1
|
Ye J, Bi Y, Ting N. How to select the initial dose for a pediatric study? J Biopharm Stat 2023; 33:844-858. [PMID: 36476267 DOI: 10.1080/10543406.2022.2149770] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 11/16/2022] [Indexed: 12/13/2022]
Abstract
In typical clinical development programs, a new drug is first developed for the adult use. Drugs are often approved for adult use or in the process of obtaining approval in adults in the target indication before pediatric development is initiated. In designing the first pediatric clinical trial, one of the challenges is to select the initial dose to be tested. The ICH E11 R1 guidance advises that chronologic age alone may not always be the most appropriate categorical determinant to define developmental subgroups in pediatric studies. In this manuscript, the approaches to utilize available data in adults related to those factors beyond age to inform the starting dose selection in pediatric drug development are discussed. Practical considerations and approaches are provided for informing pediatric starting dose. Additional considerations to use pre-clinical information are provided in the case when adult information is limited or not available.
Collapse
Affiliation(s)
- Jingjing Ye
- Global Statistics and Data Science (GSDS), Fulton, MD, USA
| | - Youwei Bi
- Division of Pharmacometrics, Office of Translational Sciences (OTS), Center for Drug Evaluation and Research (CDER), US Food and Drug Administration (FDA), Silver Spring, MD, USA
| | - Naitee Ting
- Biostatistics and Data Science, Boehringer Ingelheim Pharmaceuticals, Inc, Ridgefield, CT, USA
| |
Collapse
|
2
|
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: 6.0] [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
|
3
|
Bi Y, Liu J, Li F, Yu J, Bhattaram A, Bewernitz M, Li RJ, Ahn J, Earp J, Ma L, Zhuang L, Yang Y, Zhang X, Zhu H, Wang Y. Model-Informed Drug Development in Pediatric Dose Selection. J Clin Pharmacol 2021; 61 Suppl 1:S60-S69. [PMID: 34185906 DOI: 10.1002/jcph.1848] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 02/18/2021] [Indexed: 01/12/2023]
Abstract
Model-informed drug development (MIDD) has been a powerful and efficient tool applied widely in pediatric drug development due to its ability to integrate and leverage existing knowledge from different sources to narrow knowledge gaps. The dose selection is the most common MIDD application in regulatory submission related to pediatric drug development. This article aims to give an overview of the 3 broad categories of use of MIDD in pediatric dose selection: leveraging from adults to pediatric patients, leveraging from animals to pediatric patients, and integrating mechanism in infants and neonates. Population pharmacokinetic analyses with allometric scaling can reasonably predict the clearance in pediatric patients aged >5 years. A mechanistic-based approach, such as physiologically based pharmacokinetic accounting for ontogeny, or an allometric model with age-dependent exponent, can be applied to select the dose in pediatric patients aged ≤2 years. The exposure-response relationship from adults or from other drugs in the same class may be useful in aiding the pediatric dose selection and benefit-risk assessment. Increasing application and understanding of use of MIDD have contributed greatly to several policy developments in the pediatric field. With the increasing efforts of MIDD under the Prescription Drug User Fee Act VI, bigger impacts of MIDD approaches in pediatric dose selection can be expected. Due to the complexity of model-based analyses, early engagement between drug developers and regulatory agencies to discuss MIDD issues is highly encouraged, as it is expected to increase the efficiency and reduce the uncertainty.
