51
|
Intrinsic physicochemical profile of marketed antibody-based biotherapeutics. Proc Natl Acad Sci U S A 2021; 118:2020577118. [PMID: 34504010 PMCID: PMC8449350 DOI: 10.1073/pnas.2020577118] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/03/2021] [Indexed: 01/28/2023] Open
Abstract
Successful biologic drug discovery and development involves finding functional as well as developable candidates. Once a candidate has been demonstrated to be functional, the next step is to determine whether it can be translated into a drug product. This requires that the candidate can withstand stresses encountered during manufacturing, shipping, and storage. Additionally, it must be safe, efficacious, and possess good pharmacology. In silico analyses of the variable regions of 77 marketed antibody-based biotherapeutics have revealed five nonredundant physicochemical descriptors. Distributions of these descriptors, observed for marketed biotherapeutics, can help prioritize a drug candidate for experimental testing at early discovery stages, guide engineering efforts to further optimize it, and help increase the productivity of biologic drug discovery and development. Feeding biopharma pipelines with biotherapeutic candidates that possess desirable developability profiles can help improve the productivity of biologic drug discovery and development. Here, we have derived an in silico profile by analyzing computed physicochemical descriptors for the variable regions (Fv) found in 77 marketed antibody-based biotherapeutics. Fv regions of these biotherapeutics demonstrate significant diversities in their germlines, complementarity determining region loop lengths, hydrophobicity, and charge distributions. Furthermore, an analysis of 24 physicochemical descriptors, calculated using homology-based molecular models, has yielded five nonredundant descriptors whose distributions represent stability, isoelectric point, and molecular surface characteristics of their Fv regions. Fv regions of candidates from our internal discovery campaigns, human next-generation sequencing repertoires, and those in clinical-stages (CST) were assessed for similarity with the physicochemical profile derived here. The Fv regions in 33% of CST antibodies show physicochemical properties that are dissimilar to currently marketed biotherapeutics. In comparison, physicochemical characteristics of ∼29% of the Fv regions in human antibodies and ∼27% of our internal hits deviated significantly from those of marketed biotherapeutics. The early availability of this information can help guide hit selection, lead identification, and optimization of biotherapeutic candidates. Insights from this work can also help support portfolio risk assessment, in-licensing, and biopharma collaborations.
Collapse
|
52
|
Dyson MR, Masters E, Pazeraitis D, Perera RL, Syrjanen JL, Surade S, Thorsteinson N, Parthiban K, Jones PC, Sattar M, Wozniak-Knopp G, Rueker F, Leah R, McCafferty J. Beyond affinity: selection of antibody variants with optimal biophysical properties and reduced immunogenicity from mammalian display libraries. MAbs 2021; 12:1829335. [PMID: 33103593 PMCID: PMC7592150 DOI: 10.1080/19420862.2020.1829335] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The early phase of protein drug development has traditionally focused on target binding properties leading to a desired mode of therapeutic action. As more protein therapeutics pass through the development pipeline; however, it is clear that non-optimal biophysical properties can emerge, particularly as proteins are formulated at high concentrations, causing aggregation or polyreactivity. Such late-stage "developability" problems can lead to delay or failure in traversing the development process. Aggregation propensity is also correlated with increased immunogenicity, resulting in expensive, late-stage clinical failures. Using nucleases-directed integration, we have constructed large mammalian display libraries where each cell contains a single antibody gene/cell inserted at a single locus, thereby achieving transcriptional normalization. We show a strong correlation between poor biophysical properties and display level achieved in mammalian cells, which is not replicated by yeast display. Using two well-documented examples of antibodies with poor biophysical characteristics (MEDI-1912 and bococizumab), a library of variants was created based on surface hydrophobic and positive charge patches. Mammalian display was used to select for antibodies that retained target binding and permitted increased display level. The resultant variants exhibited reduced polyreactivity and reduced aggregation propensity. Furthermore, we show in the case of bococizumab that biophysically improved variants are less immunogenic than the parental molecule. Thus, mammalian display helps to address multiple developability issues during the earliest stages of lead discovery, thereby significantly de-risking the future development of protein drugs.
Collapse
Affiliation(s)
| | | | | | | | - Johanna L Syrjanen
- WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory , NY, USA
| | | | | | | | | | | | - Gordana Wozniak-Knopp
- Department of Biotechnology, BOKU-University of Natural Resources and Life Sciences , Vienna, Austria
| | - Florian Rueker
- Department of Biotechnology, BOKU-University of Natural Resources and Life Sciences , Vienna, Austria
| | | | | |
Collapse
|
53
|
Liu C, Kim YS, Lowe JHN, Chung S. A cell-based FcRn-dependent recycling assay for predictive pharmacokinetic assessment of therapeutic antibodies. Bioanalysis 2021; 13:1135-1144. [PMID: 34289743 DOI: 10.4155/bio-2021-0099] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 06/30/2021] [Indexed: 12/28/2022] Open
Abstract
Aim: Evaluation of suitable pharmacokinetic properties is critical for successful development of IgG-based biotherapeutics. The prolonged half-lives of IgGs depend on the intracellular trafficking function of neonatal Fc receptor, which rescues internalized IgGs from lysosomal degradation and recycles them back to circulation. Results: Here, we developed a novel cell-based assay to quantify recycling of monoclonal antibodies in a transwell culture system that uses a cell line that stably expresses human neonatal Fc receptor. We tested seven therapeutic antibodies and showed that the recycling output of the assay strongly correlated with the clearance in humans. Conclusion: This recycling assay has potential application as a pharmacokinetic prescreening tool to facilitate development and selection of IgG-based candidate therapeutic monoclonal antibodies.
Collapse
Affiliation(s)
- Chang Liu
- Department of BioAnalytical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Yeon Su Kim
- Department of BioAnalytical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - John Hok-Nin Lowe
- Department of BioAnalytical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Shan Chung
- Department of BioAnalytical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| |
Collapse
|
54
|
Desai AA, Smith MD, Zhang Y, Makowski EK, Gerson JE, Ionescu E, Starr CG, Zupancic JM, Moore SJ, Sutter AB, Ivanova MI, Murphy GG, Paulson HL, Tessier PM. Rational affinity maturation of anti-amyloid antibodies with high conformational and sequence specificity. J Biol Chem 2021; 296:100508. [PMID: 33675750 PMCID: PMC8081927 DOI: 10.1016/j.jbc.2021.100508] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 02/05/2021] [Accepted: 03/02/2021] [Indexed: 01/01/2023] Open
Abstract
The aggregation of amyloidogenic polypeptides is strongly linked to several neurodegenerative disorders, including Alzheimer's and Parkinson's diseases. Conformational antibodies that selectively recognize protein aggregates are leading therapeutic agents for selectively neutralizing toxic aggregates, diagnostic and imaging agents for detecting disease, and biomedical reagents for elucidating disease mechanisms. Despite their importance, it is challenging to generate high-quality conformational antibodies in a systematic and site-specific manner due to the properties of protein aggregates (hydrophobic, multivalent, and heterogeneous) and limitations of immunization (uncontrolled antigen presentation and immunodominant epitopes). Toward addressing these challenges, we have developed a systematic directed evolution procedure for affinity maturing antibodies against Alzheimer's Aβ fibrils and selecting variants with strict conformational and sequence specificity. We first designed a library based on a lead conformational antibody by sampling combinations of amino acids in the antigen-binding site predicted to mediate high antibody specificity. Next, we displayed this library on the surface of yeast, sorted it against Aβ42 aggregates, and identified promising clones using deep sequencing. The resulting antibodies displayed similar or higher affinities than clinical-stage Aβ antibodies (aducanumab and crenezumab). Moreover, the affinity-matured antibodies retained high conformational specificity for Aβ aggregates, as observed for aducanumab and unlike crenezumab. Notably, the affinity-maturated antibodies displayed extremely low levels of nonspecific interactions, as observed for crenezumab and unlike aducanumab. We expect that our systematic methods for generating antibodies with unique combinations of desirable properties will improve the generation of high-quality conformational antibodies specific for diverse types of aggregated conformers.
Collapse
Affiliation(s)
- Alec A Desai
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan, USA
| | - Matthew D Smith
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan, USA
| | - Yulei Zhang
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan, USA
| | - Emily K Makowski
- Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan, USA; Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, Michigan, USA
| | - Julia E Gerson
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Edward Ionescu
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan, USA
| | - Charles G Starr
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan, USA; Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, Michigan, USA
| | - Jennifer M Zupancic
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan, USA
| | - Shannon J Moore
- Protein Folding Disease Initiative, University of Michigan, Ann Arbor, Michigan, USA; Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Alexandra B Sutter
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA; Biophysics Program, University of Michigan, Ann Arbor, Michigan, USA
| | - Magdalena I Ivanova
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA; Biophysics Program, University of Michigan, Ann Arbor, Michigan, USA
| | - Geoffrey G Murphy
- Protein Folding Disease Initiative, University of Michigan, Ann Arbor, Michigan, USA; Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Henry L Paulson
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA; Protein Folding Disease Initiative, University of Michigan, Ann Arbor, Michigan, USA; Michigan Alzheimer's Disease Center, University of Michigan, Ann Arbor, Michigan, USA
| | - Peter M Tessier
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan, USA; Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, Michigan, USA; Protein Folding Disease Initiative, University of Michigan, Ann Arbor, Michigan, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA.
| |
Collapse
|
55
|
Campbell SM, DeBartolo J, Apgar JR, Mosyak L, McManus V, Beyer S, Bennett EM, Lambert M, Cunningham O. Combining random mutagenesis, structure-guided design and next-generation sequencing to mitigate polyreactivity of an anti-IL-21R antibody. MAbs 2021; 13:1883239. [PMID: 33557673 PMCID: PMC7889167 DOI: 10.1080/19420862.2021.1883239] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Despite substantial technological advances in antibody library and display platform development, the number of approved biotherapeutics from displayed libraries remains limited. In vivo, 20–50% of peripheral B cells undergo a process of receptor editing, which modifies the variable and junctional regions of light chains to delete auto-reactive clones. However, in vitro antibody evolution relies primarily on interaction with antigen, with no in-built checkpoints to ensure that the selected antibodies have not acquired additional specificities or biophysical liabilities during the optimization process. We had previously observed an enrichment of positive charge in the complementarity-determining regions of an anti-IL-21 R antibody during affinity optimization, which correlated with more potent IL-21 neutralization, but poor in vivo pharmacokinetics (PK). There is an emerging body of data that has correlated antibody nonspecificity with poor PK in vivo, and established a series of screening assays that are predictive of this behavior. In this study we revisit the challenge of developing an anti-IL-21 R antibody that can effectively compete with IL-21 for its highly negatively charged paratope while maintaining favorable biophysical properties. In vitro deselection methods that included an excess of negatively charged membrane preparations, or deoxyribonucleic acid, during phage selection of optimization libraries were unsuccessful in avoiding enrichment of highly charged, nonspecific antibody variants. However, a combination of structure-guided rational library design, next-generation sequencing of library outputs and application of linear regression models resulted in the identification of an antibody that maintained high affinity for IL-21 R and exhibited a desirable stability and biophysical profile.
Collapse
Affiliation(s)
| | | | | | | | | | - Sonia Beyer
- Biomedicine Design, Pfizer , Dublin, Ireland
| | | | | | | |
Collapse
|
56
|
Kraft TE, Richter WF, Emrich T, Knaupp A, Schuster M, Wolfert A, Kettenberger H. Heparin chromatography as an in vitro predictor for antibody clearance rate through pinocytosis. MAbs 2021; 12:1683432. [PMID: 31769731 PMCID: PMC6927760 DOI: 10.1080/19420862.2019.1683432] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The pharmacokinetic (PK) properties of therapeutic antibodies directly affect efficacy, dose and dose intervals, application route and tissue penetration. In indications where health-care providers and patients can choose between several efficacious and safe therapeutic options, convenience (determined by dosing interval or route of application), which is mainly driven by PK properties, can affect drug selection. Therapeutic antibodies can have greatly different PK even if they have identical Fc domains and show no target-mediated drug disposition. Biophysical properties like surface charge or hydrophobicity, and binding to surrogates for high abundant off-targets (e.g., baculovirus particles, Chinese hamster ovary cell membrane proteins) were proposed to be responsible for these differences. Here, we used heparin chromatography to separate a polyclonal mix of endogenous human IgGs (IVIG) into fractions that differ in their PK properties. Heparin was chosen as a surrogate for highly negatively charged glycocalyx components on endothelial cells, which are among the main contributors to nonspecific clearance. By directly correlating heparin retention time with clearance, we identified heparin chromatography as a tool to assess differences in unspecific cell-surface interaction and the likelihood for increased pinocytotic uptake and degradation. Building on these results, we combined predictors for FcRn-mediated recycling and cell-surface interaction. The combination of heparin and FcRn chromatography allow identification of antibodies with abnormal PK by mimicking the major root causes for fast, non-target-mediated, clearance of therapeutic, Fc-containing proteins.
Collapse
Affiliation(s)
- Thomas E Kraft
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| | - Wolfgang F Richter
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Thomas Emrich
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| | - Alexander Knaupp
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| | - Michaela Schuster
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| | - Andreas Wolfert
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| | - Hubert Kettenberger
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| |
Collapse
|
57
|
Makowski EK, Wu L, Desai AA, Tessier PM. Highly sensitive detection of antibody nonspecific interactions using flow cytometry. MAbs 2021; 13:1951426. [PMID: 34313552 PMCID: PMC8317921 DOI: 10.1080/19420862.2021.1951426] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 06/29/2021] [Accepted: 06/30/2021] [Indexed: 11/12/2022] Open
Abstract
The rapidly evolving nature of antibody drug development has resulted in technologies that generate vast numbers (hundreds to thousands) of lead antibody candidates during early discovery. These candidates must be rapidly pared down to identify the most drug-like candidates for in-depth analysis of their safety and efficacy, which can only be performed on a limited number of antibodies due to time and resource requirements. One key biophysical property of successful antibody therapeutics is high specificity, defined as low levels of nonspecific binding or polyspecificity. Although there has been some progress in developing assays for detecting antibody polyspecificity, most of these assays are limited by poor sensitivity or assay formats that require proprietary antibody surface display methods, and some of these assays use complex and poorly defined polyspecificity reagents. Here we report the PolySpecificity Particle (PSP) assay, a sensitive flow cytometry assay for evaluating antibody nonspecific interactions that overcomes previous limitations and can be used for evaluating diverse types of IgGs, multispecific antibodies and Fc-fusion proteins. Our approach uses micron-sized magnetic beads coated with Protein A to capture antibodies at extremely dilute concentrations (<0.02 mg/mL). Flow cytometry analysis of polyspecificity reagent binding to these conjugates results in sensitive detection of differences in nonspecific interactions for clinical-stage antibodies. Our PSP assay strongly discriminates between antibodies with different levels of polyspecificity using previously reported polyspecificity reagents that are either well-defined proteins or highly complex protein mixtures. Moreover, we also find that a unique reagent, namely ovalbumin, results in the best assay sensitivity and specificity. Importantly, our assay is much more sensitive than standard assays such as ELISAs. We expect that our simple, sensitive, and high-throughput PSP assay will accelerate the development of safe and effective antibody therapeutics.
