1
|
Shaniv D, Allegaert K. Applied pharmacokinetics to improve pharmacotherapy in neonatal and paediatric intensive care units: focus on correct dose selection. Arch Dis Child Educ Pract Ed 2024; 109:184-190. [PMID: 38408792 DOI: 10.1136/archdischild-2023-326325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 02/13/2024] [Indexed: 02/28/2024]
Abstract
Drug dosing and exposure throughout childhood are constantly affected by maturational changes like weight, age or body surface area. In neonatal and paediatric intensive care units (NICU and PICU, respectively), drug dosing and exposure are further impacted by non-maturational changes. These changes are related to factors such as sepsis, cardiac failure, acute kidney injury, extracorporeal circuits or drug-drug interactions (DDIs) resulting from polypharmacy.This potentially complex situation may alter drug pharmacokinetics to result in greater-than-usual intrapatient and interpatient drug exposure variability. These effects may call for individual dosage adjustments. Dosage adjustments may apply to both loading doses or maintenance doses, which should be used as appropriate, depending on the specific characteristics of a given drug. Phenobarbital and vancomycin dosing are hereby used as illustrations.To optimise dose selection in NICU/PICU settings, we suggest to consider therapeutic drug monitoring integrated in model-informed precision dosing, and to familiarise oneself with existing paediatric drug formularies as well as DDI databases/search engines. Paediatric clinical pharmacologists and pharmacists can hereby guide clinicians with no prior experience on how to properly apply these data sources to day-to-day practice in individual patients or specific subpopulations of NICU or PICU patients.
Collapse
Affiliation(s)
- Dotan Shaniv
- Pharmacy Services, Kaplan Medical Center, Clalit Health Services, Rehovot, Israel
- Neonatal Intensive Care Unit, Kaplan Medical Center, Clarit Health Services, Rehovot, Israel
| | - 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, Netherlands
| |
Collapse
|
2
|
Cherifi F, Da Silva A, Martins-Branco D, Awada A, Nader-Marta G. Pharmacokinetics and pharmacodynamics of antibody-drug conjugates for the treatment of patients with breast cancer. Expert Opin Drug Metab Toxicol 2024; 20:45-59. [PMID: 38214896 DOI: 10.1080/17425255.2024.2302460] [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: 09/25/2023] [Accepted: 01/03/2024] [Indexed: 01/13/2024]
Abstract
INTRODUCTION Currently three antibody-drug-conjugates (ADC) are approved by the European Medicines Agency (EMA) for treatment of breast cancer (BC) patient: trastuzumab-emtansine, trastuzumab-deruxtecan and sacituzumab-govitecan. ADC are composed of a monoclonal antibody (mAb) targeting a specific antigen, a cytotoxic payload and a linker. Pharmacokinetics (PK) and pharmacodynamics (PD) distinguish ADC from conventional chemotherapy and must be understood by clinicians. AREAS COVERED Our review delineates the PK/PD profiles of ADC approved for the treatment of BC with insight for future development. This is an expert opinion literature review based on the EMA's Assessment Reports, enriched by a comprehensive literature search performed on Medline in August 2023. EXPERT OPINION All three ADC distributions are described by a two-compartment structure: tissue and serum. Payload concentration peak is immediate but remains at low concentration. The distribution varied for all ADC only with body weight. mAb will be metabolised firstly by the saturable complex formation of ADC/Tumour-Receptor and secondly by binding of FcgRs in immune cells. They are all excreted in the bile and faeces with minimal urine elimination. Dose adjustments, apart from weight, are not recommended. Novel ADC are composed of cleavable linkers with various targets/payloads with the same PK/PD properties, but novel structures of ADC are in development.
