101
|
Piche-Nicholas NM, Avery LB, King AC, Kavosi M, Wang M, O'Hara DM, Tchistiakova L, Katragadda M. Changes in complementarity-determining regions significantly alter IgG binding to the neonatal Fc receptor (FcRn) and pharmacokinetics. MAbs 2017; 10:81-94. [PMID: 28991504 PMCID: PMC5800364 DOI: 10.1080/19420862.2017.1389355] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
A large body of data exists demonstrating that neonatal Fc receptor (FcRn) binding of an IgG via its Fc CH2-CH3 interface trends with the pharmacokinetics (PK) of IgG. We have observed that PK of IgG molecules vary widely, even when they share identical Fc domains. This led us to hypothesize that domains distal from the Fc could contribute to FcRn binding and affect PK. In this study, we explored the role of these IgG domains in altering the affinity between IgG and FcRn. Using a surface plasmon resonance-based assay developed to examine the steady-state binding affinity (KD) of IgG molecules to FcRn, we dissected the contributions of IgG domains in modulating the affinity between FcRn and IgG. Through analysis of a broad collection of therapeutic antibodies containing more than 50 unique IgG molecules, we demonstrated that variable domains, and in particular complementarity-determining regions (CDRs), significantly alter binding affinity to FcRn in vitro. Furthermore, a panel of IgG molecules differing only by 1–5 mutations in CDRs altered binding affinity to FcRn in vitro, by up to 79-fold, and the affinity values correlated with calculated isoelectric point values of both variable domains and CDR-L3. In addition, tighter affinity values trend with faster in vivo clearance of a set of IgG molecules differing only by 1–3 mutations in human FcRn transgenic mice. Understanding the role of CDRs in modulation of IgG affinity to FcRn in vitro and their effect on PK of IgG may have far-reaching implications in the optimization of IgG therapeutics.
Collapse
Affiliation(s)
| | | | - Amy C King
- a BioMedicine Design, Pfizer Inc. , Cambridge , MA , USA
| | - Mania Kavosi
- b BioMedicine Design, Pfizer Inc. , Andover , MA , USA
| | - Mengmeng Wang
- b BioMedicine Design, Pfizer Inc. , Andover , MA , USA
| | | | | | | |
Collapse
|
102
|
Klinger M. A role for macromolecular crowding in off-target binding of therapeutic antibodies. Protein Eng Des Sel 2017; 30:489-494. [PMID: 28873984 DOI: 10.1093/protein/gzx035] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 06/21/2017] [Indexed: 01/09/2023] Open
Abstract
The nonspecific binding of certain therapeutic antibodies to tissues or to soluble biomolecules can accelerate their clearance from the circulation and undermine their benefit to patients. This article proposes that tandem amino acid repeat sequences in antibody hypervariable segments, particularly the complementarity determining regions (CDRs), can enhance this off-target binding. This hypothesis is based on two sets of observations. First, in a limited number of cases, antibodies with clusters of amino acid repeats in their CDRs have significantly higher clearance rates in experimental animals than otherwise identical antibodies without the repeats. Second, tandem amino acid repeats are abundant in intracellular hub proteins where they appear to promote the promiscuous binding of these proteins to a wide variety of other molecules. These nonspecific hub protein interactions are highly favored by the intense macromolecular crowding that permeates the cytoplasm. A survey of the variable region sequences of 137 antibodies in various stages of development revealed that 26 have at least one CDR containing a cluster of three closely spaced amino acid repeats. If the overall hypothesis is valid, then it suggests strategies for site-directed mutagenesis to improve pharmacokinetic behavior and for the design of more reliable in vitro binding assays to predict off-target binding in vivo.
Collapse
Affiliation(s)
- Martin Klinger
- Hawk BioDiscovery, 7465 Highway 51, Sterrett, AL 35147, USA
| |
Collapse
|
103
|
Abstract
Efforts to develop effective antibody therapeutics are frequently hampered by issues such as aggregation and nonspecificity, often only detected in late stages of the development process. In this study, we used a high throughput cross-reactivity assay to select nonspecific clones from a naïve human repertoire scFv library displayed on the surface of yeast. Most antibody families were de-enriched; however, the rarely expressed VH6 family was highly enriched among nonspecific clones, representing almost 90% of isolated clones. Mutational analysis of this family reveals a dominant role of CDRH2 in driving nonspecific binding. Homology modeling of a panel of VH6 antibodies shows a constrained β-sheet structure in CDRH2 that is not present in other families, potentially contributing to nonspecificity of the family. These findings confirm the common decision to exclude VH6 from synthetic antibody libraries, and support VH6 polyreactivity as a possible important role for the family in early ontogeny and cause for its overabundance in cases of some forms of autoimmunity.
