1
|
Allegaert K, Mitra S, Smits A, Turner MA. Advocating for drug development in newborn infants. Early Hum Dev 2024; 199:106136. [PMID: 39488008 DOI: 10.1016/j.earlhumdev.2024.106136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 10/25/2024] [Accepted: 10/25/2024] [Indexed: 11/04/2024]
Abstract
Neonatal care needs more robust guidance on pharmacotherapy, (formulation, dosage regimen, safety and efficacy information). This requires structured advocacy. We therefore discuss advocacy related to improving information about medicines including current practices, clinical trials, the current setting, and trial preparedness. This steps can improve neonatal drug development by generating evidence, particularly if a programmatic approach (identify dosing, eligibility criteria, and outcomes) to evidence generation is followed. Trial design should be guided by the intended use of the medicine and the benefits/risks that the study participant is exposed to. Regulatory trials (explanatory, controlled environment, internal validity, endpoints reflect clinically important outcomes, strong causal evidence) are sometimes necessary. However, some research questions are best addressed with informative trials. In either case, trial design can be supported by real world data and evidence, extrapolation from other subpopulations, or physiologically-based pharmacokinetic modeling. Data management, safety reporting, and management of drugs should be specified and proportionate. Trial design and conduct also necessitate awareness of Good Clinical Practice specific to neonates. Relevant aspects include protocol and trial design, research skills and interactions with Ethics Committees or Institutional Research Boards, capacities and competences needed within the research team, and aspects related to consent and recruitment.
Collapse
Affiliation(s)
- Karel Allegaert
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium; Department of Development and Regeneration, KU Leuven, Leuven, Belgium; Department of Hospital Pharmacy, Erasmus Medical Center, 3015GD Rotterdam, the Netherlands.
| | - Souvik Mitra
- Division of Neonatology, Department of Pediatrics, University of British Columbia, Vancouver, Canada
| | - Anne Smits
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium; Neonatal Intensive Care Unit, University Hospitals Leuven, Leuven, Belgium
| | - Mark A Turner
- Institute of Lifecourse and Medical Sciences, University of Liverpool, Liverpool Health Partners, Liverpool, United Kingdom; conect4children Stichting, Utrecht, the Netherlands
| |
Collapse
|
2
|
Ahmed MA, Burnham J, Dwivedi G, AbuAsal B. Achieving big with small: quantitative clinical pharmacology tools for drug development in pediatric rare diseases. J Pharmacokinet Pharmacodyn 2023; 50:429-444. [PMID: 37140724 DOI: 10.1007/s10928-023-09863-x] [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: 03/04/2023] [Accepted: 04/26/2023] [Indexed: 05/05/2023]
Abstract
Pediatric populations represent a major fraction of rare diseases and compound the intrinsic challenges of pediatric drug development and drug development for rare diseases. The intertwined complexities of pediatric and rare disease populations impose unique challenges to clinical pharmacologists and require integration of novel clinical pharmacology and quantitative tools to overcome multiple hurdles during the discovery and development of new therapies. Drug development strategies for pediatric rare diseases continue to evolve to meet the inherent challenges and produce new medicines. Advances in quantitative clinical pharmacology research have been a key component in advancing pediatric rare disease research to accelerate drug development and inform regulatory decisions. This article will discuss the evolution of the regulatory landscape in pediatric rare diseases, the challenges encountered during the design of rare disease drug development programs and will highlight the use of innovative tools and potential solutions for future development programs.
Collapse
Affiliation(s)
- Mariam A Ahmed
- Takeda Development Center Americas Inc, 125 Binney St, Cambridge, MA, 02142-1123, USA.