Collapse
Affiliation(s)
| | - Lina Wu
- Department of Chemical Engineering, University of Michigan
| | - Alec A. Desai
- Department of Chemical Engineering, University of Michigan
| | - Peter M. Tessier
- Department of Pharmaceutical Sciences, University of Michigan
- Department of Chemical Engineering, University of Michigan
- Department of Biomedical Engineering, Biointerfaces Institute, University of Michigan, Ann Arbor, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, USA
| |
Collapse
|
58
|
Makowski EK, Wu L, Gupta P, Tessier PM. Discovery-stage identification of drug-like antibodies using emerging experimental and computational methods. MAbs 2021; 13:1895540. [PMID: 34313532 PMCID: PMC8346245 DOI: 10.1080/19420862.2021.1895540] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 02/05/2021] [Accepted: 02/22/2021] [Indexed: 11/30/2022] Open
Abstract
There is intense and widespread interest in developing monoclonal antibodies as therapeutic agents to treat diverse human disorders. During early-stage antibody discovery, hundreds to thousands of lead candidates are identified, and those that lack optimal physical and chemical properties must be deselected as early as possible to avoid problems later in drug development. It is particularly challenging to characterize such properties for large numbers of candidates with the low antibody quantities, concentrations, and purities that are available at the discovery stage, and to predict concentrated antibody properties (e.g., solubility, viscosity) required for efficient formulation, delivery, and efficacy. Here we review key recent advances in developing and implementing high-throughput methods for identifying antibodies with desirable in vitro and in vivo properties, including favorable antibody stability, specificity, solubility, pharmacokinetics, and immunogenicity profiles, that together encompass overall drug developability. In particular, we highlight impressive recent progress in developing computational methods for improving rational antibody design and prediction of drug-like behaviors that hold great promise for reducing the amount of required experimentation. We also discuss outstanding challenges that will need to be addressed in the future to fully realize the great potential of using such analysis for minimizing development times and improving the success rate of antibody candidates in the clinic.
Collapse
Affiliation(s)
- Emily K. Makowski
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Lina Wu
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Chemical Engineering
| | - Priyanka Gupta
- Department of Biochemistry and Biophysics, Rensselaer Polytechnic Institute, Troy, NY, USA
- Biotherapeutics Discovery Department, Boehringer Ingelheim, Ridgefield, CT, USA
| | - Peter M. Tessier
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Chemical Engineering
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
59
|
Sawant MS, Streu CN, Wu L, Tessier PM. Toward Drug-Like Multispecific Antibodies by Design. Int J Mol Sci 2020; 21:E7496. [PMID: 33053650 PMCID: PMC7589779 DOI: 10.3390/ijms21207496] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 10/02/2020] [Accepted: 10/02/2020] [Indexed: 12/18/2022] Open
Abstract
The success of antibody therapeutics is strongly influenced by their multifunctional nature that couples antigen recognition mediated by their variable regions with effector functions and half-life extension mediated by a subset of their constant regions. Nevertheless, the monospecific IgG format is not optimal for many therapeutic applications, and this has led to the design of a vast number of unique multispecific antibody formats that enable targeting of multiple antigens or multiple epitopes on the same antigen. Despite the diversity of these formats, a common challenge in generating multispecific antibodies is that they display suboptimal physical and chemical properties relative to conventional IgGs and are more difficult to develop into therapeutics. Here we review advances in the design and engineering of multispecific antibodies with drug-like properties, including favorable stability, solubility, viscosity, specificity and pharmacokinetic properties. We also highlight emerging experimental and computational methods for improving the next generation of multispecific antibodies, as well as their constituent antibody fragments, with natural IgG-like properties. Finally, we identify several outstanding challenges that need to be addressed to increase the success of multispecific antibodies in the clinic.
Collapse
Affiliation(s)
- Manali S. Sawant
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA; (M.S.S.); (C.N.S.)
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA;
| | - Craig N. Streu
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA; (M.S.S.); (C.N.S.)
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA;
- Department of Chemistry, Albion College, Albion, MI 49224, USA
| | - Lina Wu
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA;
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Peter M. Tessier
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA; (M.S.S.); (C.N.S.)
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA;
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
60
|
Ruano-Salguero JS, Lee KH. Adsorptive-Mediated Endocytosis of Sulfo-Cy5-Labeled IgG Causes Aberrant IgG Processing by Brain Endothelial-Like Cells. Mol Pharm 2020; 17:4280-4285. [PMID: 32986439 DOI: 10.1021/acs.molpharmaceut.0c00712] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Brain endothelial cells (BECs) hinder macromolecules from reaching brain parenchyma, necessitating the evaluation and engineering of therapeutic immunoglobulin γ (IgG) for improved brain delivery. Emerging fluorescent-based approaches to assess IgG brain exposure can expedite and complement current methods; however, alterations in IgG pharmacokinetics following fluorophore conjugation, which remain unexplained, indicate that conjugation may confound analysis of native IgG processing. Here, changes in transcytosis and intracellular processing of IgG conjugates (with sulfonated cyanine 5) were examined using human induced pluripotent stem cell-derived BECs (iBECs). Above a critical degree of labeling, transcytosis rates increased significantly but could be attenuated by nonspecific protein competition. Concurrent increases in intracellular accumulation, which was not attributable to disrupted binding by the neonatal Fc receptor (FcRn), are indicative of indirect reduction of FcRn-mediated recycling that agrees with reported aberrations in the pharmacokinetics of certain unconjugated IgGs. Overall, these findings support the notion that certain fluorophore-IgG conjugates can engage in adsorptive interactions with cell surface moieties, reminiscent of phenomena exhibited by cationized IgG, and provide in vitro criteria to identify changes in IgG processing following fluorophore conjugation.
Collapse
Affiliation(s)
- John S Ruano-Salguero
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware 19713, United States.,Delaware Biotechnology Institute, University of Delaware, Newark, Delaware 19713, United States
| | - Kelvin H Lee
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware 19713, United States.,Delaware Biotechnology Institute, University of Delaware, Newark, Delaware 19713, United States
| |
Collapse
|
61
|
Sun Y, Cai H, Hu Z, Boswell CA, Diao J, Li C, Zhang L, Shen A, Teske CA, Zhang B, Kamath AV, Jiang G. Balancing the Affinity and Pharmacokinetics of Antibodies by Modulating the Size of Charge Patches on Complementarity-Determining Regions. J Pharm Sci 2020; 109:3690-3696. [PMID: 32910947 DOI: 10.1016/j.xphs.2020.09.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 09/01/2020] [Accepted: 09/02/2020] [Indexed: 12/18/2022]
Abstract
A localized positive charge on IgG (referred to as a "charge patch") shows an adverse effect on pharmacokinetics (PK), so it would seem to be best practice to avoid charge patches during the discovery stage and closely monitor charge interactions during the development process. In certain circumstances, however, charge patches are required for target binding, in which case completely removing charge patches is not feasible. Therefore, quantitative measurement of a charge patch and its impact on PK is critical to the success of therapeutic antibody development. In this article, we generated mutations of a recombinant human antibody (referred to as mAb1) with disrupted charge patches to investigate how charge patches on IgG antibodies impact both target-binding affinity and PK-related factors. We conclude that it is important to modulate the size of the charge patch in order to balance target-binding affinity and PK.
Collapse
Affiliation(s)
- Yue Sun
- Biological Technologies, Genentech, Inc., South San Francisco, CA 94080, USA; Protein Analytical Chemistry, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Hao Cai
- DevSci PTPK, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Zhilan Hu
- Early Stage Cell Culture, Genentech, Inc., South San Francisco, CA 94080, USA
| | - C Andrew Boswell
- DevSci PTPK, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Jinpian Diao
- Purification Development, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Charlene Li
- Protein Analytical Chemistry, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Liangyi Zhang
- Protein Analytical Chemistry, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Amy Shen
- Early Stage Cell Culture, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Christopher A Teske
- Purification Development, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Boyan Zhang
- Protein Analytical Chemistry, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Amrita V Kamath
- DevSci PTPK, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Guoying Jiang
- Biological Technologies, Genentech, Inc., South San Francisco, CA 94080, USA.
| |
Collapse
|
62
|
Datta-Mannan A, Estwick S, Zhou C, Choi H, Douglass NE, Witcher DR, Lu J, Beidler C, Millican R. Influence of physiochemical properties on the subcutaneous absorption and bioavailability of monoclonal antibodies. MAbs 2020; 12:1770028. [PMID: 32486889 PMCID: PMC7531508 DOI: 10.1080/19420862.2020.1770028] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Many therapeutic monoclonal antibodies (mAbs) were initially developed for intravenous (IV) administration. As a means to improve mAb drug-ability and the patient experience, subcutaneous (SC) administration is an increasingly important delivery route for mAbs. Unlike IV administration, bioavailability limitations for antibodies have been reported following SC injection and can dictate whether a mAb is administered via this parenteral route. The SC bioavailability of antibodies has been difficult to predict, and it can be variable and partial, with values ranging from ~50% to 100%. The mechanisms leading to the incomplete bioavailability of some mAbs relative to others are not well understood. There are some limited data that suggest the physiochemical properties inherent to a mAb can contribute to its SC absorption, bioavailability, and in vivo fate. In this study, we evaluated the integrated influence of multiple mAb physiochemical factors on the SC absorption and bioavailability of six humanized mAbs in both rats and cynomolgus monkeys. We demonstrate the physiochemical properties of mAbs are critical to their rate and extent of SC absorption. The combination of high positive charge and hydrophobic interaction significantly reduced the rate of the evaluated mAb’s SC absorption and bioavailability. Reduction or balancing of both these attributes via re-engineering the mAbs restored desirable properties of the molecules assessed. This included reduced association with SC tissue, improvements in mAb absorption from the SC space and overall SC bioavailability. Our findings point to the importance of evaluating the relative balance between various physiochemical factors, including charge, hydrophobicity, and stability, to improve the SC drug-ability of mAbs for selecting or engineering mAbs with enhanced in vivo absorption and bioavailability following SC administration.
Collapse
Affiliation(s)
- Amita Datta-Mannan
- Departments of Exploratory Medicine and Pharmacology, Lilly Research Laboratories, Lilly Corporate Center , Indianapolis, IN, USA
| | - Selina Estwick
- External Innovation, Lilly Research Laboratories, Lilly Corporate Center , Indianapolis, IN, USA
| | | | - Hiuwan Choi
- Biotechnology Discovery Research, Lilly Research Laboratories, Lilly Corporate Center , Indianapolis, IN, USA
| | - Nicole E Douglass
- Clinical Design/Delivery/Analytics, Lilly Research Laboratories Lilly Corporate Center , Indianapolis, IN, USA
| | - Derrick R Witcher
- Biotechnology Discovery Research, Lilly Research Laboratories, Lilly Corporate Center , Indianapolis, IN, USA
| | - Jirong Lu
- Biotechnology Discovery Research, Lilly Research Laboratories, Lilly Corporate Center , Indianapolis, IN, USA
| | - Catherine Beidler
- Biotechnology Discovery Research, Lilly Research Laboratories, Lilly Corporate Center , Indianapolis, IN, USA
| | - Rohn Millican
- Biotechnology Discovery Research, Lilly Research Laboratories, Lilly Corporate Center , Indianapolis, IN, USA
| |
Collapse
|
63
|
Epitope Mapping and Computational Analysis of Anti-HPV16 E6 and E7 Antibodies in Single-Chain Format for Clinical Development as Antitumor Drugs. Cancers (Basel) 2020; 12:cancers12071803. [PMID: 32640530 PMCID: PMC7408665 DOI: 10.3390/cancers12071803] [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] [Received: 06/10/2020] [Revised: 07/02/2020] [Accepted: 07/03/2020] [Indexed: 11/17/2022] Open
Abstract
Human Papillomavirus 16-associated cancer, affecting primarily the uterine cervix but, increasingly, other body districts, including the head–neck area, will long be a public health problem, despite there being a vaccine. Since the virus oncogenic activity is fully ascribed to the viral E6 and E7 oncoproteins, one of the therapeutic approaches for HPV16 cancer is based on specific antibodies in single-chain format targeting the E6/E7 activity. We analyzed the Complementarity Determining Regions, repositories of antigen-binding activity, of four anti-HPV16 E6 and -HPV16 E7 scFvs, to highlight possible conformity to biophysical properties, recognized to be advantageous for therapeutic use. By epitope mapping, using E7 mutants with amino acid deletions or variations, we investigated differences among the anti-16E7 scFvs in terms of antigen-binding capacity. We also performed computational analyses to determine whether length, total net charge, surface hydrophobicity, polarity and charge distribution conformed well to those of the antibodies that had already reached clinical use, through the application of developability guidelines derived from recent literature on clinical-stage antibodies, and the Therapeutic Antibodies Profiler software. Overall, our findings show that the scFvs investigated may represent valid candidates to be developed as therapeutic molecules for clinical use, and highlight characteristics that could be improved by molecular engineering.
Collapse
|
64
|
Zhang Y, Wu L, Gupta P, Desai AA, Smith MD, Rabia LA, Ludwig SD, Tessier PM. Physicochemical Rules for Identifying Monoclonal Antibodies with Drug-like Specificity. Mol Pharm 2020; 17:2555-2569. [PMID: 32453957 PMCID: PMC7936472 DOI: 10.1021/acs.molpharmaceut.0c00257] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The ability of antibodies to recognize their target antigens with high specificity is fundamental to their natural function. Nevertheless, therapeutic antibodies display variable and difficult-to-predict levels of nonspecific and self-interactions that can lead to various drug development challenges, including antibody aggregation, abnormally high viscosity, and rapid antibody clearance. Here we report a method for predicting the overall specificity of antibodies in terms of their relative risk for displaying high levels of nonspecific or self-interactions at physiological conditions. We find that individual and combined sets of chemical rules that limit the maximum and minimum numbers of certain solvent-exposed amino acids in antibody variable regions are strong predictors of specificity for large panels of preclinical and clinical-stage antibodies. We also demonstrate how the chemical rules can be used to identify sites that mediate nonspecific interactions in suboptimal antibodies and guide the design of targeted sublibraries that yield variants with high antibody specificity. These findings can be readily used to improve the selection and engineering of antibodies with drug-like specificity.
