1
|
Bihorel S, Dingman R, Mendell J, Norman KC, George RT, Zhao XQ, Pordy R, Garcia D, Putnam WS, Raghuveer G, McCrindle BW, Fornari E, Baric I, Srinivasan S, Diamond M, Brinton EA, Davis JD, DiCioccio AT, Harnisch L. Comparison of Model-Predicted and Observed Evinacumab Pharmacokinetics and Efficacy in Children Aged < 5 Years With Homozygous Familial Hypercholesterolemia. CPT Pharmacometrics Syst Pharmacol 2025. [PMID: 40095766 DOI: 10.1002/psp4.70017] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Accepted: 02/24/2025] [Indexed: 03/19/2025] Open
Abstract
Evinacumab, an angiopoietin-like 3 inhibitor, significantly reduces low-density lipoprotein cholesterol (LDL-C) in patients with homozygous familial hypercholesterolemia (HoFH). Herein, we report pharmacokinetic and efficacy analyses of evinacumab in < 5-year-old patients with HoFH. Population pharmacometric models characterizing evinacumab exposure and LDL-C response accounting for lipoprotein apheresis effect in ≥ 5-year-old patients were adapted for growth and maturation to predict and compare evinacumab and LDL-C concentrations across age/weight groups in virtual ≥ 6-month-old patients receiving 15 mg/kg evinacumab intravenous (iv) infusions every 4 weeks (q4w). As expected from allometric theory, weight-based dosing resulted in decreasing evinacumab exposures with declining body weight. Consistent with trends observed in > 5-year-old patients, the predicted percent change from LDL-C baseline (%∆LDL-C) was generally comparable or even higher in < 5-year-old patients (63.0%-68.5%) than in 5- to < 18-year-old patients (61.3%-67.8%) or adults (51.7%), with the predicted percentages of patients achieving %∆LDL-C > 50% also higher in < 5-year-old patients (82.0%-86.9%) versus 5- to < 18-year-old patients (72.0%-84.5%) and adults (54.8%). Through a managed access program, six 1- to < 5-year-old patients received between 5 and 23 iv infusions of 15 mg/kg evinacumab q4w. Rapid and clinically meaningful LDL-C reductions were observed, with %∆LDL-C at the last reported dose ranging from 41.3% to 77.3%. Based on the actual patient dosing and plasmapheresis history, model-predicted evinacumab and LDL-C concentrations were comparable to the observed data collected in the managed access program. Overall, this analysis provides evidence for the use of evinacumab 15 mg/kg iv q4w dosing regimen in 6-month-old to 5-year-old patients.
Collapse
Affiliation(s)
| | - Robert Dingman
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, USA
| | - Jeanne Mendell
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, USA
| | - Katy C Norman
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, USA
| | | | - Xue-Qiao Zhao
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, USA
| | - Robert Pordy
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, USA
| | - Daniel Garcia
- Ultragenyx Pharmaceutical, Inc., Novato, California, USA
| | - Wendy S Putnam
- Ultragenyx Pharmaceutical, Inc., Novato, California, USA
| | - Geetha Raghuveer
- University of Missouri-Kansas City School of Medicine, Children's Mercy Hospital, Kansas City, Missouri, USA
| | - Brian W McCrindle
- Department of Pediatrics, University of Toronto, Labatt Family Heart Centre, the Hospital for Sick Children, Toronto, Canada
| | - Elena Fornari
- Department of Surgery, Dentistry, Pediatrics and Gynecology, Section of Pediatric Diabetes and Metabolism, University of Verona, Verona, Italy
| | - Ivo Baric
- Department of Paediatrics, University Hospital Centre Zagreb and University of Zagreb, School of Medicine, Zagreb, Croatia
| | - Shubha Srinivasan
- Institute of Endocrinology and Diabetes, Children's Hospital at Westmead, Sydney, Australia
| | - Melissa Diamond
- Joe DiMaggio Children's Hospital, Memorial Division of Pediatric Cardiology, Hollywood, Florida, USA
| | | | - John D Davis
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, USA
| | | | - Lutz Harnisch
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, USA
| |
Collapse
|
2
|
Clegg L, Freshwater E, Leach A, Villafana T, Hamrén UW. Population Pharmacokinetics of Nirsevimab in Preterm and Term Infants. J Clin Pharmacol 2024; 64:555-567. [PMID: 38294353 DOI: 10.1002/jcph.2401] [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: 10/19/2023] [Accepted: 12/29/2023] [Indexed: 02/01/2024]