| | | | - Gaurav Dwivedi
- Takeda Development Center Americas Inc, 125 Binney St, Cambridge, MA, 02142-1123, USA
| | - Bilal AbuAsal
- US Food and Drug Administration, 10903, New Hampshire Ave, Silver Spring, MD, 20993, USA
| |
Collapse
|
3
|
Domingues C, Jarak I, Veiga F, Dourado M, Figueiras A. Pediatric Drug Development: Reviewing Challenges and Opportunities by Tracking Innovative Therapies. Pharmaceutics 2023; 15:2431. [PMID: 37896191 PMCID: PMC10610377 DOI: 10.3390/pharmaceutics15102431] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/16/2023] [Accepted: 09/25/2023] [Indexed: 10/29/2023] Open
Abstract
The paradigm of pediatric drug development has been evolving in a "carrot-and-stick"-based tactic to address population-specific issues. However, the off-label prescription of adult medicines to pediatric patients remains a feature of clinical practice, which may compromise the age-appropriate evaluation of treatments. Therefore, the United States and the European Pediatric Formulation Initiative have recommended applying nanotechnology-based delivery systems to tackle some of these challenges, particularly applying inorganic, polymeric, and lipid-based nanoparticles. Connected with these, advanced therapy medicinal products (ATMPs) have also been highlighted, with optimistic perspectives for the pediatric population. Despite the results achieved using these innovative therapies, a workforce that congregates pediatric patients and/or caregivers, healthcare stakeholders, drug developers, and physicians continues to be of utmost relevance to promote standardized guidelines for pediatric drug development, enabling a fast lab-to-clinical translation. Therefore, taking into consideration the significance of this topic, this work aims to compile the current landscape of pediatric drug development by (1) outlining the historic regulatory panorama, (2) summarizing the challenges in the development of pediatric drug formulation, and (3) delineating the advantages/disadvantages of using innovative approaches, such as nanomedicines and ATMPs in pediatrics. Moreover, some attention will be given to the role of pharmaceutical technologists and developers in conceiving pediatric medicines.
Collapse
Affiliation(s)
- Cátia Domingues
- Univ Coimbra, Laboratory of Drug Development and Technologies, Faculty of Pharmacy, 3000-548 Coimbra, Portugal; (C.D.); (I.J.); (F.V.)
- LAQV-REQUIMTE, Laboratory of Drug Development and Technologies, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
- Univ Coimbra, Institute for Clinical and Biomedical Research (iCBR) Area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, 3000-548 Coimbra, Portugal;
| | - Ivana Jarak
- Univ Coimbra, Laboratory of Drug Development and Technologies, Faculty of Pharmacy, 3000-548 Coimbra, Portugal; (C.D.); (I.J.); (F.V.)
- Institute for Health Research and Innovation (i3s), University of Porto, 4200-135 Porto, Portugal
| | - Francisco Veiga
- Univ Coimbra, Laboratory of Drug Development and Technologies, Faculty of Pharmacy, 3000-548 Coimbra, Portugal; (C.D.); (I.J.); (F.V.)
- LAQV-REQUIMTE, Laboratory of Drug Development and Technologies, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Marília Dourado
- Univ Coimbra, Institute for Clinical and Biomedical Research (iCBR) Area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, 3000-548 Coimbra, Portugal;
- Univ Coimbra, Center for Health Studies and Research of the University of Coimbra (CEISUC), Faculty of Medicine, 3000-548 Coimbra, Portugal
- Univ Coimbra, Center for Studies and Development of Continuous and Palliative Care (CEDCCP), Faculty of Medicine, 3000-548 Coimbra, Portugal
| | - Ana Figueiras
- Univ Coimbra, Laboratory of Drug Development and Technologies, Faculty of Pharmacy, 3000-548 Coimbra, Portugal; (C.D.); (I.J.); (F.V.)