Collapse
Affiliation(s)
- Yulei Zhang
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Lina Wu
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Priyanka Gupta
- Department of Biochemistry and Biophysics, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
- Biotherapeutics Discovery Department, Boehringer Ingelheim, Ridgefield, CT 06877
| | - Alec A. Desai
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Matthew D. Smith
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Lilia A. Rabia
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
- Isermann Department of Chemical & Biological Engineering, Troy, NY 12180, USA
| | - Seth D. Ludwig
- Isermann Department of Chemical & Biological Engineering, Troy, NY 12180, USA
| | - Peter M. Tessier
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
- Isermann Department of Chemical & Biological Engineering, Troy, NY 12180, USA
- Department of Biochemistry and Biophysics, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| |
Collapse
|
65
|
Hu S, D'Argenio DZ. Predicting monoclonal antibody pharmacokinetics following subcutaneous administration via whole-body physiologically-based modeling. J Pharmacokinet Pharmacodyn 2020; 47:385-409. [PMID: 32500362 DOI: 10.1007/s10928-020-09691-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 05/14/2020] [Indexed: 12/12/2022]
Abstract
Use of the subcutaneous (SC) route for administering monoclonal antibodies (mAbs) to treat chronic conditions has been hindered because of an incomplete understanding of fundamental mechanisms controlling mAb absorption from the SC site, and due to the limited translatability of preclinical studies. In this paper, we report on the development and evaluation of a whole-body physiologically-based model to predict mAb pharmacokinetics following SC administration. The circulatory model is based on the physiological processes governing mAb transport and includes two mAb-specific parameters representing differences in pinocytosis rate and the diffusive/convective transport rates among mAbs. At the SC administration site, two additional parameters are used to represent mAb differences in lymphatic capillary uptake and in pre-systemic clearance. Model development employed clinical intravenous (IV) plasma PK data from 20 mAbs and SC plasma PK data from 12 of these mAbs, as obtained from the literature. The resulting model reliably described both the IV and SC measured plasma concentration data. In addition, a metric based on the positive charge across the mAb's complementarity determining region vicinity was found to positively correlate with the model-based estimates of the mAb-specific parameter governing organ/tissue pinocytosis transport and with estimates of the mAb's SC lymphatic capillary clearance. These two relationships were incorporated into the model and accurately predicted the SC PK profiles of three out of four separate mAbs not included in model development. The whole-body physiologically-based model reported herein, provides a platform to characterize and predict the plasma disposition of monoclonal antibodies following SC administration in humans.
Collapse
Affiliation(s)
- Shihao Hu
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA
| | - David Z D'Argenio
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
66
|
Marks C, Deane CM. How repertoire data are changing antibody science. J Biol Chem 2020; 295:9823-9837. [PMID: 32409582 DOI: 10.1074/jbc.rev120.010181] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 04/28/2020] [Indexed: 12/13/2022] Open
Abstract
Antibodies are vital proteins of the immune system that recognize potentially harmful molecules and initiate their removal. Mammals can efficiently create vast numbers of antibodies with different sequences capable of binding to any antigen with high affinity and specificity. Because they can be developed to bind to many disease agents, antibodies can be used as therapeutics. In an organism, after antigen exposure, antibodies specific to that antigen are enriched through clonal selection, expansion, and somatic hypermutation. The antibodies present in an organism therefore report on its immune status, describe its innate ability to deal with harmful substances, and reveal how it has previously responded. Next-generation sequencing technologies are being increasingly used to query the antibody, or B-cell receptor (BCR), sequence repertoire, and the amount of BCR data in public repositories is growing. The Observed Antibody Space database, for example, currently contains over a billion sequences from 68 different studies. Repertoires are available that represent both the naive state (i.e. antigen-inexperienced) and that after immunization. This wealth of data has created opportunities to learn more about our immune system. In this review, we discuss the many ways in which BCR repertoire data have been or could be exploited. We highlight its utility for providing insights into how the naive immune repertoire is generated and how it responds to antigens. We also consider how structural information can be used to enhance these data and may lead to more accurate depictions of the sequence space and to applications in the discovery of new therapeutics.
Collapse
Affiliation(s)
- Claire Marks
- Department of Statistics, University of Oxford, Oxford, United Kingdom
| | - Charlotte M Deane
- Department of Statistics, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
67
|
Goulet DR, Atkins WM. Considerations for the Design of Antibody-Based Therapeutics. J Pharm Sci 2020; 109:74-103. [PMID: 31173761 PMCID: PMC6891151 DOI: 10.1016/j.xphs.2019.05.031] [Citation(s) in RCA: 173] [Impact Index Per Article: 34.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 05/02/2019] [Accepted: 05/29/2019] [Indexed: 02/06/2023]
Abstract
Antibody-based proteins have become an important class of biologic therapeutics, due in large part to the stability, specificity, and adaptability of the antibody framework. Indeed, antibodies not only have the inherent ability to bind both antigens and endogenous immune receptors but also have proven extremely amenable to protein engineering. Thus, several derivatives of the monoclonal antibody format, including bispecific antibodies, antibody-drug conjugates, and antibody fragments, have demonstrated efficacy for treating human disease, particularly in the fields of immunology and oncology. Reviewed here are considerations for the design of antibody-based therapeutics, including immunological context, therapeutic mechanisms, and engineering strategies. First, characteristics of antibodies are introduced, with emphasis on structural domains, functionally important receptors, isotypic and allotypic differences, and modifications such as glycosylation. Then, aspects of therapeutic antibody design are discussed, including identification of antigen-specific variable regions, choice of expression system, use of multispecific formats, and design of antibody derivatives based on fragmentation, oligomerization, or conjugation to other functional moieties. Finally, strategies to enhance antibody function through protein engineering are reviewed while highlighting the impact of fundamental biophysical properties on protein developability.
Collapse
Affiliation(s)
- Dennis R Goulet
- Department of Medicinal Chemistry, University of Washington, Seattle, Washington 98195.
| | - William M Atkins
- Department of Medicinal Chemistry, University of Washington, Seattle, Washington 98195
| |
Collapse
|
68
|
Chiu ML, Goulet DR, Teplyakov A, Gilliland GL. Antibody Structure and Function: The Basis for Engineering Therapeutics. Antibodies (Basel) 2019; 8:antib8040055. [PMID: 31816964 PMCID: PMC6963682 DOI: 10.3390/antib8040055] [Citation(s) in RCA: 290] [Impact Index Per Article: 48.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 11/25/2019] [Accepted: 11/28/2019] [Indexed: 12/11/2022] Open
Abstract
Antibodies and antibody-derived macromolecules have established themselves as the mainstay in protein-based therapeutic molecules (biologics). Our knowledge of the structure–function relationships of antibodies provides a platform for protein engineering that has been exploited to generate a wide range of biologics for a host of therapeutic indications. In this review, our basic understanding of the antibody structure is described along with how that knowledge has leveraged the engineering of antibody and antibody-related therapeutics having the appropriate antigen affinity, effector function, and biophysical properties. The platforms examined include the development of antibodies, antibody fragments, bispecific antibody, and antibody fusion products, whose efficacy and manufacturability can be improved via humanization, affinity modulation, and stability enhancement. We also review the design and selection of binding arms, and avidity modulation. Different strategies of preparing bispecific and multispecific molecules for an array of therapeutic applications are included.
Collapse
Affiliation(s)
- Mark L. Chiu
- Drug Product Development Science, Janssen Research & Development, LLC, Malvern, PA 19355, USA
- Correspondence:
| | - Dennis R. Goulet
- Department of Medicinal Chemistry, University of Washington, P.O. Box 357610, Seattle, WA 98195-7610, USA;
| | - Alexey Teplyakov
- Biologics Research, Janssen Research & Development, LLC, Spring House, PA 19477, USA; (A.T.); (G.L.G.)
| | - Gary L. Gilliland
- Biologics Research, Janssen Research & Development, LLC, Spring House, PA 19477, USA; (A.T.); (G.L.G.)
| |
Collapse
|
69
|
Singh P, Roche A, van der Walle CF, Uddin S, Du J, Warwicker J, Pluen A, Curtis R. Determination of Protein-Protein Interactions in a Mixture of Two Monoclonal Antibodies. Mol Pharm 2019; 16:4775-4786. [PMID: 31613625 DOI: 10.1021/acs.molpharmaceut.9b00430] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The coformulation of monoclonal antibody (mAb) mixtures provides an attractive route to achieving therapeutic efficacy where the targeting of multiple epitopes is necessary. Controlling and predicting the behavior of such mixtures requires elucidating the molecular basis for the self- and cross-protein-protein interactions and how they depend on solution variables. While self-interactions are now beginning to be well understood, systematic studies of cross-interactions between mAbs in solution do not exist. Here, we have used static light scattering to measure the set of self- and cross-osmotic second virial coefficients in a solution containing a mixture of two mAbs, mAbA and mAbB, as a function of ionic strength and pH. mAbB exhibits strong association at a low ionic strength, which is attributed to an electrostatic attraction that is enhanced by the presence of a strong short-ranged attraction of nonelectrostatic origin. Under all solution conditions, the measured cross-interactions are intermediate self-interactions and follow similar patterns of behavior. There is a strong electrostatic attraction at higher pH values, reflecting the behavior of mAbB. Protein-protein interactions become more attractive with an increasing pH due to reducing the overall protein net charges, an effect that is attenuated with an increasing ionic strength due to the screening of electrostatic interactions. Under moderate ionic strength conditions, the reduced cross-virial coefficient, which reflects only the energetic contribution to protein-protein interactions, is given by a geometric average of the corresponding self-coefficients. We show the relationship can be rationalized using a patchy sphere model, where the interaction energy between sites i and j is given by the arithmetic mean of the i-i and j-j interactions. The geometric mean does not necessarily apply to all mAb mixtures and is expected to break down at a lower ionic strength due to the nonadditivity of electrostatic interactions.
Collapse
Affiliation(s)
- Priyanka Singh
- Manchester Pharmacy School , University of Manchester , Manchester M13 9PL , United Kingdom
| | - Aisling Roche
- School of Chemical Engineering and Analytical Science , University of Manchester , Manchester M1 7DN , United Kingdom
| | - Christopher F van der Walle
- School of Chemical Engineering and Analytical Science , University of Manchester , Manchester M1 7DN , United Kingdom.,Dosage Form Design & Development , AstraZeneca , Granta Park , Cambridge CB21 6GH , United Kingdom
| | - Shahid Uddin
- Formulation Sciences CMC , Immunocore , Milton Park , Abingdon OX14 4RW , United Kingdom
| | - Jiali Du
- Dosage Form Design & Development , AstraZeneca , Gaithersburg MD20878 , United States
| | - Jim Warwicker
- School of Chemistry , University of Manchester , Manchester M1 7DN , United Kingdom
| | - Alain Pluen
- Manchester Pharmacy School , University of Manchester , Manchester M13 9PL , United Kingdom
| | - Robin Curtis
- School of Chemical Engineering and Analytical Science , University of Manchester , Manchester M1 7DN , United Kingdom
| |
Collapse
|
70
|
Datta-Mannan A. Mechanisms Influencing the Pharmacokinetics and Disposition of Monoclonal Antibodies and Peptides. Drug Metab Dispos 2019; 47:1100-1110. [PMID: 31043438 DOI: 10.1124/dmd.119.086488] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 04/22/2019] [Indexed: 12/15/2022] Open
Abstract
Monoclonal antibodies (mAbs) and peptides are an important class of therapeutic modalities that have brought improved health outcomes in areas with limited therapeutic optionality. Presently, there more than 90 mAb and peptide therapeutics on the United States market, with over 600 more in various clinical stages of development in a broad array of therapeutic areas, including diabetes, autoimmune disorders, oncology, neuroscience, and cardiovascular and infectious diseases. Notwithstanding this potential, there is high clinical rate of attrition, with approximately 10% reaching patients. A major contributor to the failure of the molecules is often times an incomplete or poor understanding of the pharmacokinetics (PK) and disposition profiles leading to limited or diminished efficacy. Increased and thorough characterization efforts directed at disseminating mechanisms influencing the PK and disposition of mAbs and peptides can aid in improving the design for their intended pharmacological activity, and thereby their clinical success. The PK and disposition factors for mAbs and peptides are broadly influenced by target-mediated drug disposition and nontarget-related clearance mechanisms related to the interplay between the relationship of the structure and physiochemical properties of mAbs and peptides with physiologic processes. This review focuses on nontarget-related factors influencing the disposition and PK of mAbs and peptides. Contemporary considerations around the increasing in silico approaches to identify nontarget-related molecule limitations and enhancing the druggability of mAbs and peptides, including parenteral and nonparenteral delivery strategies that are geared toward improving patient experience and compliance, are also discussed.
Collapse
Affiliation(s)
- Amita Datta-Mannan
- Department of Experimental Medicine and Pharmacology, Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, Indiana
| |
Collapse
|
71
|
Foti RS, Biswas K, Aral J, Be X, Berry L, Cheng Y, Conner K, Falsey JR, Glaus C, Herberich B, Hickman D, Ikotun T, Li H, Long J, Huang L, Miranda LP, Murray J, Moyer B, Netirojjanakul C, Nixey TE, Sham K, Soto M, Tegley CM, Tran L, Wu B, Yin L, Rock DA. Use of Cryopreserved Hepatocytes as Part of an Integrated Strategy to Characterize In Vivo Clearance for Peptide-Antibody Conjugate Inhibitors of Nav1.7 in Preclinical Species. Drug Metab Dispos 2019; 47:1111-1121. [PMID: 31387871 DOI: 10.1124/dmd.119.087742] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 07/08/2019] [Indexed: 02/13/2025] Open
Abstract
The identification of nonopioid alternatives to treat chronic pain has received a great deal of interest in recent years. Recently, the engineering of a series of Nav1.7 inhibitory peptide-antibody conjugates has been reported, and herein, the preclinical efforts to identify novel approaches to characterize the pharmacokinetic properties of the peptide conjugates are described. A cryopreserved plated mouse hepatocyte assay was designed to measure the depletion of the peptide-antibody conjugates from the media, with a correlation being observed between percentage remaining in the media and in vivo clearance (Pearson r = -0.5525). Physicochemical (charge and hydrophobicity), receptor-binding [neonatal Fc receptor (FcRn)], and in vivo pharmacokinetic data were generated and compared with the results from our in vitro hepatocyte assay, which was hypothesized to encompass all of the aforementioned properties. Correlations were observed among hydrophobicity; FcRn binding; depletion rates from the hepatocyte assay; and ultimately, in vivo clearance. Subsequent studies identified potential roles for the low-density lipoprotein and mannose/galactose receptors in the association of the Nav1.7 peptide conjugates with mouse hepatocytes, although in vivo studies suggested that FcRn was still the primary receptor involved in determining the pharmacokinetics of the peptide conjugates. Ultimately, the use of the cryopreserved hepatocyte assay along with FcRn binding and hydrophobic interaction chromatography provided an efficient and integrated approach to rapidly triage molecules for advancement while reducing the number of in vivo pharmacokinetic studies. SIGNIFICANCE STATEMENT: Although multiple in vitro and in silico tools are available in small-molecule drug discovery, pharmacokinetic characterization of protein therapeutics is still highly dependent upon the use of in vivo studies in preclinical species. The current work demonstrates the combined use of cryopreserved hepatocytes, hydrophobic interaction chromatography, and neonatal Fc receptor binding to characterize a series of Nav1.7 peptide-antibody conjugates prior to conducting in vivo studies, thus providing a means to rapidly evaluate novel protein therapeutic platforms while concomitantly reducing the number of in vivo studies conducted in preclinical species.