Collapse
Affiliation(s)
- François Cherifi
- Oncology Medicine Department, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (ULB), Brussels, Belgium
- Breast Cancer Unit, CLCC François Baclesse, Institut Normand du Sein, Caen, France
| | - Angélique Da Silva
- Departments of Pharmacology and Medical Oncology, Caen-Normandy University Hospital, PICARO Cardio-Oncology Program, Normandie Univ, UNICAEN, INSERM U1086 ANTICIPE, Caen, France
| | - Diogo Martins-Branco
- Academic Trials Promoting Team (ATPT), Institut Jules Bordet, Hôpital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (U.L.B), Brussels, Belgium
| | - Ahmad Awada
- Oncology Medicine Department, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Guilherme Nader-Marta
- Academic Trials Promoting Team (ATPT), Institut Jules Bordet, Hôpital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (U.L.B), Brussels, Belgium
| |
Collapse
|
3
|
Cardoso E, Monfort A, Ferreira E, Nordeng H, Winterfeld U, Allegaert K, Gandia P, Guidi M, Panchaud A. Maternal drugs and breastfeeding: Risk assessment from pharmacokinetics to safety evidence - A contribution from the ConcePTION project. Therapie 2023; 78:149-156. [PMID: 36804048 DOI: 10.1016/j.therap.2023.01.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 01/24/2023] [Indexed: 02/01/2023]
Abstract
Human milk is the most appropriate form of nutrition for infants while taking medication during the postpartum period is common. Discontinuation of breastfeeding is sometimes wrongly recommended for fear of adverse effects in the breastfed infant whereas only a few drugs are strictly contraindicated while breastfeeding. Most drugs are transferred from the mother's blood to the milk, but the breastfed infant usually ingests a small drug amount through human milk. As population-based evidence is still scarce on safety of drugs during breastfeeding, risk assessment relies on the little clinical evidence available and on pharmacokinetic principles, as well as on specialized sources of information that are essential for clinical decision-making. Risk assessment should not only be based on the drug's potential risk for the breastfed infant but should always take into account the benefits associated to breastfeeding, the risks of untreated maternal disease and the maternal willingness to breastfeed. Identifying situations with potential for drug accumulation in the breastfed infant is decisive while assessing the risk. Health care providers should always assume that mothers will be concerned and use risk communication as a key to ensure medication adherence and prevent unnecessary interruption of breastfeeding. When a mother still expresses concerns, decision support algorithms may facilitate communication and some strategies can be offered to minimize the drug exposure in the breastfed infant even when clinically not justified.
Collapse
Affiliation(s)
- Evelina Cardoso
- Service of Pharmacy, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland.
| | - Anaëlle Monfort
- Platform of Biopharmacy, Faculty of Pharmacy, University of Montreal, Montreal, H3T 1J4 QC, Canada; CHU Sainte-Justine, Montreal, H3T 1C5 QC, Canada; Faculty of Pharmacy, University of Montreal, Montreal, H3C 3J7 QC, Canada
| | - Ema Ferreira
- CHU Sainte-Justine, Montreal, H3T 1C5 QC, Canada; Faculty of Pharmacy, University of Montreal, Montreal, H3C 3J7 QC, Canada
| | - Hedvig Nordeng
- Pharmacoepidemiology and Drug Safety Research Group, Department of Pharmacy, PharmaTox Strategic Initiative, Faculty of Mathematics and Natural Sciences, University of Oslo, 0316 Oslo, Norway; Department of Child Health and Development, Norwegian Institute of Public Health, 0473 Oslo, Norway
| | - Ursula Winterfeld
- Swiss Teratogen Information Service, Service of Clinical Pharmacology, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland
| | - Karel Allegaert
- Child and Youth Institute, KU Leuven, Herestraat 49, 3000 Leuven, Belgium; Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium; Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium; Department of Hospital Pharmacy, Erasmus MC, 3000 GA Rotterdam, the Netherlands
| | - Peggy Gandia
- Laboratory of Pharmacokinetics and Toxicology, Purpan Hospital, University Hospital of Toulouse, 31073 Toulouse, France
| | - Monia Guidi
- Service of Clinical Pharmacology, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland; Center for Research and Innovation in Clinical Pharmaceutical Sciences, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland
| | - Alice Panchaud
- Service of Pharmacy, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland; Institute of Primary Health Care (BIHAM), University of Bern, 3012 Bern, Switzerland
| |
Collapse
|
4
|
Prieto-Garcia JM, Graham L, Alkhabbaz O, Mazzari ALDA. Potential Pharmacokinetic Interactions of Common Cardiovascular Drugs and Selected European and Latin American Herbal Medicines: A Scoping Review. PLANTS (BASEL, SWITZERLAND) 2023; 12:623. [PMID: 36771707 PMCID: PMC9920503 DOI: 10.3390/plants12030623] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 01/17/2023] [Accepted: 01/20/2023] [Indexed: 06/01/2023]
Abstract
BACKGROUND Herb-drug interactions are nowadays an important decision factor in many healthcare interventions. Patients with cardiovascular risk factors such as hyperlipidemia and hypertension are usually prescribed long-term treatments. We need more informed decision tools to direct future clinical research and decision making to avoid HDI occurrences in this group. METHODS A scoping review was conducted using data from online databases such as PUBMED, the National Library of Medicine, and the electronic Medicines Compendium. Included studies consisted of the reported effects on Phase 1/2 and P-glycoprotein of herbal medicines listed in the medicines agencies of Latin America and Europe and drugs used for cardiovascular conditions (statins, diuretics, beta blockers, calcium channel blockers, and ACE inhibitors). The cross tabulation of the results allowed for finding potential HDI. RESULTS AND CONCLUSIONS as per the preclinical data reviewed here, we encourage more clinical research on whether drugs with apparently very low interaction risk, such as pravastatin, nadolol, and nimodipine/nitrendipine, may help prevent HDI when statins, beta blockers, and calcium channel blockers, respectively, are prescribed for long-term treatments.