Collapse
Affiliation(s)
- Youwei Bi
- Office of Clinical Pharmacology, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Jiang Liu
- Office of Clinical Pharmacology, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Fang Li
- Office of Clinical Pharmacology, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Jingyu Yu
- Office of Clinical Pharmacology, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Atul Bhattaram
- Office of Clinical Pharmacology, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Michael Bewernitz
- Office of Clinical Pharmacology, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Ruo-Jing Li
- Office of Clinical Pharmacology, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Jihye Ahn
- Office of Clinical Pharmacology, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Justin Earp
- Office of Clinical Pharmacology, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Lian Ma
- Office of Clinical Pharmacology, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Luning Zhuang
- Office of Clinical Pharmacology, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Yuching Yang
- Office of Clinical Pharmacology, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Xinyuan Zhang
- Office of Clinical Pharmacology, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Hao Zhu
- Office of Clinical Pharmacology, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Yaning Wang
- Office of Clinical Pharmacology, Food and Drug Administration, Silver Spring, Maryland, USA
| |
Collapse
|
4
|
Parris P, Martin EA, Stanard B, Glowienke S, Dolan DG, Li K, Binazon O, Giddings A, Whelan G, Masuda-Herrera M, Bercu J, Broschard T, Bruen U, Callis CM, Stults CL, Erexson GL, Cruz MT, Nagao LM. Considerations when deriving compound-specific limits for extractables and leachables from pharmaceutical products: Four case studies. Regul Toxicol Pharmacol 2020; 118:104802. [DOI: 10.1016/j.yrtph.2020.104802] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 09/26/2020] [Accepted: 10/06/2020] [Indexed: 12/24/2022]
|
5
|
Ye PP, Zheng Y, Du B, Liu XT, Tang BH, Kan M, Zhou Y, Hao GX, Huang X, Su LQ, Wang WQ, Yu F, Zhao W. First dose in neonates: pharmacokinetic bridging study from juvenile mice to neonates for drugs metabolized by CYP3A. Xenobiotica 2020; 50:1275-1284. [PMID: 32400275 DOI: 10.1080/00498254.2020.1768454] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
First dose prediction is challenging in neonates. Our objective in this proof-of-concept study was to perform a pharmacokinetic (PK) bridging study from juvenile mice to neonates for drugs metabolized by CYP3A. We selected midazolam and clindamycin as model drugs. We developed juvenile mice population PK models using NONMEM. The PK parameters of these two drugs in juvenile mice were used to bridge PK parameters in neonates using different correction methods. The bridging results were evaluated by the fold-error of 0.5- to 1.5-fold. Simple allometry with and without a correction factor for maximum lifespan potential could be used for a bridging of clearance (CL) and volume of distribution (Vd), respectively, from juvenile mice to neonates. Simulation results demonstrated that for midazolam, 100% of clinical studies for which both the predictive CL and Vd were within 0.5- to 1.5-fold of the observed. For clindamycin, 75% and 100% of clinical studies for which the predictive CL and Vd were within 0.5- to 1.5-fold of the observed. A PK bridging of drugs metabolized by CYP3A is feasible from juvenile mice to neonates. It could be a complement to the ADE and PBPK models to support the first dose in neonates.
Collapse
Affiliation(s)
- Pan-Pan Ye
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China.,Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Yi Zheng
- Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Bin Du
- Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xi-Ting Liu
- Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Bo-Hao Tang
- Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Min Kan
- Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yue Zhou
- Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Guo-Xiang Hao
- Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xin Huang
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Le-Qun Su
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Wen-Qi Wang
- Clinical Research Center, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Feng Yu
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Wei Zhao
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University, Jinan, China.,Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China.,Clinical Research Center, The First Affiliated Hospital of Shandong First Medical University, Jinan, China.,Department of Pediatrics, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| |
Collapse
|
6
|
Srinivas NR. Interspecies scaling of excretory amounts using allometry - retrospective analysis with rifapentine, aztreonam, carumonam, pefloxacin, miloxacin, trovafloxacin, doripenem, imipenem, cefozopran, ceftazidime, linezolid for urinary excretion and rifapentine, cabotegravir, and dolutegravir for fecal excretion. Xenobiotica 2016; 46:784-92. [PMID: 26711252 DOI: 10.3109/00498254.2015.1121554] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Revised: 11/11/2015] [Accepted: 11/15/2015] [Indexed: 11/13/2022]
Abstract
1. Interspecies allometry scaling for prediction of human excretory amounts in urine or feces was performed for numerous antibacterials. Antibacterials used for urinary scaling were: rifapentine, pefloxacin, trovafloxacin (Gr1/low; <10%); miloxacin, linezolid, PNU-142300 (Gr2/medium; 10-40%); aztreonam, carumonam, cefozopran, doripenem, imipenem, and ceftazidime (Gr3/high; >50%). Rifapentine, cabotegravir, and dolutegravir was used for fecal scaling (high; >50%). 2. The employment of allometry equation: Y = aW(b) enabled scaling of urine/fecal amounts from animal species. Corresponding predicted amounts were converted into % recovery by considering the respective human dose. Comparison of predicted/observed values enabled fold difference and error calculations (mean absolute error [MAE] and root mean square error [RMSE]). Comparisons were made for urinary/fecal data; and qualitative assessment was made amongst Gr1/Gr2/Gr3 for urine. 3. Average correlation coefficient for the allometry scaling was >0.995. Excretory amount predictions were largely within 0.75- to 1.5-fold differences. Average MAE and RMSE were within ±22% and 23%, respectively. Although robust predictions were achieved for higher urinary/fecal excretion (>50%), interspecies scaling was applicable for low/medium excretory drugs. 4. Based on the data, interspecies scaling of urine or fecal excretory amounts may be potentially used as a tool to understand the significance of either urinary or fecal routes of elimination in humans in early development.