Collapse
Affiliation(s)
- Robert S Foti
- Pharmacokinetics and Drug Metabolism, Amgen Research, Cambridge, Massachusetts (R.S.F., X.B., L.B., D.H., L.H.); Therapeutic Discovery (K.B., J.A., Y.C., J.R.F., C.G., B.H., T.I., J.L., L.P.M., J.M., C.N., T.E.N., K.S., C.M.T., B.W., L.Y.), Neuroscience (B.M.), and Pharmacokinetics and Drug Metabolism (H.L., M.S., L.T.), Amgen Research, Thousand Oaks, California; and Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, California (K.C., D.A.R.)
| | - Kaustav Biswas
- Pharmacokinetics and Drug Metabolism, Amgen Research, Cambridge, Massachusetts (R.S.F., X.B., L.B., D.H., L.H.); Therapeutic Discovery (K.B., J.A., Y.C., J.R.F., C.G., B.H., T.I., J.L., L.P.M., J.M., C.N., T.E.N., K.S., C.M.T., B.W., L.Y.), Neuroscience (B.M.), and Pharmacokinetics and Drug Metabolism (H.L., M.S., L.T.), Amgen Research, Thousand Oaks, California; and Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, California (K.C., D.A.R.)
| | - Jennifer Aral
- Pharmacokinetics and Drug Metabolism, Amgen Research, Cambridge, Massachusetts (R.S.F., X.B., L.B., D.H., L.H.); Therapeutic Discovery (K.B., J.A., Y.C., J.R.F., C.G., B.H., T.I., J.L., L.P.M., J.M., C.N., T.E.N., K.S., C.M.T., B.W., L.Y.), Neuroscience (B.M.), and Pharmacokinetics and Drug Metabolism (H.L., M.S., L.T.), Amgen Research, Thousand Oaks, California; and Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, California (K.C., D.A.R.)
| | - Xuhai Be
- Pharmacokinetics and Drug Metabolism, Amgen Research, Cambridge, Massachusetts (R.S.F., X.B., L.B., D.H., L.H.); Therapeutic Discovery (K.B., J.A., Y.C., J.R.F., C.G., B.H., T.I., J.L., L.P.M., J.M., C.N., T.E.N., K.S., C.M.T., B.W., L.Y.), Neuroscience (B.M.), and Pharmacokinetics and Drug Metabolism (H.L., M.S., L.T.), Amgen Research, Thousand Oaks, California; and Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, California (K.C., D.A.R.)
| | - Loren Berry
- Pharmacokinetics and Drug Metabolism, Amgen Research, Cambridge, Massachusetts (R.S.F., X.B., L.B., D.H., L.H.); Therapeutic Discovery (K.B., J.A., Y.C., J.R.F., C.G., B.H., T.I., J.L., L.P.M., J.M., C.N., T.E.N., K.S., C.M.T., B.W., L.Y.), Neuroscience (B.M.), and Pharmacokinetics and Drug Metabolism (H.L., M.S., L.T.), Amgen Research, Thousand Oaks, California; and Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, California (K.C., D.A.R.)
| | - Yuan Cheng
- Pharmacokinetics and Drug Metabolism, Amgen Research, Cambridge, Massachusetts (R.S.F., X.B., L.B., D.H., L.H.); Therapeutic Discovery (K.B., J.A., Y.C., J.R.F., C.G., B.H., T.I., J.L., L.P.M., J.M., C.N., T.E.N., K.S., C.M.T., B.W., L.Y.), Neuroscience (B.M.), and Pharmacokinetics and Drug Metabolism (H.L., M.S., L.T.), Amgen Research, Thousand Oaks, California; and Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, California (K.C., D.A.R.)
| | - Kip Conner
- Pharmacokinetics and Drug Metabolism, Amgen Research, Cambridge, Massachusetts (R.S.F., X.B., L.B., D.H., L.H.); Therapeutic Discovery (K.B., J.A., Y.C., J.R.F., C.G., B.H., T.I., J.L., L.P.M., J.M., C.N., T.E.N., K.S., C.M.T., B.W., L.Y.), Neuroscience (B.M.), and Pharmacokinetics and Drug Metabolism (H.L., M.S., L.T.), Amgen Research, Thousand Oaks, California; and Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, California (K.C., D.A.R.)
| | - James R Falsey
- Pharmacokinetics and Drug Metabolism, Amgen Research, Cambridge, Massachusetts (R.S.F., X.B., L.B., D.H., L.H.); Therapeutic Discovery (K.B., J.A., Y.C., J.R.F., C.G., B.H., T.I., J.L., L.P.M., J.M., C.N., T.E.N., K.S., C.M.T., B.W., L.Y.), Neuroscience (B.M.), and Pharmacokinetics and Drug Metabolism (H.L., M.S., L.T.), Amgen Research, Thousand Oaks, California; and Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, California (K.C., D.A.R.)
| | - Charles Glaus
- Pharmacokinetics and Drug Metabolism, Amgen Research, Cambridge, Massachusetts (R.S.F., X.B., L.B., D.H., L.H.); Therapeutic Discovery (K.B., J.A., Y.C., J.R.F., C.G., B.H., T.I., J.L., L.P.M., J.M., C.N., T.E.N., K.S., C.M.T., B.W., L.Y.), Neuroscience (B.M.), and Pharmacokinetics and Drug Metabolism (H.L., M.S., L.T.), Amgen Research, Thousand Oaks, California; and Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, California (K.C., D.A.R.)
| | - Brad Herberich
- Pharmacokinetics and Drug Metabolism, Amgen Research, Cambridge, Massachusetts (R.S.F., X.B., L.B., D.H., L.H.); Therapeutic Discovery (K.B., J.A., Y.C., J.R.F., C.G., B.H., T.I., J.L., L.P.M., J.M., C.N., T.E.N., K.S., C.M.T., B.W., L.Y.), Neuroscience (B.M.), and Pharmacokinetics and Drug Metabolism (H.L., M.S., L.T.), Amgen Research, Thousand Oaks, California; and Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, California (K.C., D.A.R.)
| | - Dean Hickman
- Pharmacokinetics and Drug Metabolism, Amgen Research, Cambridge, Massachusetts (R.S.F., X.B., L.B., D.H., L.H.); Therapeutic Discovery (K.B., J.A., Y.C., J.R.F., C.G., B.H., T.I., J.L., L.P.M., J.M., C.N., T.E.N., K.S., C.M.T., B.W., L.Y.), Neuroscience (B.M.), and Pharmacokinetics and Drug Metabolism (H.L., M.S., L.T.), Amgen Research, Thousand Oaks, California; and Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, California (K.C., D.A.R.)
| | - Tayo Ikotun
- Pharmacokinetics and Drug Metabolism, Amgen Research, Cambridge, Massachusetts (R.S.F., X.B., L.B., D.H., L.H.); Therapeutic Discovery (K.B., J.A., Y.C., J.R.F., C.G., B.H., T.I., J.L., L.P.M., J.M., C.N., T.E.N., K.S., C.M.T., B.W., L.Y.), Neuroscience (B.M.), and Pharmacokinetics and Drug Metabolism (H.L., M.S., L.T.), Amgen Research, Thousand Oaks, California; and Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, California (K.C., D.A.R.)
| | - Hongyan Li
- Pharmacokinetics and Drug Metabolism, Amgen Research, Cambridge, Massachusetts (R.S.F., X.B., L.B., D.H., L.H.); Therapeutic Discovery (K.B., J.A., Y.C., J.R.F., C.G., B.H., T.I., J.L., L.P.M., J.M., C.N., T.E.N., K.S., C.M.T., B.W., L.Y.), Neuroscience (B.M.), and Pharmacokinetics and Drug Metabolism (H.L., M.S., L.T.), Amgen Research, Thousand Oaks, California; and Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, California (K.C., D.A.R.)
| | - Jason Long
- Pharmacokinetics and Drug Metabolism, Amgen Research, Cambridge, Massachusetts (R.S.F., X.B., L.B., D.H., L.H.); Therapeutic Discovery (K.B., J.A., Y.C., J.R.F., C.G., B.H., T.I., J.L., L.P.M., J.M., C.N., T.E.N., K.S., C.M.T., B.W., L.Y.), Neuroscience (B.M.), and Pharmacokinetics and Drug Metabolism (H.L., M.S., L.T.), Amgen Research, Thousand Oaks, California; and Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, California (K.C., D.A.R.)
| | - Liyue Huang
- Pharmacokinetics and Drug Metabolism, Amgen Research, Cambridge, Massachusetts (R.S.F., X.B., L.B., D.H., L.H.); Therapeutic Discovery (K.B., J.A., Y.C., J.R.F., C.G., B.H., T.I., J.L., L.P.M., J.M., C.N., T.E.N., K.S., C.M.T., B.W., L.Y.), Neuroscience (B.M.), and Pharmacokinetics and Drug Metabolism (H.L., M.S., L.T.), Amgen Research, Thousand Oaks, California; and Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, California (K.C., D.A.R.)
| | - Les P Miranda
- Pharmacokinetics and Drug Metabolism, Amgen Research, Cambridge, Massachusetts (R.S.F., X.B., L.B., D.H., L.H.); Therapeutic Discovery (K.B., J.A., Y.C., J.R.F., C.G., B.H., T.I., J.L., L.P.M., J.M., C.N., T.E.N., K.S., C.M.T., B.W., L.Y.), Neuroscience (B.M.), and Pharmacokinetics and Drug Metabolism (H.L., M.S., L.T.), Amgen Research, Thousand Oaks, California; and Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, California (K.C., D.A.R.)
| | - Justin Murray
- Pharmacokinetics and Drug Metabolism, Amgen Research, Cambridge, Massachusetts (R.S.F., X.B., L.B., D.H., L.H.); Therapeutic Discovery (K.B., J.A., Y.C., J.R.F., C.G., B.H., T.I., J.L., L.P.M., J.M., C.N., T.E.N., K.S., C.M.T., B.W., L.Y.), Neuroscience (B.M.), and Pharmacokinetics and Drug Metabolism (H.L., M.S., L.T.), Amgen Research, Thousand Oaks, California; and Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, California (K.C., D.A.R.)
| | - Bryan Moyer
- Pharmacokinetics and Drug Metabolism, Amgen Research, Cambridge, Massachusetts (R.S.F., X.B., L.B., D.H., L.H.); Therapeutic Discovery (K.B., J.A., Y.C., J.R.F., C.G., B.H., T.I., J.L., L.P.M., J.M., C.N., T.E.N., K.S., C.M.T., B.W., L.Y.), Neuroscience (B.M.), and Pharmacokinetics and Drug Metabolism (H.L., M.S., L.T.), Amgen Research, Thousand Oaks, California; and Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, California (K.C., D.A.R.)
| | - Chawita Netirojjanakul
- Pharmacokinetics and Drug Metabolism, Amgen Research, Cambridge, Massachusetts (R.S.F., X.B., L.B., D.H., L.H.); Therapeutic Discovery (K.B., J.A., Y.C., J.R.F., C.G., B.H., T.I., J.L., L.P.M., J.M., C.N., T.E.N., K.S., C.M.T., B.W., L.Y.), Neuroscience (B.M.), and Pharmacokinetics and Drug Metabolism (H.L., M.S., L.T.), Amgen Research, Thousand Oaks, California; and Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, California (K.C., D.A.R.)
| | - Thomas E Nixey
- Pharmacokinetics and Drug Metabolism, Amgen Research, Cambridge, Massachusetts (R.S.F., X.B., L.B., D.H., L.H.); Therapeutic Discovery (K.B., J.A., Y.C., J.R.F., C.G., B.H., T.I., J.L., L.P.M., J.M., C.N., T.E.N., K.S., C.M.T., B.W., L.Y.), Neuroscience (B.M.), and Pharmacokinetics and Drug Metabolism (H.L., M.S., L.T.), Amgen Research, Thousand Oaks, California; and Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, California (K.C., D.A.R.)
| | - Kelvin Sham
- Pharmacokinetics and Drug Metabolism, Amgen Research, Cambridge, Massachusetts (R.S.F., X.B., L.B., D.H., L.H.); Therapeutic Discovery (K.B., J.A., Y.C., J.R.F., C.G., B.H., T.I., J.L., L.P.M., J.M., C.N., T.E.N., K.S., C.M.T., B.W., L.Y.), Neuroscience (B.M.), and Pharmacokinetics and Drug Metabolism (H.L., M.S., L.T.), Amgen Research, Thousand Oaks, California; and Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, California (K.C., D.A.R.)
| | - Marcus Soto
- Pharmacokinetics and Drug Metabolism, Amgen Research, Cambridge, Massachusetts (R.S.F., X.B., L.B., D.H., L.H.); Therapeutic Discovery (K.B., J.A., Y.C., J.R.F., C.G., B.H., T.I., J.L., L.P.M., J.M., C.N., T.E.N., K.S., C.M.T., B.W., L.Y.), Neuroscience (B.M.), and Pharmacokinetics and Drug Metabolism (H.L., M.S., L.T.), Amgen Research, Thousand Oaks, California; and Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, California (K.C., D.A.R.)
| | - Christopher M Tegley
- Pharmacokinetics and Drug Metabolism, Amgen Research, Cambridge, Massachusetts (R.S.F., X.B., L.B., D.H., L.H.); Therapeutic Discovery (K.B., J.A., Y.C., J.R.F., C.G., B.H., T.I., J.L., L.P.M., J.M., C.N., T.E.N., K.S., C.M.T., B.W., L.Y.), Neuroscience (B.M.), and Pharmacokinetics and Drug Metabolism (H.L., M.S., L.T.), Amgen Research, Thousand Oaks, California; and Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, California (K.C., D.A.R.)
| | - Linh Tran
- Pharmacokinetics and Drug Metabolism, Amgen Research, Cambridge, Massachusetts (R.S.F., X.B., L.B., D.H., L.H.); Therapeutic Discovery (K.B., J.A., Y.C., J.R.F., C.G., B.H., T.I., J.L., L.P.M., J.M., C.N., T.E.N., K.S., C.M.T., B.W., L.Y.), Neuroscience (B.M.), and Pharmacokinetics and Drug Metabolism (H.L., M.S., L.T.), Amgen Research, Thousand Oaks, California; and Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, California (K.C., D.A.R.)
