1
|
Choi S, Valente D, Virone‐Oddos A, Mauriac C. Developing a mechanistic translational PK/PD model for a trifunctional NK cell engager to predict the first-in-human dose for acute myeloid leukemia. Clin Transl Sci 2024; 17:e13689. [PMID: 37990450 PMCID: PMC10772472 DOI: 10.1111/cts.13689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 11/08/2023] [Accepted: 11/10/2023] [Indexed: 11/23/2023] Open
Abstract
Natural killer cell engagers (NKCEs), a treatment that stimulates innate immunity, have lately gained attention owing to their favorable safety profile, and their efficacy. Natural killer (NK) cell activation is driven by immune synapse formation between drugs, NK cells, and tumor cells. However, no clear translational modeling approach has been reported for first-in-human (FIH) dose estimation of humanized NKCEs. We developed the first translational mechanistic synapse-driven pharmacokinetic/pharmacodynamic (PK/PD) model for a trifunctional NKp46/CD16a-CD123 (CD123-NKCE) by integrating (i) in vitro target cell cytotoxicity in MOLM-13 tumor cell lines at varying effector-to-tumor cell ratios and incubation intervals; (ii) nonhuman primate PK and profiles of CD123+ cells and NKP46+ NK cells; and (iii) healthy human or patients with acute myeloid leukemia system-specific parameters. To depict direct tumor cell killing by the innate immunity, no transit compartment was included in PK/PD model structures. Model predictions suggested an intrapatient dose escalation of 10/30/100 μg/kg twice weekly to be selected as the starting dose in the FIH trial. However, sensitivity analyses revealed that CD123+ cell growth rate constant and maximal tumor killing rate constant were the key uncertainties to the recommended active dose. This novel translational model structure can be used as the basis to predict clinical PK/PD data for CD123-NKCE, and the translational strategy may serve as a foundation for future advancements of NKCEs.
Collapse
|
2
|
Choules MP, Bonate PL, Heo N, Weddell J. Prospective approaches to gene therapy computational modeling - spotlight on viral gene therapy. J Pharmacokinet Pharmacodyn 2023:10.1007/s10928-023-09889-1. [PMID: 37848637 DOI: 10.1007/s10928-023-09889-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 09/25/2023] [Indexed: 10/19/2023]
Abstract
Clinical studies have found there still exists a lack of gene therapy dose-toxicity and dose-efficacy data that causes gene therapy dose selection to remain elusive. Model informed drug development (MIDD) has become a standard tool implemented throughout the discovery, development, and approval of pharmaceutical therapies, and has the potential to inform dose-toxicity and dose-efficacy relationships to support gene therapy dose selection. Despite this potential, MIDD approaches for gene therapy remain immature and require standardization to be useful for gene therapy clinical programs. With the goal to advance MIDD approaches for gene therapy, in this review we first provide an overview of gene therapy types and how they differ from a bioanalytical, formulation, route of administration, and regulatory standpoint. With this biological and regulatory background, we propose how MIDD can be advanced for AAV-based gene therapies by utilizing physiological based pharmacokinetic modeling and quantitative systems pharmacology to holistically inform AAV and target protein dynamics following dosing. We discuss how this proposed model, allowing for in-depth exploration of AAV pharmacology, could be the key the field needs to treat these unmet disease populations.
Collapse
Affiliation(s)
- Mary P Choules
- Early Development, New Technologies Group, Astellas, Northbrook, IL, USA
| | - Peter L Bonate
- Early Development, New Technologies Group, Astellas, Northbrook, IL, USA.
| | - Nakyo Heo
- Early Development, New Technologies Group, Astellas, Northbrook, IL, USA
| | - Jared Weddell
- Early Development, New Technologies Group, Astellas, Northbrook, IL, USA
| |
Collapse
|
3
|
Jones G, Zeng L, Kim J. Application of Allometric Scaling to Nanochelator Pharmacokinetics. ACS OMEGA 2023; 8:27256-27263. [PMID: 37546686 PMCID: PMC10399172 DOI: 10.1021/acsomega.3c02570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 06/22/2023] [Indexed: 08/08/2023]
Abstract
Deferoxamine (DFO) is an effective FDA-approved iron chelator; however, its use is considerably limited by off-target toxicities and an extremely cumbersome dose regimen involving daily infusions. The recent development of a deferoxamine-based nanochelator (DFO-NP) with selective renal excretion has shown promise in ameliorating iron overload and associated physiological complications in rodent models with a substantially improved safety profile. While the dose- and administration route-dependent pharmacokinetics (PK) of DFO-NPs have been recently characterized, the optimized PK model was not validated, and the prior studies did not directly address the clinical translatability of DFO-NPs into humans. In the present work, these gaps were addressed by applying allometric scaling of DFO-NP PK in rats to predict those in mice and humans. First, this approach predicted serum concentration-time profiles of DFO-NPs, which were similar to those experimentally measured in mice, validating the nonlinear disposition and absorption models for DFO-NPs across the species. Subsequently, we explored the utility of allometric scaling by predicting the PK profile of DFO-NPs in humans under clinically relevant dosing schemes. These in silico efforts demonstrated that the novel nanochelator is expected to improve the PK of DFO when compared to standard infusion regimens of native DFO. Moreover, reasonable formulation strategies were identified and discussed for both early clinical development and more sophisticated formulation development.
Collapse
Affiliation(s)
- Gregory Jones
- Department
of Pharmaceutical Sciences, Northeastern
University, Boston, Massachusetts 02115, United States
| | - Lingxue Zeng
- Department
of Biomedical & Nutritional Sciences, University of Massachusetts Lowell, Lowell, Massachusetts 01854, United States
| | - Jonghan Kim
- Department
of Biomedical & Nutritional Sciences, University of Massachusetts Lowell, Lowell, Massachusetts 01854, United States
| |
Collapse
|
4
|
Pasquiers B, Benamara S, Felices M, Nguyen L, Declèves X. Review of the Existing Translational Pharmacokinetics Modeling Approaches Specific to Monoclonal Antibodies (mAbs) to Support the First-In-Human (FIH) Dose Selection. Int J Mol Sci 2022; 23:12754. [PMID: 36361546 PMCID: PMC9657028 DOI: 10.3390/ijms232112754] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 10/18/2022] [Accepted: 10/19/2022] [Indexed: 08/27/2023] Open
Abstract
The interest in therapeutic monoclonal antibodies (mAbs) has continuously growing in several diseases. However, their pharmacokinetics (PK) is complex due to their target-mediated drug disposition (TMDD) profiles which can induce a non-linear PK. This point is particularly challenging during the pre-clinical and translational development of a new mAb. This article reviews and describes the existing PK modeling approaches used to translate the mAbs PK from animal to human for intravenous (IV) and subcutaneous (SC) administration routes. Several approaches are presented, from the most empirical models to full physiologically based pharmacokinetic (PBPK) models, with a focus on the population PK methods (compartmental and minimal PBPK models). They include the translational approaches for the linear part of the PK and the TMDD mechanism of mAbs. The objective of this article is to provide an up-to-date overview and future perspectives of the translational PK approaches for mAbs during a model-informed drug development (MIDD), since the field of PK modeling has gained recently significant interest for guiding mAbs drug development.
Collapse
Affiliation(s)
- Blaise Pasquiers
- PhinC Development, 91300 Massy, France
- Université Paris Cité, Inserm UMRS-1144, Optimisation Thérapeutique en Neuropsychopharmacologie, 75006 Paris, France
| | | | | | | | - Xavier Declèves
- Université Paris Cité, Inserm UMRS-1144, Optimisation Thérapeutique en Neuropsychopharmacologie, 75006 Paris, France
| |
Collapse
|
5
|
Zhang W, Xiang Y, Wang L, Wang F, Li G, Zhuang X. Translational pharmacokinetics of a novel bispecific antibody against Ebola virus (MBS77E) from animal to human by PBPK modeling & simulation. Int J Pharm 2022; 626:122160. [PMID: 36089211 DOI: 10.1016/j.ijpharm.2022.122160] [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: 06/28/2022] [Revised: 08/11/2022] [Accepted: 08/28/2022] [Indexed: 11/17/2022]
Abstract
The goal of this study was to construct a PBPK model to accelerate the translation of MBS77E, a humanized bispecific antibody against the Ebola virus. In-depth nonclinical pharmacokinetic studies in rats, monkeys, wild-type mice and transgenic mice were conducted. The pH-dependent affinities (KD) of MBS77E to recombinant FcRn of different species were determined by surface plasmon resonance analysis. A mechanistic whole-body PBPK model of MBS77E was developed and validated in the assessment of PK profiles and tissue distributions in preclinical models. This PBPK model was finally used to predict human PK behaviors of MBS77E. Simulations from the PBPK model with measured and fitted parameters were able to yield good predictions of the serum and tissue pharmacokinetic parameters of MBS77E within 2-fold errors. The predicted serum concentration in humans was able to maintain a sufficiently high level for more than 14 days after 50 mg/kg i.v. administrating. This achievement unlocks that PBPK modeling is a powerful tool to gain insights into the properties of antibody drugs. It guided experimental efforts to obtain necessary information before entry into humans.