Collapse
Affiliation(s)
- Nuggehally R Srinivas
- a Department of Integrated Drug Development , Suramus Bio , Bangalore , Karnataka , India
| |
Collapse
|
7
|
Prediction of Antimalarial Drug Clearance in Children: A Comparison of Three Different Interspecies Scaling Methods. Eur J Drug Metab Pharmacokinet 2015; 41:767-775. [DOI: 10.1007/s13318-015-0305-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
8
|
Samant TS, Mangal N, Lukacova V, Schmidt S. Quantitative clinical pharmacology for size and age scaling in pediatric drug development: A systematic review. J Clin Pharmacol 2015; 55:1207-17. [DOI: 10.1002/jcph.555] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 05/19/2015] [Indexed: 01/24/2023]
Affiliation(s)
- Tanay S. Samant
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics; College of Pharmacy, University of Florida; Lake Nona (Orlando) FL USA
| | - Naveen Mangal
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics; College of Pharmacy, University of Florida; Lake Nona (Orlando) FL USA
| | | | - Stephan Schmidt
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics; College of Pharmacy, University of Florida; Lake Nona (Orlando) FL USA
| |
Collapse
|
9
|
First dose in neonates: are juvenile mice, adults and in vitro-in silico data predictive of neonatal pharmacokinetics of fluconazole. Clin Pharmacokinet 2015; 53:1005-18. [PMID: 25154507 DOI: 10.1007/s40262-014-0169-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
BACKGROUND AND OBJECTIVES Selection of the first-dose-in-neonates is challenging. The objective of this proof-of-concept study was to evaluate a pharmacokinetic bridging approach to predict a neonatal dosing regimen. METHODS We selected fluconazole as a paradigm compound. We used data from studies in juvenile mice and adults to develop population pharmacokinetic models using NONMEM. We also develop a physiologically-based pharmacokinetic model from in vitro-in silico data using Simcyp. These three models were then used to predict neonatal pharmacokinetics and dosing regimens for fluconazole. RESULTS From juvenile mice to neonates, a correction factor of maximum lifespan potential should be used for extrapolation, while a "renal factor" taking into account renal maturation was required for successful bridging based on adult and in vitro-in silico data. Simulations results demonstrated that the predicted drug exposure based on bridging approach was comparable to the observed value in neonates. The prediction errors were -2.2, +10.1 and -4.6 % for juvenile mice, adults and in vitro-in silico data, respectively. CONCLUSION A model-based bridging approach provided consistent predictions of fluconazole pharmacokinetic parameters in neonates and demonstrated the feasibility of this approach to justify the first-dose-in-neonates, based on all data available from different sources (including physiological informations, preclinical studies and adult data), allowing evidence-based decisions of neonatal dose rather than empiricism.
Collapse
|
10
|
Foissac F, Bouazza N, Valade E, De Sousa Mendes M, Fauchet F, Benaboud S, Hirt D, Tréluyer JM, Urien S. Prediction of drug clearance in children. J Clin Pharmacol 2015; 55:739-47. [DOI: 10.1002/jcph.488] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 02/24/2015] [Indexed: 12/20/2022]
Affiliation(s)
- Frantz Foissac
- EA 08; Université Paris Descartes; Sorbonne Paris Cité France
- Unité de Recherche Clinique; Assistance Publique Hôpitaux de Paris (APHP); Hôpital Tarnier; Paris France
- CIC-1419 Inserm; Cochin-Necker; Paris France
| | - Naïm Bouazza
- EA 08; Université Paris Descartes; Sorbonne Paris Cité France
- Unité de Recherche Clinique; Assistance Publique Hôpitaux de Paris (APHP); Hôpital Tarnier; Paris France
- CIC-1419 Inserm; Cochin-Necker; Paris France
| | - Elodie Valade
- EA 08; Université Paris Descartes; Sorbonne Paris Cité France
- Unité de Recherche Clinique; Assistance Publique Hôpitaux de Paris (APHP); Hôpital Tarnier; Paris France
- CIC-1419 Inserm; Cochin-Necker; Paris France
| | - Mailys De Sousa Mendes
- EA 08; Université Paris Descartes; Sorbonne Paris Cité France
- Unité de Recherche Clinique; Assistance Publique Hôpitaux de Paris (APHP); Hôpital Tarnier; Paris France
- CIC-1419 Inserm; Cochin-Necker; Paris France
| | - Floris Fauchet
- EA 08; Université Paris Descartes; Sorbonne Paris Cité France
- Unité de Recherche Clinique; Assistance Publique Hôpitaux de Paris (APHP); Hôpital Tarnier; Paris France
- CIC-1419 Inserm; Cochin-Necker; Paris France
| | - Sihem Benaboud
- EA 08; Université Paris Descartes; Sorbonne Paris Cité France
- Unité de Recherche Clinique; Assistance Publique Hôpitaux de Paris (APHP); Hôpital Tarnier; Paris France
- CIC-1419 Inserm; Cochin-Necker; Paris France
- Laboratoire de Pharmacologie; Hôpital Cochin; APHP; Paris France
| | - Déborah Hirt
- EA 08; Université Paris Descartes; Sorbonne Paris Cité France