| | - Bin Wu
- Pharmacokinetics and Drug Metabolism, Amgen Research, Cambridge, Massachusetts (R.S.F., X.B., L.B., D.H., L.H.); Therapeutic Discovery (K.B., J.A., Y.C., J.R.F., C.G., B.H., T.I., J.L., L.P.M., J.M., C.N., T.E.N., K.S., C.M.T., B.W., L.Y.), Neuroscience (B.M.), and Pharmacokinetics and Drug Metabolism (H.L., M.S., L.T.), Amgen Research, Thousand Oaks, California; and Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, California (K.C., D.A.R.)
| | - Lin Yin
- Pharmacokinetics and Drug Metabolism, Amgen Research, Cambridge, Massachusetts (R.S.F., X.B., L.B., D.H., L.H.); Therapeutic Discovery (K.B., J.A., Y.C., J.R.F., C.G., B.H., T.I., J.L., L.P.M., J.M., C.N., T.E.N., K.S., C.M.T., B.W., L.Y.), Neuroscience (B.M.), and Pharmacokinetics and Drug Metabolism (H.L., M.S., L.T.), Amgen Research, Thousand Oaks, California; and Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, California (K.C., D.A.R.)
| | - Dan A Rock
- Pharmacokinetics and Drug Metabolism, Amgen Research, Cambridge, Massachusetts (R.S.F., X.B., L.B., D.H., L.H.); Therapeutic Discovery (K.B., J.A., Y.C., J.R.F., C.G., B.H., T.I., J.L., L.P.M., J.M., C.N., T.E.N., K.S., C.M.T., B.W., L.Y.), Neuroscience (B.M.), and Pharmacokinetics and Drug Metabolism (H.L., M.S., L.T.), Amgen Research, Thousand Oaks, California; and Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, California (K.C., D.A.R.)
| |
Collapse
|
72
|
Shehata L, Maurer DP, Wec AZ, Lilov A, Champney E, Sun T, Archambault K, Burnina I, Lynaugh H, Zhi X, Xu Y, Walker LM. Affinity Maturation Enhances Antibody Specificity but Compromises Conformational Stability. Cell Rep 2019; 28:3300-3308.e4. [PMID: 31553901 DOI: 10.1016/j.celrep.2019.08.056] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 06/04/2019] [Accepted: 08/16/2019] [Indexed: 11/16/2022] Open
Abstract
Monoclonal antibodies (mAbs) have recently emerged as one of the most promising classes of biotherapeutics. A potential advantage of B cell-derived mAbs as therapeutic agents is that they have been subjected to natural filtering mechanisms, which may enrich for B cell receptors (BCRs) with favorable biophysical properties. Here, we evaluated 400 human mAbs for polyreactivity, hydrophobicity, and thermal stability using high-throughput screening assays. Overall, mAbs derived from memory B cells and long-lived plasma cells (LLPCs) display reduced levels of polyreactivity, hydrophobicity, and thermal stability compared with naive B cell-derived mAbs. Somatic hypermutation (SHM) is inversely associated with all three biophysical properties, as well as BCR expression levels. Finally, the developability profiles of the human B cell-derived mAbs are comparable with those observed for clinical mAbs, suggesting their high therapeutic potential. The results provide insight into the biophysical consequences of affinity maturation and have implications for therapeutic antibody engineering and development.
Collapse
|
73
|
Lecerf M, Kanyavuz A, Lacroix-Desmazes S, Dimitrov JD. Sequence features of variable region determining physicochemical properties and polyreactivity of therapeutic antibodies. Mol Immunol 2019; 112:338-346. [DOI: 10.1016/j.molimm.2019.06.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 06/17/2019] [Accepted: 06/17/2019] [Indexed: 12/17/2022]
|
74
|
Chen G, Gerrior A, Hale G, Ghosh R. Feasibility study of the fractionation of monoclonal antibody charge variants using a cuboid packed-bed device. Biochem Eng J 2019. [DOI: 10.1016/j.bej.2019.04.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
75
|
Datta-Mannan A, Brown RM, Fitchett J, Heng AR, Balasubramaniam D, Pereira J, Croy JE. Modulation of the Biophysical Properties of Bifunctional Antibodies as a Strategy for Mitigating Poor Pharmacokinetics. Biochemistry 2019; 58:3116-3132. [DOI: 10.1021/acs.biochem.9b00074] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
| | | | - Jonathan Fitchett
- Biotechnology Discovery Research, Lilly Research Laboratories, Lilly Biotechnology Center, San Diego, California 92121, United States
| | - Aik Roy Heng
- Biotechnology Discovery Research, Lilly Research Laboratories, Lilly Biotechnology Center, San Diego, California 92121, United States
| | - Deepa Balasubramaniam
- Biotechnology Discovery Research, Lilly Research Laboratories, Lilly Biotechnology Center, San Diego, California 92121, United States
| | | | | |
Collapse
|
76
|
Rabia LA, Zhang Y, Ludwig SD, Julian MC, Tessier PM. Net charge of antibody complementarity-determining regions is a key predictor of specificity. Protein Eng Des Sel 2019; 31:409-418. [PMID: 30770934 DOI: 10.1093/protein/gzz002] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 12/23/2018] [Accepted: 01/18/2019] [Indexed: 11/14/2022] Open
Abstract
Specificity is one of the most important and complex properties that is central to both natural antibody function and therapeutic antibody efficacy. However, it has proven extremely challenging to define robust guidelines for predicting antibody specificity. Here we evaluated the physicochemical determinants of antibody specificity for multiple panels of antibodies, including >100 clinical-stage antibodies. Surprisingly, we find that the theoretical net charge of the complementarity-determining regions (CDRs) is a strong predictor of antibody specificity. Antibodies with positively charged CDRs have a much higher risk of low specificity than antibodies with negatively charged CDRs. Moreover, the charge of the entire set of six CDRs is a much better predictor of antibody specificity than the charge of individual CDRs, variable domains (VH or VL) or the entire variable fragment (Fv). The best indicators of antibody specificity in terms of CDR amino acid composition are reduced levels of arginine and lysine and increased levels of aspartic and glutamic acid. Interestingly, clinical-stage antibodies with negatively charged CDRs also have a lower risk for poor biophysical properties in general, including a reduced risk for high levels of self-association. These findings provide powerful guidelines for predicting antibody specificity and for identifying safe and potent antibody therapeutics.
Collapse
Affiliation(s)
- Lilia A Rabia
- Isermann Department of Chemical & Biological Engineering, Center for Biotechnology & Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA.,Department of Pharmaceutical Sciences.,Department of Chemical Engineering
| | | | - Seth D Ludwig
- Isermann Department of Chemical & Biological Engineering, Center for Biotechnology & Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Mark C Julian
- Isermann Department of Chemical & Biological Engineering, Center for Biotechnology & Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Peter M Tessier
- Isermann Department of Chemical & Biological Engineering, Center for Biotechnology & Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA.,Department of Pharmaceutical Sciences.,Department of Chemical Engineering.,Department of Biomedical Engineering, Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
77
|
Walker KW, Salimi-Moosavi H, Arnold GE, Chen Q, Soto M, Jacobsen FW, Hui J. Pharmacokinetic comparison of a diverse panel of non-targeting human antibodies as matched IgG1 and IgG2 isotypes in rodents and non-human primates. PLoS One 2019; 14:e0217061. [PMID: 31120944 PMCID: PMC6533040 DOI: 10.1371/journal.pone.0217061] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 05/03/2019] [Indexed: 11/24/2022] Open
Abstract
In this study we compared the pharmacokinetic profile of four unrelated antibodies, which do not bind to mammalian antigens, in IgG1 and IgG2 frameworks in both rats and non-human primates (NHP). This allowed for extensive cross comparison of the impact of antibody isotype, complementarity determining regions (CDR) and model species on pharmacokinetics without the confounding influence of antigen binding in the hosts. While antibody isotype had no significant impact on the pharmacokinetics, the CDRs do alter the profile, and there is an inverse correlation between the neonatal Fc receptor (FcRn) affinity and pharmacokinetic performance. Faster clearance rates were also associated with higher isoelectric points; however, although this panel of antibodies all possess basic isoelectric points, ranging from 8.44 to 9.18, they also have exceptional in vivo half-lives, averaging 369 hours, and low clearance rates, averaging 0.18 ml/h/kg in NHPs. This pattern of pharmacokinetic characteristics was conserved between rats and NHPs.
Collapse
Affiliation(s)
- Kenneth W. Walker
- Amgen Research, Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA, United States of America
- * E-mail:
| | - Hossein Salimi-Moosavi
- Amgen Research, Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA, United States of America
| | - Gregory E. Arnold
- Amgen Research, Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA, United States of America
| | - Qing Chen
- Amgen Research, Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA, United States of America
| | - Marcus Soto
- Amgen Research, Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA, United States of America
| | - Frederick W. Jacobsen
- Amgen Research, Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA, United States of America
| | - John Hui
- Amgen Research, Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA, United States of America
| |
Collapse
|
78
|
Chung S, Nguyen V, Lin YL, Lafrance-Vanasse J, Scales SJ, Lin K, Deng R, Williams K, Sperinde G, Li JJ, Zheng K, Sukumaran S, Tesar D, Ernst JA, Fischer S, Lazar GA, Prabhu S, Song A. An in vitro FcRn- dependent transcytosis assay as a screening tool for predictive assessment of nonspecific clearance of antibody therapeutics in humans. MAbs 2019; 11:942-955. [PMID: 30982394 PMCID: PMC6601550 DOI: 10.1080/19420862.2019.1605270] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
A cell-based assay employing Madin–Darby canine kidney cells stably expressing human neonatal Fc receptor (FcRn) heavy chain and β2-microglobulin genes was developed to measure transcytosis of monoclonal antibodies (mAbs) under conditions relevant to the FcRn-mediated immunoglobulin G (IgG) salvage pathway. The FcRn-dependent transcytosis assay is modeled to reflect combined effects of nonspecific interactions between mAbs and cells, cellular uptake via pinocytosis, pH-dependent interactions with FcRn, and dynamics of intracellular trafficking and sorting mechanisms. Evaluation of 53 mAbs, including 30 marketed mAb drugs, revealed a notable correlation between the transcytosis readouts and clearance in humans. FcRn was required to promote efficient transcytosis of mAbs and contributed directly to the observed correlation. Furthermore, the transcytosis assay correctly predicted rank order of clearance of glycosylation and Fv charge variants of Fc-containing proteins. These results strongly support the utility of this assay as a cost-effective and animal-sparing screening tool for evaluation of mAb-based drug candidates during lead selection, optimization, and process development for desired pharmacokinetic properties.
Collapse
Affiliation(s)
- Shan Chung
- a Department of BioAnalytical Sciences , Genentech Inc ., South San Francisco , CA , USA
| | - Van Nguyen
- a Department of BioAnalytical Sciences , Genentech Inc ., South San Francisco , CA , USA
| | - Yuwen Linda Lin
- a Department of BioAnalytical Sciences , Genentech Inc ., South San Francisco , CA , USA
| | | | - Suzie J Scales
- c Department of Molecular Biology , Genentech Inc ., South San Francisco , CA , USA
| | - Kevin Lin
- d Department of Analytical Operations , Genentech Inc ., South San Francisco , CA , USA
| | - Rong Deng
- e Department of Clinical Pharmacology , Genentech Inc ., South San Francisco , CA , USA
| | - Kathi Williams
- a Department of BioAnalytical Sciences , Genentech Inc ., South San Francisco , CA , USA
| | - Gizette Sperinde
- a Department of BioAnalytical Sciences , Genentech Inc ., South San Francisco , CA , USA
| | - Juan Jenny Li
- f Department of Biochemistry and Cellular Pharmacology , Genentech Inc ., South San Francisco , CA , USA
| | - Kai Zheng
- g Department of Late Stage Pharmaceutical Development , Genentech Inc ., South San Francisco , CA , USA
| | - Siddharth Sukumaran
- h Department of Pharmacokinetics & Pharmacodynamics , Genentech Inc ., South San Francisco , CA , USA
| | - Devin Tesar
- i Department of Drug Delivery , Genentech Inc ., South San Francisco , CA , USA
| | - James A Ernst
- b Department of Protein Chemistry , Genentech Inc ., South San Francisco , CA , USA
| | - Saloumeh Fischer
- a Department of BioAnalytical Sciences , Genentech Inc ., South San Francisco , CA , USA
| | - Greg A Lazar
- j Department of Antibody Engineering , Genentech Inc ., South San Francisco , CA , USA
| | - Saileta Prabhu
- h Department of Pharmacokinetics & Pharmacodynamics , Genentech Inc ., South San Francisco , CA , USA
| | - An Song
- a Department of BioAnalytical Sciences , Genentech Inc ., South San Francisco , CA , USA
| |
Collapse
|
79
|
Julian MC, Rabia LA, Desai AA, Arsiwala A, Gerson JE, Paulson HL, Kane RS, Tessier PM. Nature-inspired design and evolution of anti-amyloid antibodies. J Biol Chem 2019; 294:8438-8451. [PMID: 30918024 DOI: 10.1074/jbc.ra118.004731] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 03/21/2019] [Indexed: 12/17/2022] Open
Abstract
Antibodies that recognize amyloidogenic aggregates with high conformational and sequence specificity are important for detecting and potentially treating a wide range of neurodegenerative disorders, including Alzheimer's and Parkinson's diseases. However, these types of antibodies are challenging to generate because of the large size, hydrophobicity, and heterogeneity of protein aggregates. To address this challenge, we developed a method for generating antibodies specific for amyloid aggregates. First, we grafted amyloidogenic peptide segments from the target polypeptide [Alzheimer's amyloid-β (Aβ) peptide] into the complementarity-determining regions (CDRs) of a stable antibody scaffold. Next, we diversified the grafted and neighboring CDR sites using focused mutagenesis to sample each WT or grafted residue, as well as one to five of the most commonly occurring amino acids at each site in human antibodies. Finally, we displayed these antibody libraries on the surface of yeast cells and selected antibodies that strongly recognize Aβ-amyloid fibrils and only weakly recognize soluble Aβ. We found that this approach enables the generation of monovalent and bivalent antibodies with nanomolar affinity for Aβ fibrils. These antibodies display high conformational and sequence specificity as well as low levels of nonspecific binding and recognize a conformational epitope at the extreme N terminus of human Aβ. We expect that this systematic approach will be useful for generating antibodies with conformational and sequence specificity against a wide range of peptide and protein aggregates associated with neurodegenerative disorders.