Collapse
Affiliation(s)
- Ryan L Kelly
- a Department of Biological Engineering, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology , Cambridge , MA , U.S.A
| | - Jessie Zhao
- b Department of Chemical Engineering, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology , Cambridge , MA , U.S.A
| | - Doris Le
- b Department of Chemical Engineering, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology , Cambridge , MA , U.S.A
| | - K Dane Wittrup
- a Department of Biological Engineering, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology , Cambridge , MA , U.S.A.,b Department of Chemical Engineering, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology , Cambridge , MA , U.S.A
| |
Collapse
|
104
|
Hutt M, Marquardt L, Seifert O, Siegemund M, Müller I, Kulms D, Pfizenmaier K, Kontermann RE. Superior Properties of Fc-comprising scTRAIL Fusion Proteins. Mol Cancer Ther 2017; 16:2792-2802. [PMID: 28904131 DOI: 10.1158/1535-7163.mct-17-0551] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 08/11/2017] [Accepted: 09/05/2017] [Indexed: 11/16/2022]
Abstract
The TNF-related apoptosis-inducing ligand (TRAIL) has been considered as a promising molecule for cancer treatment. However, clinical studies with soluble TRAIL failed to show therapeutic activity, which resulted in subsequent development of more potent TRAIL-based therapeutics. In this study, we applied defined oligomerization and tumor targeting as strategies to further improve the activity of a single-chain version of TRAIL (scTRAIL). We compared three different formats of EGF receptor (EGFR)-targeting dimeric scTRAIL fusion proteins [Diabody (Db)-scTRAIL, scFv-IgE heavy chain domain 2 (EHD2)-scTRAIL, scFv-Fc-scTRAIL] as well as two nontargeted dimeric scTRAIL molecules (EHD2-scTRAIL, Fc-scTRAIL) to reveal the influence of targeting and protein format on antitumor activity. All EGFR-targeted dimeric scTRAIL molecules showed similar binding properties and comparable cell death induction in vitro, exceeding the activity of the respective nontargeted dimeric format and monomeric scTRAIL. Superior properties were observed for the Fc fusion proteins with respect to production and in vivo half-life. In vivo studies using a Colo205 xenograft model revealed potent antitumor activity of all EGFR-targeting formats and Fc-scTRAIL and furthermore highlighted the higher efficacy of fusion proteins comprising an Fc part. Despite enhanced in vitro cell death induction of targeted scTRAIL molecules, however, comparable antitumor activities were found for the EGFR-targeting scFv-Fc-scTRAIL and the nontargeting Fc-scTRAIL in vivoMol Cancer Ther; 16(12); 2792-802. ©2017 AACR.
Collapse
Affiliation(s)
- Meike Hutt
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Lisa Marquardt
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Oliver Seifert
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Martin Siegemund
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Ines Müller
- Experimental Dermatology, Department of Dermatology, Technische Universität Dresden, Dresden, Germany
- Center for Regenerative Therapies Technische Universität Dresden, Dresden, Germany
| | - Dagmar Kulms
- Experimental Dermatology, Department of Dermatology, Technische Universität Dresden, Dresden, Germany
- Center for Regenerative Therapies Technische Universität Dresden, Dresden, Germany
| | - Klaus Pfizenmaier
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
- Stuttgart Research Center Systems Biology, University of Stuttgart, Stuttgart, Germany
| | - Roland E Kontermann
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany.
- Stuttgart Research Center Systems Biology, University of Stuttgart, Stuttgart, Germany
| |
Collapse
|
105
|
Dostalek M, Prueksaritanont T, Kelley RF. Pharmacokinetic de-risking tools for selection of monoclonal antibody lead candidates. MAbs 2017; 9:756-766. [PMID: 28463063 DOI: 10.1080/19420862.2017.1323160] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023] Open
Abstract
Pharmacokinetic studies play an important role in all stages of drug discovery and development. Recent advancements in the tools for discovery and optimization of therapeutic proteins have created an abundance of candidates that may fulfill target product profile criteria. Implementing a set of in silico, small scale in vitro and in vivo tools can help to identify a clinical lead molecule with promising properties at the early stages of drug discovery, thus reducing the labor and cost in advancing multiple candidates toward clinical development. In this review, we describe tools that should be considered during drug discovery, and discuss approaches that could be included in the pharmacokinetic screening part of the lead candidate generation process to de-risk unexpected pharmacokinetic behaviors of Fc-based therapeutic proteins, with an emphasis on monoclonal antibodies.
