1
|
Cui H, Lyu L, Bian J, Xu S, Chen R, Cai C, Chen Y, Xu ZR. LC-MS/MS quantification of ropivacaine and local analgesic and adverse effects of Long-acting Ropivacaine Injection based on pharmacokinetic-pharmacodynamic modelling in Bama minipigs. J Chromatogr B Analyt Technol Biomed Life Sci 2023; 1223:123716. [PMID: 37084699 DOI: 10.1016/j.jchromb.2023.123716] [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: 10/26/2022] [Revised: 03/28/2023] [Accepted: 04/08/2023] [Indexed: 04/23/2023]
Abstract
The local analgesic efficacy and adverse effects of a new Long-acting Ropivacaine formulation were examined based on pharmacokinetic-pharmacodynamic (PK-PD) modelling in Bama minipigs. 24 Bama minipigs, 12 males and 12 females, were randomly and equally divided into the following treatment groups: normal saline injection, drug vehicle injection, Long-acting Ropivacaine Injection and Ropivacaine Hydrochloride Injection. After routine disinfection, a skin incision about 3 cm long and 3 cm deep was produced in the leg of each pig, and mechanical withdrawal threshold (MWT) measured at various times pre- and post-injection as an index of analgesia against incision pain. Plasma ropivacaine concentrations were also measured at the same times using a novel liquid chromatography-tandem mass spectroscopy (LC-MS/MS) method. Minipigs were sacrificed 24 h post-injection and hearts collected for drug concentration measurements by LC-MS/MS. The LC-MS/MS method demonstrated high sensitivity, linearity and precision. The Long-acting Ropivacaine formulation produced a longer analgesic effect (∼12 h) at a lower plasma concentration than Ropivacaine Hydrochloride (∼4h), suggesting a better side-effects profile. A PK-PD model revealed a direct relationship between plasma ropivacaine concentration and MWT, with peak analgesia at about 1000 ng/mL and behaved good prediction ability. Long-acting Ropivacaine Injection is a superior local anaesthetic-analgesic treatment due to longer-lasting efficacy at lower concentrations compared to Ropivacaine Hydrochloride, which will reduce the risk of side effects such as cardiotoxicity.
Collapse
Affiliation(s)
- Huixin Cui
- College of Chemistry and Chemical Engineering, Shanghai University of Engineering Science, NO.333, Longteng Road, Songjiang District, Shanghai City, Shanghai 201620, China; State Key Laboratory of New Drug and Pharmaceutical Process, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, Shanghai 200437, China; Shanghai Professional and Technical Service Center for Biological Material Drug-ability Evaluation, Shanghai 200437, China
| | - Lihong Lyu
- State Key Laboratory of New Drug and Pharmaceutical Process, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, Shanghai 200437, China; Shanghai Professional and Technical Service Center for Biological Material Drug-ability Evaluation, Shanghai 200437, China; School of Medicine, Tianjin Tianshi College, Tianjin 301700, China
| | - Jing Bian
- State Key Laboratory of New Drug and Pharmaceutical Process, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, Shanghai 200437, China; Shanghai Professional and Technical Service Center for Biological Material Drug-ability Evaluation, Shanghai 200437, China
| | - Shan Xu
- State Key Laboratory of New Drug and Pharmaceutical Process, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, Shanghai 200437, China; Shanghai Professional and Technical Service Center for Biological Material Drug-ability Evaluation, Shanghai 200437, China
| | - Rui Chen
- State Key Laboratory of New Drug and Pharmaceutical Process, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, Shanghai 200437, China; Shanghai Professional and Technical Service Center for Biological Material Drug-ability Evaluation, Shanghai 200437, China
| | - Chunxiao Cai
- State Key Laboratory of New Drug and Pharmaceutical Process, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, Shanghai 200437, China; Shanghai Professional and Technical Service Center for Biological Material Drug-ability Evaluation, Shanghai 200437, China
| | - Yao Chen
- State Key Laboratory of New Drug and Pharmaceutical Process, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, Shanghai 200437, China; Shanghai Professional and Technical Service Center for Biological Material Drug-ability Evaluation, Shanghai 200437, China.
| | - Zhi-Ru Xu
- State Key Laboratory of New Drug and Pharmaceutical Process, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, Shanghai 200437, China; Shanghai Professional and Technical Service Center for Biological Material Drug-ability Evaluation, Shanghai 200437, China.