Collapse
Affiliation(s)
- Mark C Julian
- Isermann Department of Chemical and Biological Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York 12180
| | - Lilia A Rabia
- Isermann Department of Chemical and Biological Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York 12180; Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, Michigan 48109; Department of Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan 48109; Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan 48109
| | - Alec A Desai
- Department of Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan 48109; Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan 48109
| | - Ammar Arsiwala
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332
| | - Julia E Gerson
- Department of Neurology, University of Michigan, Ann Arbor, Michigan 48109
| | - Henry L Paulson
- Department of Neurology, University of Michigan, Ann Arbor, Michigan 48109; Department of Protein Folding Disease Initiative, University of Michigan, Ann Arbor, Michigan 48109; Department of Michigan Alzheimer's Disease Center, University of Michigan, Ann Arbor, Michigan 48109
| | - Ravi S Kane
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332
| | - Peter M Tessier
- Isermann Department of Chemical and Biological Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York 12180; Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, Michigan 48109; Department of Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan 48109; Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan 48109; Department of Protein Folding Disease Initiative, University of Michigan, Ann Arbor, Michigan 48109; Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan 48109.
| |
Collapse
|
80
|
Congdon EE, Chukwu JE, Shamir DB, Deng J, Ujla D, Sait HBR, Neubert TA, Kong XP, Sigurdsson EM. Tau antibody chimerization alters its charge and binding, thereby reducing its cellular uptake and efficacy. EBioMedicine 2019; 42:157-173. [PMID: 30910484 PMCID: PMC6492224 DOI: 10.1016/j.ebiom.2019.03.033] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 03/11/2019] [Accepted: 03/12/2019] [Indexed: 12/28/2022] Open
Abstract
Background Bringing antibodies from pre-clinical studies to human trials requires humanization, but this process may alter properties that are crucial for efficacy. Since pathological tau protein is primarily intraneuronal in Alzheimer's disease, the most efficacious antibodies should work both intra- and extracellularly. Thus, changes which impact uptake or antibody binding will affect antibody efficacy. Methods Initially, we examined four tau mouse monoclonal antibodies with naturally differing charges. We quantified their neuronal uptake, and efficacy in preventing toxicity and pathological seeding induced by human-derived pathological tau. Later, we generated a human chimeric 4E6 (h4E6), an antibody with well documented efficacy in multiple tauopathy models. We compared the uptake and efficacy of unmodified and chimeric antibodies in neuronal and differentiated neuroblastoma cultures. Further, we analyzed tau binding using ELISA assays. Findings Neuronal uptake of tau antibodies and their efficacy strongly depends on antibody charge. Additionally, their ability to prevent tau toxicity and seeding of tau pathology does not necessarily go together. Particularly, chimerization of 4E6 increased its charge from 6.5 to 9.6, which blocked its uptake into human and mouse cells. Furthermore, h4E6 had altered binding characteristics despite intact binding sites, compared to the mouse antibody. Importantly, these changes in uptake and binding substantially decreased its efficacy in preventing tau toxicity, although under certain conditions it did prevent pathological seeding of tau. Conclusions These results indicate that efficacy of chimeric/humanized tau antibodies should be thoroughly characterized prior to clinical trials, which may require further engineering to maintain or improve their therapeutic potential. Fund National Institutes of Health (NS077239, AG032611, R24OD18340, R24OD018339 and RR027990, Alzheimer's Association (2016-NIRG-397228) and Blas Frangione Foundation.
Collapse
Affiliation(s)
- Erin E Congdon
- New York University School of Medicine, Department of Neuroscience and Physiology, and The Neuroscience Institute, 435 E 30th St. SB1123, New York, NY 10016, United States of America
| | - Jessica E Chukwu
- New York University School of Medicine, Department of Biochemistry and Molecular Pharmacology, 550 First Ave, MSB 398, New York, NY 10016, United States of America
| | - Dov B Shamir
- New York University School of Medicine, Department of Neuroscience and Physiology, and The Neuroscience Institute, 435 E 30th St. SB1123, New York, NY 10016, United States of America
| | - Jingjing Deng
- New York University School of Medicine, Department of Cell Biology, 540 First Avenue, Skirball Institute Lab 5-18, New York, NY 10016, United States of America
| | - Devyani Ujla
- New York University School of Medicine, Department of Neuroscience and Physiology, and The Neuroscience Institute, 435 E 30th St. SB1123, New York, NY 10016, United States of America
| | - Hameetha B R Sait
- New York University School of Medicine, Department of Neuroscience and Physiology, and The Neuroscience Institute, 435 E 30th St. SB1123, New York, NY 10016, United States of America
| | - Thomas A Neubert
- New York University School of Medicine, Department of Cell Biology, 540 First Avenue, Skirball Institute Lab 5-18, New York, NY 10016, United States of America
| | - Xiang-Peng Kong
- New York University School of Medicine, Department of Biochemistry and Molecular Pharmacology, 550 First Ave, MSB 398, New York, NY 10016, United States of America
| | - Einar M Sigurdsson
- New York University School of Medicine, Department of Neuroscience and Physiology, and The Neuroscience Institute, 435 E 30th St. SB1123, New York, NY 10016, United States of America; New York University School of Medicine, Department of Psychiatry, 435 E 30th St. Science Building SB1115, New York, NY 10016, United States of America.
| |
Collapse
|
81
|
Starr CG, Tessier PM. Selecting and engineering monoclonal antibodies with drug-like specificity. Curr Opin Biotechnol 2019; 60:119-127. [PMID: 30822699 DOI: 10.1016/j.copbio.2019.01.008] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Revised: 11/16/2018] [Accepted: 01/19/2019] [Indexed: 11/19/2022]
Abstract
Despite the recent explosion in the use of monoclonal antibodies (mAbs) as drugs, it remains a significant challenge to generate antibodies with a combination of physicochemical properties that are optimal for therapeutic applications. We argue that one of the most important and underappreciated drug-like antibody properties is high specificity - defined here as low levels of antibody non-specific and self-interactions - which is linked to low off-target binding and slow antibody clearance in vivo and high solubility and low viscosity in vitro. Here, we review the latest advances in characterizing antibody specificity and elucidating its molecular determinants as well as using these findings to improve the selection and engineering of antibodies with extremely high, drug-like specificity.
Collapse
Affiliation(s)
- Charles G Starr
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Peter M Tessier
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
82
|
Faber MS, Whitehead TA. Data-driven engineering of protein therapeutics. Curr Opin Biotechnol 2019; 60:104-110. [PMID: 30822697 DOI: 10.1016/j.copbio.2019.01.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 11/16/2018] [Accepted: 01/21/2019] [Indexed: 12/26/2022]
Abstract
Protein therapeutics requires a series of properties beyond biochemical activity, including serum stability, low immunogenicity, and manufacturability. Mutations that improve one property often decrease one or more of the other essential requirements for therapeutic efficacy, making the protein engineering challenge difficult. The past decade has seen an explosion of new techniques centered around cheaply reading and writing DNA. This review highlights the recent use of such high throughput technologies for engineering protein therapeutics. Examples include the use of human antibody repertoire sequence data to pair antibody heavy and light chains, comprehensive mutational analysis for engineering antibody specificity, and the use of ancestral and inter-species sequence data to engineer simultaneous improvements in enzyme catalytic efficiency and stability. We conclude with a perspective on further ways to integrate mature protein engineering pipelines with the exponential increases in the volume of sequencing data expected in the forthcoming decade.
Collapse
Affiliation(s)
- Matthew S Faber
- Dept. Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI 48824, United States
| | - Timothy A Whitehead
- Dept. of Chemical Engineering & Materials Science, Michigan State University, East Lansing, MI 48824, United States; Dept. of Biosystems Engineering, Michigan State University, East Lansing, MI 48824, United States; Dept. of Biomedical Engineering, Michigan State University, East Lansing, MI 48824, United States; Institute for Quantitative Biology, Michigan State University, East Lansing, MI 48824, United States.
| |
Collapse
|
83
|
Abstract
Immunogenicity, instability, self-association, high viscosity, polyspecificity, or poor expression can all preclude an antibody from becoming a therapeutic. Early identification of these negative characteristics is essential. Akin to the Lipinski guidelines, which measure druglikeness in small molecules, our Therapeutic Antibody Profiler highlights antibodies that possess characteristics that are rare/unseen in clinical-stage mAb therapeutics. The only required input is the variable domain sequence. We show examples where our approach would have advised against manufacturing antibodies that were found to aggregate or have poor expression. Therapeutic mAbs must not only bind to their target but must also be free from “developability issues” such as poor stability or high levels of aggregation. While small-molecule drug discovery benefits from Lipinski’s rule of five to guide the selection of molecules with appropriate biophysical properties, there is currently no in silico analog for antibody design. Here, we model the variable domain structures of a large set of post-phase-I clinical-stage antibody therapeutics (CSTs) and calculate in silico metrics to estimate their typical properties. In each case, we contextualize the CST distribution against a snapshot of the human antibody gene repertoire. We describe guideline values for five metrics thought to be implicated in poor developability: the total length of the complementarity-determining regions (CDRs), the extent and magnitude of surface hydrophobicity, positive charge and negative charge in the CDRs, and asymmetry in the net heavy- and light-chain surface charges. The guideline cutoffs for each property were derived from the values seen in CSTs, and a flagging system is proposed to identify nonconforming candidates. On two mAb drug discovery sets, we were able to selectively highlight sequences with developability issues. We make available the Therapeutic Antibody Profiler (TAP), a computational tool that builds downloadable homology models of variable domain sequences, tests them against our five developability guidelines, and reports potential sequence liabilities and canonical forms. TAP is freely available at opig.stats.ox.ac.uk/webapps/sabdab-sabpred/TAP.php.
Collapse
|
84
|
Noh J, Kim O, Jung Y, Han H, Kim JE, Kim S, Lee S, Park J, Jung RH, Kim SI, Park J, Han J, Lee H, Yoo DK, Lee AC, Kwon E, Ryu T, Chung J, Kwon S. High-throughput retrieval of physical DNA for NGS-identifiable clones in phage display library. MAbs 2019; 11:532-545. [PMID: 30735467 DOI: 10.1080/19420862.2019.1571878] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
In antibody discovery, in-depth analysis of an antibody library and high-throughput retrieval of clones in the library are crucial to identifying and exploiting rare clones with different properties. However, existing methods have technical limitations, such as low process throughput from the laborious cloning process and waste of the phenotypic screening capacity from unnecessary repetitive tests on the dominant clones. To overcome the limitations, we developed a new high-throughput platform for the identification and retrieval of clones in the library, TrueRepertoire™. This new platform provides highly accurate sequences of the clones with linkage information between heavy and light chains of the antibody fragment. Additionally, the physical DNA of clones can be retrieved in high throughput based on the sequence information. We validated the high accuracy of the sequences and demonstrated that there is no platform-specific bias. Moreover, the applicability of TrueRepertoire™ was demonstrated by a phage-displayed single-chain variable fragment library targeting human hepatocyte growth factor protein.
Collapse
Affiliation(s)
- Jinsung Noh
- a Department of Electrical Engineering and Computer Science , Seoul National University , Seoul , Republic of Korea
| | - Okju Kim
- a Department of Electrical Engineering and Computer Science , Seoul National University , Seoul , Republic of Korea.,b Bioengineering Research Institute, Celemics, Inc , Seoul , Republic of Korea
| | - Yushin Jung
- b Bioengineering Research Institute, Celemics, Inc , Seoul , Republic of Korea
| | - Haejun Han
- b Bioengineering Research Institute, Celemics, Inc , Seoul , Republic of Korea
| | - Jung-Eun Kim
- b Bioengineering Research Institute, Celemics, Inc , Seoul , Republic of Korea
| | - Soohyun Kim
- c Department of Biochemistry and Molecular Biology , Seoul National University College of Medicine , Seoul , Republic of Korea.,d Cancer Research Institute , Seoul National University College of Medicine , Seoul , Republic of Korea
| | - Sanghyub Lee
- b Bioengineering Research Institute, Celemics, Inc , Seoul , Republic of Korea
| | - Jaeseong Park
- b Bioengineering Research Institute, Celemics, Inc , Seoul , Republic of Korea
| | - Rae Hyuck Jung
- e Inter-University Semiconductor Research Center , Seoul National University , Seoul , Republic of Korea
| | - Sang Il Kim
- c Department of Biochemistry and Molecular Biology , Seoul National University College of Medicine , Seoul , Republic of Korea.,d Cancer Research Institute , Seoul National University College of Medicine , Seoul , Republic of Korea
| | - Jaejun Park
- b Bioengineering Research Institute, Celemics, Inc , Seoul , Republic of Korea
| | - Jerome Han
- c Department of Biochemistry and Molecular Biology , Seoul National University College of Medicine , Seoul , Republic of Korea.,f Department of Biomedical Science , Seoul National University College of Medicine , Seoul , Republic of Korea
| | - Hyunho Lee
- a Department of Electrical Engineering and Computer Science , Seoul National University , Seoul , Republic of Korea
| | - Duck Kyun Yoo
- c Department of Biochemistry and Molecular Biology , Seoul National University College of Medicine , Seoul , Republic of Korea.,f Department of Biomedical Science , Seoul National University College of Medicine , Seoul , Republic of Korea.,g Neuro-Immune Information Storage Network Research Center , Seoul National University College of Medicine , Seoul , Republic of Korea
| | - Amos C Lee
- h Interdisciplinary Program in Bioengineering , Seoul National University , Seoul , Republic of Korea
| | - Euijin Kwon
- b Bioengineering Research Institute, Celemics, Inc , Seoul , Republic of Korea
| | - Taehoon Ryu
- b Bioengineering Research Institute, Celemics, Inc , Seoul , Republic of Korea
| | - Junho Chung
- c Department of Biochemistry and Molecular Biology , Seoul National University College of Medicine , Seoul , Republic of Korea.,d Cancer Research Institute , Seoul National University College of Medicine , Seoul , Republic of Korea.,f Department of Biomedical Science , Seoul National University College of Medicine , Seoul , Republic of Korea
| | - Sunghoon Kwon
- a Department of Electrical Engineering and Computer Science , Seoul National University , Seoul , Republic of Korea.,e Inter-University Semiconductor Research Center , Seoul National University , Seoul , Republic of Korea.,h Interdisciplinary Program in Bioengineering , Seoul National University , Seoul , Republic of Korea.,i Institutes of Entrepreneurial BioConvergence , Seoul National University , Seoul , Republic of Korea.,j Seoul National University Hospital Biomedical Research Institute , Seoul National University Hospital , Seoul , Republic of Korea
| |
Collapse
|
85
|
Glassman PM, Balthasar JP. Physiologically-based modeling of monoclonal antibody pharmacokinetics in drug discovery and development. Drug Metab Pharmacokinet 2019; 34:3-13. [PMID: 30522890 PMCID: PMC6378116 DOI: 10.1016/j.dmpk.2018.11.002] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 09/11/2018] [Accepted: 11/19/2018] [Indexed: 12/20/2022]
Abstract
Over the past few decades, monoclonal antibodies (mAbs) have become one of the most important and fastest growing classes of therapeutic molecules, with applications in a wide variety of disease areas. As such, understanding of the determinants of mAb pharmacokinetic (PK) processes (absorption, distribution, metabolism, and elimination) is crucial in developing safe and efficacious therapeutics. In the present review, we discuss the use of physiologically-based pharmacokinetic (PBPK) models as an approach to characterize the in vivo behavior of mAbs, in the context of the key PK processes that should be considered in these models. Additionally, we discuss current and potential future applications of PBPK in the drug discovery and development timeline for mAbs, spanning from identification of potential target molecules to prediction of potential drug-drug interactions. Finally, we conclude with a discussion of currently available PBPK models for mAbs that could be implemented in the drug development process.