- LAQV-REQUIMTE, Laboratory of Drug Development and Technologies, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| |
Collapse
|
4
|
Johnson TN, Abduljalil K, Pan X, Emoto C. Development and Verification of a Japanese Pediatric Physiologically Based Pharmacokinetic Model with Emphasis on Drugs Eliminated by Cytochrome P450 or Renal Excretion. J Clin Pharmacol 2023; 63:1156-1168. [PMID: 37496106 DOI: 10.1002/jcph.2317] [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: 05/02/2023] [Accepted: 07/23/2023] [Indexed: 07/28/2023]
Abstract
Physiologically based pharmacokinetic (PBPK) models are useful in bridging drug exposure in different ethnic groups, and there is increasing regulatory application of this approach in adults. Reported pediatric PBPK models tend to focus on the North European population, with few examples in other ethnic groups. This study describes the development and verification of a Japanese pediatric PBPK population. The development of the model was based on the existing North European pediatric population. Japanese systems and clinical data were collated from public databases and the literature, and the underlying demographics and equations were optimized so that physiological outputs represented the Japanese pediatric population. The model was tested using 14 different small molecule drugs, eliminated by a variety of pathways, including cytochrome P450 3A4 (CYP3A4) metabolism and renal excretion. Given the limitations of the clinical data, the overall performance of the model was good, with 44/62 predictions for PK parameters (area under the plasma drug concentration-time curve, AUC; maximum serum concentration, Cmax ; clearance, CL) being within 0.8- to 1.25-fold, 56/62 within 0.67- to 1.5-fold, and 61/62 within 0.5- to 2.0-fold of the observed values. Specific results for the 5 CYP3A4 substrates showed 20/31 cases were predicted within 0.8- to 1.25-fold, 27/31 within 0.67- to 1.5-fold, and all were within 0.5- to 2.0-fold of the observed values. Given the increased regulatory use of pediatric PBPK in drug development, expanding these models to other ethnic groups are important. Considering qualifying these models based on the context of use, there is a need to expand on the current research to include a larger range of drugs with different elimination pathways. Collaboration among academic, industry, model providers, and regulators will facilitate further development.
Collapse
Affiliation(s)
| | | | - Xian Pan
- Simcyp Division, Certara UK Limited, Sheffield, UK
| | - Chie Emoto
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Tokyo, Japan
- Translational Research Division, Chugai Pharmaceutical Co., Ltd, Tokyo, Japan
| |
Collapse
|
5
|
Smeets NJL, Raaijmakers LPM, van der Zanden TM, Male C, de Wildt SN. Guiding future paediatric drug studies based on existing pharmacokinetic and efficacy data: Cardiovascular drugs as a proof of concept. Br J Clin Pharmacol 2023; 89:2888-2901. [PMID: 37160686 DOI: 10.1111/bcp.15781] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 03/29/2023] [Accepted: 04/13/2023] [Indexed: 05/11/2023] Open
Abstract
INTRODUCTION Off-label drug use in the paediatric population is common, and the lack of high-quality efficacy studies poses patients at risk for failing pharmacotherapy. Next to efficacy studies, pharmacokinetic (PK) studies are increasingly used to inform paediatric dose selection. As resources for paediatric trials are limited, we aimed to summarize existing PK and efficacy studies to identify knowledge gaps in available evidence supporting paediatric dosing recommendations, thereby taking paediatric cardiovascular drugs as proof of concept. METHODS For each cardiovascular drug, paediatric indication and prespecified age group, together comprising one record, the authorized state was assessed. Next, for off-label records, the highest level of evidence was scored. High-quality efficacy studies were defined as meta-analysis or randomized controlled trials. Other comparative research, noncomparative research or consensus-based expert opinions were considered low quality. The level of evidence for PK studies was scored per drug and per age group, but regardless of indication. RESULTS A total of 58 drugs included 417 records, of which 279 (67%) were off-label. Of all off-label records, the majority (81%) were not supported by high-quality efficacy studies, but for 140 of these records (62%) high-quality PK studies were available. CONCLUSION We demonstrated that for the majority of off-label cardiovascular drugs, only low-quality efficacy studies were available. However, high-quality PK studies were frequently available. Combining these PK data with extrapolation of efficacy data from adults may help to close the current information gap and prioritize the drugs for which clinical studies and safety data are urgently needed.