| |
Collapse
|
2
|
Egnell AC, Johansson S, Chen C, Berges A. Clinical Pharmacology Modeling and Simulation in Drug Development. SYSTEMS MEDICINE 2021. [DOI: 10.1016/b978-0-12-801238-3.11546-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
3
|
Leach MW, Clarke DO, Dudal S, Han C, Li C, Yang Z, Brennan FR, Bailey WJ, Chen Y, Deslandes A, Loberg LI, Mayawala K, Rogge MC, Todd M, Chemuturi NV. Strategies and Recommendations for Using a Data-Driven and Risk-Based Approach in the Selection of First-in-Human Starting Dose: An International Consortium for Innovation and Quality in Pharmaceutical Development (IQ) Assessment. Clin Pharmacol Ther 2020; 109:1395-1415. [PMID: 32757299 DOI: 10.1002/cpt.2009] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 07/02/2020] [Indexed: 01/27/2023]
Abstract
Various approaches to first-in-human (FIH) starting dose selection for new molecular entities (NMEs) are designed to minimize risk to trial subjects. One approach uses the minimum anticipated biological effect level (MABEL), which is a conservative method intended to maximize subject safety and designed primarily for NMEs having high perceived safety risks. However, there is concern that the MABEL approach is being inappropriately used for lower risk molecules with negative impacts on drug development and time to patient access. In addition, ambiguity exists in how MABEL is defined and the methods used to determine it. The International Consortium for Innovation and Quality in Pharmaceutical Development convened a working group to understand current use of MABEL and its impact on FIH starting dose selection, and to make recommendations for FIH dose selection going forward. An industry-wide survey suggested the achieved or estimated maximum tolerated dose, efficacious dose, or recommended phase II dose was > 100-fold higher than the MABEL-based starting dose for approximately one third of NMEs, including trials in patients. A decision tree and key risk factor table were developed to provide a consistent, data driven-based, and risk-based approach for selecting FIH starting doses.
Collapse
Affiliation(s)
- Michael W Leach
- Drug Safety Research and Development, Pfizer, Inc., Cambridge, Massachusetts, USA
| | - David O Clarke
- Nonclinical Safety Assessment, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana, USA
| | - Sherri Dudal
- DMPK Project Leads and Translational M&S, Pharmaceutical Sciences, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Chao Han
- Biologics Development Sciences, Janssen Research and Development, LLC, Spring House, Pennsylvania, USA
| | - Chunze Li
- Department of Clinical Pharmacology, Genentech, Inc., South San Francisco, California, USA
| | - Zheng Yang
- Metabolism and Pharmacokinetics, Bristol-Myers Squibb Co., Princeton, New Jersey, USA
| | | | - Wendy J Bailey
- Safety Assessment and Laboratory Animal Resources, Merck & Co., Inc., West Point, Pennsylvania, USA
| | - Yingxue Chen
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Boston, Massachusetts, USA
| | - Antoine Deslandes
- Translational Medicine & Early Development, Sanofi R&D, Centre de Recherche Vitry-sur-Seine 13, Vitry-sur-Seine Cedex, France
| | - Lise I Loberg
- Preclinical Safety, AbbVie, North Chicago, Illinois, USA
| | - Kapil Mayawala
- Quantitative Pharmacology and Pharmacometrics, PPDM, Merck & Co., Inc., Kenilworth, New Jersey, USA
| | - Mark C Rogge
- Quantitative and Translational Science, Takeda Pharmaceuticals, Cambridge, Massachusetts, USA
| | - Marque Todd
- Drug Safety Research and Development, Pfizer, Inc., San Diego, California, USA
| | - Nagendra V Chemuturi
- Pharmacokinetic Sciences, Novartis Institute of BioMedical Research, Inc., Cambridge, Massachusetts, USA
| |
Collapse
|
4
|
Model-based drug development: application of modeling and simulation in drug development. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2017. [DOI: 10.1007/s40005-017-0371-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
5
|
Wong H, Bohnert T, Damian-Iordache V, Gibson C, Hsu CP, Krishnatry AS, Liederer BM, Lin J, Lu Q, Mettetal JT, Mudra DR, Nijsen MJ, Schroeder P, Schuck E, Suryawanshi S, Trapa P, Tsai A, Wang H, Wu F. Translational pharmacokinetic-pharmacodynamic analysis in the pharmaceutical industry: an IQ Consortium PK-PD Discussion Group perspective. Drug Discov Today 2017; 22:1447-1459. [DOI: 10.1016/j.drudis.2017.04.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 04/03/2017] [Accepted: 04/25/2017] [Indexed: 02/06/2023]