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; Department of Pharmacology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104 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
|
86
|
Leipold D, Prabhu S. Pharmacokinetic and Pharmacodynamic Considerations in the Design of Therapeutic Antibodies. Clin Transl Sci 2018; 12:130-139. [PMID: 30414357 PMCID: PMC6440574 DOI: 10.1111/cts.12597] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 10/01/2018] [Indexed: 12/12/2022] Open
Abstract
The design and development of therapeutic monoclonal antibodies (mAbs) through optimizing their pharmacokinetic (PK) and pharmacodynamic (PD) properties is crucial to improve efficacy while minimizing adverse events. Many of these properties are interdependent, which highlights the inherent challenges in therapeutic antibody design, where improving one antibody property can sometimes lead to changes in others. Here, we discuss optimization approaches for PK/PD properties of therapeutic mAbs.
Collapse
Affiliation(s)
- Douglas Leipold
- Preclinical and Translational Pharmacokinetics/Pharmacodynamics, Genentech, South San Francisco, California, USA
| | - Saileta Prabhu
- Preclinical and Translational Pharmacokinetics/Pharmacodynamics, Genentech, South San Francisco, California, USA
| |
Collapse
|
87
|
Xu Y, Wang D, Mason B, Rossomando T, Li N, Liu D, Cheung JK, Xu W, Raghava S, Katiyar A, Nowak C, Xiang T, Dong DD, Sun J, Beck A, Liu H. Structure, heterogeneity and developability assessment of therapeutic antibodies. MAbs 2018; 11:239-264. [PMID: 30543482 DOI: 10.1080/19420862.2018.1553476] [Citation(s) in RCA: 177] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Increasing attention has been paid to developability assessment with the understanding that thorough evaluation of monoclonal antibody lead candidates at an early stage can avoid delays during late-stage development. The concept of developability is based on the knowledge gained from the successful development of approximately 80 marketed antibody and Fc-fusion protein drug products and from the lessons learned from many failed development programs over the last three decades. Here, we reviewed antibody quality attributes that are critical to development and traditional and state-of-the-art analytical methods to monitor those attributes. Based on our collective experiences, a practical workflow is proposed as a best practice for developability assessment including in silico evaluation, extended characterization and forced degradation using appropriate analytical methods that allow characterization with limited material consumption and fast turnaround time.
Collapse
Affiliation(s)
- Yingda Xu
- a Protein Analytics , Adimab , Lebanon , NH , USA
| | - Dongdong Wang
- b Analytical Department , Bioanalytix, Inc ., Cambridge , MA , USA
| | - Bruce Mason
- c Product Characterization , Alexion Pharmaceuticals, Inc ., New Haven , CT , USA
| | - Tony Rossomando
- c Product Characterization , Alexion Pharmaceuticals, Inc ., New Haven , CT , USA
| | - Ning Li
- d Analytical Chemistry , Regeneron Pharmaceuticals, Inc ., Tarrytown , NY , USA
| | - Dingjiang Liu
- e Formulation Development , Regeneron Pharmaceuticals, Inc ., Tarrytown , NY , USA
| | - Jason K Cheung
- f Pharmaceutical Sciences , MRL, Merck & Co., Inc ., Kenilworth , NJ , USA
| | - Wei Xu
- g Analytical Method Development , MRL, Merck & Co., Inc ., Kenilworth , NJ , USA
| | - Smita Raghava
- h Sterile Formulation Sciences , MRL, Merck & Co., Inc ., Kenilworth , NJ , USA
| | - Amit Katiyar
- i Analytical Development , Bristol-Myers Squibb , Pennington , NJ , USA
| | - Christine Nowak
- c Product Characterization , Alexion Pharmaceuticals, Inc ., New Haven , CT , USA
| | - Tao Xiang
- j Manufacturing Sciences , Abbvie Bioresearch Center , Worcester , MA , USA
| | - Diane D Dong
- j Manufacturing Sciences , Abbvie Bioresearch Center , Worcester , MA , USA
| | - Joanne Sun
- k Product development , Innovent Biologics , Suzhou Industrial Park , China
| | - Alain Beck
- l Analytical chemistry , NBEs, Center d'immunologie Pierre Fabre , St Julien-en-Genevois Cedex , France
| | - Hongcheng Liu
- c Product Characterization , Alexion Pharmaceuticals, Inc ., New Haven , CT , USA
| |
Collapse
|
88
|
Goulet DR, Watson MJ, Tam SH, Zwolak A, Chiu ML, Atkins WM, Nath A. Toward a Combinatorial Approach for the Prediction of IgG Half-Life and Clearance. Drug Metab Dispos 2018; 46:1900-1907. [PMID: 30232177 PMCID: PMC7370997 DOI: 10.1124/dmd.118.081893] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 09/10/2018] [Indexed: 12/27/2022] Open
Abstract
The serum half-life and clearance of therapeutic monoclonal antibodies (mAbs) are critical factors that impact their efficacy and optimal dosing regimen. The pH-dependent binding of an mAb to the neonatal Fc receptor (FcRn) has long been recognized as an important determinant of its pharmacokinetics. However, FcRn affinity alone is not a reliable predictor of mAb half-life, suggesting that other biologic or biophysical mechanisms must be accounted for. mAb thermal stability, which reflects its unfolding and aggregation propensities, may also relate to its pharmacokinetic properties. However, no rigorous statistical regression methods have been used to identify combinations of physical parameters that best predict biologic properties. In this work, a panel of eight mAbs with published human pharmacokinetic data were selected for biophysical analyses of FcRn binding and thermal stability. Biolayer interferometry was used to characterize FcRn/mAb binding at acidic and neutral pH, while differential scanning calorimetry was used to determine thermodynamic unfolding parameters. Individual binding or stability parameters were generally weakly correlated with half-life and clearance values. Least absolute shrinkage and selection operator regression was used to identify the combination of two parameters with the best correlation to half-life and clearance as being the FcRn binding response at pH 7.0 and the change in heat capacity. Leave-one-out subsampling yielded a root mean square difference between observed and predicted half-life of just 2.7 days (16%). Thus, the incorporation of multiple biophysical parameters into a cohesive model may facilitate early-stage prediction of in vivo half-life and clearance based on simple in vitro experiments.
Collapse
Affiliation(s)
- Dennis R Goulet
- Department of Medicinal Chemistry, University of Washington, Seattle, Washington (D.R.G., M.J.W., W.M.A., A.N.); and Biologics Research, Janssen Research and Development, LLC, Spring House, Pennsylvania (S.H.T., A.Z., M.L.C.)
| | - Michael J Watson
- Department of Medicinal Chemistry, University of Washington, Seattle, Washington (D.R.G., M.J.W., W.M.A., A.N.); and Biologics Research, Janssen Research and Development, LLC, Spring House, Pennsylvania (S.H.T., A.Z., M.L.C.)
| | - Susan H Tam
- Department of Medicinal Chemistry, University of Washington, Seattle, Washington (D.R.G., M.J.W., W.M.A., A.N.); and Biologics Research, Janssen Research and Development, LLC, Spring House, Pennsylvania (S.H.T., A.Z., M.L.C.)
| | - Adam Zwolak
- Department of Medicinal Chemistry, University of Washington, Seattle, Washington (D.R.G., M.J.W., W.M.A., A.N.); and Biologics Research, Janssen Research and Development, LLC, Spring House, Pennsylvania (S.H.T., A.Z., M.L.C.)
| | - Mark L Chiu
- Department of Medicinal Chemistry, University of Washington, Seattle, Washington (D.R.G., M.J.W., W.M.A., A.N.); and Biologics Research, Janssen Research and Development, LLC, Spring House, Pennsylvania (S.H.T., A.Z., M.L.C.)
| | - William M Atkins
- Department of Medicinal Chemistry, University of Washington, Seattle, Washington (D.R.G., M.J.W., W.M.A., A.N.); and Biologics Research, Janssen Research and Development, LLC, Spring House, Pennsylvania (S.H.T., A.Z., M.L.C.)
| | - Abhinav Nath
- Department of Medicinal Chemistry, University of Washington, Seattle, Washington (D.R.G., M.J.W., W.M.A., A.N.); and Biologics Research, Janssen Research and Development, LLC, Spring House, Pennsylvania (S.H.T., A.Z., M.L.C.)
| |
Collapse
|
89
|
Improvement of pharmacokinetic properties of therapeutic antibodies by antibody engineering. Drug Metab Pharmacokinet 2018; 34:25-41. [PMID: 30472066 DOI: 10.1016/j.dmpk.2018.10.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 09/13/2018] [Accepted: 10/23/2018] [Indexed: 01/17/2023]
Abstract
Monoclonal antibodies (mAbs) have become an important therapeutic option for several diseases. Since several mAbs have shown promising efficacy in clinic, the competition to develop mAbs has become severe. In efforts to gain a competitive advantage over other mAbs and provide significant benefits to patients, innovations in antibody engineering have aimed at improving the pharmacokinetic properties of mAbs. Because engineering can provide therapeutics that are more convenient, safer, and more efficacious for patients in several disease areas, it is an attractive approach to provide significant benefits to patients. Further advances in engineering mAbs to modulate their pharmacokinetics were driven by the increase of total soluble target antigen concentration that is often observed after injecting a mAb, which then requires a high dosage to antagonize. To decrease the required dosage, several antibody engineering techniques have been invented that reduce the total concentration of soluble target antigen. Here, we review the various ways that antibody engineering can improve the pharmacokinetic properties of mAbs.
Collapse
|
90
|
Targeting FcRn to Generate Antibody-Based Therapeutics. Trends Pharmacol Sci 2018; 39:892-904. [PMID: 30143244 DOI: 10.1016/j.tips.2018.07.007] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 07/24/2018] [Accepted: 07/27/2018] [Indexed: 01/01/2023]
Abstract
The MHC class I-related receptor FcRn serves multiple roles ranging from the regulation of levels of IgG isotype antibodies and albumin throughout the body to the delivery of antigen into antigen loading compartments in specialized antigen-presenting cells. In parallel with studies directed towards understanding FcRn at the molecular and cellular levels, there has been an enormous expansion in the development of engineering strategies involving FcRn to modulate the dynamic behavior of antibodies, antigens, and albumin. In this review article, we focus on a discussion of FcRn-targeted approaches that have resulted in the production of novel antibody-based platforms with considerable potential for use in the clinic.
Collapse
|
91
|
Witten J, Samad T, Ribbeck K. Selective permeability of mucus barriers. Curr Opin Biotechnol 2018; 52:124-133. [PMID: 29674157 PMCID: PMC7132988 DOI: 10.1016/j.copbio.2018.03.010] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Revised: 03/23/2018] [Accepted: 03/26/2018] [Indexed: 12/18/2022]
Abstract
Mucus is a hydrogel that exhibits complex selective permeability, permitting the passage of some particles while restricting the passage of other particles including important therapeutics. In this review, we discuss biochemical mechanisms underlying mucus penetration and mucus binding, emphasizing the importance of steric, electrostatic, and hydrophobic interactions. We discuss emerging techniques for engineering nanoparticle surface chemistries for mucus penetration as well as recent advances in tuning mucus interactions with small molecule, peptide, or protein therapeutics. Finally, we highlight recent work suggesting that mucus permeability can serve as a biomarker for disease and physiological states such as pregnancy.
Collapse
Affiliation(s)
- Jacob Witten
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Computational and Systems Biology Initiative, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Tahoura Samad
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Katharina Ribbeck
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
92
|
Hardiansyah D, Ng CM. Effects of the FcRn developmental pharmacology on the pharmacokinetics of therapeutic monoclonal IgG antibody in pediatric subjects using minimal physiologically-based pharmacokinetic modelling. MAbs 2018; 10:1144-1156. [PMID: 29969360 DOI: 10.1080/19420862.2018.1494479] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The aim of this study was to investigate neonatal Fc receptor (FcRn) concentration developmental pharmacology in adult and pediatric subjects using minimal physiologically-based pharmacokinetic (mPBPK) modelling. Three types of pharmacokinetic (PK) data for three agents (endogenous/exogenous native IgG, bevacizumab and palivizumab) were used. The adult group contained six subjects with weights from 50 to 100 kg. For pediatric subjects, seven age groups were assumed, with five subjects each having the weight of 95%, 75%, 50%, 25% and 5% percentile of the population. A first evidence-based rating system to evaluate the quality of the source data used to derive pediatric-specific mPBPK model parameter was proposed. A stepwise approach was used to examine the best combination of age/weight effect on the parameters of the mPBPK model in adult and pediatric subjects. IgG synthesis rate (Ksyn), extravasation rate (ER) and FcRn were fitted simultaneously to the PK of bevacizumab and native-IgG in both adult and pediatric. All fitting showed good fits based on the graphs and the coefficient of variation of the fitted parameters (< 50%). Estimated weight-normalized Ksyn increased while weight-normalized FcRn and ER decreased with increasing age. The age and weight effect on FcRn were successfully estimated from the data. The final mPBPK model developed with native IgG and bevacizumab was able to predict the PK of palivizumab in pediatric subjects. Implementation of the mPBPK model enables us to analyze the relationships of age, weight, FcRn, ER and Ksyn in both adult and pediatric subject. This information may benefit the understanding of complex interaction between the FcRn developmental pharmacology and PK parameters, and improve the prediction of the antibody disposition in pediatric subjects.