Collapse
Affiliation(s)
- Wenpeng Zhang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Yanan Xiang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Lingchao Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Furun Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Guanglu Li
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Xiaomei Zhuang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China.
| |
Collapse
|
6
|
Haraya K, Tsutsui H, Komori Y, Tachibana T. Recent Advances in Translational Pharmacokinetics and Pharmacodynamics Prediction of Therapeutic Antibodies Using Modeling and Simulation. Pharmaceuticals (Basel) 2022; 15:ph15050508. [PMID: 35631335 PMCID: PMC9145563 DOI: 10.3390/ph15050508] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 04/18/2022] [Accepted: 04/20/2022] [Indexed: 02/05/2023] Open
Abstract
Therapeutic monoclonal antibodies (mAbs) have been a promising therapeutic approach for several diseases and a wide variety of mAbs are being evaluated in clinical trials. To accelerate clinical development and improve the probability of success, pharmacokinetics and pharmacodynamics (PKPD) in humans must be predicted before clinical trials can begin. Traditionally, empirical-approach-based PKPD prediction has been applied for a long time. Recently, modeling and simulation (M&S) methods have also become valuable for quantitatively predicting PKPD in humans. Although several models (e.g., the compartment model, Michaelis–Menten model, target-mediated drug disposition model, and physiologically based pharmacokinetic model) have been established and used to predict the PKPD of mAbs in humans, more complex mechanistic models, such as the quantitative systemics pharmacology model, have been recently developed. This review summarizes the recent advances and future direction of M&S-based approaches to the quantitative prediction of human PKPD for mAbs.
Collapse
Affiliation(s)
- Kenta Haraya
- Discovery Biologics Department, Research Division, Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba 412-8513, Japan;
- Correspondence:
| | - Haruka Tsutsui
- Discovery Biologics Department, Research Division, Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba 412-8513, Japan;
| | - Yasunori Komori
- Pharmaceutical Science Department, Translational Research Division, Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba 412-8513, Japan; (Y.K.); (T.T.)
| | - Tatsuhiko Tachibana
- Pharmaceutical Science Department, Translational Research Division, Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba 412-8513, Japan; (Y.K.); (T.T.)
| |
Collapse
|
7
|
Ball K, Bruin G, Escandon E, Funk C, Pereira JN, Yang TY, Yu H. Characterizing the pharmacokinetics and biodistribution of therapeutic proteins: an industry white paper. Drug Metab Dispos 2022; 50:858-866. [PMID: 35149542 DOI: 10.1124/dmd.121.000463] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 01/06/2022] [Indexed: 11/22/2022] Open
Abstract
Characterization of the pharmacokinetics (PK) and biodistribution of therapeutic proteins (TPs) is a hot topic within the pharmaceutical industry, particularly with an ever-increasing catalog of novel modality TPs. Here, we review the current practices, and provide a summary of extensive cross-company discussions as well as a survey completed by International Consortium for Innovation and Quality (IQ consortium) members on this theme. A wide variety of in vitro, in vivo and in silico techniques are currently used to assess PK and biodistribution of TPs, and we discuss the relevance of these from an industry perspective, focusing on PK/PD understanding at the preclinical stage of development, and translation to human. We consider that the 'traditional in vivo biodistribution study' is becoming insufficient as a standalone tool, and thorough characterization of the interaction of the TP with its target(s), target biology, and off-target interactions at a microscopic scale are key to understand the overall biodistribution at a full-body scale. Our summary of the current challenges and our recommendations to address these issues could provide insight into the implementation of best practices in this area of drug development, and continued cross-company collaboration will be of tremendous value. Significance Statement The Innovation & Quality Consortium (IQ) Translational and ADME Sciences Leadership Group (TALG) working group for the ADME of therapeutic proteins evaluates the current practices, recent advances, and challenges in characterizing the PK and biodistribution of therapeutic proteins during drug development, and proposes recommendations to address these issues. Incorporating the in vitro, in vivo and in silico approaches discussed herein may provide a pragmatic framework to increase early understanding of PK/PD relationships, and aid translational modelling for first-in-human dose predictions.
Collapse
Affiliation(s)
| | - Gerard Bruin
- Novartis Institutes for Biomedical Research, Switzerland
| | | | - Christoph Funk
- Dept. of Drug Metabolism and Pharmacokinetics, F. Hoffmann-La Roche Ltd., Switzerland
| | | | | | - Hongbin Yu
- Boehringer Ingelheim Pharmaceuticals, Inc, United States
| |
Collapse
|
8
|
Ménochet K, Yu H, Wang B, Tibbitts J, Hsu CP, Kamath AV, Richter WF, Baumann A. Non-human primates in the PKPD evaluation of biologics: Needs and options to reduce, refine, and replace. A BioSafe White Paper. MAbs 2022; 14:2145997. [PMID: 36418217 PMCID: PMC9704389 DOI: 10.1080/19420862.2022.2145997] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 11/07/2022] [Indexed: 11/27/2022] Open
Abstract
Monoclonal antibodies (mAbs) deliver great benefits to patients with chronic and/or severe diseases thanks to their strong specificity to the therapeutic target. As a result of this specificity, non-human primates (NHP) are often the only preclinical species in which therapeutic antibodies cross-react with the target. Here, we highlight the value and limitations that NHP studies bring to the design of safe and efficient early clinical trials. Indeed, data generated in NHPs are integrated with in vitro information to predict the concentration/effect relationship in human, and therefore the doses to be tested in first-in-human trials. The similarities and differences in the systems defining the pharmacokinetics and pharmacodynamics (PKPD) of mAbs in NHP and human define the nature and the potential of the preclinical investigations performed in NHPs. Examples have been collated where the use of NHP was either pivotal to the design of the first-in-human trial or, inversely, led to the termination of a project prior to clinical development. The potential impact of immunogenicity on the results generated in NHPs is discussed. Strategies to optimize the use of NHPs for PKPD purposes include the addition of PD endpoints in safety assessment studies and the potential re-use of NHPs after non-terminal studies or cassette dosing several therapeutic agents of interest. Efforts are also made to reduce the use of NHPs in the industry through the use of in vitro systems, alternative in vivo models, and in silico approaches. In the case of prediction of ocular PK, the body of evidence gathered over the last two decades renders the use of NHPs obsolete. Expert perspectives, advantages, and pitfalls with these alternative approaches are shared in this review.
Collapse
Affiliation(s)
| | - Hongbin Yu
- R&D Project Management and Development Strategies, Boehringer Ingelheim Pharmaceuticals, Inc, Ridgefield, CT, USA
| | - Bonnie Wang
- Nonclinical Disposition and Bioanalysis, Bristol Myers Squibb, Inc, Princeton, NJ, USA
| | - Jay Tibbitts
- Nonclinical Development, South San Francisco, CA, USA
| | - Cheng-Pang Hsu
- Preclinical Development and Clinical Pharmacology, AskGene Pharma Inc, Camarillo, CA, USA
| | - Amrita V. Kamath
- Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech Inc, South San Francisco, CA, USA
| | - Wolfgang F. Richter
- Roche Pharma Research and Early Development, Roche Innovation, Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Andreas Baumann
- R&D, Bayer Pharma AG, Berlin, Germany & Non-clinical Biotech Consulting, Potsdam, Germany °(° present affiliation)
| |
Collapse
|
9
|
Singh R, Moreno M, Stanimirovic D. Comparison of Various Approaches to Translate Non-Linear Pharmacokinetics of Monoclonal Antibodies from Cynomolgus Monkey to Human. Eur J Drug Metab Pharmacokinet 2021; 46:555-567. [PMID: 34120326 DOI: 10.1007/s13318-021-00691-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/19/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND AND OBJECTIVES The prediction of pharmacokinetics of monoclonal antibodies (mAbs) exhibiting non-linear pharmacokinetics in preclinical species to human is challenging, and very limited scientific work has been published in this field of research. Therefore, we have conducted an elaborate comparative assessment to determine the most reliable preclinical to clinical scaling strategy for mAbs with non-linear pharmacokinetics. METHODS We have compared three different scaling approaches to predict human pharmacokinetics from cynomolgus monkey. In the first approach, cynomolgus monkey pharmacokinetic parameters estimated using a two-compartment model with parallel linear and non-linear elimination were allometrically scaled to simulate human pharmacokinetics. In the second approach, allometric exponents were integrated with a minimal physiologically based pharmacokinetic (mPBPK) model to translate human pharmacokinetics. In the third approach, we have employed a species time-invariant method, wherein a two-compartment model with parallel linear and non-linear elimination was used as a framework model for simulation of the human profile. RESULTS Human exposure parameters projected by an integrated allometric method were only within two fold for approximately 45-70% of predictions at different doses of five mAbs evaluated, while approximately 70-80% of Cmax and AUC predictions by integrated mPBPK modelling as well as the species time-invariant method were within two-fold error. The average fold error for clearance predictions by the integrated mPBPK method was 1.10-1.45 fold, whilst for the species time-variant and integrated allometric methods, the average fold error was between 1.04 and 1.37 fold and 1.24 and 2.13 fold, respectively. CONCLUSIONS Our findings suggest that the species time-variant method and mPBPK proposed by us can be employed to reliably translate non-linear pharmacokinetics of mAbs from cynomolgus monkey to human.