|
6
|
Stein AM, Ramakrishna R. AFIR: A Dimensionless Potency Metric for Characterizing the Activity of Monoclonal Antibodies. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2017; 6:258-266. [PMID: 28375563 PMCID: PMC5397564 DOI: 10.1002/psp4.12169] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 12/06/2016] [Accepted: 01/03/2017] [Indexed: 12/14/2022]
Abstract
For monoclonal antibody (mAb) drugs, soluble targets may accumulate several thousand fold after binding to the drug. Time course data of mAb and total target is often collected and, although free target is more closely related to clinical effect, it is difficult to measure. Therefore, mathematical models of this data are used to predict target engagement. In this article, a “potency factor” is introduced as an approximation for the model‐predicted target inhibition. This potency factor is defined to be the time‐Averaged Free target concentration to Initial target concentration Ratio (AFIR), and it depends on three key quantities: the average drug concentration at steady state; the binding affinity; and the degree of target accumulation. AFIR provides the intuition for how changes in dosing regimen and binding affinity affect target capture and AFIR can be used to predict the druggability of new targets and the expected benefits of more potent, second‐generation mAbs.
Collapse
Affiliation(s)
- A M Stein
- Novartis Institute for BioMedical Research, Cambridge, Massachusetts, USA
| | - R Ramakrishna
- Novartis Institute for BioMedical Research, Cambridge, Massachusetts, USA
| |
Collapse
|
7
|
A First-in-Human Study To Assess the Safety and Pharmacokinetics of Monoclonal Antibodies against Human Cytomegalovirus in Healthy Volunteers. Antimicrob Agents Chemother 2016; 60:2881-7. [PMID: 26926639 DOI: 10.1128/aac.02698-15] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 02/20/2016] [Indexed: 11/20/2022] Open
Abstract
Human cytomegalovirus (HCMV) can cause significant disease in immunocompromised patients and treatment options are limited by toxicities. CSJ148 is a combination of two anti-HCMV human monoclonal antibodies (LJP538 and LJP539) that bind to and inhibit the function of viral HCMV glycoprotein B (gB) and the pentameric complex, consisting of glycoproteins gH, gL, UL128, UL130, and UL131. Here, we evaluated the safety, tolerability, and pharmacokinetics of a single intravenous dose of LJP538 or LJP539 or their combination in healthy volunteers. Adverse events and laboratory abnormalities occurred sporadically with similar incidence between antibody and placebo groups and without any apparent relationship to dose. No subject who received antibody developed a hypersensitivity, infusion-related reaction or anti-drug antibodies. After intravenous administration, both LJP538 and LJP539 demonstrated typical human IgG1 pharmacokinetic properties, with slow clearances, limited volumes of distribution, and long terminal half-lives. The pharmacokinetic parameters were linear and dose proportional for both antibodies across the 50-fold range of doses evaluated in the study. There was no apparent impact on pharmacokinetics when the antibodies were administered alone or in combination. CSJ148 and the individual monoclonal antibodies were safe and well tolerated, with pharmacokinetics as expected for human immunoglobulin.
Collapse
|
8
|
Dua P, Hawkins E, van der Graaf PH. A Tutorial on Target-Mediated Drug Disposition (TMDD) Models. CPT Pharmacometrics Syst Pharmacol 2015; 4:324-37. [PMID: 26225261 PMCID: PMC4505827 DOI: 10.1002/psp4.41] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Accepted: 04/07/2015] [Indexed: 12/16/2022] Open
Abstract
Target-mediated drug disposition (TMDD) is the phenomenon in which a drug binds with high affinity to its pharmacological target site (such as a receptor) to such an extent that this affects its pharmacokinetic characteristics.1 The aim of this Tutorial is to provide an introductory guide to the mathematical aspects of TMDD models for pharmaceutical researchers. Examples of Berkeley Madonna2 code for some models discussed in this Tutorial are provided in the Supplementary Materials.