Collapse
Affiliation(s)
- Miroslav Dostalek
- a Drug Metabolism and Pharmacokinetics, Global Nonclinical Development, Shire , Lexington , MA , USA
| | | | - Robert F Kelley
- c Department of Drug Delivery , Genentech Inc. , South San Francisco , CA , USA
| |
Collapse
|
106
|
Jaramillo CAC, Belli S, Cascais AC, Dudal S, Edelmann MR, Haak M, Brun ME, Otteneder MB, Ullah M, Funk C, Schuler F, Simon S. Toward in vitro-to-in vivo translation of monoclonal antibody pharmacokinetics: Application of a neonatal Fc receptor-mediated transcytosis assay to understand the interplaying clearance mechanisms. MAbs 2017; 9:781-791. [PMID: 28440708 DOI: 10.1080/19420862.2017.1320008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
Monoclonal antibodies (mAbs) are a rapidly growing drug class for which great efforts have been made to optimize certain molecular features to achieve the desired pharmacokinetic (PK) properties. One approach is to engineer the interactions of the mAb with the neonatal Fc receptor (FcRn) by introducing specific amino acid sequence mutations, and to assess their effect on the PK profile with in vivo studies. Indeed, FcRn protects mAbs from intracellular degradation, thereby prolongs antibody circulation time in plasma and modulates its systemic clearance. To allow more efficient and focused mAb optimization, in vitro input that helps to identify and quantitatively predict the contribution of different processes driving non-target mediated mAb clearance in vivo and supporting translational PK modeling activities is essential. With this aim, we evaluated the applicability and in vivo-relevance of an in vitro cellular FcRn-mediated transcytosis assay to explain the PK behavior of 25 mAbs in rat or monkey. The assay was able to capture species-specific differences in IgG-FcRn interactions and overall correctly ranked Fc mutants according to their in vivo clearance. However, it could not explain the PK behavior of all tested IgGs, indicating that mAb disposition in vivo is a complex interplay of additional processes besides the FcRn interaction. Overall, the transcytosis assay was considered suitable to rank mAb candidates for their FcRn-mediated clearance component before extensive in vivo testing, and represents a first step toward a multi-factorial in vivo clearance prediction approach based on in vitro data.
Collapse
Affiliation(s)
| | - Sara Belli
- a Roche Pharmaceutical Research and Early Development, Roche Innovation Center , Basel , Switzerland
| | - Anne-Christine Cascais
- a Roche Pharmaceutical Research and Early Development, Roche Innovation Center , Basel , Switzerland
| | | | - Martin R Edelmann
- a Roche Pharmaceutical Research and Early Development, Roche Innovation Center , Basel , Switzerland
| | - Markus Haak
- a Roche Pharmaceutical Research and Early Development, Roche Innovation Center , Basel , Switzerland
| | - Marie-Elise Brun
- a Roche Pharmaceutical Research and Early Development, Roche Innovation Center , Basel , Switzerland
| | - Michael B Otteneder
- a Roche Pharmaceutical Research and Early Development, Roche Innovation Center , Basel , Switzerland
| | - Mohammed Ullah
- a Roche Pharmaceutical Research and Early Development, Roche Innovation Center , Basel , Switzerland
| | - Christoph Funk
- a Roche Pharmaceutical Research and Early Development, Roche Innovation Center , Basel , Switzerland
| | - Franz Schuler
- a Roche Pharmaceutical Research and Early Development, Roche Innovation Center , Basel , Switzerland
| | - Silke Simon
- a Roche Pharmaceutical Research and Early Development, Roche Innovation Center , Basel , Switzerland
| |
Collapse
|
107
|
Kelly RL, Yu Y, Sun T, Caffry I, Lynaugh H, Brown M, Jain T, Xu Y, Wittrup KD. Target-independent variable region mediated effects on antibody clearance can be FcRn independent. MAbs 2016; 8:1269-1275. [PMID: 27610650 DOI: 10.1080/19420862.2016.1208330] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
The importance of the neonatal Fc receptor (FcRn) in extending the serum half-life of monoclonal antibodies (mAbs) is well demonstrated, and has led to the development of multiple engineering approaches designed to alter Fc interactions with FcRn. Recent reports have additionally highlighted the effect of nonspecific interactions on antibody pharmacokinetics (PK), suggesting an FcRn-independent mechanism for mAb clearance. In this report we examine a case study of 2 anti-interleukin-12/23 antibodies, ustekinumab and briakinumab, which share the same target and Fc, but differ in variable region sequences. Ustekinumab displayed near baseline signal in a wide range of early stage developability assays for undesirable protein/protein interactions, while briakinumab showed significant propensity for self- and cross-interactions. This phenotypic difference correlates with faster clearance rates for briakinumab in both human FcRn transgenic and FcRn knockout mice. These findings support a dominant contribution for FcRn-independent clearance for antibodies with high nonspecificity, and highlight a key role for early stage developability screening to eliminate clones with such high nonspecific disposition PK.