Abstract
Nirsevimab, a monoclonal antibody with an extended half-life, is approved for the prevention of respiratory syncytial virus (RSV) disease in all infants in Canada, the EU, Great Britain, and the USA. A population pharmacokinetics (PK) model was built to describe the PK of nirsevimab in preterm and term infants, and to evaluate the influence of covariates, including body weight and age, in infants. Nirsevimab PK was characterized by a 2-compartment model with first-order clearance (CL) and first-order absorption following intramuscular (IM) administration. The typical CL in a 5 kg infant was 3.4 mL/day. Body weight and postmenstrual age were the primary covariates on CL, with minor effects for race, second RSV season, and antidrug antibody status (deemed not clinically relevant). Congenital heart disease (CHD) and chronic lung disease (CLD) did not significantly impact nirsevimab PK. The final population PK model, based on 8987 PK observations from 2683 participants across 5 clinical trials, successfully predicted PK in an additional cohort of 967 healthy infants. Weight-banded dosing (50 mg in infants <5 kg; 100 mg in infants ≥5 kg) was predicted to be appropriate for infants ≥1 kg in their first RSV season. Together, these data support weight-banded dosing of nirsevimab in all infants in their first RSV season, including in healthy infants, infants with CHD or CLD, and in infants born prematurely.
Collapse
Affiliation(s)
- Lindsay Clegg
- Clinical Pharmacology and Quantitative Pharmacology, R&D, AstraZeneca, Gaithersburg, MD, USA
| | | | - Amanda Leach
- Clinical Development, Vaccines and Immune Therapies, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Tonya Villafana
- Clinical Development, Vaccines and Immune Therapies, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Ulrika Wählby Hamrén
- Clinical Pharmacology and Quantitative Pharmacology, R&D, AstraZeneca, Gothenburg, Sweden
| |
Collapse
|
3
|
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: 1.0] [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
|
4
|
Faraj A, Le Moan N, Gorina E, Blouse GE, Knudsen T, Simonsson USH. Model-Informed Support of Dose Selection for Prophylactic Treatment with Dalcinonacog Alfa in Adult and Paediatric Hemophilia B Patients. Adv Ther 2023; 40:3739-3750. [PMID: 37341915 PMCID: PMC10427527 DOI: 10.1007/s12325-023-02570-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 05/26/2023] [Indexed: 06/22/2023]
Abstract
INTRODUCTION Dalcinonacog alfa (DalcA), a novel subcutaneously administered recombinant human factor IX (FIX) variant is being developed for adult and paediatric patients with hemophilia B (HB). DalcA has been shown to raise FIX to clinically meaningful levels in adults with HB. This work aimed to support dosing regimen selection in adults and perform first-in-paediatric dose extrapolations using a model-based pharmacokinetic (PK) approach. METHODS A population PK model was built using adult data from two clinical trials (NCT03186677, NCT03995784). With allometry in the model, clinical trial simulations were performed to study alternative dosing regimens in adults and children. Steady-state trough levels and the time-to-reach target were derived to inform dose selection. RESULTS Almost 90% of the adults were predicted to achieve desirable FIX levels, i.e. 10% FIX activity, following daily 100 IU/kg dosing, with 90% of the subjects reaching target within 1.6-7.1 days. No every-other-day regimen met the target. A dose of 125 IU/kg resulted in adequate FIX levels down to 6 years, whereas a 150 IU/kg dose was needed below 6 down to 2 years of age. For subjects down to 6 years that did not reach target with 125 IU/kg, a dose escalation to 150 IU/kg was appropriate. The children below 6 to 2 years were shown to need a dose escalation to 200 IU/kg if 150 IU/kg given daily was insufficient. CONCLUSION This study supported the adult dose selection for DalcA in the presence of sparse data and enabled first-in-paediatric dose selection to achieve FIX levels that reduce risk of spontaneous bleeds.