Collapse
Affiliation(s)
- Nori J L Smeets
- Department of Pharmacology and Toxicology, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Intensive Care and Department of Paediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Lieke P M Raaijmakers
- Department of Pharmacology and Toxicology, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Tjitske M van der Zanden
- Department of Pharmacology and Toxicology, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Intensive Care and Department of Paediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
- Dutch Knowledge Center Pharmacotherapy for Children, The Hague, The Netherlands
| | - Christoph Male
- Department of Paediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Saskia N de Wildt
- Department of Pharmacology and Toxicology, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Intensive Care and Department of Paediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
- Dutch Knowledge Center Pharmacotherapy for Children, The Hague, The Netherlands
| |
Collapse
|
6
|
Bansal S, Ladumor MK, Paine MF, Unadkat JD. A Physiologically-Based Pharmacokinetic Model for Cannabidiol in Healthy Adults, Hepatically-Impaired Adults, and Children. Drug Metab Dispos 2023; 51:743-752. [PMID: 36972999 PMCID: PMC10197200 DOI: 10.1124/dmd.122.001128] [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: 09/21/2022] [Revised: 03/08/2023] [Accepted: 03/10/2023] [Indexed: 03/29/2023] Open
Abstract
Cannabidiol (CBD) is available as a prescription oral drug that is indicated for the treatment of some types of epilepsy in children and adults. CBD is also available over-the-counter and is used to self-treat a variety of other ailments, including pain, anxiety, and insomnia. Accordingly, CBD may be consumed with other medications, resulting in possible CBD-drug interactions. Such interactions can be predicted in healthy and hepatically-impaired (HI) adults and in children through physiologically based pharmacokinetic (PBPK) modeling and simulation. These PBPK models must be populated with CBD-specific parameters, including the enzymes that metabolize CBD in adults. In vitro reaction phenotyping experiments showed that UDP-glucuronosyltransferases (UGTs, 80%), particularly UGT2B7 (64%), were the major contributors to CBD metabolism in adult human liver microsomes. Among the cytochrome P450s (CYPs) tested, CYP2C19 (5.7%) and CYP3A (6.5%) were the major CYPs responsible for CBD metabolism. Using these and other physicochemical parameters, a CBD PBPK model was developed and validated for healthy adults. This model was then extended to predict CBD systemic exposure in HI adults and children. Our PBPK model successfully predicted CBD systemic exposure in both populations within 0.5- to 2-fold of the observed values. In conclusion, we developed and validated a PBPK model to predict CBD systemic exposure in healthy and HI adults and children. This model can be used to predict CBD-drug or CBD-drug-disease interactions in these populations. SIGNIFICANCE STATEMENT: Our PBPK model successfully predicted CBD systemic exposure in healthy and hepatically-impaired adults, as well as children with epilepsy. This model could be used in the future to predict CBD-drug or CBD-drug-disease interactions in these special populations.
Collapse
Affiliation(s)
- Sumit Bansal
- Department of Pharmaceutics, University of Washington, Seattle, Washington (S.B., M.K.L., J.D.U.); Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington (M.F.P.); and Center of Excellence for Natural Product Drug Interaction Research (M.F.P., J.D.U.)
| | - Mayur K Ladumor
- Department of Pharmaceutics, University of Washington, Seattle, Washington (S.B., M.K.L., J.D.U.); Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington (M.F.P.); and Center of Excellence for Natural Product Drug Interaction Research (M.F.P., J.D.U.)
| | - Mary F Paine
- Department of Pharmaceutics, University of Washington, Seattle, Washington (S.B., M.K.L., J.D.U.); Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington (M.F.P.); and Center of Excellence for Natural Product Drug Interaction Research (M.F.P., J.D.U.)
| | - Jashvant D Unadkat
- Department of Pharmaceutics, University of Washington, Seattle, Washington (S.B., M.K.L., J.D.U.); Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington (M.F.P.); and Center of Excellence for Natural Product Drug Interaction Research (M.F.P., J.D.U.)