Collapse
Affiliation(s)
- Deni Hardiansyah
- a College of Pharmacy , University of Kentucky , Lexington , USA
| | - Chee Meng Ng
- a College of Pharmacy , University of Kentucky , Lexington , USA
| |
Collapse
|
93
|
Datta-Mannan A, Boyles J, Huang L, Jin ZY, Peariso A, Murphy AT, Ellis B, Douglass N, Norouziyan-Cooper F, Witcher DR. Engineered FcRn Binding Fusion Peptides Significantly Enhance the Half-Life of a Fab Domain in Cynomolgus Monkeys. Biotechnol J 2018; 14:e1800007. [DOI: 10.1002/biot.201800007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 04/25/2018] [Indexed: 01/04/2023]
Affiliation(s)
- Amita Datta-Mannan
- Department of Drug Disposition, Development/Commercialization; Lilly Research Laboratories; Lilly Corporate Center; Indianapolis Indiana
| | - Jeffrey Boyles
- Department of Biotechnology Discovery Research; Lilly Research Laboratories; Lilly Corporate Center; Indianapolis Indiana
| | - Lihua Huang
- Department of Bioproduct Research/Development; Lilly Research Laboratories; Lilly Corporate Center; Indianapolis Indiana
| | - Zhaoyan Y. Jin
- Department of Drug Disposition, Development/Commercialization; Lilly Research Laboratories; Lilly Corporate Center; Indianapolis Indiana
| | - Amber Peariso
- Department of Biotechnology Discovery Research; Lilly Research Laboratories; Lilly Corporate Center; Indianapolis Indiana
| | - Anthony T. Murphy
- Department of Drug Disposition, Development/Commercialization; Lilly Research Laboratories; Lilly Corporate Center; Indianapolis Indiana
| | - Bernice Ellis
- Department of Drug Disposition, Development/Commercialization; Lilly Research Laboratories; Lilly Corporate Center; Indianapolis Indiana
| | - Nicole Douglass
- Department of Drug Disposition, Development/Commercialization; Lilly Research Laboratories; Lilly Corporate Center; Indianapolis Indiana
| | - Fariba Norouziyan-Cooper
- Department of Drug Disposition, Development/Commercialization; Lilly Research Laboratories; Lilly Corporate Center; Indianapolis Indiana
| | - Derrick R. Witcher
- Department of Biotechnology Discovery Research; Lilly Research Laboratories; Lilly Corporate Center; Indianapolis Indiana
| |
Collapse
|
94
|
Rabia LA, Desai AA, Jhajj HS, Tessier PM. Understanding and overcoming trade-offs between antibody affinity, specificity, stability and solubility. Biochem Eng J 2018; 137:365-374. [PMID: 30666176 DOI: 10.1016/j.bej.2018.06.003] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The widespread use of monoclonal antibodies for therapeutic applications has led to intense interest in optimizing several of their natural properties (affinity, specificity, stability, solubility and effector functions) as well as introducing non-natural activities (bispecificity and cytotoxicity mediated by conjugated drugs). A common challenge during antibody optimization is that improvements in one property (e.g., affinity) can lead to deficits in other properties (e.g., stability). Here we review recent advances in understanding trade-offs between different antibody properties, including affinity, specificity, stability and solubility. We also review new approaches for co-optimizing multiple antibody properties and discuss how these methods can be used to rapidly and systematically generate antibodies for a wide range of applications.
Collapse
Affiliation(s)
- Lilia A Rabia
- Center for Biotechnology & Interdisciplinary Studies, Isermann Dept. of Chemical & Biological Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180
| | - Alec A Desai
- Department of Chemical Engineering, Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Harkamal S Jhajj
- Department of Biomedical Engineering, Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Peter M Tessier
- Center for Biotechnology & Interdisciplinary Studies, Isermann Dept. of Chemical & Biological Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180
- Department of Chemical Engineering, Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Biomedical Engineering, Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Pharmaceutical Sciences, Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
95
|
Two-Pore Minimum Physiologically-based Pharmacokinetic Model to Describe the Disposition of Therapeutic Monoclonal IgG Antibody in Humans. Pharm Res 2018; 35:47. [PMID: 29411151 DOI: 10.1007/s11095-017-2292-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 10/23/2017] [Indexed: 12/26/2022]
Abstract
PURPOSE The aim of this study was to develop a two-pore minimum physiologically-based pharmacokinetic (mPBPK) model in describing the pharmacokinetic (PK) of therapeutic monoclonal antibody (TMAb) in human subjects. METHODS PK data used in this study were endogenous/exogenous native IgG and two TMAbs (palivizumab and Motavizumab-YTE) in normal volunteer or familial hypercatabolic hypoproteinemia (FIHH) patient. Several important components were implemented to overcome the limitations of the early mPBPK model, e.g. two-pore model to describe the transcapillary transport of IgG from vascular to interstitial space. Six mPBPK models with different osmotic reflection coefficient (OFC) of transcapillary transport, endocytosis rates (ETR) and plasma clearance for the TMAbs/IgG were tested and the best model was selected using AICc values. RESULTS The final model consisted of different OFC and ETR values for native IgG and TMAbs, supporting the hypothesis that the dynamics in the endosomal space had an important role in the compliant FcRn salvage mechanism to determine the clearance of TMAbs. The estimated FcRn concentration of FIHH subjects was 2.72 μmol/l. The final two-pore mPBPK model has a better performance for native IgG than previously developed mPBPK model. CONCLUSIONS The final two-pore mPBPK model not only overcome the limitations of the early mPBPK model but also has a better performance to describe the disposition of the IgG antibody in human subjects.
Collapse
|
96
|
Avery LB, Wade J, Wang M, Tam A, King A, Piche-Nicholas N, Kavosi MS, Penn S, Cirelli D, Kurz JC, Zhang M, Cunningham O, Jones R, Fennell BJ, McDonnell B, Sakorafas P, Apgar J, Finlay WJ, Lin L, Bloom L, O'Hara DM. Establishing in vitro in vivo correlations to screen monoclonal antibodies for physicochemical properties related to favorable human pharmacokinetics. MAbs 2018; 10:244-255. [PMID: 29271699 DOI: 10.1080/19420862.2017.1417718] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Implementation of in vitro assays that correlate with in vivo human pharmacokinetics (PK) would provide desirable preclinical tools for the early selection of therapeutic monoclonal antibody (mAb) candidates with minimal non-target-related PK risk. Use of these tools minimizes the likelihood that mAbs with unfavorable PK would be advanced into costly preclinical and clinical development. In total, 42 mAbs varying in isotype and soluble versus membrane targets were tested in in vitro and in vivo studies. MAb physicochemical properties were assessed by measuring non-specific interactions (DNA- and insulin-binding ELISA), self-association (affinity-capture self-interaction nanoparticle spectroscopy) and binding to matrix-immobilized human FcRn (surface plasmon resonance and column chromatography). The range of scores obtained from each in vitro assay trended well with in vivo clearance (CL) using both human FcRn transgenic (Tg32) mouse allometrically projected human CL and observed human CL, where mAbs with high in vitro scores resulted in rapid CL in vivo. Establishing a threshold value for mAb CL in human of 0.32 mL/hr/kg enabled refinement of thresholds for each in vitro assay parameter, and using a combinatorial triage approach enabled the successful differentiation of mAbs at high risk for rapid CL (unfavorable PK) from those with low risk (favorable PK), which allowed mAbs requiring further characterization to be identified. Correlating in vitro parameters with in vivo human CL resulted in a set of in vitro tools for use in early testing that would enable selection of mAbs with the greatest likelihood of success in the clinic, allowing costly late-stage failures related to an inadequate exposure profile, toxicity or lack of efficacy to be avoided.
Collapse
Affiliation(s)
| | - Jason Wade
- b BioMedicine Design, Pfizer Inc. , Cambridge , MA , USA
| | - Mengmeng Wang
- a BioMedicine Design, Pfizer Inc. , Andover , MA , USA
| | - Amy Tam
- b BioMedicine Design, Pfizer Inc. , Cambridge , MA , USA
| | - Amy King
- b BioMedicine Design, Pfizer Inc. , Cambridge , MA , USA
| | | | | | - Steve Penn
- a BioMedicine Design, Pfizer Inc. , Andover , MA , USA.,c Medicine Design, Pfizer Inc. , Cambridge , MA , USA
| | - David Cirelli
- d Pharmaceutical Sciences, Pfizer Inc. , Andover , MA , USA
| | | | - Minlei Zhang
- a BioMedicine Design, Pfizer Inc. , Andover , MA , USA
| | | | - Rhys Jones
- b BioMedicine Design, Pfizer Inc. , Cambridge , MA , USA.,f Currently Medicine Design, Pfizer Inc. , La Jolla , CA , USA
| | | | | | - Paul Sakorafas
- d Pharmaceutical Sciences, Pfizer Inc. , Andover , MA , USA
| | - James Apgar
- b BioMedicine Design, Pfizer Inc. , Cambridge , MA , USA
| | - William J Finlay
- e Biomedicine Design, Pfizer Inc. , Dublin , Ireland.,g Currently CodeBase , Edinburgh , UK
| | - Laura Lin
- b BioMedicine Design, Pfizer Inc. , Cambridge , MA , USA
| | - Laird Bloom
- b BioMedicine Design, Pfizer Inc. , Cambridge , MA , USA
| | | |
Collapse
|
97
|
Abstract
As of May 1, 2017, 74 antibody-based molecules have been approved by a regulatory authority in a major market. Additionally, there are 70 and 575 antibody-based molecules in phase III and phase I/II clinical trials, respectively. These total 719 antibody-based clinical stage molecules include 493 naked IgGs, 87 antibody-drug conjugates, 61 bispecific antibodies, 37 total Fc fusion proteins, 17 radioimmunoglobulins, 13 antibody fragments, and 11 immunocytokines. New uses for these antibodies are being discovered each year. For oncology, many of the exciting new approaches involve antibody modulation of T-cells. There are over 80 antibodies in clinical trials targeting T cell checkpoints, 26 T-cell-redirected bispecific antibodies, and 145 chimeric antigen receptor (CAR) cell-based candidates (all currently in phase I or II clinical trials), totaling more than 250 T cell interacting clinical stage antibody-based candidates. Finally, significant progress has been made recently on routes of delivery, including delivery of proteins across the blood-brain barrier, oral delivery to the gut, delivery to the cellular cytosol, and gene- and viral-based delivery of antibodies. Thus, there are currently at least 864 antibody-based clinical stage molecules or cells, with incredible diversity in how they are constructed and what activities they impart. These are followed by a next wave of novel molecules, approaches, and new methods and routes of delivery, demonstrating that the field of antibody-based biologics is very innovative and diverse in its approaches to fulfill their promise to treat unmet medical needs.
Collapse
|
98
|
Carter PJ, Lazar GA. Next generation antibody drugs: pursuit of the 'high-hanging fruit'. Nat Rev Drug Discov 2017; 17:197-223. [DOI: 10.1038/nrd.2017.227] [Citation(s) in RCA: 447] [Impact Index Per Article: 55.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
99
|
Kelly RL, Le D, Zhao J, Wittrup KD. Reduction of Nonspecificity Motifs in Synthetic Antibody Libraries. J Mol Biol 2017; 430:119-130. [PMID: 29183788 DOI: 10.1016/j.jmb.2017.11.008] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 11/12/2017] [Accepted: 11/16/2017] [Indexed: 12/26/2022]
Abstract
Successful antibody development requires both functional binding and desirable biophysical characteristics. In the current study, we analyze the causes of one hurdle to clinical development, off-target reactivity, or nonspecificity. We used a high-throughput nonspecificity assay to isolate panels of nonspecific antibodies from two synthetic single-chain variable fragment libraries expressed on the surface of yeast, identifying both individual amino acids and motifs within the complementarity-determining regions which contribute to the phenotype. We find enrichment of glycine, valine, and arginine as both individual amino acids and as a part of motifs, and additionally enrichment of motifs containing tryptophan. Insertion of any of these motifs into the complementarity-determining region H3 of a "clean" antibody increased its nonspecificity, with greatest increases in antibodies containing Trp or Val motifs. We next applied these rules to the creation of a synthetic diversity library based on natural frameworks with significantly decreased incorporation of such motifs and demonstrated its ability to isolate binders to a wide panel of antigens. This work both provides a greater understanding of the drivers of nonspecificity and provides design rules to increase efficiency in the isolation of antibodies with drug-like properties.
Collapse
Affiliation(s)
- Ryan L Kelly
- Department of Biological, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, 02142, MA, USA
| | - Doris Le
- Department of Chemical Engineering, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, 02142, MA, USA
| | - Jessie Zhao
- Department of Chemical Engineering, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, 02142, MA, USA
| | - K Dane Wittrup
- Department of Biological, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, 02142, MA, USA; Department of Chemical Engineering, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, 02142, MA, USA.
| |
Collapse
|
100
|
Datta-Mannan A, Croy JE, Schirtzinger L, Torgerson S, Breyer M, Wroblewski VJ. Aberrant bispecific antibody pharmacokinetics linked to liver sinusoidal endothelium clearance mechanism in cynomolgus monkeys. MAbs 2017; 8:969-82. [PMID: 27111637 DOI: 10.1080/19420862.2016.1178435] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Bispecific antibodies (BsAbs) can affect multiple disease pathways, thus these types of constructs potentially provide promising approaches to improve efficacy in complex disease indications. The specific and non-specific clearance mechanisms/biology that affect monoclonal antibody (mAb) pharmacokinetics are likely involved in the disposition of BsAbs. Despite these similarities, there are a paucity of studies on the in vivo biology that influences the biodistribution and pharmacokinetics of BsAbs. The present case study evaluated the in vivo disposition of 2 IgG-fusion BsAb formats deemed IgG-ECD (extracellular domain) and IgG-scFv (single-chain Fv) in cynomolgus monkeys. These BsAb molecules displayed inferior in vivo pharmacokinetic properties, including a rapid clearance (> 0.5 mL/hr/kg) and short half-life relative to their mAb counterparts. The current work evaluated factors in vivo that result in the aberrant clearance of these BsAb constructs. Results showed the rapid clearance of the BsAbs that was not attributable to target binding, reduced neonatal Fc receptor (FcRn) interactions or poor molecular/biochemical properties. Evaluation of the cellular distribution of the constructs suggested that the major clearance mechanism was linked to binding/association with liver sinusoidal endothelial cells (LSECs) versus liver macrophages. The role of LSECs in facilitating the clearance of the IgG-ECD and IgG-scFv BsAb constructs described in these studies was consistent with the minimal influence of clodronate-mediated macrophage depletion on the pharmacokinetics of the constructs in cynomolgus monkeys The findings in this report are an important demonstration that the elucidation of clearance mechanisms for some IgG-ECD and IgG-scFv BsAb molecules can be unique and complicated, and may require increased attention due to the proliferation of these more complex mAb-like structures.
Collapse
Affiliation(s)
- Amita Datta-Mannan
- a Department of Drug Disposition, Development/ Commercialization , Lilly Research Laboratories, Lilly Corporate Center , Indianapolis , Indiana , USA
| | - Johnny E Croy
- b Department of Biotechnology Discovery Research , Lilly Research Laboratories, Lilly Corporate Center , Indianapolis , Indiana , USA
| | - Linda Schirtzinger
- a Department of Drug Disposition, Development/ Commercialization , Lilly Research Laboratories, Lilly Corporate Center , Indianapolis , Indiana , USA
| | - Stacy Torgerson
- a Department of Drug Disposition, Development/ Commercialization , Lilly Research Laboratories, Lilly Corporate Center , Indianapolis , Indiana , USA
| | - Matthew Breyer
- b Department of Biotechnology Discovery Research , Lilly Research Laboratories, Lilly Corporate Center , Indianapolis , Indiana , USA
| | - Victor J Wroblewski
- a Department of Drug Disposition, Development/ Commercialization , Lilly Research Laboratories, Lilly Corporate Center , Indianapolis , Indiana , USA
| |
Collapse
|