Collapse
Affiliation(s)
- Alan Faraj
- Department of Pharmaceutical Biosciences, BMC, Uppsala University, Box 591, 751 24, Uppsala, Sweden
| | | | | | | | - Tom Knudsen
- Catalyst Biosciences, South San Francisco, CA, USA
| | - Ulrika S H Simonsson
- Department of Pharmaceutical Biosciences, BMC, Uppsala University, Box 591, 751 24, Uppsala, Sweden.
| |
Collapse
|
5
|
Challenger JD, van Beek SW, ter Heine R, van der Boor SC, Charles GD, Smit MJ, Ockenhouse C, Aponte JJ, McCall MBB, Jore MM, Churcher TS, Bousema T. Modeling the Impact of a Highly Potent Plasmodium falciparum Transmission-Blocking Monoclonal Antibody in Areas of Seasonal Malaria Transmission. J Infect Dis 2023; 228:212-223. [PMID: 37042518 PMCID: PMC10345482 DOI: 10.1093/infdis/jiad101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 02/06/2023] [Accepted: 04/11/2023] [Indexed: 04/13/2023] Open
Abstract
Transmission-blocking interventions can play an important role in combating malaria worldwide. Recently, a highly potent Plasmodium falciparum transmission-blocking monoclonal antibody (TB31F) was demonstrated to be safe and efficacious in malaria-naive volunteers. Here we predict the potential public health impact of large-scale implementation of TB31F alongside existing interventions. We developed a pharmaco-epidemiological model, tailored to 2 settings of differing transmission intensity with already established insecticide-treated nets and seasonal malaria chemoprevention interventions. Community-wide annual administration (at 80% coverage) of TB31F over a 3-year period was predicted to reduce clinical incidence by 54% (381 cases averted per 1000 people per year) in a high-transmission seasonal setting, and 74% (157 cases averted per 1000 people per year) in a low-transmission seasonal setting. Targeting school-aged children gave the largest reduction in terms of cases averted per dose. An annual administration of the transmission-blocking monoclonal antibody TB31F may be an effective intervention against malaria in seasonal malaria settings.
Collapse
Affiliation(s)
- Joseph D Challenger
- Medical Research Council Centre for Global Infections Disease Analysis, Department of Infectious Disease Epidemiology, Imperial College London, London, United Kingdom
| | | | - Rob ter Heine
- Department of Pharmacy, Radboud Institute for Health Sciences
| | - Saskia C van der Boor
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Giovanni D Charles
- Medical Research Council Centre for Global Infections Disease Analysis, Department of Infectious Disease Epidemiology, Imperial College London, London, United Kingdom
| | - Merel J Smit
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Chris Ockenhouse
- PATH Center for Vaccine Innovation and Access, Washington, District of Columbia, USA
| | - John J Aponte
- PATH Center for Vaccine Innovation and Access, Geneva, Switzerland
| | - Matthew B B McCall
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Matthijs M Jore
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Thomas S Churcher
- Medical Research Council Centre for Global Infections Disease Analysis, Department of Infectious Disease Epidemiology, Imperial College London, London, United Kingdom
| | - Teun Bousema
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
6
|
Faraj A, van Wijk RC, Neuman L, Desai S, Blouse GE, Knudsen T, Simonsson USH. Model-informed pediatric dose selection of marzeptacog alfa (activated): An exposure matching strategy. CPT Pharmacometrics Syst Pharmacol 2023; 12:977-987. [PMID: 37042339 PMCID: PMC10349190 DOI: 10.1002/psp4.12967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 02/13/2023] [Accepted: 03/20/2023] [Indexed: 04/13/2023] Open
Abstract
Marzeptacog alfa (activated) (MarzAA) is an activated recombinant human rFVII variant intended for subcutaneous (s.c.) administration to treat or prevent bleeding in individuals with hemophilia A (HA) or B (HB) with inhibitors, and other rare bleeding disorders. The s.c. administration provides benefits over i.v. injections. The objective of the study was to support the first-in-pediatric dose selection for s.c. MarzAA to treat episodic bleeding episodes in children up through 11 years in a registrational phase III trial. Assuming the same exposure-response relationship as in adults, an exposure matching strategy was used with a population pharmacokinetics model. A sensitivity analysis evaluating the impact of doubling in absorption rate and age-dependent allometric exponents on dose selection was performed. Subsequently, the probability of trial success, defined as the number of successful trials for a given pediatric dose divided by the number of simulated trials (n = 1000) was studied. A successful trial was defined as outcome where four, three, or two out of 24 pediatric subjects per trial were allowed to fall outside the adult exposures after s.c. administration of 60 μg/kg. A dose of 60 μg/kg in children with HA/HB was supported by the clinical trial simulations to match exposures in adults. The sensitivity analyses further supported selection of the 60 μg/kg dose level in all age groups. Moreover, the probability of trial success evaluations given a plausible design confirmed the potential of a 60 μg/kg dose level. Taken together, this work demonstrates the utility of model-informed drug development and could be helpful for other pediatric development programs for rare diseases.
