1
|
Onita T, Ishihara N, Yano T. PK/PD-Guided Strategies for Appropriate Antibiotic Use in the Era of Antimicrobial Resistance. Antibiotics (Basel) 2025; 14:92. [PMID: 39858377 PMCID: PMC11759776 DOI: 10.3390/antibiotics14010092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 01/11/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
Antimicrobial resistance (AMR) poses a critical global health threat, necessitating the optimal use of existing antibiotics. Pharmacokinetic/pharmacodynamic (PK/PD) principles provide a scientific framework for optimizing antimicrobial therapy, particularly to respond to evolving resistance patterns. This review examines PK/PD strategies for antimicrobial dosing optimization, focusing on three key aspects. First, we discuss the importance of drug concentration management for enhancing efficacy while preventing toxicity, considering various patient populations, including pediatric and elderly patients with their unique physiological characteristics. Second, we analyze different PK modeling approaches: the classic top-down approach exemplified by population PK analysis, the bottom-up approach represented by physiologically based PK modeling, and hybrid models combining both approaches for enhanced predictive performance. Third, we explore clinical applications, including nomogram-based dosing strategies, Bayesian estimation, and emerging artificial intelligence applications, for real-time dose optimization. Critical challenges in implementing PK/PD simulation are addressed, particularly the selection of appropriate PK models, the optimization of PK/PD indices, and considerations concerning antimicrobial concentrations at infection sites. Understanding these principles and challenges is crucial for optimizing antimicrobial therapy and combating AMR through improved dosing strategies.
Collapse
Affiliation(s)
| | | | - Takahisa Yano
- Department of Pharmacy, Shimane University Hospital, 89-1 Enya, Izumo 693-8501, Shimane, Japan
| |
Collapse
|
2
|
Huang H, Zhao W, Qin N, Duan X. Recent Progress on Physiologically Based Pharmacokinetic (PBPK) Model: A Review Based on Bibliometrics. TOXICS 2024; 12:433. [PMID: 38922113 PMCID: PMC11209072 DOI: 10.3390/toxics12060433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 06/04/2024] [Accepted: 06/13/2024] [Indexed: 06/27/2024]
Abstract
Physiologically based pharmacokinetic/toxicokinetic (PBPK/PBTK) models are designed to elucidate the mechanism of chemical compound action in organisms based on the physiological, biochemical, anatomical, and thermodynamic properties of organisms. After nearly a century of research and practice, good results have been achieved in the fields of medicine, environmental science, and ecology. However, there is currently a lack of a more systematic review of progress in the main research directions of PBPK models, especially a more comprehensive understanding of the application in aquatic environmental research. In this review, a total of 3974 articles related to PBPK models from 1996 to 24 March 2024 were collected. Then, the main research areas of the PBPK model were categorized based on the keyword co-occurrence maps and cluster maps obtained by CiteSpace. The results showed that research related to medicine is the main application area of PBPK. Four major research directions included in the medical field were "drug assessment", "cross-species prediction", "drug-drug interactions", and "pediatrics and pregnancy drug development", in which "drug assessment" accounted for 55% of the total publication volume. In addition, bibliometric analyses indicated a rapid growth trend in the application in the field of environmental research, especially in predicting the residual levels in organisms and revealing the relationship between internal and external exposure. Despite facing the limitation of insufficient species-specific parameters, the PBPK model is still an effective tool for improving the understanding of chemical-biological effectiveness and will provide a theoretical basis for accurately assessing potential risks to ecosystems and human health. The combination with the quantitative structure-activity relationship model, Bayesian method, and machine learning technology are potential solutions to the previous research gaps.
Collapse
Affiliation(s)
| | | | - Ning Qin
- School of Energy and Environmental Engineering, University of Science and Technology Beijing, Beijing 100083, China; (H.H.); (W.Z.)
| | - Xiaoli Duan
- School of Energy and Environmental Engineering, University of Science and Technology Beijing, Beijing 100083, China; (H.H.); (W.Z.)