Collapse
Affiliation(s)
- Renu Singh
- Translation Bioscience, Human Health Therapeutics, National Research Council of Canada, Ottawa, ON, Canada. .,Global DMPK, Research IVIVT, 1250 S. Collegeville Road, Collegeville, PA, 19426, USA. .,Translation Bioscience, Human Health Therapeutics, National Research Council of Canada, Montreal, QC, Canada.
| | - Maria Moreno
- Translation Bioscience, Human Health Therapeutics, National Research Council of Canada, Ottawa, ON, Canada
| | - Danica Stanimirovic
- Translation Bioscience, Human Health Therapeutics, National Research Council of Canada, Ottawa, ON, Canada
| |
Collapse
|
10
|
Ayyar VS, Jaiprasart P, Geist B, Huang Devine Z, Case M, Hazra A, Hsu CH, Chintala M, Wang W. Translational PK/PD and model-informed development of JNJ-67842125, a F ab reversal agent for JNJ-64179375, a long-acting thrombin inhibitor. Br J Pharmacol 2021; 178:3943-3958. [PMID: 34008170 DOI: 10.1111/bph.15533] [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: 05/07/2020] [Revised: 04/30/2021] [Accepted: 05/06/2021] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND AND PURPOSE Antigen-binding fragment (Fab ) reversal agents were developed to reverse, in bleeding emergency, the long-acting anticoagulant effect of JNJ-64179375 (JNJ-9375), a monoclonal antibody that binds exosite-1 on thrombin. EXPERIMENTAL APPROACH The pharmacokinetic and pharmacodynamic (PK/PD) activities of three reversal agents of varying in vitro binding affinities to JNJ-9375 were characterised in cynomolgus monkeys. The time course of JNJ-9375 anticoagulant activity and reversal effects of each agent were evaluated. A mechanism-based PK/PD model, which integrated free serum concentrations of reversal agent, total and free serum concentrations of JNJ-9375, and thrombin time, was developed to quantitatively relate JNJ-9375 neutralisation to reversal of induced thrombin time prolongation. Model-based allometric scale-up of the lead reversal agent and the PK/PD relationship of JNJ-9375 in healthy volunteers were utilised to predict clinical dosing regimens. KEY RESULTS Lowering of free JNJ-9375 by the reversal agents corresponded with reversal of thrombin time prolongation. Total JNJ-9375 displayed typical mAb clearance at 2.75 ml·day-1 ·kg-1 , whereas reversal agents cleared faster between 1400 and 2400 ml·day-1 ·kg-1 . The model-estimated in vivo KD values for JNJ-9375 reversal agents were 9 nM (ICHB-256), 0.4 nM (ICHB-281) and 13.7 pM (ICHB-164), in rank-ordered agreement of their KD values determined in vitro. The three reversal agents exhibited different neutralisation characteristics in vivo, governed primarily by their binding kinetics to JNJ-9375. The model predicted a priori free JNJ-9375 kinetics after dosing ICHB-164 (JNJ-67842125) and JNJ-9375 under a different regimen. CONCLUSION AND IMPLICATIONS The results enabled selection of JNJ-67842125 as the reversal agent for JNJ-9375.
Collapse
Affiliation(s)
- Vivaswath S Ayyar
- Biologics Development Sciences, Janssen BioTherapeutics, Janssen Research & Development, LLC, Spring House, Pennsylvania, USA.,Clinical Pharmacology and Pharmacometrics, Janssen Research & Development, LLC, Spring House, Pennsylvania, USA
| | - Pharavee Jaiprasart
- Biologics Development Sciences, Janssen BioTherapeutics, Janssen Research & Development, LLC, Spring House, Pennsylvania, USA.,Clinical Pharmacology and Pharmacometrics, Janssen Research & Development, LLC, Spring House, Pennsylvania, USA
| | - Brian Geist
- Biologics Development Sciences, Janssen BioTherapeutics, Janssen Research & Development, LLC, Spring House, Pennsylvania, USA
| | - Zheng Huang Devine
- Cardiovascular and Metabolism, Janssen Research & Development, LLC, Spring House, Pennsylvania, USA
| | - Martin Case
- New Platforms and Technology, Janssen BioTherapeutics, Janssen Research & Development, LLC, San Diego, California, USA
| | - Anasuya Hazra
- Clinical Pharmacology and Pharmacometrics, Janssen Research & Development, LLC, Spring House, Pennsylvania, USA
| | - Chyi-Hung Hsu
- Clinical Pharmacology and Pharmacometrics, Janssen Research & Development, LLC, Spring House, Pennsylvania, USA
| | - Madhu Chintala
- Cardiovascular and Metabolism, Janssen Research & Development, LLC, Spring House, Pennsylvania, USA
| | - Weirong Wang
- Clinical Pharmacology and Pharmacometrics, Janssen Research & Development, LLC, Spring House, Pennsylvania, USA
| |
Collapse
|
11
|
Conner KP, Pastuskovas CV, Soto M, Thomas VA, Wagner M, Rock DA. Preclinical characterization of the ADME properties of a surrogate anti-IL-36R monoclonal antibody antagonist in mouse serum and tissues. MAbs 2021; 12:1746520. [PMID: 32310023 PMCID: PMC7188401 DOI: 10.1080/19420862.2020.1746520] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The decision to pursue a monoclonal antibody (mAb) as a therapeutic for disease intervention requires the assessment of many factors, such as target-biology, including the total target burden and its accessibility at the intended site of action, as well as mAb-specific properties like binding affinity and the pharmacokinetics in serum and tissue. Interleukin-36 receptor (IL-36 R) is a member of the IL-1 family cytokine receptors and an attractive target to treat numerous epithelial-mediated inflammatory conditions, including psoriatic and rheumatoid arthritis, asthma, and chronic obstructive pulmonary disease. However, information concerning the expression profile of IL-36 R at the protein level is minimal, so the feasibility of developing a therapeutic mAb against this target is uncertain. Here, we present a characterization of the properties associated with absorption, distribution, metabolism, and excretion of a high-affinity IL-36 R-targeted surrogate rat (IgG2a) mAb antagonist in preclinical mouse models. The presence of IL-36 R in the periphery was confirmed unequivocally as the driver of non-linear pharmacokinetics in blood/serum, although a predominant site of tissue accumulation was not observed based upon the kinetics of radiotracer. Additionally, the contribution of IL-36 R-mediated catabolism of mAb in kidney was tested in a 5/6 nephrectomized mouse model where minimal effects on serum pharmacokinetics were observed, although analysis of functional mAb in urine suggests that target can influence the amount of mAb excreted. Our data highlight an interesting case of target-mediated drug disposition (TMDD) where low, yet broadly expressed levels of membrane-bound target result in a cumulative effect to drive TMDD behavior typical of a large, saturable target sink. The potential differences between our mouse model and IL-36 R target profile in humans are also presented.
Collapse
Affiliation(s)
- Kip P Conner
- Department Pharmacokinetics and Drug Metabolism, Amgen, South San Francisco, CA, USA
| | - Cinthia V Pastuskovas
- Department Pharmacokinetics and Drug Metabolism, Amgen, South San Francisco, CA, USA
| | - Marcus Soto
- Department Pharmacokinetics and Drug Metabolism, Amgen, Thousand Oaks, CA, USA
| | - Veena A Thomas
- Department Pharmacokinetics and Drug Metabolism, Amgen, South San Francisco, CA, USA
| | - Mylo Wagner
- Department Pharmacokinetics and Drug Metabolism, Amgen, Thousand Oaks, CA, USA
| | - Dan A Rock
- Department Pharmacokinetics and Drug Metabolism, Amgen, South San Francisco, CA, USA
| |
Collapse
|
12
|
Nakamura G, Ozeki K, Nagayasu M, Nambu T, Nemoto T, Hosoya KI. Predicting Method for the Human Plasma Concentration-Time Profile of a Monoclonal Antibody from the Half-life of Non-human Primates. Biol Pharm Bull 2021; 43:823-830. [PMID: 32378559 DOI: 10.1248/bpb.b19-01042] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Efficiency (speed and cost) and animal welfare are important factors in the development of new drugs. A novel method (the half-life method) was developed to predict the human plasma concentration-time profile of a monoclonal antibody (mAb) after intravenous (i.v.) administration using less data compared to the conventional approach; moreover, predicted results were comparable to conventional method. This new method use human geometric means of pharmacokinetics (PK) parameters and the non-human primates (NHP) half-life of each mAb. PK data on mAbs in humans and NHPs were collected from literature focusing on linear elimination, and the two-compartment model was used for analysis. The following features were revealed in humans: 1) the coefficient of variation in the distribution volume of the central compartment and at steady state of mAbs was small (22.6 and 23.8%, respectively) and 2) half-life at the elimination phase (t1/2β) was the main contributor to plasma clearance. Moreover, distribution volume showed no significant correlation between humans and NHPs, and human t1/2β showed a good correlation with allometrically scaled t1/2β of NHP. Based on the features revealed in this study, we propose a new method for predicting the human plasma concentration-time profile of mAbs after i.v. dosing. When tested, this half-life method showed reasonable human prediction compared with a conventional empirical approach. The half-life method only requires t1/2β to predict human PK, and is therefore able to improve animal welfare and potentially accelerate the drug development process.
Collapse
Affiliation(s)
- Genki Nakamura
- Research Division, Chugai Pharmaceutical Co., Ltd.,Department of Pharmaceutics, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama
| | | | | | - Takeru Nambu
- Research Division, Chugai Pharmaceutical Co., Ltd
| | | | - Ken-Ichi Hosoya
- Department of Pharmaceutics, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama
| |
Collapse
|
13
|
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: 15] [Impact Index Per Article: 5.0] [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
|
14
|
Chung TK, Lee HA, Park SI, Oh DY, Lee KW, Kim JW, Kim JH, Woo A, Lee SJ, Bang YJ, Lee H. A target-mediated drug disposition population pharmacokinetic model of GC1118, a novel anti-EGFR antibody, in patients with solid tumors. Clin Transl Sci 2021; 14:990-1001. [PMID: 33382918 PMCID: PMC8212746 DOI: 10.1111/cts.12963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 11/18/2020] [Accepted: 12/03/2020] [Indexed: 11/29/2022] Open
Abstract
Abstract GC1118 is a monoclonal antibody for epidermal growth factor receptor (EGFR) that is currently under clinical development to treat patients with solid tumors. In this study, the pharmacokinetics (PKs) of GC1118 were modeled in solid tumor patients who received a 2‐h intravenous infusion of GC1118 at 0.3, 1, 3, 5, or 4 mg/kg once‐weekly (Q1W) on days 1, 8, 15, and 22 or 8 mg/kg every other week on days 1 and 15. A target‐mediated drug disposition population PK model adequately described the concentration‐time profiles of GC1118. Monte‐Carlo simulation experiments of the PK profiles and EGFR occupancies (ROs) by GC1118 based on the final model showed that Q1W at 4 or 5 mg/kg will produce a better antitumor effect than Q2W at 8 mg/kg. Because GC1118 was safer at 4 mg/kg than 5 mg/kg in the phase I study, we suggest to test the 4 mg/kg Q1W regimen in further clinical trials with GC1118. Study Highlights WHAT IS THE CURRENT KNOWLEDGE ON THE TOPIC?