Collapse
Affiliation(s)
- Ryan L Kelly
- a Department of Biological Engineering , Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology , Cambridge , MA , USA
| | - Yao Yu
- b Protein Analytics , Adimab , Lebanon , NH , USA
| | - Tingwan Sun
- b Protein Analytics , Adimab , Lebanon , NH , USA
| | | | | | | | - Tushar Jain
- c Computational Biology , Adimab , Palo Alto , CA , USA
| | - Yingda Xu
- d Department of Chemical Engineering, Koch Institute for Integrative Cancer Research , Massachusetts Institute of Technology , Cambridge , MA , USA
| | - K Dane Wittrup
- a Department of Biological Engineering , Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology , Cambridge , MA , USA.,d Department of Chemical Engineering, Koch Institute for Integrative Cancer Research , Massachusetts Institute of Technology , Cambridge , MA , USA
| |
Collapse
|
108
|
Stapleton NM, Einarsdóttir HK, Stemerding AM, Vidarsson G. The multiple facets of FcRn in immunity. Immunol Rev 2016; 268:253-68. [PMID: 26497526 DOI: 10.1111/imr.12331] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The neonatal Fc receptor, FcRn, is best known for its role in transporting IgG in various tissues, providing newborns with humoral immunity, and for prolonging the half-life of IgG. Recent findings implicate the involvement of FcRn in a far wider range of biological and immunological processes, as FcRn has been found to bind and extend the half-life of albumin; to be involved in IgG transport and antigen sampling at mucosal surfaces; and to be crucial for efficient IgG-mediated phagocytosis. Herein, the function of FcRn will be reviewed, with emphasis on its recently documented significance for IgG polymorphisms affecting the half-life and biodistribution of IgG3, on its role in phagocyte biology, and the subsequent role for the presentation of antigens to lymphocytes.
Collapse
Affiliation(s)
- Nigel M Stapleton
- Sanquin Research and Landsteiner Laboratory, Amsterdam Medical Centre, Amsterdam, The Netherlands
| | - Helga K Einarsdóttir
- Sanquin Research and Landsteiner Laboratory, Amsterdam Medical Centre, Amsterdam, The Netherlands
| | | | - Gestur Vidarsson
- Sanquin Research and Landsteiner Laboratory, Amsterdam Medical Centre, Amsterdam, The Netherlands
| |
Collapse
|
109
|
Souders CA, Nelson SC, Wang Y, Crowley AR, Klempner MS, Thomas W. A novel in vitro assay to predict neonatal Fc receptor-mediated human IgG half-life. MAbs 2016; 7:912-21. [PMID: 26018774 PMCID: PMC4622054 DOI: 10.1080/19420862.2015.1054585] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Immunoglobulin G (IgG) has an unusually long serum half-life in comparison to proteins of a similar size. It is well-known that this phenomenon is due to IgG's ability to bind the neonatal Fc receptor (FcRn) in a pH-dependent manner. FcRn binding properties can vary among IgGs, resulting in altered in vivo half-lives, and therefore it would be beneficial to accurately predict the FcRn binding properties of therapeutic IgG monoclonal antibodies (mAbs). Here we describe the development of an in vitro model capable of predicting the in vivo half-life of human IgG. Using a high-throughput biolayer interferometry (BLI) platform, the human FcRn association rate at acidic pH and subsequent dissociation rate at physiological pH was determined for 5 human IgG1 mAbs. Comparing the combined FcRn association and dissociation rates to the Phase 1 clinical study half-lives of the mAbs resulted in a strong correlation. The correlation was also verified in vivo using mice transgenic for human FcRn. The model was used to characterize various factors that may influence FcRn-mAb binding, including mAb variable region sequence differences and constant region glycosylation patterns. Results indicated that the complementarity-determining regions of the heavy chain significantly influence the mAb's FcRn binding properties, while the absence of glycosylation does not alter mAb-FcRn binding. Development of this high-throughput FcRn binding model could potentially predict the half-life of therapeutic IgGs and aid in selection of lead candidates while also serving as a screening tool for the development of mAbs with desired pharmacokinetic properties.