| |
Collapse
|
3
|
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
|
4
|
Litjens CHC, Verscheijden LFM, Svensson EM, van den Broek PHH, van Hove H, Koenderink JB, Russel FGM, Aarnoutse RE, te Brake LHM. Physiologically-Based Pharmacokinetic Modelling to Predict the Pharmacokinetics and Pharmacodynamics of Linezolid in Adults and Children with Tuberculous Meningitis. Antibiotics (Basel) 2023; 12:antibiotics12040702. [PMID: 37107064 PMCID: PMC10135070 DOI: 10.3390/antibiotics12040702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 03/23/2023] [Accepted: 03/30/2023] [Indexed: 04/07/2023] Open
Abstract
Linezolid is used off-label for treatment of central nervous system infections. However, its pharmacokinetics and target attainment in cranial cerebrospinal fluid (CSF) in tuberculous meningitis patients is unknown. This study aimed to predict linezolid cranial CSF concentrations and assess attainment of pharmacodynamic (PD) thresholds (AUC:MIC of >119) in plasma and cranial CSF of adults and children with tuberculous meningitis. A physiologically based pharmacokinetic (PBPK) model was developed to predict linezolid cranial CSF profiles based on reported plasma concentrations. Simulated steady-state PK curves in plasma and cranial CSF after linezolid doses of 300 mg BID, 600 mg BID, and 1200 mg QD in adults resulted in geometric mean AUC:MIC ratios in plasma of 118, 281, and 262 and mean cranial CSF AUC:MIC ratios of 74, 181, and 166, respectively. In children using ~10 mg/kg BID linezolid, AUC:MIC values at steady-state in plasma and cranial CSF were 202 and 135, respectively. Our model predicts that 1200 mg per day in adults, either 600 mg BID or 1200 mg QD, results in reasonable (87%) target attainment in cranial CSF. Target attainment in our simulated paediatric population was moderate (56% in cranial CSF). Our PBPK model can support linezolid dose optimization efforts by simulating target attainment close to the site of TBM disease.
Collapse
Affiliation(s)
- Carlijn H. C. Litjens
- Department of Pharmacy, Radboud Institute for Health Sciences, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands
| | - Laurens F. M. Verscheijden
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands
| | - Elin M. Svensson
- Department of Pharmacy, Radboud Institute for Health Sciences, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands
- Department of Pharmacy, Uppsala University, 75123 Uppsala, Sweden
| | - Petra H. H. van den Broek
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands
| | - Hedwig van Hove
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands
| | - Jan B. Koenderink
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands
| | - Frans G. M. Russel
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands
| | - Rob E. Aarnoutse
- Department of Pharmacy, Radboud Institute for Health Sciences, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands
| | - Lindsey H. M. te Brake
- Department of Pharmacy, Radboud Institute for Health Sciences, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands
| |
Collapse
|
5
|
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: 0.5] [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
|
6
|
Humphries H, Almond L, Berg A, Gardner I, Hatley O, Pan X, Small B, Zhang M, Jamei M, Romero K. Development of physiologically-based pharmacokinetic models for standard of care and newer tuberculosis drugs. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2021; 10:1382-1395. [PMID: 34623770 PMCID: PMC8592506 DOI: 10.1002/psp4.12707] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 08/12/2021] [Accepted: 08/22/2021] [Indexed: 12/19/2022]
Abstract
Tuberculosis (TB) remains a global health problem and there is an ongoing effort to develop more effective therapies and new combination regimes that can reduce duration of treatment. The purpose of this study was to demonstrate utility of a physiologically‐based pharmacokinetic modeling approach to predict plasma and lung concentrations of 11 compounds used or under development as TB therapies (bedaquiline [and N‐desmethyl bedaquiline], clofazimine, cycloserine, ethambutol, ethionamide, isoniazid, kanamycin, linezolid, pyrazinamide, rifampicin, and rifapentine). Model accuracy was assessed by comparison of simulated plasma pharmacokinetic parameters with healthy volunteer data for compounds administered alone or in combination. Eighty‐four percent (area under the curve [AUC]) and 91% (maximum concentration [Cmax]) of simulated mean values were within 1.5‐fold of the observed data and the simulated drug‐drug interaction ratios were within 1.5‐fold (AUC) and twofold (Cmax) of the observed data for nine (AUC) and eight (Cmax) of the 10 cases. Following satisfactory recovery of plasma concentrations in healthy volunteers, model accuracy was assessed further (where patients’ with TB data were available) by comparing clinical data with simulated lung concentrations (9 compounds) and simulated lung: plasma concentration ratios (7 compounds). The 5th–95th percentiles for the simulated lung concentration data recovered between 13% (isoniazid and pyrazinamide) and 88% (pyrazinamide) of the observed data points (Am J Respir Crit Care Med, 198, 2018, 1208; Nat Med, 21, 2015, 1223; PLoS Med, 16, 2019, e1002773). The impact of uncertain model parameters, such as the fraction of drug unbound in lung tissue mass (fumass), is discussed. Additionally, the variability associated with the patient lung concentration data, which was sparse and included extensive within‐subject, interlaboratory, and experimental variability (as well interindividual variability) is reviewed. All presented models are transparently documented and are available as open‐source to aid further research.