Collapse
Affiliation(s)
- P Dua
- Pharmatherapeutics Research Clinical Pharmacology, Pfizer NeusentisCambridge, UK
| | - E Hawkins
- Pharmatherapeutics Research Clinical Pharmacology, Pfizer NeusentisCambridge, UK
- Department of Mathematics, University of SurreyGuildford, UK
| | - PH van der Graaf
- Leiden Academic Centre for Drug Research (LACDR), Systems PharmacologyLeiden, The Netherlands
| |
Collapse
|
9
|
Li H, Köck K, Wisler JA, Rees WA, Prince PJ, Reynhardt KO, Hsu H, Yu Z, Borie DC, Salinger DH, Pan WJ. Prediction of clinical pharmacokinetics of AMG 181, a human anti-α 4 β 7 monoclonal antibody for treating inflammatory bowel diseases. Pharmacol Res Perspect 2014; 3:e00098. [PMID: 25692016 PMCID: PMC4317229 DOI: 10.1002/prp2.98] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 09/12/2014] [Indexed: 01/05/2023] Open
Abstract
The purpose of this study was to predict a safe starting dose of AMG 181, a human anti-α4β7 antibody for treating inflammatory bowel diseases, based on cynomolgus monkey pharmacokinetic (PK) and pharmacodynamic (PD) data. A two-compartment model with parallel linear and target-mediated drug disposition for AMG 181 PK in cynomolgus monkey was developed. The estimated parameters were allometrically scaled to predict human PK. An Emax PD model was used to relate AMG 181 concentration and free α4β7 receptor data in cynomolgus monkey. AMG 181 clinical doses were selected based on observed exposures at the no adverse effect level of 80 mg·kg−1 in monkeys, the predicted human exposures, and AMG 181 concentration expected to produce greater than 50% α4β7 receptor occupancy in humans. The predicted human AMG 181 clearance and central volume of distribution were 144 mL·day−1 and 2900 mL, respectively. The estimated EC50 for free α4β7 receptor was 14 ng·mL−1. At the 0.7 mg starting dose in humans, the predicted exposure margins were greater than 490,000 and AMG 181 concentrations were predicted to only briefly cover the free α4β7 receptor EC10. Predictions for both Cmax and AUC matched with those observed in the first-in-human study within the 7 mg subcutaneous to 420 mg intravenous dose range. The developed model aided in selection of a safe starting dose and a pharmacological relevant dose escalation strategy for testing of AMG 181 in humans. The clinically observed human AMG 181 PK data validated the modeling approach based on cynomolgus monkey data alone.
Collapse
Affiliation(s)
- Hong Li
- Pharmacokinetics and Drug Metabolism, Amgen Inc. Seattle, Washington
| | - Kathleen Köck
- Pharmacokinetics and Drug Metabolism, Amgen Inc. Seattle, Washington
| | - John A Wisler
- Comparative Biology and Safety Sciences, Amgen Inc. Thousand Oaks, California
| | | | - Peter J Prince
- Pharmacokinetics and Drug Metabolism, Amgen Inc. Seattle, Washington
| | | | - Hailing Hsu
- Inflammation Discovery Research, Amgen Inc. Thousand Oaks, California
| | - Zhigang Yu
- Medical Sciences, Amgen Inc. Thousand Oaks, California
| | - Dominic C Borie
- Global Development, Amgen Inc. South San Francisco, California
| | - David H Salinger
- Pharmacokinetics and Drug Metabolism, Amgen Inc. Seattle, Washington
| | - Wei-Jian Pan
- Pharmacokinetics and Drug Metabolism, Amgen Inc. Seattle, Washington
| |
Collapse
|
10
|
Model-based drug discovery: implementation and impact. Drug Discov Today 2013; 18:764-75. [DOI: 10.1016/j.drudis.2013.05.012] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Revised: 04/03/2013] [Accepted: 05/20/2013] [Indexed: 01/15/2023]
|
11
|
Pharmacokinetic studies of protein drugs: past, present and future. Adv Drug Deliv Rev 2013; 65:1065-73. [PMID: 23541379 DOI: 10.1016/j.addr.2013.03.007] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Revised: 03/18/2013] [Accepted: 03/18/2013] [Indexed: 12/11/2022]
Abstract
Among the growing number of therapeutic proteins on the market, there is an emergence of biotherapeutics designed from our comprehension of the physiological mechanisms responsible for their peripheral and tissue pharmacokinetics. Most of them have been optimized to increase their half-life through glycosylation engineering, polyethylene glycol conjugation or Fc fusion. However, our understanding of biological drug behaviors is still its infancy compared to the huge amount of data regarding small molecular weight drugs accumulated over half a century. Unfortunately, therapeutic proteins share few resemblances with these drugs. For instance drug-targeted-mediated disposition, binding to glycoreceptors, lysosomal recycling, large hydrodynamic volume and electrostatic charge are typical critical characteristics that cannot be derived from our anterior knowledge of classical drugs. However, the numerous discoveries made in the two last decades have driven and will continue to drive new options in biochemical engineering and support the design of complex delivery systems. Most of these new developments will be supported by novel analytical methods for assessing in vitro or in vivo metabolism parameters.
