1
|
Gjølberg TT, Mester S, Calamera G, Telstad JS, Sandlie I, Andersen JT. Targeting the Neonatal Fc Receptor in Autoimmune Diseases: Pipeline and Progress. BioDrugs 2025; 39:373-409. [PMID: 40156757 PMCID: PMC12031853 DOI: 10.1007/s40259-025-00708-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/31/2025] [Indexed: 04/01/2025]
Abstract
Autoimmune diseases are highly prevalent and affect people at all ages, women more often than men. The most prominent immunological manifestation is the production of antibodies directed against self-antigens. In many cases, these antibodies (Abs) drive the pathogenesis by attacking the body's own healthy cells, causing serious health problems that may be life threatening. Most autoantibodies are of the immunoglobulin G (IgG) isotype, which has a long plasma half-life and potent effector functions. Thus, there is a need for specific treatment options that rapidly eliminate these pathogenic IgG auto-Abs. In this review, we discuss how the neonatal Fc receptor (FcRn) acts as a regulator of the high levels of not only IgG Abs, but also albumin, by rescuing both these soluble proteins from cellular catabolism, and how a molecular and cellular understanding of this complex biology has spurred an intense interest in the development of FcRn-targeting strategies for the treatment of IgG-driven autoimmune diseases. We find that this emerging therapeutic class demonstrates efficacy within several autoimmune diseases with distinct pathophysiology. This offers hope for both new therapeutic avenues for highly prevalent diseases currently treated by other means, and rare diseases with no approved therapies to date. In addition, we elaborate on studies that have led to approval of the first FcRn antagonists, the clinical progress and structural design of molecules in the pipeline, their position in the overall therapeutic landscape of autoimmunity, the design of next-generation antagonists as well as the use of this receptor-targeting principle for other therapeutic applications.
Collapse
Affiliation(s)
- Torleif Tollefsrud Gjølberg
- Authera AS, 0349, Oslo, Norway.
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, 0372, Oslo, Norway.
- Department of Immunology, Oslo University Hospital and University of Oslo, 0372, Oslo, Norway.
- Precision Immunotherapy Alliance (PRIMA), University of Oslo, Oslo, Norway.
| | - Simone Mester
- Authera AS, 0349, Oslo, Norway
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, 0372, Oslo, Norway
- Department of Immunology, Oslo University Hospital and University of Oslo, 0372, Oslo, Norway
- Precision Immunotherapy Alliance (PRIMA), University of Oslo, Oslo, Norway
| | | | | | - Inger Sandlie
- Department of Biosciences, University of Oslo, 0316, Oslo, Norway
| | - Jan Terje Andersen
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, 0372, Oslo, Norway.
- Department of Immunology, Oslo University Hospital and University of Oslo, 0372, Oslo, Norway.
- Precision Immunotherapy Alliance (PRIMA), University of Oslo, Oslo, Norway.
| |
Collapse
|
2
|
Li N, Gong N, Duan B, Zhang Y, Jian Y, Xu Y, Liu J, Wang X, Zhang X, Du M, Zhou F, Zhao J, Guan X, Peng X, Wang S, Zhang H, Li X. Reduction of circulating IgE and allergens by a pH-sensitive antibody with enhanced FcγRIIb binding. Mol Ther 2024; 32:3729-3742. [PMID: 39228125 PMCID: PMC11489548 DOI: 10.1016/j.ymthe.2024.08.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 06/29/2024] [Accepted: 08/30/2024] [Indexed: 09/05/2024] Open
Abstract
Allergen-crosslinked IgE triggers allergy by interacting with its receptor on basophils and mast cells. The anti-IgE monoclonal antibody omalizumab can alleviate allergy by competing with the receptor for IgE binding. However, along with neutralization, omalizumab also inhibits IgE degradation, which is clinically associated with high-dose and total IgE accumulation problems. In this study, we have developed an IgE-eliminating antibody on the basis of omalizumab, which has pH-dependent Fabs and an Fc with high affinity for FcγRIIb. In mice, the antibody rapidly eliminated total serum IgE to baseline levels and caused lower free IgE levels than omalizumab. At low dosages, the antibody also exhibited favorable IgE elimination effects. In addition, the antibody can degrade the corresponding allergen with the removal of IgE, addressing the allergy from its source. Introduction of the M252Y/S254T/T256E (YTE) mutation into this antibody prolongs its serum half-life without reducing potency. Thus, this engineered antibody holds a promising therapeutic option for allergy patients. Mechanistic insights are also included in this study.