GC1118, a fully human IgG1 monoclonal antibody (mAb) for epidermal growth factor receptor (EGFR), showed a nonlinear pharmacokinetic (PK) profile in monkeys and humans. The total clearance of GC1118 decreased as the dose was increased up to 3–4 mg/kg in humans, beyond which it remained stable. The recommended phase II dose for GC1118 was 4 mg/kg intravenously infused over 2 h once weekly.
WHAT QUESTION DID THIS STUDY ADDRESS?
We developed a target‐mediated drug disposition (TMDD) population PK model that described the nonlinear PK profile of GC1118 in patients with solid tumors. We also simulated the PK profiles and receptor occupancies for different dosage regimens.
WHAT DOES THIS STUDY ADD TO OUR KNOWLEDGE?
The TMDD population PK model adequately described the nonlinear and multiphasic PK profiles of GC1118 in humans. The simulation experiment showed that once‐weekly GC1118 at 4–5 mg/kg could be more efficacious than the biweekly regimen at 8 mg/kg.
HOW MIGHT THIS CHANGE CLINICAL PHARMACOLOGY OR TRANSLATIONAL SCIENCE?
The pharmacometrics analysis could support better informed drug development decisions for GC1118, particularly for determining an optimal dosage regimen.
Collapse
Affiliation(s)
- Tae Kyu Chung
- Department of Transdisciplinary Studies, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea
| | - Hyun A Lee
- Department of Transdisciplinary Studies, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea
| | - Sang-In Park
- Department of Pharmacology, College of Medicine, Kangwon National University, Chuncheon, Korea
| | - Do-Youn Oh
- Department of Internal Medicine, Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Keun-Wook Lee
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Jin Won Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Jee Hyun Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | | | | | - Yung-Jue Bang
- Department of Internal Medicine, Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Howard Lee
- Department of Transdisciplinary Studies, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea.,Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Korea.,Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea.,Center for Convergence Approaches in Drug Development, Seoul National University, Seoul, Korea.,Advanced Institute of Convergence Technology, Suwon, Korea
| |
Collapse
|
15
|
Singh AP, Seigel GM, Guo L, Verma A, Wong GGL, Cheng HP, Shah DK. Evolution of the Systems Pharmacokinetics-Pharmacodynamics Model for Antibody-Drug Conjugates to Characterize Tumor Heterogeneity and In Vivo Bystander Effect. J Pharmacol Exp Ther 2020; 374:184-199. [PMID: 32273304 DOI: 10.1124/jpet.119.262287] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Accepted: 03/30/2020] [Indexed: 12/18/2022] Open
Abstract
The objective of this work was to develop a systems pharmacokinetics-pharmacodynamics (PK-PD) model that can characterize in vivo bystander effect of antibody-drug conjugate (ADC) in a heterogeneous tumor. To accomplish this goal, a coculture xenograft tumor with 50% GFP-MCF7 (HER2-low) and 50% N87 (HER2-high) cells was developed. The relative composition of a heterogeneous tumor for each cell type was experimentally determined by immunohistochemistry analysis. Trastuzumab-vc-MMAE (T-vc-MMAE) was used as a tool ADC. Plasma and tumor PK of T-vc-MMAE was analyzed in N87, GFP-MCF7, and coculture tumor-bearing mice. In addition, tumor growth inhibition (TGI) studies were conducted in all three xenografts at different T-vc-MMAE dose levels. To characterize the PK of ADC in coculture tumors, our previously published tumor distribution model was evolved to account for different cell populations. The evolved tumor PK model was able to a priori predict the PK of all ADC analytes in the coculture tumors reasonably well. The tumor PK model was subsequently integrated with a PD model that used intracellular tubulin occupancy to drive ADC efficacy in each cell type. The final systems PK-PD model was able to simultaneously characterize all the TGI data reasonably well, with a common set of parameters for MMAE-induced cytotoxicity. The model was later used to simulate the effect of different dosing regimens and tumor compositions on the bystander effect of ADC. The model simulations suggested that dose-fractionation regimen may further improve overall efficacy and bystander effect of ADCs by prolonging the tubulin occupancy in each cell type. SIGNIFICANCE STATEMENT: A PK-PD analysis is presented to understand bystander effect of Trastuzumab-vc-MMAE ADC in antigen (Ag)-low, Ag-high, and coculture (i.e., Ag-high + Ag-low) xenograft mice. This study also describes a novel single cell-level systems PK-PD model to characterize in vivo bystander effect of ADCs. The proposed model can serve as a platform to mathematically characterize multiple cell populations and their interactions in tumor tissues. Our analysis also suggests that fractionated dosing regimen may help improve the bystander effect of ADCs.
Collapse
Affiliation(s)
- Aman P Singh
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences (A.P.S., L.G., A.V., H.-P.C., D.K.S.), Center for Hearing and Deafness, SUNY Eye Institute (G.M.S.), and Department of Biological Sciences (G.G.-L.W.), The State University of New York at Buffalo, Buffalo, New York
| | - Gail M Seigel
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences (A.P.S., L.G., A.V., H.-P.C., D.K.S.), Center for Hearing and Deafness, SUNY Eye Institute (G.M.S.), and Department of Biological Sciences (G.G.-L.W.), The State University of New York at Buffalo, Buffalo, New York
| | - Leiming Guo
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences (A.P.S., L.G., A.V., H.-P.C., D.K.S.), Center for Hearing and Deafness, SUNY Eye Institute (G.M.S.), and Department of Biological Sciences (G.G.-L.W.), The State University of New York at Buffalo, Buffalo, New York
| | - Ashwni Verma
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences (A.P.S., L.G., A.V., H.-P.C., D.K.S.), Center for Hearing and Deafness, SUNY Eye Institute (G.M.S.), and Department of Biological Sciences (G.G.-L.W.), The State University of New York at Buffalo, Buffalo, New York
| | - Gloria Gao-Li Wong
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences (A.P.S., L.G., A.V., H.-P.C., D.K.S.), Center for Hearing and Deafness, SUNY Eye Institute (G.M.S.), and Department of Biological Sciences (G.G.-L.W.), The State University of New York at Buffalo, Buffalo, New York
| | - Hsuan-Ping Cheng
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences (A.P.S., L.G., A.V., H.-P.C., D.K.S.), Center for Hearing and Deafness, SUNY Eye Institute (G.M.S.), and Department of Biological Sciences (G.G.-L.W.), The State University of New York at Buffalo, Buffalo, New York
| | - Dhaval K Shah
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences (A.P.S., L.G., A.V., H.-P.C., D.K.S.), Center for Hearing and Deafness, SUNY Eye Institute (G.M.S.), and Department of Biological Sciences (G.G.-L.W.), The State University of New York at Buffalo, Buffalo, New York
| |
Collapse
|
16
|
Ahlberg J, Giragossian C, Li H, Myzithras M, Raymond E, Caviness G, Grimaldi C, Brown SE, Perez R, Yang D, Kroe-Barrett R, Joseph D, Pamulapati C, Coble K, Ruus P, Woska JR, Ganesan R, Hansel S, Mbow ML. Retrospective analysis of model-based predictivity of human pharmacokinetics for anti-IL-36R monoclonal antibody MAB92 using a rat anti-mouse IL-36R monoclonal antibody and RNA expression data (FANTOM5). MAbs 2019; 11:956-964. [PMID: 31068073 PMCID: PMC6601564 DOI: 10.1080/19420862.2019.1615345] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Accurate prediction of the human pharmacokinetics (PK) of a candidate monoclonal antibody from nonclinical data is critical to maximize the success of clinical trials. However, for monoclonal antibodies exhibiting nonlinear clearance due to target-mediated drug disposition, PK predictions are particularly challenging. That challenge is further compounded for molecules lacking cross-reactivity in a nonhuman primate, in which case a surrogate antibody selective for the target in rodent may be required. For these cases, prediction of human PK must account for any interspecies differences in binding kinetics, target expression, target turnover, and potentially epitope. We present here a model-based method for predicting the human PK of MAB92 (also known as BI 655130), a humanized IgG1 κ monoclonal antibody directed against human IL-36R. Preclinical PK was generated in the mouse with a chimeric rat anti-mouse IgG2a surrogate antibody cross-reactive against mouse IL-36R. Target-specific parameters such as antibody binding affinity (KD), internalization rate of the drug target complex (kint), target degradation rate (kdeg), and target abundance (R0) were integrated into the model. Two different methods of assigning human R0 were evaluated: the first assumed comparable expression between human and mouse and the second used high-resolution mRNA transcriptome data (FANTOM5) as a surrogate for expression. Utilizing the mouse R0 to predict human PK, AUC0-∞ was substantially underpredicted for nonsaturating doses; however, after correcting for differences in RNA transcriptome between species, AUC0-∞ was predicted largely within 1.5-fold of observations in first-in-human studies, demonstrating the validity of the modeling approach. Our results suggest that semi-mechanistic models incorporating RNA transcriptome data and target-specific parameters may improve the predictivity of first-in-human PK.