Collapse
Affiliation(s)
- Jose M. Prieto-Garcia
- Centre for Natural Products Discovery, School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool L3 3AF, UK
| | - Louise Graham
- Centre for Natural Products Discovery, School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool L3 3AF, UK
| | - Osamah Alkhabbaz
- Centre for Natural Products Discovery, School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool L3 3AF, UK
| | | |
Collapse
|
5
|
Burroughs DL, Lorch G, Guo Y, Hill K, Schroeder EL, Cole LK, Phelps MA. Noncompartmental pharmacokinetics of three intravenous mycophenolate mofetil concentrations in healthy Standardbred mares. Vet Dermatol 2022; 34:222-234. [PMID: 35929548 DOI: 10.1111/vde.13109] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 05/10/2022] [Accepted: 05/24/2022] [Indexed: 11/28/2022]
Abstract
BACKGROUND Mycophenolate mofetil (MMF) is the prodrug of mycophenolic acid (MPA) which acts as an immunosuppressive agent. During the biotransformation of MMF to MPA, additional metabolites including MPA phenol glucuronide (MPAG), MPA acyl glucuronide (AcMPAG) and MPA phenol glucoside (MPG) are formed. OBJECTIVE To define the noncompartmental pharmacokinetic (PK) parameters of three single doses of intravenous (i.v.) MMF and its downstream metabolites in healthy horses. ANIMALS Six healthy Standardbred mares. MATERIALS AND METHODS Generic MMF (Par Pharmaceuticals; Chestnut Ridge, NY, USA) was reconstituted and administered as a single i.v. bolus at 1.0 mg/kg, 5.0 mg/kg and 10.0 mg/kg with an eight day washout between treatments. Blood samples were collected immediately before MMF administration and over 24 h. A liquid chromatography-tandem mass spectrometry assay was developed following FDA guidance to determine plasma MMF, MPA, MPAG, AcMPAG and MPG concentrations. Plasma concentrations were analysed independently, followed by calculation of geometric mean and coefficient of variation. RESULTS Noncompartmental PK parameters were determined for MMF and all metabolites at all doses. MMF was rapidly converted to MPA in all horses. Each incremental dose of MMF resulted in increases in Cmax and AUCinf _obs for MPA and the three additional metabolites. Within the 10-fold dose range, the increase in Cmax and AUCinf _obs for MMF and its metabolites was nonlinear. CONCLUSIONS AND CLINICAL RELEVANCE Horses biotransform MMF into MPA, MPAG, AcMPAG and MPG via the glucuronidation and glucosidation clearance pathways. Equine reference PK profiles for MPA and the metabolites, MPAG, AcMPAG and MPG were established.
