would provide a valuable tool to apply in specific pediatric formulation design by reducing time and costs and avoiding unnecessary pediatric experiments restricted due to ethical reasons. Additionally, pBCS could minimize the associated risks to the use of adult medicines or pharmaceutical compound formulations.
Collapse
|
42
|
Korth-Bradley JM. The Path to Perfect Pediatric Posology - Drug Development in Pediatrics. J Clin Pharmacol 2019; 58 Suppl 10:S48-S57. [PMID: 30248197 DOI: 10.1002/jcph.1081] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 12/21/2017] [Indexed: 11/06/2022]
Abstract
Reluctance to enroll pediatric subjects in clinical trials has left gaps in information about dosing, safety, and efficacy of medications. Pharmacotherapeutic information for pediatric patients may be available for only a small range of ages and may be deficient, as children respond differently as they grow and mature from prematurity to adolescence. Current regulations, however, require early planning for the participation of children in drug development, as pediatric plans must be submitted at the end of phase 1 (European Union) or the end of phase 2 (United States). These plans are extensive, outlining planned studies, subjects to be enrolled, dose and dosage form justification, planned observations, and statistical analysis as well as planned modeling, simulation, and extrapolation analyses. The extent to which efficacy information in adults can be extrapolated to children depends on how similar the disease is in adults and each of the 5 pediatric age groups. Extrapolation may not be possible for conditions that do not occur in adults, requiring a complete development plan in adults, or extrapolation may be complete because of similar pathology and response to treatment. Pharmacokinetic and safety information cannot be extrapolated and must be collected in children of all ages, unless a waiver is granted. Physiologically based pharmacokinetic modeling, optimal design, population pharmacokinetics, and scavenged samples are all examples of new methodologies being used to study pediatric therapeutics. Clinicaltrials.gov and EU Clinical Trials registry are good sources of results of pediatric trials, although sponsors are also working toward prompt publication of study results in peer-reviewed journals.
Collapse
Affiliation(s)
- Joan M Korth-Bradley
- Clinical Pharmacology, Global Product Development, Pfizer Inc., Collegeville, PA, USA
| |
Collapse
|
43
|
Waalewijn H, Turkova A, Rakhmanina N, Cressey TR, Penazzato M, Colbers A, Burger DM. Optimizing Pediatric Dosing Recommendations and Treatment Management of Antiretroviral Drugs Using Therapeutic Drug Monitoring Data in Children Living With HIV. Ther Drug Monit 2019; 41:431-443. [PMID: 31008997 PMCID: PMC6636807 DOI: 10.1097/ftd.0000000000000637] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 03/03/2019] [Indexed: 12/27/2022]
Abstract
INTRODUCTION This review summarizes the current dosing recommendations for antiretroviral (ARV) drugs in the international pediatric guidelines of the World Health Organization (WHO), US Department of Health and Human Services (DHHS), and Pediatric European Network for Treatment of AIDS (PENTA), and evaluates the research that informed these approaches. We further explore the role of data generated through therapeutic drug monitoring in optimizing the dosing of ARVs in children. METHODS A PubMed search was conducted for the literature on ARV dosing published in English. In addition, the registration documentation of European Medicines Agency and the US Food and Drug Administration for currently used ARVs and studies referenced by the WHO, DHHS, and EMA guidelines were screened. Resulting publications were screened for papers containing data on the area under the concentration-time curve, trough concentration, and peak concentration. Studies with enrolled participants with a median or mean age of ≥18 years were excluded. No restriction on publishing date was applied. DISCUSSION AND CONCLUSION Pediatric ARV dosing is frequently based on data obtained from small studies and is often simplified to facilitate dosing in the context of a public health approach. Pharmacokinetic parameters of pediatric ARVs are subject to high interpatient variation and this leads to a potential risk of underdosing or overdosing when drugs are used in real life. To ensure optimal use of ARVs and validate dosing recommendations for children, it is essential to monitor ARV dosing more thoroughly with larger sample sizes and to include diverse subpopulations. Therapeutic drug monitoring data generated in children, where available and affordable, have the potential to enhance our understanding of the appropriateness of simplified pediatric dosing strategies recommended using a public health approach and to uncover suboptimal dosing or other unanticipated issues postmarketing, further facilitating the ultimate goal of optimizing pediatric ARV treatment.