Collapse
Affiliation(s)
- Jennifer Ahlberg
- a Biotherapeutics Disccovery Research , Boehringer Ingelheim Pharmaceuticals, Inc , Ridgefield , CT , USA
| | - Craig Giragossian
- a Biotherapeutics Disccovery Research , Boehringer Ingelheim Pharmaceuticals, Inc , Ridgefield , CT , USA
| | - Hua Li
- a Biotherapeutics Disccovery Research , Boehringer Ingelheim Pharmaceuticals, Inc , Ridgefield , CT , USA
| | - Maria Myzithras
- a Biotherapeutics Disccovery Research , Boehringer Ingelheim Pharmaceuticals, Inc , Ridgefield , CT , USA
| | - Ernie Raymond
- a Biotherapeutics Disccovery Research , Boehringer Ingelheim Pharmaceuticals, Inc , Ridgefield , CT , USA
| | - Gary Caviness
- a Biotherapeutics Disccovery Research , Boehringer Ingelheim Pharmaceuticals, Inc , Ridgefield , CT , USA
| | - Christine Grimaldi
- a Biotherapeutics Disccovery Research , Boehringer Ingelheim Pharmaceuticals, Inc , Ridgefield , CT , USA
| | - Su-Ellen Brown
- a Biotherapeutics Disccovery Research , Boehringer Ingelheim Pharmaceuticals, Inc , Ridgefield , CT , USA
| | - Rocio Perez
- a Biotherapeutics Disccovery Research , Boehringer Ingelheim Pharmaceuticals, Inc , Ridgefield , CT , USA
| | - Danlin Yang
- a Biotherapeutics Disccovery Research , Boehringer Ingelheim Pharmaceuticals, Inc , Ridgefield , CT , USA
| | - Rachel Kroe-Barrett
- a Biotherapeutics Disccovery Research , Boehringer Ingelheim Pharmaceuticals, Inc , Ridgefield , CT , USA
| | - David Joseph
- a Biotherapeutics Disccovery Research , Boehringer Ingelheim Pharmaceuticals, Inc , Ridgefield , CT , USA
| | - Chandrasena Pamulapati
- a Biotherapeutics Disccovery Research , Boehringer Ingelheim Pharmaceuticals, Inc , Ridgefield , CT , USA
| | - Kelly Coble
- a Biotherapeutics Disccovery Research , Boehringer Ingelheim Pharmaceuticals, Inc , Ridgefield , CT , USA
| | - Peter Ruus
- b Translational Medicine, Clinical Pharmacology , Boehringer Ingelheim Pharma GmbH & Co. KG , Ingelheim am Rein , Germany
| | - Joseph R Woska
- a Biotherapeutics Disccovery Research , Boehringer Ingelheim Pharmaceuticals, Inc , Ridgefield , CT , USA
| | - Rajkumar Ganesan
- a Biotherapeutics Disccovery Research , Boehringer Ingelheim Pharmaceuticals, Inc , Ridgefield , CT , USA
| | - Steven Hansel
- a Biotherapeutics Disccovery Research , Boehringer Ingelheim Pharmaceuticals, Inc , Ridgefield , CT , USA
| | - M Lamine Mbow
- a Biotherapeutics Disccovery Research , Boehringer Ingelheim Pharmaceuticals, Inc , Ridgefield , CT , USA
| |
Collapse
|
17
|
A Cell-Level Systems PK-PD Model to Characterize In Vivo Efficacy of ADCs. Pharmaceutics 2019; 11:pharmaceutics11020098. [PMID: 30823607 PMCID: PMC6409735 DOI: 10.3390/pharmaceutics11020098] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 02/18/2019] [Accepted: 02/20/2019] [Indexed: 01/13/2023] Open
Abstract
Here, we have presented the development of a systems pharmacokinetics-pharmacodynamics (PK-PD) model for antibody-drug conjugates (ADCs), which uses intracellular target occupancy to drive in-vivo efficacy. The model is built based on PK and efficacy data generated using Trastuzumab-Valine-Citrulline-Monomethyl Auristatin E (T-vc-MMAE) ADC in N87 (high-HER2) and GFP-MCF7 (low-HER2) tumor bearing mice. It was observed that plasma PK of all ADC analytes was similar between the two tumor models; however, total trastuzumab, unconjugated MMAE, and total MMAE exposures were >10-fold, ~1.6-fold, and ~1.8-fold higher in N87 tumors. In addition, a prolonged retention of MMAE was observed within the tumors of both the mouse models, suggesting intracellular binding of MMAE to tubulin. A systems PK model, developed by integrating single-cell PK model with tumor distribution model, was able to capture all in vivo PK data reasonably well. Intracellular occupancy of tubulin predicted by the PK model was used to drive the efficacy of ADC using a novel PK-PD model. It was found that the same set of PD parameters was able to capture MMAE induced killing of GFP-MCF7 and N87 cells in vivo. These observations highlight the benefit of adopting a systems approach for ADC and provide a robust and predictive framework for successful clinical translation of ADCs.
Collapse
|
18
|
Ishii-Watabe A, Kuwabara T. Biosimilarity assessment of biosimilar therapeutic monoclonal antibodies. Drug Metab Pharmacokinet 2019; 34:64-70. [DOI: 10.1016/j.dmpk.2018.11.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 11/27/2018] [Accepted: 11/28/2018] [Indexed: 12/11/2022]
|
19
|
Shen J, Swift B, Mamelok R, Pine S, Sinclair J, Attar M. Design and Conduct Considerations for First-in-Human Trials. Clin Transl Sci 2019; 12:6-19. [PMID: 30048046 PMCID: PMC6342261 DOI: 10.1111/cts.12582] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 07/12/2018] [Indexed: 12/14/2022] Open
Abstract
A milestone step in translational science to transform basic scientific discoveries into therapeutic applications is the advancement of a drug candidate from preclinical studies to initial human testing. First-in-human (FIH) trials serve as the link to advance new promising drug candidates and are conducted primarily to determine the safe dose range for further clinical development. Cross-functional collaboration is essential to ensure efficient and successful FIH trials. The aim of this publication is to serve as a tutorial for conducting FIH trials for both small molecule and biological drug candidates with topics covering regulatory requirements, preclinical safety testing, study design considerations, safety monitoring, biomarker assessment, and global considerations. An emphasis is placed on FIH trial design considerations, including starting dose selection, study size and population, dose escalation scheme, and implementation of adaptive designs. In light of the recent revision of the European Medicines Agency (EMA) guideline on FIH trials to promote safety and mitigate risk, we also discuss new measures introduced in the guideline that impact FIH trial design.
Collapse
Affiliation(s)
- Jie Shen
- Clinical PharmacologyAllerganCaliforniaUSA
| | - Brandon Swift
- Clinical PharmacologyRoivant SciencesDurhamNorth CarolinaUSA
| | | | - Samuel Pine
- Bioanalytical OperationsBioAgilytixHamburgGermany
| | - John Sinclair
- Nonclinical DevelopmentKodiak Sciences Inc.Palo AltoCaliforniaUSA
| | - Mayssa Attar
- Non‐Clinical and Translational SciencesAllerganCaliforniaUSA
| |
Collapse
|
20
|
A pre-clinical quantitative model predicts the pharmacokinetics/pharmacodynamics of an anti-BDCA2 monoclonal antibody in humans. J Pharmacokinet Pharmacodyn 2018; 45:817-827. [DOI: 10.1007/s10928-018-9609-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 10/20/2018] [Indexed: 12/12/2022]
|
21
|
Hecht M, Veigure R, Couchman L, S Barker CI, Standing JF, Takkis K, Evard H, Johnston A, Herodes K, Leito I, Kipper K. Utilization of data below the analytical limit of quantitation in pharmacokinetic analysis and modeling: promoting interdisciplinary debate. Bioanalysis 2018; 10:1229-1248. [PMID: 30033744 DOI: 10.4155/bio-2018-0078] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Traditionally, bioanalytical laboratories do not report actual concentrations for samples with results below the LOQ (BLQ) in pharmacokinetic studies. BLQ values are outside the method calibration range established during validation and no data are available to support the reliability of these values. However, ignoring BLQ data can contribute to bias and imprecision in model-based pharmacokinetic analyses. From this perspective, routine use of BLQ data would be advantageous. We would like to initiate an interdisciplinary debate on this important topic by summarizing the current concepts and use of BLQ data by regulators, pharmacometricians and bioanalysts. Through introducing the limit of detection and evaluating its variability, BLQ data could be released and utilized appropriately for pharmacokinetic research.