Collapse
Affiliation(s)
- Na Li
- College of Life Sciences and State Key Laboratory of Medicinal Chemical Biology, Nankai University, 94 Weijin Road, Tianjin 300071, P.R. China
| | - Nanxin Gong
- College of Life Sciences and State Key Laboratory of Medicinal Chemical Biology, Nankai University, 94 Weijin Road, Tianjin 300071, P.R. China; College of Life Science, Institute of Life Science and Green Development, Hebei University, Baoding 071002, P.R. China
| | - Baoxin Duan
- College of Life Sciences and State Key Laboratory of Medicinal Chemical Biology, Nankai University, 94 Weijin Road, Tianjin 300071, P.R. China
| | - Yongyan Zhang
- College of Life Sciences and State Key Laboratory of Medicinal Chemical Biology, Nankai University, 94 Weijin Road, Tianjin 300071, P.R. China
| | - Yi Jian
- College of Life Sciences and State Key Laboratory of Medicinal Chemical Biology, Nankai University, 94 Weijin Road, Tianjin 300071, P.R. China
| | - Yanqin Xu
- College of Life Sciences and State Key Laboratory of Medicinal Chemical Biology, Nankai University, 94 Weijin Road, Tianjin 300071, P.R. China
| | - Jinming Liu
- College of Life Sciences and State Key Laboratory of Medicinal Chemical Biology, Nankai University, 94 Weijin Road, Tianjin 300071, P.R. China
| | - Xiaoqian Wang
- College of Life Sciences and State Key Laboratory of Medicinal Chemical Biology, Nankai University, 94 Weijin Road, Tianjin 300071, P.R. China
| | - Xiaoqi Zhang
- College of Life Sciences and State Key Laboratory of Medicinal Chemical Biology, Nankai University, 94 Weijin Road, Tianjin 300071, P.R. China
| | - Mingjuan Du
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, P.R. China
| | - Feilong Zhou
- College of Life Sciences and State Key Laboratory of Medicinal Chemical Biology, Nankai University, 94 Weijin Road, Tianjin 300071, P.R. China
| | - Jiliang Zhao
- College of Life Sciences and State Key Laboratory of Medicinal Chemical Biology, Nankai University, 94 Weijin Road, Tianjin 300071, P.R. China
| | - Xiangchen Guan
- College of Life Sciences and State Key Laboratory of Medicinal Chemical Biology, Nankai University, 94 Weijin Road, Tianjin 300071, P.R. China
| | - Xiangda Peng
- Shanghai Zelixir Biotech, Shanghai 200030, P.R. China
| | - Sheng Wang
- Shanghai Zelixir Biotech, Shanghai 200030, P.R. China
| | - Hongkai Zhang
- College of Life Sciences and State Key Laboratory of Medicinal Chemical Biology, Nankai University, 94 Weijin Road, Tianjin 300071, P.R. China; Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, P.R. China.
| | - Xin Li
- College of Life Sciences and State Key Laboratory of Medicinal Chemical Biology, Nankai University, 94 Weijin Road, Tianjin 300071, P.R. China.
| |
Collapse
|
3
|
Tulika T, Ruso-Julve F, Ahmadi S, Ljungars A, Rivera-de-Torre E, Wade J, Fernández-Quintero ML, Jenkins TP, Belfakir SB, Ross GMS, Boyens-Thiele L, Buell AK, Sakya SA, Sørensen CV, Bohn MF, Ledsgaard L, Voldborg BG, Francavilla C, Schlothauer T, Lomonte B, Andersen JT, Laustsen AH. Engineering of pH-dependent antigen binding properties for toxin-targeting IgG1 antibodies using light-chain shuffling. Structure 2024; 32:1404-1418.e7. [PMID: 39146931 PMCID: PMC11385703 DOI: 10.1016/j.str.2024.07.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 06/07/2024] [Accepted: 07/19/2024] [Indexed: 08/17/2024]
Abstract
Immunoglobulin G (IgG) antibodies that bind their cognate antigen in a pH-dependent manner (acid-switched antibodies) can release their bound antigen for degradation in the acidic environment of endosomes, while the IgGs are rescued by the neonatal Fc receptor (FcRn). Thus, such IgGs can neutralize multiple antigens over time and therefore be used at lower doses than their non-pH-responsive counterparts. Here, we show that light-chain shuffling combined with phage display technology can be used to discover IgG1 antibodies with increased pH-dependent antigen binding properties, using the snake venom toxins, myotoxin II and α-cobratoxin, as examples. We reveal differences in how the selected IgG1s engage their antigens and human FcRn and show how these differences translate into distinct cellular handling properties related to their pH-dependent antigen binding phenotypes and Fc-engineering for improved FcRn binding. Our study showcases the complexity of engineering pH-dependent antigen binding IgG1s and demonstrates the effects on cellular antibody-antigen recycling.