Collapse
Affiliation(s)
| | - Lisa Almond
- Certara UK Limited, Simcyp Division, Sheffield, UK
| | | | - Iain Gardner
- Certara UK Limited, Simcyp Division, Sheffield, UK
| | | | - Xian Pan
- Certara UK Limited, Simcyp Division, Sheffield, UK
| | - Ben Small
- Certara UK Limited, Simcyp Division, Sheffield, UK
| | - Mian Zhang
- Certara UK Limited, Simcyp Division, Sheffield, UK
| | - Masoud Jamei
- Certara UK Limited, Simcyp Division, Sheffield, UK
| | | |
Collapse
|
7
|
Wang K, Jiang K, Wei X, Li Y, Wang T, Song Y. Physiologically Based Pharmacokinetic Models Are Effective Support for Pediatric Drug Development. AAPS PharmSciTech 2021; 22:208. [PMID: 34312742 PMCID: PMC8312709 DOI: 10.1208/s12249-021-02076-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 06/16/2021] [Indexed: 12/30/2022] Open
Abstract
Pediatric drug development faces many difficulties. Traditionally, pediatric drug doses are simply calculated linearly based on the body weight, age, and body surface area of adults. Due to the ontogeny of children, this simple linear scaling may lead to drug overdose in pediatric patients. The physiologically based pharmacokinetic (PBPK) model, as a mathematical model, contributes to the research and development of pediatric drugs. An example of a PBPK model guiding drug dose selection in pediatrics has emerged and has been approved by the relevant regulatory agencies. In this review, we discuss the principle of the PBPK model, emphasize the necessity of establishing a pediatric PBPK model, introduce the absorption, distribution, metabolism, and excretion of the pediatric PBPK model, and understand the various applications and related prospects of the pediatric PBPK model.
Collapse
|
8
|
Chen F, Liu H, Wang B, Yang Z, Chen Y, Yang L, Wang B, Jiao Z, Lin HS, Quan Y, Wang H, Xiang X. Evaluation of the Impacts of Formulation Parameters on the Pharmacokinetics and Bioequivalence of Risperidone Orodispersible Film: a Physiologically Based Pharmacokinetic Modeling Approach. AAPS PharmSciTech 2020; 21:245. [PMID: 32856178 DOI: 10.1208/s12249-020-01728-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 06/08/2020] [Indexed: 12/27/2022] Open
Abstract
The purpose of this study was to investigate the impacts of the formulation parameters on the pharmacokinetics and bioequivalence of risperidone orodispersible film (ODF) using physiologically based pharmacokinetic model. The pharmacokinetic profiles of two risperidone ODFs, which exhibit different in vitro dissolution, were examined in Beagle dogs after supralingual administration. Subsequently, a physiologically based pharmacokinetic (PBPK) model was constructed to evaluate the in vivo performance of risperidone ODF. The parameter sensitivity analysis (PSA) was used to access the impacts of formulation parameters on the pharmacokinetics of risperidone. Moreover, the validated PBPK model was applied to predict human pharmacokinetic profiles and examine the bioequivalence of these two ODFs. These two ODFs displayed similar risperidone pharmacokinetic profiles in dogs. The parameter sensitivity analysis indicated that the changes in the solubility, particle size, particle density, and diffusion coefficient did not have obvious influence on the in vivo properties of risperidone ODF. Alternation of the in vitro complete dissolution time in water from 15 to 30 min led to a 30% decrease in Cmax and 20% of increase in Tmax. AUC0-∞ would be decreased if risperidone was not fully released within 1 h. As both ODFs completely released risperidone within 15 min, the difference in the extent of in vivo absorption, intestinal regional absorption location, and plasma concentration-time curves between these two ODFs was almost negligible. Consequently, a bioequivalence was foreseen in humans. The in vitro cumulative dissolution percentage in water at 15 min was found to be the major determinant on the in vivo properties of risperidone ODF. PBPK modeling appears to be an innovative strategy to guide the development of risperidone ODF.