Collapse
Affiliation(s)
- Max Hecht
- Chair of Analytical Chemistry, Institute of Chemistry, University of Tartu, 14a Ravila Street, 50411 Tartu, Estonia
- Analytical Services International, St George's University of London, Cranmer Terrace, London, SW17 0RE, UK
| | - Rūta Veigure
- Chair of Analytical Chemistry, Institute of Chemistry, University of Tartu, 14a Ravila Street, 50411 Tartu, Estonia
| | - Lewis Couchman
- Analytical Services International, St George's University of London, Cranmer Terrace, London, SW17 0RE, UK
| | - Charlotte I S Barker
- Paediatric Infectious Diseases Research Group, Institute for Infection & Immunity, St George's University of London, London, SW17 0RE, UK
- Inflammation, Infection & Rheumatology Section, UCL Great Ormond Street Institute of Child Health, London, WC1N 1EH, UK
- Paediatric Infectious Diseases Unit, St George's University Hospitals NHS Foundation Trust, London, SW17 0RE, UK
| | - Joseph F Standing
- Paediatric Infectious Diseases Research Group, Institute for Infection & Immunity, St George's University of London, London, SW17 0RE, UK
- Inflammation, Infection & Rheumatology Section, UCL Great Ormond Street Institute of Child Health, London, WC1N 1EH, UK
| | - Kalev Takkis
- Analytical Services International, St George's University of London, Cranmer Terrace, London, SW17 0RE, UK
| | - Hanno Evard
- Chair of Analytical Chemistry, Institute of Chemistry, University of Tartu, 14a Ravila Street, 50411 Tartu, Estonia
| | - Atholl Johnston
- Analytical Services International, St George's University of London, Cranmer Terrace, London, SW17 0RE, UK
- Clinical Pharmacology, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Koit Herodes
- Chair of Analytical Chemistry, Institute of Chemistry, University of Tartu, 14a Ravila Street, 50411 Tartu, Estonia
| | - Ivo Leito
- Chair of Analytical Chemistry, Institute of Chemistry, University of Tartu, 14a Ravila Street, 50411 Tartu, Estonia
| | - Karin Kipper
- Chair of Analytical Chemistry, Institute of Chemistry, University of Tartu, 14a Ravila Street, 50411 Tartu, Estonia
- Analytical Services International, St George's University of London, Cranmer Terrace, London, SW17 0RE, UK
| |
Collapse
|
22
|
Figueroa I, Leipold D, Leong S, Zheng B, Triguero-Carrasco M, Fourie-O'Donohue A, Kozak KR, Xu K, Schutten M, Wang H, Polson AG, Kamath AV. Prediction of non-linear pharmacokinetics in humans of an antibody-drug conjugate (ADC) when evaluation of higher doses in animals is limited by tolerability: Case study with an anti-CD33 ADC. MAbs 2018; 10:738-750. [PMID: 29757698 PMCID: PMC6150628 DOI: 10.1080/19420862.2018.1465160] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 04/03/2018] [Accepted: 04/09/2018] [Indexed: 11/01/2022] Open
Abstract
For antibody-drug conjugates (ADCs) that carry a cytotoxic drug, doses that can be administered in preclinical studies are typically limited by tolerability, leading to a narrow dose range that can be tested. For molecules with non-linear pharmacokinetics (PK), this limited dose range may be insufficient to fully characterize the PK of the ADC and limits translation to humans. Mathematical PK models are frequently used for molecule selection during preclinical drug development and for translational predictions to guide clinical study design. Here, we present a practical approach that uses limited PK and receptor occupancy (RO) data of the corresponding unconjugated antibody to predict ADC PK when conjugation does not alter the non-specific clearance or the antibody-target interaction. We used a 2-compartment model incorporating non-specific and specific (target mediated) clearances, where the latter is a function of RO, to describe the PK of anti-CD33 ADC with dose-limiting neutropenia in cynomolgus monkeys. We tested our model by comparing PK predictions based on the unconjugated antibody to observed ADC PK data that was not utilized for model development. Prospective prediction of human PK was performed by incorporating in vitro binding affinity differences between species for varying levels of CD33 target expression. Additionally, this approach was used to predict human PK of other previously tested anti-CD33 molecules with published clinical data. The findings showed that, for a cytotoxic ADC with non-linear PK and limited preclinical PK data, incorporating RO in the PK model and using data from the corresponding unconjugated antibody at higher doses allowed the identification of parameters to characterize monkey PK and enabled human PK predictions.
Collapse
Affiliation(s)
| | - Doug Leipold
- Preclinical Translational Pharmacokinetics Department
| | | | | | | | | | | | | | - Melissa Schutten
- Safety Assessment Department Genentech Inc., South San Francisco, CA, USA
| | - Hong Wang
- Safety Assessment Department Genentech Inc., South San Francisco, CA, USA
| | | | | |
Collapse
|
23
|
Betts A, Keunecke A, van Steeg TJ, van der Graaf PH, Avery LB, Jones H, Berkhout J. Linear pharmacokinetic parameters for monoclonal antibodies are similar within a species and across different pharmacological targets: A comparison between human, cynomolgus monkey and hFcRn Tg32 transgenic mouse using a population-modeling approach. MAbs 2018; 10:751-764. [PMID: 29634430 PMCID: PMC6150614 DOI: 10.1080/19420862.2018.1462429] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The linear pharmacokinetics (PK) of therapeutic monoclonal antibodies (mAbs) can be considered a class property with values that are similar to endogenous IgG. Knowledge of these parameters across species could be used to avoid unnecessary in vivo PK studies and to enable early PK predictions and pharmacokinetic/pharmacodynamic (PK/PD) simulations. In this work, population-pharmacokinetic (popPK) modeling was used to determine a single set of ‘typical’ popPK parameters describing the linear PK of mAbs in human, cynomolgus monkey and transgenic mice expressing the human neonatal Fc receptor (hFcRn Tg32), using a rich dataset of 27 mAbs. Non-linear PK was excluded from the datasets and a 2-compartment model was applied to describe mAb disposition. Typical human popPK estimates compared well with data from comparator mAbs with linear PK in the clinic. Outliers with higher than typical clearance were found to have non-specific interactions in an affinity-capture self-interaction nanoparticle spectroscopy assay, offering a potential tool to screen out these mAbs at an early stage. Translational strategies were investigated for prediction of human linear PK of mAbs, including use of typical human popPK parameters and allometric exponents from cynomolgus monkey and Tg32 mouse. Each method gave good prediction of human PK with parameters predicted within 2-fold. These strategies offer alternative options to the use of cynomolgus monkeys for human PK predictions of linear mAbs, based on in silico methods (typical human popPK parameters) or using a rodent species (Tg32 mouse), and call into question the value of completing extensive in vivo preclinical PK to inform linear mAb PK.
Collapse
Affiliation(s)
- Alison Betts
- a Department of Biomedicine Design , Pfizer Inc. , Cambridge , MA , USA
| | | | | | - Piet H van der Graaf
- c Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research , RA Leiden , The Netherlands
| | - Lindsay B Avery
- d Department of Biomedicine Design , Pfizer Inc. , Andover , MA , USA
| | - Hannah Jones
- a Department of Biomedicine Design , Pfizer Inc. , Cambridge , MA , USA
| | | |
Collapse
|
24
|
de Vries Schultink AHM, Doornbos RP, Bakker ABH, Bol K, Throsby M, Geuijen C, Maussang D, Schellens JHM, Beijnen JH, Huitema ADR. Translational PK-PD modeling analysis of MCLA-128, a HER2/HER3 bispecific monoclonal antibody, to predict clinical efficacious exposure and dose. Invest New Drugs 2018; 36:1006-1015. [PMID: 29728897 PMCID: PMC6244972 DOI: 10.1007/s10637-018-0593-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 03/19/2018] [Indexed: 01/15/2023]
Abstract
Introduction MCLA-128 is a bispecific monoclonal antibody targeting the HER2 and HER3 receptors. Pharmacokinetics (PK) and pharmacodynamics (PD) of MCLA-128 have been evaluated in preclinical studies in cynomolgus monkeys and mice. The aim of this study was to characterize the PK and PD of MCLA-128 and to predict a safe starting dose and efficacious clinical dose for the First-In-Human study. Methods A PK-PD model was developed based on PK data from cynomolgus monkeys and tumor growth data from a mouse JIMT-1 xenograft model. Allometric scaling was used to scale PK parameters between species. Simulations were performed to predict the safe and efficacious clinical dose, based on AUCs, receptor occupancies and PK-PD model simulations. Results MCLA-128 PK in cynomolgus monkeys was described by a two-compartment model with parallel linear and nonlinear clearance. The xenograft tumor growth model consisted of a tumor compartment with a zero-order growth rate and a first-order dying rate, both affected by MCLA-128. Human doses of 10 to 480 mg q3wk were predicted to show a safety margin of >10-fold compared to the cynomolgus monkey AUC at the no-observed-adverse-effect-level (NOAEL). Doses of ≥360 mg resulted in predicted receptor occupancies above 99% (Cmax and Cave). These doses showed anti-tumor efficacy in the PK-PD model. Conclusions This analysis predicts that a flat dose of 10 to 480 mg q3wk is suitable as starting dose for a First-in-Human study with MCLA-128. Flat doses ≥360 mg q3wk are expected to be efficacious in human, based on receptor occupancies and PK-PD model simulations.