Collapse
Affiliation(s)
- Dylan L Burroughs
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| | - Gwendolen Lorch
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| | - Yizhen Guo
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Kasey Hill
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Eric L Schroeder
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| | - Lynette K Cole
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| | - Mitch A Phelps
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
6
|
Negru PA, Radu AF, Vesa CM, Behl T, Abdel-Daim MM, Nechifor AC, Endres L, Stoicescu M, Pasca B, Tit DM, Bungau SG. Therapeutic dilemmas in addressing SARS-CoV-2 infection: Favipiravir versus Remdesivir. Biomed Pharmacother 2022; 147:112700. [PMID: 35131656 PMCID: PMC8813547 DOI: 10.1016/j.biopha.2022.112700] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 01/27/2022] [Accepted: 02/02/2022] [Indexed: 02/07/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) represents an unmet clinical need, due to a high mortality rate, rapid mutation rate in the virus, increased chances of reinfection, lack of effectiveness of repurposed drugs and economic damage. COVID-19 pandemic has created an urgent need for effective molecules. Clinically proven efficacy and safety profiles have made favipiravir (FVP) and remdesivir (RDV) promising therapeutic options for use against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Even though both are prodrug molecules with an antiviral role based on a similar mechanism of action, differences in pharmacological, pharmacokinetic and pharmacotoxicological mechanisms have been identified. The present study aims to provide a comprehensive comparative assessment of FVP and RDV against SARS-CoV-2 infections, by centralizing medical data provided by significant literature and authorized clinical trials, focusing on the importance of a better understanding of the interactions between drug molecules and infectious agents in order to improve the global management of COVID-19 patients and to reduce the risk of antiviral resistance.
Collapse
Affiliation(s)
- Paul Andrei Negru
- Doctoral School of Biological and Biomedical Sciences, University of Oradea, 410087 Oradea, Romania.
| | - Andrei-Flavius Radu
- Doctoral School of Biological and Biomedical Sciences, University of Oradea, 410087 Oradea, Romania.
| | - Cosmin Mihai Vesa
- Department of Preclinical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania.
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India.
| | - Mohamed M. Abdel-Daim
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jedah 21442, Saudi Arabia,Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt
| | - Aurelia Cristina Nechifor
- Analytical Chemistry and Environmental Engineering Department, Polytechnic University of Bucharest, 011061 Bucharest, Romania.
| | - Laura Endres
- Department of Psycho-Neuroscience and Recovery, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania.
| | - Manuela Stoicescu
- Department of Medical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania.
| | - Bianca Pasca
- Doctoral School of Biological and Biomedical Sciences, University of Oradea, 410087 Oradea, Romania.
| | - Delia Mirela Tit
- Doctoral School of Biological and Biomedical Sciences, University of Oradea, 410087 Oradea, Romania; Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028 Oradea, Romania.
| | - Simona Gabriela Bungau
- Doctoral School of Biological and Biomedical Sciences, University of Oradea, 410087 Oradea, Romania; Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028 Oradea, Romania.
| |
Collapse
|
7
|
Patel A, Meesters K. Macrolides in children: judicious use, avoiding resistance and reducing adverse effects. Arch Dis Child Educ Pract Ed 2021; 106:216-219. [PMID: 33883190 DOI: 10.1136/archdischild-2020-320357] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 02/20/2021] [Accepted: 03/22/2021] [Indexed: 11/04/2022]
Abstract
Macrolides, a group of antibiotics molecularly characterised by a macrocyclic ring, are among the most frequently prescribed antibiotics for children. Beyond their antibacterial action, macrolides exert immunomodulatory effects. Prophylactic use is increasing. Macrolides are usually well tolerated in children, and dosing schedules are convenient. Furthermore, increasing data suggests that their prophylactic usage reduces the exacerbation frequency in children with bronchiectasis. Yet, to preserve their antibacterial action, each macrolide prescription should be judiciously considered. For prophylactic use, the indication should be regularly reviewed, and usage longer than 6 months should be avoided to reduce antimicrobial resistance.
Collapse
Affiliation(s)
- Anisha Patel
- Pharmacy, Evelina London Children's Hospital, London, UK
| | - Kevin Meesters
- Paediatric Infectious Diseases and Immunology, Evelina London Children's Hospital, London, UK
| |
Collapse
|
8
|
Tauzin M, Ouldali N, Béchet S, Caeymaex L, Cohen R. Pharmacokinetic and pharmacodynamic considerations of cephalosporin use in children. Expert Opin Drug Metab Toxicol 2019; 15:869-880. [PMID: 31597049 DOI: 10.1080/17425255.2019.1678585] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Introduction: Cephalosporins are a major class of antibiotics, frequently used in children because of their remarkable antibacterial activity and excellent safety profile. Time above the minimal inhibitory concentration of the non-protein-bound fraction (fT>MIC) is the pharmacokinetic/pharmacodynamic parameter that correlates with the therapeutic efficacy. In the pediatric population, the inter-individual variability in cephalosporin pharmacokinetics is large because of maturational changes. However, the prescription of cephalosporins promotes emergence of Enterobacteriaceae producing broad-spectrum ß-lactamases.Areas covered: Here we describe in vitro activities and the main pharmacokinetic characteristics of cephalosporins in children. On the basis of these characteristics, we propose an estimation of the fT>MIC for each molecule as a tool to help optimize the use of cephalosporins. We also provide an inventory of the clinical use of cephalosporins and present prospects for the development of new molecules or associations to address the emergence of resistant strains.Expert opinion: Cephalosporins represent a heterogeneous group of antibiotics with various pharmacokinetics and in vitro antimicrobial activity that the clinician needs to master to optimize their use. However, their broad use plays a role in the emergence of broad-spectrum ß-lactamase-producing strains and must thus be restricted to probabilistic broad-spectrum therapy and situations without therapeutic alternatives.