Collapse
Affiliation(s)
- Alan Faraj
- Department of Pharmaceutical BiosciencesUppsala UniversityUppsalaSweden
| | - Rob C. van Wijk
- Department of Pharmaceutical BiosciencesUppsala UniversityUppsalaSweden
| | - Linda Neuman
- Catalyst BiosciencesSouth San FranciscoCaliforniaUSA
| | | | | | - Tom Knudsen
- Catalyst BiosciencesSouth San FranciscoCaliforniaUSA
| | | |
Collapse
|
7
|
Mahmood I. A Simple Method for the Prediction of Therapeutic Proteins (Monoclonal and Polyclonal Antibodies and Non-Antibody Proteins) for First-in-Pediatric Dose Selection: Application of Salisbury Rule. Antibodies (Basel) 2022; 11:antib11040066. [PMID: 36278619 PMCID: PMC9590058 DOI: 10.3390/antib11040066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 10/18/2022] [Accepted: 10/19/2022] [Indexed: 11/29/2022] Open
Abstract
In order to conduct a pediatric clinical trial, it is important to optimize pediatric dose as accurately as possible. In this study, a simple weight-based method known as ‘Salisbury Rule’ was used to predict pediatric dose for therapeutic proteins and was then compared with the observed pediatric dose. The observed dose was obtained mainly from the FDA package insert and if dosing information was not available from the FDA package insert then the observed dose was based on the dose given to an age group in a particular study. It was noted that the recommended doses of most of the therapeutic proteins were extrapolated to pediatrics from adult dose based on per kilogram (kg) body weight basis. Since it is widely believed that pediatric dose should be selected based on the pediatric clearance (CL), a CL based pediatric dose was projected from the following equation: Dose in children = Adult dose × (Observed CL in children/Observed adult CL). In this study, this dose was also considered observed pediatric dose for comparison. A ±30% prediction error (predicted vs. observed) was considered acceptable. There were 21 monoclonal antibodies, 5 polyclonal antibodies in children ≥ 2 years of age, 4 polyclonal antibodies in preterm and term neonates, and 11 therapeutic proteins (non-antibodies) in the study. In children < 30 kg body weight, the predicted doses were within 0.5−1.5-fold prediction error for 87% (monoclonal antibody), 100% (polyclonal antibody), and 92% (non-antibodies) observations. In children > 30 kg body weight, the predicted doses were within 0.5−1.5-fold prediction error for 96% (monoclonal antibody), 100% (polyclonal antibody), and 100% (non-antibodies) observations. The Salisbury Rule mimics more to CL-based dose rather than per kg body weight-based extrapolated dose from adults. The Salisbury Rule for the pediatric dose prediction can be used to select first-in-children dose in pediatric clinical trials and may be in clinical settings.
Collapse
Affiliation(s)
- Iftekhar Mahmood
- Mahmood Clinical Pharmacology Consultancy, LLC 1709, Piccard DR, Rockville, MD 20850, USA
| |
Collapse
|
8
|
Mahmood I, Tegenge MA. Spreadsheet-Based Minimal Physiological Models for the Prediction of Clearance of Therapeutic Proteins in Pediatric Patients. J Clin Pharmacol 2021; 61 Suppl 1:S108-S116. [PMID: 34185903 DOI: 10.1002/jcph.1846] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 02/19/2021] [Indexed: 12/15/2022]
Abstract
There is a growing interest in the use of physiologically based pharmacokinetic (PBPK) models as clinical pharmacology drug development tools. In PBPK modeling, not every organ or physiological parameter is required, leading to the development of a minimal PBPK (mPBPK) model, which is simple and efficient. The objective of this study was to streamline mPBPK modeling approaches and enable straightforward prediction of clearance of protein-based products in children. Four mPBPK models for scaling clearance from adult to children were developed and evaluated on Excel spreadsheets using (1) liver and kidneys; (2) liver, kidneys, and skin; (3) liver, kidneys, skin, and lymph; and (4) interstitial, lymph, and plasma volume. There were 35 therapeutic proteins with a total of 113 observations across different age groups (premature neonates to adolescents). For monoclonal and polyclonal antibodies, more than 90% of observations were within a 0.5- to 2-fold prediction error for all 4 methods. For nonantibodies, 79% to 100% of observations were within the 0.5- to 2-fold prediction error for the 4 different methods. Methods 1 and 4 provided the best results, >90% of the total observations were within the 0.5- to 2-fold prediction error for all 3 classes of protein-based products across a wide age range. The precision of clearance prediction was comparatively lower in children ≤2 years of age vs older children (>2 years of age) with methods 1 and 4 predicting 80% to 100% and 75% to 90% of observations within the 0.5- to 2-fold prediction error, respectively. The results of the study indicated that mPBPK models can be developed on spreadsheets, with acceptable performance for prediction of clearance.