Collapse
Affiliation(s)
- Colby A Souders
- a MassBiologics of the University of Massachusetts Medical School ; Boston , MA USA
| | | | | | | | | | | |
Collapse
|
110
|
Glassman PM, Balthasar JP. Application of a catenary PBPK model to predict the disposition of "catch and release" anti-PCSK9 antibodies. Int J Pharm 2016; 505:69-78. [PMID: 27041125 DOI: 10.1016/j.ijpharm.2016.03.066] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 03/14/2016] [Accepted: 03/31/2016] [Indexed: 10/22/2022]
Abstract
The development of 'catch and release', or pH-sensitive, monoclonal antibodies (mAb) has become of interest to groups seeking to reduce the influence of target-mediated elimination on pharmacokinetics and pharmacodynamics. In this work, a catenary physiologically-based pharmacokinetic (PBPK) model is described to predict the pharmacokinetic benefit conferred by engineering mAbs for 'catch and release' binding. Our previously published PBPK model was adapted for consideration of the production and elimination of proprotein convertase subtilisin/kexin type 9 (PCSK9) in mice, and the model was then applied to predict the pharmacokinetics of anti-PCSK9 mAb with pH-stable (J10) and pH-sensitive binding (J17). The model was able to generate reasonable predictions of both J10 and J17 plasma pharmacokinetics. For J10, mean (±standard deviation) predicted vs. observed areas under the plasma concentration curve (AUCinf) were: 217 (77.1) vs. 103nMday (1mg/kg), 1.14×10(3) (858) vs. 769nMday (3mg/kg), and 6.60×10(3) (5.58×10(3)) vs. 2.86×10(3)nMday (10mg/kg), and for J17 the values were: 838 (161) vs. 818nMday (1mg/kg), 2.30×10(3) (441) vs. 2.57×10(3)nMday (3mg/kg), and 8.42×10(3) (1.50×10(3)) vs. 9.17×10(3)nMday (10mg/kg). Further simulations with the model predicted that target turnover and the magnitude of change in the complex dissociation rate constant between pH 7.4 and pH 6.0 are key determinants of the improvements in pharmacokinetics associated with 'catch and release' mAbs. The model described here may be useful for prediction of the pharmacokinetics of 'catch and release' mAbs directed against other targets.
Collapse
Affiliation(s)
- Patrick M Glassman
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, NY 14214, United States
| | - Joseph P Balthasar
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, NY 14214, United States.
| |
Collapse
|
111
|
Tibbitts J, Canter D, Graff R, Smith A, Khawli LA. Key factors influencing ADME properties of therapeutic proteins: A need for ADME characterization in drug discovery and development. MAbs 2015; 8:229-45. [PMID: 26636901 PMCID: PMC4966629 DOI: 10.1080/19420862.2015.1115937] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Protein therapeutics represent a diverse array of biologics including antibodies, fusion proteins, and therapeutic replacement enzymes. Since their inception, they have revolutionized the treatment of a wide range of diseases including respiratory, vascular, autoimmune, inflammatory, infectious, and neurodegenerative diseases, as well as cancer. While in vivo pharmacokinetic, pharmacodynamic, and efficacy studies are routinely carried out for protein therapeutics, studies that identify key factors governing their absorption, distribution, metabolism, and excretion (ADME) properties have not been fully investigated. Thorough characterization and in-depth study of their ADME properties are critical in order to support drug discovery and development processes for the production of safer and more effective biotherapeutics. In this review, we discuss the main factors affecting the ADME characteristics of these large macromolecular therapies. We also give an overview of the current tools, technologies, and approaches available to investigate key factors that influence the ADME of recombinant biotherapeutic drugs, and demonstrate how ADME studies will facilitate their future development.