Collapse
|
9
|
Physiologically-based pharmacokinetic models for children: Starting to reach maturation? Pharmacol Ther 2020; 211:107541. [DOI: 10.1016/j.pharmthera.2020.107541] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 03/19/2020] [Indexed: 12/13/2022]
|
10
|
Physiologically-Based Pharmacokinetic (PBPK) Modeling of Buprenorphine in Adults, Children and Preterm Neonates. Pharmaceutics 2020; 12:pharmaceutics12060578. [PMID: 32585880 PMCID: PMC7355427 DOI: 10.3390/pharmaceutics12060578] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 06/18/2020] [Accepted: 06/21/2020] [Indexed: 12/15/2022] Open
Abstract
Buprenorphine plays a crucial role in the therapeutic management of pain in adults, adolescents and pediatric subpopulations. However, only few pharmacokinetic studies of buprenorphine in children, particularly neonates, are available as conducting clinical trials in this population is especially challenging. Physiologically-based pharmacokinetic (PBPK) modeling allows the prediction of drug exposure in pediatrics based on age-related physiological differences. The aim of this study was to predict the pharmacokinetics of buprenorphine in pediatrics with PBPK modeling. Moreover, the drug-drug interaction (DDI) potential of buprenorphine with CYP3A4 and P-glycoprotein perpetrator drugs should be elucidated. A PBPK model of buprenorphine and norbuprenorphine in adults has been developed and scaled to children and preterm neonates, accounting for age-related changes. One-hundred-percent of the predicted AUClast values in adults (geometric mean fold error (GMFE): 1.22), 90% of individual AUClast predictions in children (GMFE: 1.54) and 75% in preterm neonates (GMFE: 1.57) met the 2-fold acceptance criterion. Moreover, the adult model was used to simulate DDI scenarios with clarithromycin, itraconazole and rifampicin. We demonstrate the applicability of scaling adult PBPK models to pediatrics for the prediction of individual plasma profiles. The novel PBPK models could be helpful to further investigate buprenorphine pharmacokinetics in various populations, particularly pediatric subgroups.
Collapse
|
11
|
Tan YM, Chan M, Chukwudebe A, Domoradzki J, Fisher J, Hack CE, Hinderliter P, Hirasawa K, Leonard J, Lumen A, Paini A, Qian H, Ruiz P, Wambaugh J, Zhang F, Embry M. PBPK model reporting template for chemical risk assessment applications. Regul Toxicol Pharmacol 2020; 115:104691. [PMID: 32502513 PMCID: PMC8188465 DOI: 10.1016/j.yrtph.2020.104691] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 05/18/2020] [Accepted: 05/28/2020] [Indexed: 12/04/2022]
Abstract
Physiologically-based pharmacokinetic (PBPK) modeling analysis does not stand on its own for regulatory purposes but is a robust tool to support drug/chemical safety assessment. While the development of PBPK models have grown steadily since their emergence, only a handful of models have been accepted to support regulatory purposes due to obstacles such as the lack of a standardized template for reporting PBPK analysis. Here, we expand the existing guidances designed for pharmaceutical applications by recommending additional elements that are relevant to environmental chemicals. This harmonized reporting template can be adopted and customized by public health agencies receiving PBPK model submission, and it can also serve as general guidance for submitting PBPK-related studies for publication in journals or other modeling sharing purposes. The current effort represents one of several ongoing collaborations among the PBPK modeling and risk assessment communities to promote, when appropriate, incorporating PBPK modeling to characterize the influence of pharmacokinetics on safety decisions made by regulatory agencies.