| |
Collapse
|
7
|
Small BG, Johnson TN, Rowland Yeo K. Another Step Toward Qualification of Pediatric Physiologically Based Pharmacokinetic Models to Facilitate Inclusivity and Diversity in Pediatric Clinical Studies. Clin Pharmacol Ther 2023; 113:735-745. [PMID: 36306419 DOI: 10.1002/cpt.2777] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 10/23/2022] [Indexed: 11/06/2022]
Abstract
Robust prediction of pharmacokinetics (PKs) in pediatric subjects of diverse ages, ethnicities, and morbidities is critical. Qualification of pediatric physiologically-based pharmacokinetic (P-PBPK) models is an essential step toward enabling precision dosing of these vulnerable groups. Twenty-two manuscripts involving P-PBPK predictions and corresponding observed PK data (e.g., area under the curve and clearance) for 22 small-molecule compounds metabolized by CYP (3A4, 1A2, and 2C9), UGT (1A9 and 2B7), FMO3, renal, non-renal, and complex routes were identified; ratios of mean predicted/observed (P/O) PK parameters were calculated. Seventy-eight of 115 mean predicted PK parameters were within 0.8 to 1.25-fold of observed data, 98 within 0.67 to 1.5-fold, 109 within 2-fold, and only 6 P/O ratios were outside of these bounds. A set of 12 CYP3A4-metabolized compounds and a set of 6 metabolized by other enzymes, CYP1A2 (1 compound), CYP2C9 (2 compounds), UGT1A9 (1 compound) and UGT2B7 (2 compounds) had 56 of 59 and 22 of 25 mean P/O ratios, respectively, that fell within the > 0.5 and < 2.0-fold boundaries. For compounds covering renal, non-renal, complex, and FM03 routes of elimination, 29 of 31 mean P/O ratios fell within the 0.67 to 1.5-fold bounds, including 4 of 5 P/O ratios from newborns. P-PBPK modeling and simulation is a strategic component of the complement of precision dosing methods and has a vital role to play in dose adjustment in vulnerable pediatric populations, such as those with disease or in different ethnic groups. Qualification of such models is an essential step toward acceptance of this methodology by regulators.
Collapse
Affiliation(s)
- Ben G Small
- Certara UK Limited (Simcyp Division), Sheffield, UK
| | | | | |
Collapse
|
8
|
Freriksen JJM, van der Heijden JEM, de Hoop-Sommen MA, Greupink R, de Wildt SN. Physiologically Based Pharmacokinetic (PBPK) Model-Informed Dosing Guidelines for Pediatric Clinical Care: A Pragmatic Approach for a Special Population. Paediatr Drugs 2023; 25:5-11. [PMID: 36201128 PMCID: PMC9534738 DOI: 10.1007/s40272-022-00535-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/05/2022] [Indexed: 01/06/2023]
Abstract
Physiologically based pharmacokinetic (PBPK) modeling can be an attractive tool to increase the evidence base of pediatric drug dosing recommendations by making optimal use of existing pharmacokinetic (PK) data. A pragmatic approach of combining available compound models with a virtual pediatric physiology model can be a rational solution to predict PK and hence support dosing guidelines for children in real-life clinical care, when it can also be employed by individuals with little experience in PBPK modeling. This comes within reach as user-friendly PBPK modeling platforms exist and, for many drugs and populations, models are ready for use. We have identified a list of drugs that can serve as a starting point for pragmatic PBPK modeling to address current clinical dosing needs.
Collapse
Affiliation(s)
- Jolien J M Freriksen
- Department of Pharmacology and Toxicology, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - Joyce E M van der Heijden
- Department of Pharmacology and Toxicology, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Marika A de Hoop-Sommen
- Department of Pharmacology and Toxicology, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Rick Greupink
- Department of Pharmacology and Toxicology, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Saskia N de Wildt
- Department of Pharmacology and Toxicology, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Intensive Care and Department of Pediatrics Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
| |
Collapse
|
9
|
Catozzi S, Hill R, Li X, Dulong S, Collard E, Ballesta A. Interspecies and in vitro-in vivo scaling for quantitative modeling of whole-body drug pharmacokinetics in patients: Application to the anticancer drug oxaliplatin. CPT Pharmacometrics Syst Pharmacol 2022; 12:221-235. [PMID: 36537068 PMCID: PMC9931436 DOI: 10.1002/psp4.12895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 11/02/2022] [Accepted: 11/05/2022] [Indexed: 12/24/2022] Open
Abstract
Quantitative systems pharmacology holds the promises of integrating results from laboratory animals or in vitro human systems into the design of human pharmacokinetic/pharmacodynamic (PK/PD) models allowing for precision and personalized medicine. However, reliable and general in vitro-to-in vivo extrapolation and interspecies scaling methods are still lacking. Here, we developed a translational strategy for the anticancer drug oxaliplatin. Using ex vivo PK data in the whole blood of the mouse, rat, and human, a model representing the amount of platinum (Pt) in the plasma and in the red blood cells was designed and could faithfully fit each dataset independently. A "purely physiologically-based (PB)" scaling approach solely based on preclinical data failed to reproduce human observations, which were then included in the calibration. Investigating approaches in which one parameter was set as species-specific, whereas the others were computed by PB scaling laws, we concluded that allowing the Pt binding rate to plasma proteins to be species-specific permitted to closely fit all data, and guaranteed parameter identifiability. Such a strategy presenting the drawback of including all clinical datasets, we further identified a minimal subset of human data ensuring accurate model calibration. Next, a "whole body" model of oxaliplatin human PK was inferred from the ex vivo study. Its three remaining parameters were estimated, using one third of the available patient data. Remarkably, the model achieved a good fit to the training dataset and successfully reproduced the unseen observations. Such validation endorsed the legitimacy of our scaling methodology calling for its testing with other drugs.