Collapse
Affiliation(s)
- Iftekhar Mahmood
- Mahmood Clinical Pharmacology Consultancy, Rockville, Maryland, USA
| | - Million A Tegenge
- Division of Clinical Evaluation and Pharmacology/Toxicology, Center for Biologics Evaluation and Research (CBER), Office of Tissues and Advanced Therapies (OTAT), Food and Drug Administration (FDA), Silver Spring, Maryland, USA
| |
Collapse
|
9
|
Green FG, Park K, Burckart GJ. Methods Used for Pediatric Dose Selection in Drug Development Programs Submitted to the US FDA 2012-2020. J Clin Pharmacol 2021; 61 Suppl 1:S28-S35. [PMID: 34185898 DOI: 10.1002/jcph.1853] [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: 01/21/2021] [Accepted: 03/08/2021] [Indexed: 11/08/2022]
Abstract
Dosing is a critical aspect of drug development in pediatrics that has led to trial failures and the inability to label the drug for pediatric use by the US Food and Drug Administration. Developing a structured approach for pediatric dose selection requires knowledge of the current approaches and their success or failure. This study describes the current experience with pediatric dosing methods from 2012 to 2020 and had 2 primary objectives: (1) to identify how the initial pediatric dose was selected and (2) to identify the pivotal dosing strategy used to identify the initially selected dose for safety and efficacy for pediatric clinical trials. Through September 2020, a total of 275 pediatric drug development programs were characterized for initial and pivotal dosing strategies. The success rate for labeling for pediatric use was 76.4%. The most common initial dosing strategy was previous experience with the product, followed by allometric scaling and exposure matching with adults. The most common pivotal dosing strategy was titration to target response in 33% of programs, with the second and third most common being pharmacokinetic/pharmacodynamic studies (30%) and exposure matching (20%), respectively. Additionally, about one-half of pediatric programs incorporated model-informed drug development. The emergence of titration to target response may signal a shift toward precision medicine in pediatric patients. Future work in pediatric drug dose selection should move toward the development of a structured pediatric dose selection approach.
Collapse
Affiliation(s)
- Francis G Green
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Kyunghun Park
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Gilbert J Burckart
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| |
Collapse
|
10
|
Germovsek E, Cheng M, Giragossian C. Allometric scaling of therapeutic monoclonal antibodies in preclinical and clinical settings. MAbs 2021; 13:1964935. [PMID: 34530672 PMCID: PMC8463036 DOI: 10.1080/19420862.2021.1964935] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/19/2021] [Accepted: 08/03/2021] [Indexed: 02/06/2023] Open
Abstract
Constant technological advancement enabled the production of therapeutic monoclonal antibodies (mAbs) and will continue to contribute to their rapid expansion. Compared to small-molecule drugs, mAbs have favorable characteristics, but also more complex pharmacokinetics (PK), e.g., target-mediated nonlinear elimination and recycling by neonatal Fc-receptor. This review briefly discusses mAb biology, similarities and differences in PK processes across species and within human, and provides a detailed overview of allometric scaling approaches for translating mAb PK from preclinical species to human and extrapolating from adults to children. The approaches described here will remain vital in mAb drug development, although more data are needed, for example, from very young patients and mAbs with nonlinear PK, to allow for more confident conclusions and contribute to further growth of this field. Improving mAb PK predictions will facilitate better planning of (pediatric) clinical studies and enable progression toward the ultimate goal of expediting drug development.
Collapse
Affiliation(s)
- Eva Germovsek
- Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Ingelheim, Germany
| | - Ming Cheng
- Development Biologicals, Drug Metabolism And Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, US
| | - Craig Giragossian
- Biotherapeutics Discovery, Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, US
| |
Collapse
|