Collapse
Affiliation(s)
- Yu-Mei Tan
- U.S. Environmental Protection Agency, Office of Pesticide Programs, Health Effects Division, 109 TW Alexander Dr, Research Triangle Park, NC, 27709, USA.
| | - Melissa Chan
- Corteva Agriscience, Haskell R&D Center, 1090 Elkton Road, Newark, DE, 19714, USA.
| | - Amechi Chukwudebe
- BASF Corporation, 26 Davis Drive, Research Triangle Park, NC, 27709, USA.
| | - Jeanne Domoradzki
- Corteva Agriscience, Haskell R&D Center, 1090 Elkton Road, Newark, DE, 19714, USA
| | - Jeffrey Fisher
- National Center for Toxicological Research, US Food and Drug Administration, 3900 NCTR Rd, Jefferson, AR, 72079, USA.
| | - C Eric Hack
- ScitoVation, 100 Capitola Drive, Durham, NC, 27713, USA.
| | - Paul Hinderliter
- Syngenta Crop Protection, LLC, 410 Swing Rd, Greensboro, NC, 27409, USA.
| | - Kota Hirasawa
- Sumitomo Chemical Co, Ltd, 1-98, Kasugadenaka 3-chome, Konohana-ku, Osaka, 554-8558, Japan.
| | - Jeremy Leonard
- Oak Ridge Institute for Science and Education, 100 ORAU Way, Oak Ridge, TN, 37830, USA.
| | - Annie Lumen
- National Center for Toxicological Research, US Food and Drug Administration, 3900 NCTR Rd, Jefferson, AR, 72079, USA.
| | - Alicia Paini
- European Commission Joint Research Centre, Via E. Fermi 2749, Ispra I, 21027, Italy.
| | - Hua Qian
- ExxonMobil Biomedical Sciences, Inc, 1545 US Hwy 22 East, Annandale, NJ, 08801, USA.
| | - Patricia Ruiz
- CDC-ATSDR, 4770 Buford Hwy, Mailstop S102-1, Chamblee, GA, 3034, USA.
| | - John Wambaugh
- US Environmental Protection Agency, Center for Computational Toxicology and Exposure, 109 TW Alexander Dr, Research Triangle Park, NC, 27711, USA.
| | - Fagen Zhang
- The Dow Chemical Company, 1803 Building, Midland, MI, 48674, USA.
| | - Michelle Embry
- Health and Environmental Sciences Institute, 740 15th Street, NW, Suite 600, Washington, DC, 20005, USA.
| |
Collapse
|
12
|
Li SC, Ye Q, Xu H, Zhang L, Wang Y. Population Pharmacokinetics and Dosing Optimization of Linezolid in Pediatric Patients. Antimicrob Agents Chemother 2019; 63:e02387-18. [PMID: 30642929 PMCID: PMC6437496 DOI: 10.1128/aac.02387-18] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 01/08/2019] [Indexed: 01/26/2023] Open
Abstract
Linezolid is a synthetic antibiotic very effective in the treatment of infections caused by Gram-positive pathogens. Although the clinical application of linezolid in children has increased progressively, data on linezolid pharmacokinetics in pediatric patients are very limited. The aim of this study was to develop a population pharmacokinetic model for linezolid in children and optimize the dosing strategy in order to improve therapeutic efficacy. We performed a prospective pharmacokinetic study of pediatric patients aged 0 to 12 years. The population pharmacokinetic model was developed using the NONMEM program. Goodness-of-fit plots, nonparametric bootstrap analysis, normalized prediction distribution errors, and a visual predictive check were employed to evaluate the final model. The dosing regimen was optimized based on the final model. The pharmacokinetic data from 112 pediatric patients ages 0.03 to 11.9 years were analyzed. The pharmacokinetics could best be described by a one-compartment model with first-order elimination along with body weight and the estimated glomerular filtration rate as significant covariates. Simulations demonstrated that the currently approved dosage of 10 mg/kg of body weight every 8 h (q8h) would lead to a high risk of underdosing for children in the presence of bacteria with MICs of ≥2 mg/liter. To reach the pharmacokinetic target, an elevated dosage of 15 or 20 mg/kg q8h may be required for them. The population pharmacokinetics of linezolid were characterized in pediatric patients, and simulations provide an evidence-based approach for linezolid dosage individualization.