Collapse
Affiliation(s)
- Manon Tauzin
- Neonatal Intensive Care Unit, Centre Hospitalier Intercommunal de Créteil, Créteil, France
| | - Naim Ouldali
- ACTIV, Association Clinique et Thérapeutique Infantile du Val de Marne, Créteil, France.,GPIP, Groupe de Pathologie Infectieuse Pédiatrique, Paris, France.,Unité d'Epidémiologie Clinique, Assistance Publique-Hôpitaux de Paris, Hôpital Robert Debré, ECEVE INSERM UMR, Paris, France
| | - Stéphane Béchet
- ACTIV, Association Clinique et Thérapeutique Infantile du Val de Marne, Créteil, France
| | - Laurence Caeymaex
- Neonatal Intensive Care Unit, Centre Hospitalier Intercommunal de Créteil, Créteil, France.,Paris-Est Créteil University, Créteil, France.,Clinical Research Center (CRC), Centre Hospitalier Intercommunal de Créteil, Créteil, France
| | - Robert Cohen
- ACTIV, Association Clinique et Thérapeutique Infantile du Val de Marne, Créteil, France.,GPIP, Groupe de Pathologie Infectieuse Pédiatrique, Paris, France.,Clinical Research Center (CRC), Centre Hospitalier Intercommunal de Créteil, Créteil, France.,Université Paris Est, IMRB- GRC GEMINI, Créteil, France.,Unité Court séjour Petits Nourrissons, Service de Néonatologie, Centre Hospitalier Intercommunal de Créteil, Créteil, France
| |
Collapse
|
9
|
Ketharanathan N, Yamamoto Y, Rohlwink UK, Wildschut ED, Mathôt RAA, de Lange ECM, de Wildt SN, Argent AC, Tibboel D, Figaji AA. Combining Brain Microdialysis and Translational Pharmacokinetic Modeling to Predict Drug Concentrations in Pediatric Severe Traumatic Brain Injury: The Next Step Toward Evidence-Based Pharmacotherapy? J Neurotrauma 2018; 36:111-117. [PMID: 30019622 DOI: 10.1089/neu.2017.5588] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Evidence-based analgosedation in severe pediatric traumatic brain injury (pTBI) management is lacking, and improved pharmacological understanding is needed. This starts with increased knowledge of factors controlling the pharmacokinetics (PK) of unbound drug at the target site (brain) and related drug effect(s). This prospective, descriptive study tested a pediatric physiology-based pharmacokinetic software model by comparing actual plasma and brain extracellular fluid (brainECF) morphine concentrations with predicted concentration-time profiles in severe pTBI patients (Glasgow Coma Scale [GCS], ≤8). Plasma and brainECF samples were obtained after legal guardian written consent and were collected from 8 pTBI patients (75% male; median age, 96 months [34.0-155.5]; median weight, 24 kg [14.5-55.0]) with a need for intracranial pressure monitoring (GCS, ≤8) and receiving continuous morphine infusion (10-40 μg/kg/h). BrainECF samples were obtained by microdialysis. BrainECF samples were taken from "injured" and "uninjured" regions as determined by microdialysis catheter location on computed head tomography. A previously developed physiology-based software model to predict morphine concentrations in the brain was adapted to children using pediatric physiological properties. The model predicted plasma morphine concentrations well for individual patients (97% of data points within the 90% prediction interval). In addition, predicted brainECF concentration-time profiles fell within a 90% prediction interval of microdialysis brainECF drug concentrations when sampled from an uninjured area. Prediction was less accurate in injured areas. This approach of translational physiology-based PK modeling allows prediction of morphine concentration-time profiles in uninjured brain of individual patients and opens promising avenues towards evidence-based pharmacotherapies in pTBI.