Collapse
Affiliation(s)
- Aurelia H M de Vries Schultink
- Department of Pharmacy & Pharmacology, Antoni van Leeuwenhoek - The Netherlands Cancer Institute and MC Slotervaart, Louwesweg 6, 1066, EC, Amsterdam, the Netherlands.
| | | | | | - Kees Bol
- Merus N.V, Yalelaan 62, 3584, CM, Utrecht, the Netherlands
| | - Mark Throsby
- Merus N.V, Yalelaan 62, 3584, CM, Utrecht, the Netherlands
| | - Cecile Geuijen
- Merus N.V, Yalelaan 62, 3584, CM, Utrecht, the Netherlands
| | - David Maussang
- Merus N.V, Yalelaan 62, 3584, CM, Utrecht, the Netherlands
| | - Jan H M Schellens
- Department of Clinical Pharmacology, Antoni van Leeuwenhoek - The Netherlands Cancer Institute, P.O Box 90203, 1006, BE, Amsterdam, the Netherlands.,Science Faculty, Utrecht Institute for Pharmaceutical Sciences (UIPS), Division of Pharmacoepidemiology & Clinical Pharmacology, Utrecht University, P.O. Box 80082, 3508, TB, Utrecht, the Netherlands
| | - Jos H Beijnen
- Department of Pharmacy & Pharmacology, Antoni van Leeuwenhoek - The Netherlands Cancer Institute and MC Slotervaart, Louwesweg 6, 1066, EC, Amsterdam, the Netherlands. .,Science Faculty, Utrecht Institute for Pharmaceutical Sciences (UIPS), Division of Pharmacoepidemiology & Clinical Pharmacology, Utrecht University, P.O. Box 80082, 3508, TB, Utrecht, the Netherlands.
| | - Alwin D R Huitema
- Department of Pharmacy & Pharmacology, Antoni van Leeuwenhoek - The Netherlands Cancer Institute and MC Slotervaart, Louwesweg 6, 1066, EC, Amsterdam, the Netherlands.,Department of Clinical Pharmacy, University Medical Center Utrecht, P.O. Box 85500, 3508, GA, Utrecht, the Netherlands
| |
Collapse
|
25
|
Apgar JF, Tang JP, Singh P, Balasubramanian N, Burke J, Hodges MR, Lasaro MA, Lin L, Millard BL, Moore K, Jun LS, Sobolov S, Wilkins AK, Gao X. Quantitative Systems Pharmacology Model of hUGT1A1-modRNA Encoding for the UGT1A1 Enzyme to Treat Crigler-Najjar Syndrome Type 1. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2018; 7:404-412. [PMID: 29637732 PMCID: PMC6391595 DOI: 10.1002/psp4.12301] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 03/19/2018] [Accepted: 03/21/2018] [Indexed: 12/15/2022]
Abstract
Crigler‐Najjar syndrome type 1 (CN1) is an autosomal recessive disease caused by a marked decrease in uridine‐diphosphate‐glucuronosyltransferase (UGT1A1) enzyme activity. Delivery of hUGT1A1‐modRNA (a modified messenger RNA encoding for UGT1A1) as a lipid nanoparticle is anticipated to restore hepatic expression of UGT1A1, allowing normal glucuronidation and clearance of bilirubin in patients. To support translation from preclinical to clinical studies, and first‐in‐human studies, a quantitative systems pharmacology (QSP) model was developed. The QSP model was calibrated to plasma and liver mRNA, and total serum bilirubin in Gunn rats, an animal model of CN1. This QSP model adequately captured the observed plasma and liver biomarker behavior across a range of doses and dose regimens in Gunn rats. First‐in‐human dose projections made using the translated model indicated that 0.5 mg/kg Q4W dose should provide a clinically meaningful and sustained reduction of >5 mg/dL in total bilirubin levels.
Collapse
Affiliation(s)
| | - Jian-Ping Tang
- Alexion Pharmaceuticals, Inc., New Haven, Connecticut, USA
| | - Pratap Singh
- Alexion Pharmaceuticals, Inc., New Haven, Connecticut, USA
| | | | - John Burke
- Applied BioMath, Lincoln, Massachusetts, USA
| | | | | | - Lin Lin
- Applied BioMath, Lincoln, Massachusetts, USA
| | | | - Kristi Moore
- Alexion Pharmaceuticals, Inc., New Haven, Connecticut, USA
| | - Lucy S Jun
- Alexion Pharmaceuticals, Inc., New Haven, Connecticut, USA
| | - Susan Sobolov
- Alexion Pharmaceuticals, Inc., New Haven, Connecticut, USA
| | | | - Xiang Gao
- Alexion Pharmaceuticals, Inc., New Haven, Connecticut, USA
| |
Collapse
|
26
|
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: 1.3] [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
|
27
|
Singh AP, Shah DK. Application of a PK-PD Modeling and Simulation-Based Strategy for Clinical Translation of Antibody-Drug Conjugates: a Case Study with Trastuzumab Emtansine (T-DM1). AAPS JOURNAL 2017; 19:1054-1070. [PMID: 28374319 DOI: 10.1208/s12248-017-0071-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 02/28/2017] [Indexed: 02/06/2023]
Abstract
Successful clinical translation of antibody-drug conjugates (ADCs) can be challenging due to complex pharmacokinetics and differences between preclinical and clinical tumors. To facilitate this translation, we have developed a general pharmacokinetic-pharmacodynamic (PK-PD) modeling and simulation (M&S)-based strategy for ADCs. Here we present the validation of this strategy using T-DM1 as a case study. A previously developed preclinical tumor disposition model for T-DM1 (Singh and Shah, AAPSJ. 2015; 18(4):861-875) was used to develop a PK-PD model that can characterize in vivo efficacy of T-DM1 in preclinical tumor models. The preclinical data was used to estimate the efficacy parameters for T-DM1. Human PK of T-DM1 was a priori predicted using allometric scaling of monkey PK parameters. The predicted human PK, preclinically estimated efficacy parameters, and clinically observed volume and growth parameters for breast cancer were combined to develop a translated clinical PK-PD model for T-DM1. Clinical trial simulations were performed using the translated PK-PD model to predict progression-free survival (PFS) and objective response rates (ORRs) for T-DM1. The model simulated PFS rates for HER2 1+ and 3+ populations were comparable to the rates observed in three different clinical trials. The model predicted only a modest improvement in ORR with an increase in clinically approved dose of T-DM1. However, the model suggested that a fractionated dosing regimen (e.g., front loading) may provide an improvement in the efficacy. In general, the PK-PD M&S-based strategy presented here is capable of a priori predicting the clinical efficacy of ADCs, and this strategy has been now retrospectively validated for all clinically approved ADCs.
Collapse
Affiliation(s)
- Aman P Singh
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, The State University of New York at Buffalo, 455 Kapoor Hall, Buffalo, New York, 14214-8033, USA
| | - Dhaval K Shah
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, The State University of New York at Buffalo, 455 Kapoor Hall, Buffalo, New York, 14214-8033, USA.
| |
Collapse
|
28
|
Tiwari A, Abraham AK, Harrold JM, Zutshi A, Singh P. Optimal Affinity of a Monoclonal Antibody: Guiding Principles Using Mechanistic Modeling. AAPS JOURNAL 2016; 19:510-519. [PMID: 28004347 DOI: 10.1208/s12248-016-0004-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 10/07/2016] [Indexed: 12/21/2022]
Abstract
Affinity optimization of monoclonal antibodies (mAbs) is essential for developing drug candidates with the highest likelihood of clinical success; however, a quantitative approach for setting affinity requirements is often lacking. In this study, we computationally analyzed the in vivo mAb-target binding kinetics to delineate general principles for defining optimal equilibrium dissociation constant ([Formula: see text]) of mAbs against soluble and membrane-bound targets. Our analysis shows that in general [Formula: see text] to achieve 90% coverage for a soluble target is one tenth of its baseline concentration ([Formula: see text]), and is independent of the dosing interval, target turnover rate or the presence of competing ligands. For membrane-bound internalizing targets, it is equal to the ratio of internalization rate of mAb-target complex and association rate constant ([Formula: see text]). In cases where soluble and membrane-bound forms of the target co-exist, [Formula: see text] lies within a range determined by the internalization rate ([Formula: see text]) of the mAb-membrane target complex and the ratio of baseline concentrations of soluble and membrane-bound forms ([Formula: see text]). Finally, to demonstrate practical application of these general rules, we collected target expression and turnover data to project [Formula: see text] for a number of marketed mAbs against soluble (TNFα, RANKL, and VEGF) and membrane-bound targets (CD20, EGFR, and HER2).
Collapse
Affiliation(s)
- Abhinav Tiwari
- Pharmacokinetics, Dynamics and Metabolism, Pfizer, Cambridge, Massachusetts, USA
| | - Anson K Abraham
- Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck, West Point, Pennsylvania, USA
| | - John M Harrold
- Department of Pharmacokinetics and Drug Metabolism, Amgen, South San Francisco, California, USA
| | | | - Pratap Singh
- Pharmacokinetics, Dynamics and Metabolism, Pfizer, Cambridge, Massachusetts, USA.