Collapse
Affiliation(s)
- Si-Chan Li
- Department of Clinical Pharmacy, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Qi Ye
- Department of Clinical Pharmacy, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Hua Xu
- Department of Clinical Pharmacy, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Long Zhang
- Department of Intensive Care Unit, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Yang Wang
- Department of Clinical Pharmacy, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| |
Collapse
|
13
|
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 2019; 47:296-313. [PMID: 30567878 DOI: 10.1124/dmd.118.084418] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 12/14/2018] [Indexed: 02/13/2025] Open
Abstract
A thorough knowledge of the newborn (age, birth to 1 month postpartum) infant's gastrointestinal tract (GIT) is critical to the evaluation of the absorption, distribution, metabolism, and excretion (ADME) of orally administered drugs in this population. Developmental changes in the GIT during the newborn period are important for nutrient uptake as well as the disposition of orally administered medications. Some aspects of gastrointestinal function do not mature until driven by increased dietary complexity and nutritional demands later in the postnatal period. The functionalities present at birth, and subsequent maturation, can also impact the ADME parameters of orally administered compounds. This review will examine some specific contributors to the ADME processes in human neonates, as well as what is currently understood about the drivers for their maturation. Key species differences will be highlighted, with a focus on laboratory animals used in juvenile toxicity studies. Because of the gaps and inconsistencies in our knowledge, we will also highlight areas where additional study is warranted to better inform the appropriate use of medicines specifically intended for neonates.
Collapse
Affiliation(s)
- April Neal-Kluever
- US Food and Drug Administration, Center for Food Safety and Applied Nutrition, College Park, Maryland (A.N.-K.); US Food and Drug Administration, National Center for Toxicological Research, Jefferson, Arkansas (J.F.); Independent Consultant, Vienna, Virginia (L.G.); and Genentech Inc., South San Francisco, California (S.K.-K., W.H.)
| | - Jeffrey Fisher
- US Food and Drug Administration, Center for Food Safety and Applied Nutrition, College Park, Maryland (A.N.-K.); US Food and Drug Administration, National Center for Toxicological Research, Jefferson, Arkansas (J.F.); Independent Consultant, Vienna, Virginia (L.G.); and Genentech Inc., South San Francisco, California (S.K.-K., W.H.)
| | - Lawrence Grylack
- US Food and Drug Administration, Center for Food Safety and Applied Nutrition, College Park, Maryland (A.N.-K.); US Food and Drug Administration, National Center for Toxicological Research, Jefferson, Arkansas (J.F.); Independent Consultant, Vienna, Virginia (L.G.); and Genentech Inc., South San Francisco, California (S.K.-K., W.H.)
| | - Satoko Kakiuchi-Kiyota
- US Food and Drug Administration, Center for Food Safety and Applied Nutrition, College Park, Maryland (A.N.-K.); US Food and Drug Administration, National Center for Toxicological Research, Jefferson, Arkansas (J.F.); Independent Consultant, Vienna, Virginia (L.G.); and Genentech Inc., South San Francisco, California (S.K.-K., W.H.)
| | - Wendy Halpern
- US Food and Drug Administration, Center for Food Safety and Applied Nutrition, College Park, Maryland (A.N.-K.); US Food and Drug Administration, National Center for Toxicological Research, Jefferson, Arkansas (J.F.); Independent Consultant, Vienna, Virginia (L.G.); and Genentech Inc., South San Francisco, California (S.K.-K., W.H.)
| |
Collapse
|
14
|
Smits A, De Cock P, Vermeulen A, Allegaert K. Physiologically based pharmacokinetic (PBPK) modeling and simulation in neonatal drug development: how clinicians can contribute. Expert Opin Drug Metab Toxicol 2018; 15:25-34. [PMID: 30554542 DOI: 10.1080/17425255.2019.1558205] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Introduction: Legal initiatives to stimulate neonatal drug development should be accompanied by development of valid research tools. Physiologically based (PB)-pharmacokinetic (PK) modeling and simulation are established tools, accepted by regulatory authorities. Consequently, PBPK holds promise to be a strong research tool to support neonatal drug development. Area covered: The currently available PBPK models still have poor predictive performance in neonates. Using an illustrative approach on distinct PK processes of absorption, distribution, metabolism, excretion, and real-world data in neonates, we provide evidence on the need to further refine available PBPK system parameters through generation and integration of new knowledge. This necessitates cross talk between clinicians and modelers to integrate knowledge (PK datasets, system knowledge, maturational physiology) or test and refine PBPK models. Expert opinion: Besides refining these models for 'small molecules', PBPK model development should also be more widely applied for therapeutic proteins and to determine exposure through breastfeeding. Researchers should also be aware that PBPK modeling in combination with clinical observations can also be used to elucidate age-related changes that are almost impossible to study based on in vivo or in vitro data. This approach has been explored for hepatic biliary excretion, renal tubular activity, and central nervous system exposure.