Collapse
Affiliation(s)
- Simona Catozzi
- Institut Curie, Inserm U900, MINES ParisTech, CBIO ‐ Centre for Computational BiologyPSL Research UniversitySaint‐CloudFrance
| | - Roger Hill
- EPSRC and MRC Centre for Doctoral Training in Mathematics for Real‐World SystemsUniversity of WarwickCoventryUK
| | - Xiao‐Mei Li
- UPR “Chronotherapy, Cancers and Transplantation,” Faculty of MedicineUniversité Paris‐SaclayVillejuifFrance
| | - Sandrine Dulong
- Institut Curie, Inserm U900, MINES ParisTech, CBIO ‐ Centre for Computational BiologyPSL Research UniversitySaint‐CloudFrance,UPR “Chronotherapy, Cancers and Transplantation,” Faculty of MedicineUniversité Paris‐SaclayVillejuifFrance
| | - Elodie Collard
- CEA, CNRS, NIMBEUniversité Paris‐SaclayGif‐sur‐YvetteFrance
| | - Annabelle Ballesta
- Institut Curie, Inserm U900, MINES ParisTech, CBIO ‐ Centre for Computational BiologyPSL Research UniversitySaint‐CloudFrance
| |
Collapse
|
10
|
Leeder JS, Gaedigk A, Wright KJ, Staggs VS, Soden SE, Lin YS, Pearce RE. A longitudinal study of cytochrome P450 2D6 (CYP2D6) activity during adolescence. Clin Transl Sci 2022; 15:2514-2527. [PMID: 35997001 PMCID: PMC9579386 DOI: 10.1111/cts.13380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 07/07/2022] [Accepted: 07/09/2022] [Indexed: 01/25/2023] Open
Abstract
CYP2D6 substrates are among the most highly prescribed medications in teenagers and also commonly associated with serious adverse events. To investigate the relative contributions of genetic variation, growth, and development on CYP2D6 activity during puberty, healthy children and adolescents 7-15 years of age at enrollment participated in a longitudinal phenotyping study involving administration of 0.3 mg/kg dextromethorphan (DM) and 4-h urine collection every 6 months for 3 years (7 total visits). At each visit, height, weight, and sexual maturity were recorded, and CYP2D6 activity was determined as the urinary molar ratio of DM to its metabolite dextrorphan (DX). A total of 188 participants completed at least one visit, and 102 completed all seven study visits. Following univariate analysis, only CYP2D6 activity score (p < 0.001), urinary pH (p < 0.001), weight (p = 0.018), and attention-deficit/hyperactivity disorder (ADHD) diagnosis (p < 0.001) were significantly correlated with log(DM/DX). Results of linear mixed model analysis with random intercept, random slope covariance structure revealed that CYP2D6 activity score had the strongest effect on log(DM/DX), with model-estimated average log(DM/DX) being 3.8 SDs higher for poor metabolizers than for patients with activity score 3. A moderate effect on log(DM/DX) was observed for sex, and smaller effects were observed for ADHD diagnosis and urinary pH. The log(DM/DX) did not change meaningfully with age or pubertal development. CYP2D6 genotype remains the single, largest determinant of variability in CYP2D6 activity during puberty. Incorporation of genotype-based dosing guidelines should be considered for CYP2D6 substrates given the prevalent use of these agents in this pediatric age group.