Collapse
|
12
|
Chimalakonda AP, Yadav R, Marathe P. Factors influencing magnitude and duration of target inhibition following antibody therapy: implications in drug discovery and development. AAPS JOURNAL 2013; 15:717-27. [PMID: 23588584 DOI: 10.1208/s12248-013-9477-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Accepted: 03/20/2013] [Indexed: 01/01/2023]
Abstract
Antibodies or antibody-related fusion proteins binding to soluble antigens in plasma form an important subclass of approved therapeutics. Pharmaceutical companies are constantly trying to accelerate the pace of drug discovery and development of these antibodies and identify superior candidates in face of significant attrition rates. Understanding the interplay between drug- and target-related factors on magnitude and duration of target inhibition is imperative for successful advancement of these therapeutics. Simulations using a target-mediated drug disposition model were performed to evaluate the influence of antibody-target binding affinity, baseline target concentration, and target turnover on magnitude and duration of soluble target inhibition. These simulations assumed intravenous dosing of the antibody and evaluated multiple parameters over a wide range. These simulations reveal that improvement in affinity reaches a point of diminishing returns following which further improvement in affinity does not alter the magnitude and more importantly the duration of target inhibition. Evaluation of unbound antibody and target kinetics indicated that point of diminishing returns in duration of inhibition was due to target-mediated binding and subsequent elimination of antibody at later time points. Similarly, influence of baseline target concentration and target turnover on magnitude and duration of target inhibition in plasma is shown. Additionally, the fraction of dose eliminated via target mediated elimination (Fel(™)) can be a useful tool to enable selection of strategies to increase duration of target inhibition. The implications of these simulations in drug discovery and development with regard to target identification, antibody optimization, and backup candidate selection are discussed.
Collapse
Affiliation(s)
- Anjaneya P Chimalakonda
- Metabolism and Pharmacokinetics, Department of Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Co., Mail Stop: 17-2.04, Pennington, NJ 08534, USA.
| | | | | |
Collapse
|
13
|
Vugmeyster Y, Rohde C, Perreault M, Gimeno RE, Singh P. Agonistic TAM-163 antibody targeting tyrosine kinase receptor-B: applying mechanistic modeling to enable preclinical to clinical translation and guide clinical trial design. MAbs 2013; 5:373-83. [PMID: 23529133 DOI: 10.4161/mabs.23826] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
TAM-163, an agonist monoclonal antibody targeting tyrosine receptor kinase-B (TrkB), is currently being investigated as a potential body weight modulatory agent in humans. To support the selection of the dose range for the first-in-human (FIH) trial of TAM-163, we conducted a mechanistic analysis of the pharmacokinetic (PK) and pharmacodynamic (PD) data (e.g., body weight gain) obtained in lean cynomolgus and obese rhesus monkeys following single doses ranging from 0.3 to 60 mg/kg. A target-mediated drug disposition (TMDD) model was used to describe the observed nonlinear PK and Emax approach was used to describe the observed dose-dependent PD effect. The TMDD model development was supported by the experimental determination of the binding affinity constant (9.4 nM) and internalization rate of the drug-target complex (2.08 h(-1)). These mechanistic analyses enabled linking of exposure, target (TrkB) coverage, and pharmacological activity (e.g., PD) in monkeys, and indicated that ≥ 38% target coverage (time-average) was required to achieve significant body weight gain in monkeys. Based on the scaling of the TMDD model from monkeys to humans and assuming similar relationship between the target coverage and pharmacological activity between monkey and humans, subcutaneous (SC) doses of 1 and 15 mg/kg in humans were projected to be the minimally and the fully pharmacologically active doses, respectively. Based on the minimal anticipated biological effect level (MABEL) approach for starting dose selection, the dose of 0.05 mg/kg (3 mg for a 60 kg human) SC was recommended as the starting dose for FIH trials, because at this dose level<10% target coverage was projected at Cmax (and all other time points). This study illustrates a rational mechanistic approach for the selection of FIH dose range for a therapeutic protein with a complex model of action.