Collapse
Affiliation(s)
- Naomi Ketharanathan
- 1 Intensive Care and Department of Pediatric Surgery, Erasmus MC, Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Yumi Yamamoto
- 2 Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands
| | - Ursula K Rohlwink
- 3 Division of Neurosurgery and Neuroscience Institute, Red Cross War Memorial Children's Hospital, University of Cape Town, Cape Town, South Africa
| | - Enno D Wildschut
- 1 Intensive Care and Department of Pediatric Surgery, Erasmus MC, Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Ron A A Mathôt
- 4 Department of Pharmacology, Academic Medical Center, Amsterdam, The Netherlands
| | | | - Saskia N de Wildt
- 1 Intensive Care and Department of Pediatric Surgery, Erasmus MC, Sophia Children's Hospital, Rotterdam, The Netherlands.,5 Department of Pharmacology and Toxicology, Radboud University, Nijmegen, The Netherlands
| | - Andrew C Argent
- 6 School of Child and Adolescent Health, University of Cape Town, Paediatric Intensive Care, Red Cross War Memorial Children's Hospital, Cape Town, South Africa
| | - Dick Tibboel
- 1 Intensive Care and Department of Pediatric Surgery, Erasmus MC, Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Anthony A Figaji
- 3 Division of Neurosurgery and Neuroscience Institute, Red Cross War Memorial Children's Hospital, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
10
|
Vermeulen E, van den Anker JN, Della Pasqua O, Hoppu K, van der Lee JH. How to optimise drug study design: pharmacokinetics and pharmacodynamics studies introduced to paediatricians. J Pharm Pharmacol 2017; 69:439-447. [PMID: 27671925 PMCID: PMC6084327 DOI: 10.1111/jphp.12637] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 08/10/2016] [Indexed: 02/06/2023]
Abstract
OBJECTIVES In children, there is often lack of sufficient information concerning the pharmacokinetics (PK) and pharmacodynamics (PD) of a study drug to support dose selection and effective evaluation of efficacy in a randomised clinical trial (RCT). Therefore, one should consider the relevance of relatively small PKPD studies, which can provide the appropriate data to optimise the design of an RCT. METHODS Based on the experience of experts collaborating in the EU-funded Global Research in Paediatrics consortium, we aimed to inform clinician-scientists working with children on the design of investigator-initiated PKPD studies. KEY FINDINGS The importance of the identification of an optimal dose for the paediatric population is explained, followed by the differences and similarities of dose-ranging and efficacy studies. The input of clinical pharmacologists with modelling expertise is essential for an efficient dose-finding study. CONCLUSIONS The emergence of new laboratory techniques and statistical tools allows for the collection and analysis of sparse and unbalanced data, enabling the implementation of (observational) PKPD studies in the paediatric clinic. Understanding of the principles and methods discussed in this study is essential to improve the quality of paediatric PKPD investigations, and to prevent the conduct of paediatric RCTs that fail because of inadequate dosing.
Collapse
Affiliation(s)
- Eric Vermeulen
- Pediatric Clinical Research OfficeEmma Children's HospitalAcademic Medical CenterAmsterdamThe Netherlands
| | - John N. van den Anker
- Division of Pediatric Clinical PharmacologyChildren's National Health SystemWashingtonDCUSA
- Division of Paediatric Pharmacology and PharmacometricsUniversity of Basel Children's HospitalBaselSwitzerland
- Intensive Care and Department of Pediatric SurgeryErasmus Medical CenterSophia Children's HospitalRotterdamThe Netherlands
| | - Oscar Della Pasqua
- Clinical Pharmacology Modelling & SimulationGlaxoSmithKlineStockley ParkUK
- Clinical Pharmacology & TherapeuticsUniversity College LondonLondonUK
| | - Kalle Hoppu
- Poison Information CentreHelsinki University Central HospitalHelsinkiFinland
| | - Johanna H. van der Lee
- Pediatric Clinical Research OfficeEmma Children's HospitalAcademic Medical CenterAmsterdamThe Netherlands
| | | |
Collapse
|