Collapse
Affiliation(s)
- J. Steven Leeder
- Division of Clinical Pharmacology, Toxicology and Therapeutic Innovation, Department of PediatricsChildren's Mercy Kansas CityKansas CityMissouriUSA,School of MedicineUniversity of Missouri‐Kansas CityKansas CityMissouriUSA
| | - Andrea Gaedigk
- Division of Clinical Pharmacology, Toxicology and Therapeutic Innovation, Department of PediatricsChildren's Mercy Kansas CityKansas CityMissouriUSA,School of MedicineUniversity of Missouri‐Kansas CityKansas CityMissouriUSA
| | - Krista J. Wright
- Division of Clinical Pharmacology, Toxicology and Therapeutic Innovation, Department of PediatricsChildren's Mercy Kansas CityKansas CityMissouriUSA
| | - Vincent S. Staggs
- School of MedicineUniversity of Missouri‐Kansas CityKansas CityMissouriUSA,Biostatistics & Epidemiology Core, Division of Health Services and Outcomes Research, Department of PediatricsChildren's Mercy Kansas CityKansas CityMissouriUSA,Division of Developmental and Behavioral Sciences, Department of PediatricsChildren's Mercy Kansas CityKansas CityMissouriUSA
| | - Sarah E. Soden
- Division of Clinical Pharmacology, Toxicology and Therapeutic Innovation, Department of PediatricsChildren's Mercy Kansas CityKansas CityMissouriUSA,School of MedicineUniversity of Missouri‐Kansas CityKansas CityMissouriUSA
| | - Yvonne S. Lin
- Department of PharmaceuticsUniversity of WashingtonSeattleWashingtonUSA
| | - Robin E. Pearce
- Division of Clinical Pharmacology, Toxicology and Therapeutic Innovation, Department of PediatricsChildren's Mercy Kansas CityKansas CityMissouriUSA,School of MedicineUniversity of Missouri‐Kansas CityKansas CityMissouriUSA
| |
Collapse
|
11
|
Allegaert K, Abbasi MY, Annaert P, Olafuyi O. Current and future physiologically based pharmacokinetic (PBPK) modeling approaches to optimize pharmacotherapy in preterm neonates. Expert Opin Drug Metab Toxicol 2022; 18:301-312. [PMID: 35796504 DOI: 10.1080/17425255.2022.2099836] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION There is a need for structured approaches to inform on pharmacotherapy in preterm neonates. With their proven track record up to regulatory acceptance, physiologically based pharmacokinetic (PBPK) modeling and simulation provide such a structured approach, and hold the promise to support drug development in preterm neonates. AREAS COVERED Compared to the general and pediatric use of PBPK modeling, its use to inform pharmacotherapy in preterms is limited. Using a systematic search (PBPK + preterm), we retained 25 records (20 research papers, 2 letters, 3 abstracts). We subsequently collated the published information on PBPK software packages (PK-Sim®, Simcyp®), and their applications and optimization efforts in preterm neonates. It is encouraging that these applications cover a broad range of scenarios (pharmacokinetic-dynamic analyses, drug-drug interactions, developmental pharmacogenetics, lactation related exposure) and compounds (small molecules, proteins). Furthermore, specific compartments (cerebrospinal fluid, tissue) or (patho)physiologic processes (cardiac output, biliary excretion, first pass metabolism) are considered. EXPERT OPINION Knowledge gaps exist, giving rise to various levels of model uncertainty in PBPK applications in preterm neonates. To improve this setting, we need cross talk between clinicians and modelers to generate and integrate knowledge (PK datasets, system knowledge, maturational physiology and pathophysiology) to further refine PBPK models.
Collapse
Affiliation(s)
- Karel Allegaert
- Department of Pharmaceutical and Pharmacological Sciences.,Department of Development and Regeneration, and.,Leuven Child and Youth Institute, KU Leuven, Leuven Belgium.,Department of Clinical Pharmacy, Erasmus MC, Rotterdam, the Netherlands
| | - Mohammad Yaseen Abbasi
- Division of Clinical Pharmacology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Pieter Annaert
- Department of Pharmaceutical and Pharmacological Sciences
| | - Olusola Olafuyi
- School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| |
Collapse
|