Collapse
Affiliation(s)
- Yulia Vugmeyster
- Pharmacokinetics, Dynamics and Metabolism; Pfizer, Inc.; Cambridge, MA USA
| | | | - Mylene Perreault
- Cardiovascular and Metabolic Diseases Research Unit; Pfizer, Inc.; Cambridge, MA USA
| | - Ruth E Gimeno
- Chief Scientific Officer; Eli Lilly & Co; Indianapolis, IN USA
| | - Pratap Singh
- Pharmacokinetics, Dynamics and Metabolism; Pfizer, Inc.; Cambridge, MA USA
| |
Collapse
|
14
|
Hu L, Hansen RJ. Issues, challenges, and opportunities in model-based drug development for monoclonal antibodies. J Pharm Sci 2013; 102:2898-908. [PMID: 23508847 DOI: 10.1002/jps.23504] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Revised: 02/04/2013] [Accepted: 02/20/2013] [Indexed: 12/13/2022]
Abstract
Over the last two decades, there has been a simultaneous explosion in the levels of activity and capability in both monoclonal antibody (mAb) drug development and in the use of quantitative pharmacologic models to facilitate drug development. Both of these topics are currently areas of great interest to academia, the pharmaceutical and biotechnology industries, and to regulatory authorities. In this article, we summarize convergence of these two areas and discuss some of the current and historical applications of the use of mathematical-model-based techniques to facilitate the discovery and development of mAb therapeutics. We also consider some of the current issues and limitations in model-based antibody discovery/development and highlight areas of further opportunity.
Collapse
Affiliation(s)
- Leijun Hu
- Eli Lilly and Company, Drug Disposition and PK/PD, Indianapolis, Indiana
| | | |
Collapse
|
15
|
Hansen RJ, Brown RM, Lu J, Wroblewski VJ. Qualification of a free ligand assay in the presence of anti-ligand antibody Fab fragments. MAbs 2013; 5:288-96. [PMID: 23396084 DOI: 10.4161/mabs.23508] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The aim of this work was to develop and characterize an ELISA to measure free ligand concentrations in rat serum in the presence of a Fab to the same ligand. A variety of experiments were conducted to understand optimal assay conditions and to verify that only free ligand was detected. The parameters explored included sample incubation time on plate, the initial concentrations of Fab and ligand, and the pre-incubation time required for the Fab-ligand complex concentrations to reach equilibrium. We found the optimal experimental conditions to include a 10-minute on-plate incubation of ligand-containing samples, with a 24-hour pre-incubation time for test samples of Fab and ligand to reach equilibrium. An alternative approach, involving removal of Fab-ligand complexes from the solution prior to measuring concentrations of the ligand, was also used to verify that the assay only measured free ligand. Rats were dosed subcutaneously with Fab and the assay was used to demonstrate dose-dependent suppression of endogenous free ligand levels in vivo.
Collapse
Affiliation(s)
- Ryan J Hansen
- Drug Disposition, Eli Lilly and Company, Indianapolis, IN, USA.
| | | | | | | |
Collapse
|
16
|
Abstract
Biologics, including monoclonal antibodies (mAbs) and other therapeutic proteins such as cytokines and growth hormones, have unique characteristics compared to small molecules. This paper starts from an overview of the pharmacokinetics (PK) of biologics from a mechanistic perspective, the determination of a starting dose for first-in-human (FIH) studies, and dosing regimen optimisation for phase II/III clinical trials. Subsequently, typical clinical pharmacology issues along the corresponding pathways for biologics development are summarised, including drug-drug interactions, QTc prolongation, immunogenicity, and studies in specific populations. The relationships between the molecular structure of biologics, their pharmacokinetic and pharmacodynamic characteristics, and the corresponding clinical pharmacology strategies are summarised and depicted in a schematic diagram.
Collapse
|
17
|
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: 2.2] [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
|