Collapse
Affiliation(s)
- Hylke Waalewijn
- Department of Pharmacy, Radboud Institute for Health Sciences (RIHS), Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Anna Turkova
- MRC Clinical Trials Unit, Institute of Clinical Trials and Methodology, University College London
- Department of Paediatric Infectious Diseases, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
| | - Natella Rakhmanina
- Department of Pediatrics, School of Medicine and Health Sciences, The George Washington University
- Division of Infectious Diseases, Children's National Medical Center
- Elizabeth Glaser Pediatric AIDS Foundation, Washington, District of Columbia
| | - Tim R. Cressey
- PHPT/IRD UMI 174, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
- Department of Immunology and Infectious Diseases, Harvard T.H Chan School of Public Health, Boston, Massachusetts
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, United Kingdom; and
| | - Martina Penazzato
- Treatment and Care, Department of HIV/AIDS, World Health Organization, Geneva, Switzerland
| | - Angela Colbers
- Department of Pharmacy, Radboud Institute for Health Sciences (RIHS), Radboud University Medical Centre, Nijmegen, the Netherlands
| | - David M. Burger
- Department of Pharmacy, Radboud Institute for Health Sciences (RIHS), Radboud University Medical Centre, Nijmegen, the Netherlands
| |
Collapse
|
44
|
Illamola SM, Birnbaum AK, Sherwin CM. Generic drug products in paediatrics: Where are the data? Br J Clin Pharmacol 2019; 85:1871-1873. [PMID: 31281980 DOI: 10.1111/bcp.14036] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 05/19/2019] [Accepted: 06/12/2019] [Indexed: 12/12/2022] Open
Affiliation(s)
- Sílvia M Illamola
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN, USA
| | - Angela K Birnbaum
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN, USA
| | - Catherine M Sherwin
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN, USA.,Department of Pediatrics, Wright State University Boonshoft School of Medicine, Dayton, OH, USA.,Dayton Children's Hospital, Dayton, OH, USA
| |
Collapse
|
45
|
O'Brien F, Clapham D, Krysiak K, Batchelor H, Field P, Caivano G, Pertile M, Nunn A, Tuleu C. Making Medicines Baby Size: The Challenges in Bridging the Formulation Gap in Neonatal Medicine. Int J Mol Sci 2019; 20:E2688. [PMID: 31159216 PMCID: PMC6600135 DOI: 10.3390/ijms20112688] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 05/17/2019] [Accepted: 05/24/2019] [Indexed: 12/12/2022] Open
Abstract
The development of age-appropriate formulations should focus on dosage forms that can deliver variable yet accurate doses that are safe and acceptable to the child, are matched to his/her development and ability, and avoid medication errors. However, in the past decade, the medication needs of neonates have largely been neglected. The aim of this review is to expand on what differentiates the needs of preterm and term neonates from those of the older paediatric subsets, in terms of environment of care, ability to measure and administer the dose (from the perspective of the patient and carer, the routes of administration, the device and the product), neonatal biopharmaceutics and regulatory challenges. This review offers insight into those challenges posed by the formulation of medicinal products for neonatal patients in order to support the development of clinically relevant products.
Collapse
Affiliation(s)
- Fiona O'Brien
- School of Pharmacy, Royal College of Surgeons in Ireland, 111 St Stephens Green Dublin 2, Ireland.
| | | | - Kamelia Krysiak
- School of Pharmacy, Royal College of Surgeons in Ireland, 111 St Stephens Green Dublin 2, Ireland.
| | - Hannah Batchelor
- College of Medical and Dental Sciences, Institute of Clinical Sciences, University of Birmingham, Birmingham B15 2TT, UK.
| | - Peter Field
- University College London School of Pharmacy, 29-39 Brunswick Square, London WC1N 1AX, UK.
| | - Grazia Caivano
- Chiesi Farmaceutici S.p.A. Largo Francesco Belloli 11/A-43122 Parma, Italy.
| | - Marisa Pertile
- Chiesi Farmaceutici S.p.A. Largo Francesco Belloli 11/A-43122 Parma, Italy.
| | - Anthony Nunn
- Department of Women's and Children's Health, University of Liverpool, Liverpool Women's Hospital, Liverpool L8 7SS, UK.
| | - Catherine Tuleu
- University College London School of Pharmacy, 29-39 Brunswick Square, London WC1N 1AX, UK.
| |
Collapse
|
46
|
Nicolas JM, de Lange ECM. Mind the Gaps: Ontogeny of Human Brain P-gp and Its Impact on Drug Toxicity. AAPS JOURNAL 2019; 21:67. [PMID: 31140038 DOI: 10.1208/s12248-019-0340-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 05/10/2019] [Indexed: 12/18/2022]
Abstract
Available data on human brain P-glycoprotein ontogeny during infancy and childhood are limited. This review discusses the current body of data relating to maturation of human brain P-glycoprotein including transporter expression levels in post-mortem human brain samples, in vivo transporter activity using probe substrates, surrogate marker endpoints, and extrapolations from animal models. Overall, the data tend to confirm that human brain P-glycoprotein activity keeps developing after birth, although with a developmental time frame that remains unclear. This knowledge gap is a concern given the critical role of brain P-glycoprotein in drug safety and efficacy, and the vulnerable nature of the pediatric population. Future research could include the measurement of brain P-glycoprotein activity across age groups using positron emission tomography or central pharmacodynamic responses. For now, caution is advised when extrapolating adult data to children aged younger than 2 years for drugs with P-glycoprotein-dependent central nervous system activity.