Collapse
|
112
|
Glassman PM, Chen Y, Balthasar JP. Scale-up of a physiologically-based pharmacokinetic model to predict the disposition of monoclonal antibodies in monkeys. J Pharmacokinet Pharmacodyn 2015; 42:527-40. [PMID: 26364301 DOI: 10.1007/s10928-015-9444-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 09/04/2015] [Indexed: 01/06/2023]
Abstract
Preclinical assessment of monoclonal antibody (mAb) disposition during drug development often includes investigations in non-human primate models. In many cases, mAb exhibit non-linear disposition that relates to mAb-target binding [i.e., target-mediated disposition (TMD)]. The goal of this work was to develop a physiologically-based pharmacokinetic (PBPK) model to predict non-linear mAb disposition in plasma and in tissues in monkeys. Physiological parameters for monkeys were collected from several sources, and plasma data for several mAbs associated with linear pharmacokinetics were digitized from prior literature reports. The digitized data displayed great variability; therefore, parameters describing inter-antibody variability in the rates of pinocytosis and convection were estimated. For prediction of the disposition of individual antibodies, we incorporated tissue concentrations of target proteins, where concentrations were estimated based on categorical immunohistochemistry scores, and with assumed localization of target within the interstitial space of each organ. Kinetics of target-mAb binding and target turnover, in the presence or absence of mAb, were implemented. The model was then employed to predict concentration versus time data, via Monte Carlo simulation, for two mAb that have been shown to exhibit TMD (2F8 and tocilizumab). Model predictions, performed a priori with no parameter fitting, were found to provide good prediction of dose-dependencies in plasma clearance, the areas under plasma concentration versu time curves, and the time-course of plasma concentration data. This PBPK model may find utility in predicting plasma and tissue concentration versus time data and, potentially, the time-course of receptor occupancy (i.e., mAb-target binding) to support the design and interpretation of preclinical pharmacokinetic-pharmacodynamic investigations in non-human primates.
Collapse
Affiliation(s)
- Patrick M Glassman
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, The State University of New York, 452 Kapoor Hall, Buffalo, NY, 14214, USA
| | - Yang Chen
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, The State University of New York, 452 Kapoor Hall, Buffalo, NY, 14214, USA
| | - Joseph P Balthasar
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, The State University of New York, 452 Kapoor Hall, Buffalo, NY, 14214, USA.
| |
Collapse
|
113
|
Datta-Mannan A, Lu J, Witcher DR, Leung D, Tang Y, Wroblewski VJ. The interplay of non-specific binding, target-mediated clearance and FcRn interactions on the pharmacokinetics of humanized antibodies. MAbs 2015; 7:1084-93. [PMID: 26337808 DOI: 10.1080/19420862.2015.1075109] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The application of protein engineering technologies toward successfully improving antibody pharmacokinetics has been challenging due to the multiplicity of biochemical factors that influence monoclonal antibody (mAb) disposition in vivo. Physiological factors including interactions with the neonatal Fc receptor (FcRn) and specific antigen binding properties of mAbs, along with biophysical properties of the mAbs themselves play a critical role. It has become evident that applying an integrated approach to understand the relative contribution of these factors is critical to rationally guide and apply engineering strategies to optimize mAb pharmacokinetics. The study presented here evaluated the influence of unintended non-specific interactions on the disposition of mAbs whose clearance rates are governed predominantly by either non-specific (FcRn) or target-mediated processes. The pharmacokinetics of 8 mAbs representing a diverse range of these properties was evaluated in cynomolgus monkeys. Results revealed complementarity-determining region (CDR) charge patch engineering to decrease charge-related non-specific binding can have a significant impact on improving the clearance. In contrast, the influence of enhanced in vitro FcRn binding was mixed, and related to both the strength of charge interaction and the general mechanism predominant in governing the clearance of the particular mAb. Overall, improved pharmacokinetics through enhanced FcRn interactions were apparent for a CDR charge-patch normalized mAb which was affected by non-specific clearance. The findings in this report are an important demonstration that mAb pharmacokinetics requires optimization on a case-by-case basis to improve the design of molecules with increased therapeutic application.
Collapse
Affiliation(s)
- Amita Datta-Mannan
- a Departments of Drug Disposition and Development/Commercialization, Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center , Indianapolis , IN , USA
| | - Jirong Lu
- b Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center , Indianapolis , IN , USA
| | - Derrick R Witcher
- b Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center , Indianapolis , IN , USA
| | - Donmienne Leung
- c Biotechnology Discovery Research, Applied Molecular Evolution, Eli Lilly and Company ; San Diego , CA , USA
| | - Ying Tang
- c Biotechnology Discovery Research, Applied Molecular Evolution, Eli Lilly and Company ; San Diego , CA , USA
| | - Victor J Wroblewski
- a Departments of Drug Disposition and Development/Commercialization, Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center , Indianapolis , IN , USA
| |
Collapse
|