| |
Collapse
|
29
|
Park W, Han S, Lee J, Hong T, Won J, Lim Y, Lee K, Byun HY, Yim D. Use of a Target‐Mediated Drug Disposition Model to Predict the Human Pharmacokinetics and Target Occupancy of
GC
1118, an Anti‐epidermal Growth Factor Receptor Antibody. Basic Clin Pharmacol Toxicol 2016; 120:243-249. [DOI: 10.1111/bcpt.12675] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 09/05/2016] [Indexed: 12/12/2022]
Affiliation(s)
- Wan‐Su Park
- Department of Clinical Pharmacology and Therapeutics Seoul St. Mary's Hospital Seoul Korea
- PIPET (Pharmacometrics Institute for Practical Education and Training) College of Medicine The Catholic University of Korea Seoul Korea
| | - Seunghoon Han
- Department of Clinical Pharmacology and Therapeutics Seoul St. Mary's Hospital Seoul Korea
- PIPET (Pharmacometrics Institute for Practical Education and Training) College of Medicine The Catholic University of Korea Seoul Korea
| | - Jongtae Lee
- Department of Clinical Pharmacology and Therapeutics Seoul St. Mary's Hospital Seoul Korea
- PIPET (Pharmacometrics Institute for Practical Education and Training) College of Medicine The Catholic University of Korea Seoul Korea
| | - Taegon Hong
- Department of Clinical Pharmacology Severance Hospital, Yonsei University College of Medicine Seoul Korea
| | - Jonghwa Won
- MOGAM Biotechnology Institute Yongin Gyeonggi‐do Korea
| | - Yangmi Lim
- MOGAM Biotechnology Institute Yongin Gyeonggi‐do Korea
| | - Kyuhyun Lee
- MOGAM Biotechnology Institute Yongin Gyeonggi‐do Korea
| | | | - Dong‐Seok Yim
- Department of Clinical Pharmacology and Therapeutics Seoul St. Mary's Hospital Seoul Korea
- PIPET (Pharmacometrics Institute for Practical Education and Training) College of Medicine The Catholic University of Korea Seoul Korea
| |
Collapse
|
30
|
Singh P, Rong H, Gordi T, Bosley J, Bhattacharya I. Translational Pharmacokinetic/Pharmacodynamic Analysis of MYO-029 Antibody for Muscular Dystrophy. Clin Transl Sci 2016; 9:302-310. [PMID: 27700008 PMCID: PMC5351001 DOI: 10.1111/cts.12420] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 08/29/2016] [Indexed: 12/30/2022] Open
Abstract
Suppression of the myostatin (GDF‐8) pathway has emerged as an important therapeutic paradigm for muscle‐wasting disorders. In this study, we conducted a translational pharmacokinetic/pharmacodynamic (PK/PD) analysis of MYO‐029, an anti‐myostatin monoclonal antibody, using PK data in mice, rats, monkeys, humans, mouse tissue distribution data with 125I‐labeled MYO‐029, muscle weight increase in SCID mice, and muscle circumference changes in monkeys. This analysis revealed significant in vivo potency shift between mice and monkeys (72 nM vs. 1.3 μM for 50% effect on quadriceps). Estimated central clearance of MYO‐029 (0.38 mL/h/kg) in humans was greater than twofold higher than typical IgG mAbs. Peak and trough steady‐state exposures of MYO‐029 in patients at biweekly intravenous doses of 10 mg/kg MYO‐029 are predicted to achieve only 50% and 10% of the maximum effect seen in monkeys, respectively. These retrospective analyses results suggest that the MYO‐029 exposures in this trial had a low probability of producing robust efficacy.
Collapse
Affiliation(s)
- P Singh
- Pharmacokinetics, Dynamics, and Metabolism, Pfizer, Inc, Cambridge, Massachusetts, USA
| | - H Rong
- Shire Pharmaceutical, Lexington, Massachusetts, USA
| | - T Gordi
- Nektar Therapeutics, Inc, San Francisco, California, USA
| | - J Bosley
- Clermont Bosley LLC, Kennett Square, Pennsylvania, USA
| | - I Bhattacharya
- Clinical Pharmacology, Pfizer, Inc, Cambridge, Massachusetts, USA
| |
Collapse
|
31
|
FG-3019, a Human Monoclonal Antibody Recognizing Connective Tissue Growth Factor, is Subject to Target-Mediated Drug Disposition. Pharm Res 2016; 33:1833-49. [PMID: 27059922 PMCID: PMC4942499 DOI: 10.1007/s11095-016-1918-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 03/30/2016] [Indexed: 12/27/2022]
Abstract
Purpose To evaluate and model the pharmacokinetic and pharmacodynamic behavior in rats of FG-3019, a human monoclonal antibody targeting connective tissue growth factor (CTGF). Methods FG-3019, human CTGF (rhCTGF), or the N-terminal domain of rhCTGF were administered intravenously to rats and concentrations of these proteins as well as endogenous CTGF were determined by immunoassays. FG-3019, or 125I-labeled FG-3019, and human CTGF (rhCTGF) were co-administered to assess the impact of CTGF on the elimination rate and tissue localization of FG-3019, which was further characterized by immunohistochemical analysis. A PK/PD model for target-mediated elimination of FG-3019 was developed to fit the kinetic data. Results FG-3019 exhibited non-linear pharmacokinetics in rats. Circulating concentrations of the N-terminal half of CTGF increased after dosing with FG-3019, reached maximal levels after 1–5 days, and returned toward baseline levels as FG-3019 cleared from the circulation, whereas the concentration of intact CTGF was unaffected by administration of FG-3019. Co-administration of rhCTGF dramatically enhanced the rate of FG-3019 elimination, redistributing the majority of 125I-labeled FG-3019 from the blood to the liver, kidney, spleen and adrenal gland. FG-3019 co-administered with CTGF was found along the sinusoids of the liver and adrenal glands, the capillaries of the kidney glomeruli and in the spleen. A pharmacokinetic model for target-mediated elimination of FG-3019 was used to fit the time courses of FG-3019 and endogenous CTGF plasma concentrations, as well as time courses of rhCTGF and rhCTGF N-fragment after intravenous administration of these species. Conclusions FG-3019 is subject to target mediated elimination in rats. Electronic supplementary material The online version of this article (doi:10.1007/s11095-016-1918-0) contains supplementary material, which is available to authorized users.
Collapse
|
32
|
Spilker ME, Singh P, Vicini P. Mathematical modeling of receptor occupancy data: A valuable technology for biotherapeutic drug development. CYTOMETRY PART B-CLINICAL CYTOMETRY 2015; 90:230-6. [PMID: 26296748 DOI: 10.1002/cyto.b.21318] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Revised: 07/31/2015] [Accepted: 08/18/2015] [Indexed: 12/16/2022]
Abstract
BACKGROUND In drug development, in vivo assessment of target engagement provides confidence when testing the drug's mechanism of action and improves the likelihood of clinical success. For biologics, receptor occupancy (RO) determined from circulating cells can provide evidence of target engagement. Integrating this information with mathematical modeling can further enhance the understanding of drug-target interactions and the biological factors that are critical to the successful modulation of the target and ultimately the disease state. METHODS This mini-review presents two specific types of mathematical models used to describe antibody-receptor systems and highlights how experimental data can inform the model parameters. Simulations are used to illustrate how various mechanisms influence RO, PK and total cellular receptor profiles. RESULTS The simulations demonstrate the effect antibody-receptor internalization, affinity and receptor turnover have on commonly acquired data in drug development. CONCLUSIONS Integrating RO data with mathematical models such as the two presented here (target-mediated drug disposition and site-of-action models) can provide a more comprehensive view of the biological system, which can be used to test hypotheses, extrapolate preclinical findings to humans and impact clinical study designs and risk assessments for the successful development of biotherapeutics.
Collapse
Affiliation(s)
- Mary E Spilker
- Department of Pharmacokinetics, Dynamics and Metabolism-New Biological Entities, Pfizer Worldwide Research and Development, San Diego, California
| | - Pratap Singh
- Department of Pharmacokinetics, Dynamics and Metabolism-New Biological Entities, Pfizer Worldwide Research and Development, Cambridge, Massachusetts
| | - Paolo Vicini
- Department of Pharmacokinetics, Dynamics and Metabolism-New Biological Entities, Pfizer Worldwide Research and Development, San Diego, California
- Clinical Pharmacology, Drug Metabolism and Pharmacokinetics, MedImmune, Cambridge, UK
| |
Collapse
|
33
|
Talbot JJ, Calamba D, Pai M, Ma M, Thway TM. Measurement of Free Versus Total Therapeutic Monoclonal Antibody in Pharmacokinetic Assessment is Modulated by Affinity, Incubation Time, and Bioanalytical Platform. AAPS JOURNAL 2015; 17:1446-54. [PMID: 26265093 DOI: 10.1208/s12248-015-9807-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 07/15/2015] [Indexed: 01/13/2023]
Abstract
Decisions about efficacy and safety of therapeutic proteins (TP) designed to target soluble ligands are made in part by their ex vivo quantification. Ligand binding assays (LBAs) are critical tools in measuring serum TP levels in pharmacokinetic, toxicokinetic, and pharmacodynamic studies. This study evaluated the impact of reagent antibody affinities, assay incubation times, and analytical platform on free or total TP quantitation. An ELISA-based LBA that measures monoclonal anti-sclerostin antibody (TPx) was used as the model system. To determine whether the method measures free or total TPx, the effects of K on, K off, and K D were determined. An 8:1 molar ratio of sclerostin (Scl) to TPx compared to a 1:1 molar ratio produced by rabbit polyclonal antibodies to TPx was required to achieve IC50, a measure of TPx interference effectiveness, making it unclear whether the ELISA truly measured free TPx. Kinetic analysis revealed that Scl had a rapid dissociation rate (K off) from TPx and that capture and detection antibodies had significantly higher binding affinities (K D) to TPx. These kinetic limitations along with long ELISA incubation times lead to the higher molar ratios (8:1) required for achieving 50% inhibition of TPx. However, a microfluidic platform with the same reagent pairs required shorter incubations to achieve a lower Scl IC50 molar ratio (1:1). The findings from this study provide the bioanalytical community with a deeper understanding of how reagent and platform selection for LBAs can affect what a particular method measures, either free or total TP concentrations.
Collapse
Affiliation(s)
- Jeffrey J Talbot
- Department of Pharmacokinetic and Drug Metabolism, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California, 91320, USA.,Regeneron Pharmaceuticals, 81 Columbia Turnpike, Rensselaer, New York, 12144, USA
| | - Dominador Calamba
- Department of Pharmacokinetic and Drug Metabolism, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California, 91320, USA
| | - Melody Pai
- Department of Pharmacokinetic and Drug Metabolism, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California, 91320, USA.,Department of Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California, USA
| | - Mark Ma
- Department of Pharmacokinetic and Drug Metabolism, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California, 91320, USA
| | - Theingi M Thway
- Department of Pharmacokinetic and Drug Metabolism, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California, 91320, USA.
| |
Collapse
|