Collapse
Affiliation(s)
- Jean-Marie Nicolas
- Quantitative Pharmacology DMPK Department, UCB BioPharma, Chemin du Foriest, 1420, Braine L'Alleud, Belgium.
| | - Elizabeth C M de Lange
- Research Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| |
Collapse
|
47
|
Lavan M, Byrn SR, Knipp G. Pediatric Formulations: Knowledge Gaps Limiting the Expedited Preclinical to Clinical Translation in Children. AAPS PharmSciTech 2019; 20:73. [PMID: 30631973 DOI: 10.1208/s12249-018-1253-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Accepted: 11/18/2018] [Indexed: 11/30/2022] Open
Abstract
Traditionally, drug discovery and development research have been primarily focused on the mitigation of disease treatment for the general adult population, often overlooking the medical needs of pediatric patients. While remarkable progress toward the discovery of better medicines has been made, the pharmacological differences between children and adults are often neglected as part of the translation process. In fact, until recently, children have been considered therapeutic orphans due to the lack of significant drug discovery, formulation development, and dosage form design specifically tailored for pediatric patients. Perhaps the least understood is the significant physiological changes that occur during the maturation process from birth to adulthood. It requires careful considerations to achieve age-specific-desired therapeutic outcomes with minimal toxicity. This introduces considerable risk into the preclinical and clinical testing of new medicaments, which until recently, was avoided based on the conventional approach where a demonstration of safe and efficacious use in adults over several years potentially would minimize the chance of adverse juvenile responses. However, the lack of appropriate drug products for children has led to off-label use of adult medicines with potential life-threatening adverse reactions and health complications. Recent developments and future considerations regarding pediatric drug discovery and development using a patient-centric approach in the context of ontogenic biopharmaceutical considerations are discussed below.
Collapse
|
48
|
Neal-Kluever A, Fisher J, Grylack L, Kakiuchi-Kiyota S, Halpern W. Physiology of the Neonatal Gastrointestinal System Relevant to the Disposition of Orally Administered Medications. Drug Metab Dispos 2018; 47:296-313. [DOI: 10.1124/dmd.118.084418] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 12/14/2018] [Indexed: 12/13/2022] Open
|
49
|
Van Den Abeele J, Rayyan M, Hoffman I, Van de Vijver E, Zhu W, Augustijns P. Gastric fluid composition in a paediatric population: Age-dependent changes relevant for gastrointestinal drug disposition. Eur J Pharm Sci 2018; 123:301-311. [DOI: 10.1016/j.ejps.2018.07.022] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Revised: 06/18/2018] [Accepted: 07/09/2018] [Indexed: 11/30/2022]
|
50
|
van den Anker J, Reed MD, Allegaert K, Kearns GL. Developmental Changes in Pharmacokinetics and Pharmacodynamics. J Clin Pharmacol 2018; 58 Suppl 10:S10-S25. [DOI: 10.1002/jcph.1284] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 06/21/2018] [Indexed: 12/22/2022]
Affiliation(s)
- John van den Anker
- Division of Clinical Pharmacology; Children's National Health System; Washington DC USA
- Division of Paediatric Pharmacology and Pharmacometrics; University of Basel Children's Hospital; Basel Switzerland
- Intensive Care and Department of Pediatric Surgery; Erasmus Medical Center-Sophia Children's Hospital; Rotterdam the Netherlands
| | - Michael D. Reed
- Emeritus Professor of Pediatrics; School of Medicine; Case Western Reserve University; Cleveland OH USA
| | - Karel Allegaert
- Intensive Care and Department of Pediatric Surgery; Erasmus Medical Center-Sophia Children's Hospital; Rotterdam the Netherlands
- Department of Pediatrics; Division of Neonatology; Erasmus Medical Center-Sophia Children's Hospital; Rotterdam the Netherlands
- Department of Development and Regeneration; KU Leuven; Leuven Belgium
| | | |
Collapse
|