- Unité de Recherche Clinique; Assistance Publique Hôpitaux de Paris (APHP); Hôpital Tarnier; Paris France
- CIC-1419 Inserm; Cochin-Necker; Paris France
- Laboratoire de Pharmacologie; Hôpital Cochin; APHP; Paris France
| | - Jean-Marc Tréluyer
- EA 08; Université Paris Descartes; Sorbonne Paris Cité France
- Unité de Recherche Clinique; Assistance Publique Hôpitaux de Paris (APHP); Hôpital Tarnier; Paris France
- CIC-1419 Inserm; Cochin-Necker; Paris France
- Laboratoire de Pharmacologie; Hôpital Cochin; APHP; Paris France
| | - Saïk Urien
- EA 08; Université Paris Descartes; Sorbonne Paris Cité France
- Unité de Recherche Clinique; Assistance Publique Hôpitaux de Paris (APHP); Hôpital Tarnier; Paris France
- CIC-1419 Inserm; Cochin-Necker; Paris France
| |
Collapse
|
11
|
Mahmood I. Prediction of drug clearance in children: a review of different methodologies. Expert Opin Drug Metab Toxicol 2015; 11:573-87. [DOI: 10.1517/17425255.2015.1019463] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
12
|
Interspecies allometric scaling of antimalarial drugs and potential application to pediatric dosing. Antimicrob Agents Chemother 2014; 58:6068-78. [PMID: 25092696 DOI: 10.1128/aac.02538-14] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Pharmacopeial recommendations for administration of antimalarial drugs are the same weight-based (mg/kg of body weight) doses for children and adults. However, linear calculations are known to underestimate pediatric doses; therefore, interspecies allometric scaling data may have a role in predicting doses in children. We investigated the allometric scaling relationships of antimalarial drugs using data from pharmacokinetic studies in mammalian species. Simple allometry (Y = a × W(b)) was utilized and compared to maximum life span potential (MLP) correction. All drugs showed a strong correlation with clearance (CL) in healthy controls. Insufficient data from malaria-infected species other than humans were available for allometric scaling. The allometric exponents (b) for CL of artesunate, dihydroartemisinin (from intravenous artesunate), artemether, artemisinin, clindamycin, piperaquine, mefloquine, and quinine were 0.71, 0.85, 0.66, 0.83, 0.62, 0.96, 0.52, and 0.40, respectively. Clearance was significantly lower in malaria infection than in healthy (adult) humans for quinine (0.07 versus 0.17 liter/h/kg; P = 0.0002) and dihydroartemisinin (0.81 versus 1.11 liters/h/kg; P = 0.04; power = 0.6). Interpolation of simple allometry provided better estimates of CL for children than MLP correction, which generally underestimated CL values. Pediatric dose calculations based on simple allometric exponents were 10 to 70% higher than pharmacopeial (mg/kg) recommendations. Interpolation of interspecies allometric scaling could provide better estimates than linear scaling of adult to pediatric doses of antimalarial drugs; however, the use of a fixed exponent for CL was not supported in the present study. The variability in allometric exponents for antimalarial drugs also has implications for scaling of fixed-dose combinations.
Collapse
|
13
|
Mahmood I. Dosing in Children: A Critical Review of the Pharmacokinetic Allometric Scaling and Modelling Approaches in Paediatric Drug Development and Clinical Settings. Clin Pharmacokinet 2014; 53:327-46. [DOI: 10.1007/s40262-014-0134-5] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
14
|
Zhao L, Shang EY, Sahajwalla CG. Application of pharmacokinetics-pharmacodynamics/clinical response modeling and simulation for biologics drug development. J Pharm Sci 2012; 101:4367-82. [PMID: 23018763 DOI: 10.1002/jps.23330] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2012] [Revised: 08/27/2012] [Accepted: 09/07/2012] [Indexed: 01/21/2023]
Abstract
Biologics, specifically monoclonal antibody (mAb) drugs, have unique pharmacokinetic (PK) and pharmacodynamic (PD) characteristics as opposed to small molecules. Under the paradigm of model-based drug development, PK-PD/clinical response models offer critical insight in guiding biologics development at various stages. On the basis of the molecular structure and corresponding properties of biologics, typical mechanism-based [target-mediated drug disposition (TMDD)], physiologically based PK, PK-PD, and dose-response meta-analysis models are summarized. Examples of using TMDD, PK-PD, and meta-analysis in helping starting dose determination in first-in-human studies and dosing regimen optimization in phase II/III trials are discussed. Instead of covering the entirety of model-based biologics development, this review focuses on the guiding principles and the core mathematical descriptions underlying the PK or PK-PD models most used.
Collapse
Affiliation(s)
- Liang Zhao
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA.
| | | | | |
Collapse
|
15
|
Journal Watch. Pharmaceut Med 2010. [DOI: 10.1007/bf03256823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|