Collapse
Affiliation(s)
- Anne Smits
- a Neonatal Intensive Care Unit , University Hospitals Leuven , Leuven , Belgium.,b Department of Development and Regeneration , KU Leuven , Leuven , Belgium
| | - Pieter De Cock
- c Department of Pharmacy , Ghent University Hospital , Ghent , Belgium.,d Heymans Institute of Pharmacology , Ghent University , Ghent , Belgium.,e Department of Pediatric Intensive Care , Ghent University , Ghent , Belgium
| | - An Vermeulen
- f Laboratory of Medical Biochemistry and Clinical Analysis, Faculty of Pharmaceutical Sciences , Ghent University , Ghent , Belgium
| | - Karel Allegaert
- b Department of Development and Regeneration , KU Leuven , Leuven , Belgium.,g Department of Pediatrics, Division of Neonatology , Erasmus MC Sophia Children's Hospital, University Medical Center Rotterdam , Rotterdam , The Netherlands
| |
Collapse
|
15
|
Duan P, Wu F, Moore JN, Fisher J, Crentsil V, Gonzalez D, Zhang L, Burckart GJ, Wang J. Assessing CYP2C19 Ontogeny in Neonates and Infants Using Physiologically Based Pharmacokinetic Models: Impact of Enzyme Maturation Versus Inhibition. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2018; 8:158-166. [PMID: 30520273 PMCID: PMC6430158 DOI: 10.1002/psp4.12350] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 08/13/2018] [Indexed: 12/18/2022]
Abstract
The objective of this study was to develop pediatric physiologically based pharmacokinetic (PBPK) models for pantoprazole and esomeprazole. Pediatric PBPK models were developed by Simcyp version 15 by incorporating cytochrome P450 (CYP)2C19 maturation and auto-inhibition. The predicted-to-observed pantoprazole clearance (CL) ratio ranged from 0.96-1.35 in children 1-17 years of age and 0.43-0.70 in term infants. The predicted-to-observed esomeprazole CL ratio ranged from 1.08-1.50 for children 6-17 years of age, and 0.15-0.33 for infants. The prediction was markedly improved by assuming no auto-inhibition of esomeprazole in infants in the PBPK model. Our results suggested that the CYP2C19 auto-inhibition model was appropriate for esomeprazole in adults and older children but could not be directly extended to infants. A better understanding of the complex interplay of enzyme maturation, inhibition, and compensatory mechanisms for CYP2C19 is necessary for PBPK modeling in infants.
Collapse
Affiliation(s)
- Peng Duan
- Office of New Drug Product, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Fang Wu
- Office of New Drug Product, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Jason N Moore
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Jeffrey Fisher
- Division of Biochemical Toxicology, National Center for Toxicological Research, Jefferson, Arkansas, USA
| | - Victor Crentsil
- Office of Drug Evaluation III, Office of New Drugs, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Daniel Gonzalez
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Lei Zhang
- Office of Research and Standards, Office of Generic Drugs, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Gilbert J Burckart
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Jian Wang
- Office of Drug Evaluation IV, Office of New Drugs, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| |
Collapse
|
16
|
Mahmood I, Tegenge MA. A Comparative Study Between Allometric Scaling and Physiologically Based Pharmacokinetic Modeling for the Prediction of Drug Clearance From Neonates to Adolescents. J Clin Pharmacol 2018; 59:189-197. [DOI: 10.1002/jcph.1310] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 08/09/2018] [Indexed: 12/20/2022]
Affiliation(s)
- Iftekhar Mahmood
- Office of Tissue & Advanced Therapies; Center for Biologics Evaluation and Research; Food & Drug Administration; Silver Spring MD USA
| | - Million A. Tegenge
- Office of Biostatistics & Epidemiology; Center for Biologics Evaluation and Research; Food & Drug Administration; Silver Spring MD USA
| |
Collapse
|