Collapse
Affiliation(s)
- Tulika Tulika
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Fulgencio Ruso-Julve
- Department of Pharmacology, University of Oslo, Oslo, Norway; Department of Immunology, Oslo University Hospital Rikshospitalet, Oslo, Norway; Precision Immunotherapy Alliance (PRIMA), University of Oslo, Oslo, Norway
| | - Shirin Ahmadi
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Anne Ljungars
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | | | - Jack Wade
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | | | - Timothy P Jenkins
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Selma B Belfakir
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark; VenomAid Diagnostics ApS, Lyngby, Denmark
| | | | - Lars Boyens-Thiele
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Alexander K Buell
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Siri A Sakya
- Department of Pharmacology, University of Oslo, Oslo, Norway; Department of Immunology, Oslo University Hospital Rikshospitalet, Oslo, Norway; Precision Immunotherapy Alliance (PRIMA), University of Oslo, Oslo, Norway
| | - Christoffer V Sørensen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Markus-Frederik Bohn
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Line Ledsgaard
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Bjørn G Voldborg
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Chiara Francavilla
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Tilman Schlothauer
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, Penzberg, Germany
| | - Bruno Lomonte
- Instituto Clodomiro Picado, Facultad de Microbiologia, Universidad de Costa Rica, San Jose, Costa Rica
| | - Jan Terje Andersen
- Department of Pharmacology, University of Oslo, Oslo, Norway; Department of Immunology, Oslo University Hospital Rikshospitalet, Oslo, Norway; Precision Immunotherapy Alliance (PRIMA), University of Oslo, Oslo, Norway.
| | - Andreas H Laustsen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark.
| |
Collapse
|
4
|
Vaughan HJ, Est-Witte S, Dockery LT, Urello MA, Boyd J, Keyser BD, Zhuang L, Marelli M, Christie RJ. A high-throughput lysosome trafficking assay guides ligand selection and elucidates differences in CD22-targeted nanodelivery. SCIENCE AND TECHNOLOGY OF ADVANCED MATERIALS 2024; 25:2351791. [PMID: 38817250 PMCID: PMC11138227 DOI: 10.1080/14686996.2024.2351791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 04/30/2024] [Indexed: 06/01/2024]
Abstract
Targeted nanoparticles offer potential to selectively deliver therapeutics to cells; however, their subcellular fate following endocytosis must be understood to properly design mechanisms of drug release. Here we describe a nanoparticle platform and associated cell-based assay to observe lysosome trafficking of targeted nanoparticles in live cells. The nanoparticle platform utilizes two fluorescent dyes loaded onto PEG-poly(glutamic acid) and PEG-poly(Lysine) block co-polymers that also comprise azide reactive handles on PEG termini to attach antibody-based targeting ligands. Fluorophores were selected to be pH-sensitive (pHrodo Red) or pH-insensitive (Alexafluor 488) to report when nanoparticles enter low pH lysosomes. Dye-labelled block co-polymers were further assembled into polyion complex micelle nanoparticles and crosslinked through amide bond formation to form stable nano-scaffolds for ligand attachment. Cell binding and lysosome trafficking was determined in live cells by fluorescence imaging in 96-well plates and quantification of red- and green-fluorescence signals over time. The platform and assay was validated for selection of optimal antibody-derived targeting ligands directed towards CD22 for nanoparticle delivery. Kinetic analysis of uptake and lysosome trafficking indicated differences between ligand types and the ligand with the highest lysosome trafficking efficiency translated into effective DNA delivery with nanoparticles bearing the optimal ligand.
Collapse
Affiliation(s)
- Hannah J. Vaughan
- Biologics Engineering, Oncology R&D, AstraZeneca, Gaithersburg, MD, USA
| | | | - Lance T. Dockery
- Biologics Engineering, Oncology R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Morgan A. Urello
- Biologics Engineering, Oncology R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Jonathan Boyd
- Discovery Sciences, BioPharma R&D, AstraZeneca, Gaithersburg, MD, USA
| | | | - Li Zhuang
- Biologics Engineering, Oncology R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Marcello Marelli
- Biologics Engineering, Oncology R&D, AstraZeneca, Gaithersburg, MD, USA
| | - R. James Christie
- Biologics Engineering, Oncology R&D, AstraZeneca, Gaithersburg, MD, USA
| |
Collapse
|
5
|
Daurat M, Gauthier C, El Cheikh K, Ali LMA, Morère E, Bettache N, Gary-Bobo M, Morère A, Garcia M, Maynadier M, Basile I. Engineered therapeutic antibodies with mannose 6-phosphate analogues as a tool to degrade extracellular proteins. Front Immunol 2024; 15:1273280. [PMID: 38533506 PMCID: PMC10964947 DOI: 10.3389/fimmu.2024.1273280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Accepted: 02/14/2024] [Indexed: 03/28/2024] Open
Abstract
Inducing the degradation of pathological soluble antigens could be the key to greatly enhancing the efficacy of therapeutic monoclonal antibodies (mAbs), extensively used in the treatment of autoimmune and inflammatory disorders or cancer. Lysosomal targeting has gained increasing interest in recent years due to its pharmaceutical applications far beyond the treatment of lysosomal diseases, as a way to address proteins to the lysosome for eventual degradation. Mannose 6-phosphonate derivatives (M6Pn), called AMFA, are unique glycovectors that can significantly enhance the cellular internalization of the proteins conjugated to AMFA via the cation-independent mannose 6-phosphate receptor (M6PR) pathway. AMFA engineering of mAbs results in the generation of a bifunctional antibody that is designed to bind both the antigen and the M6PR. The improvement of the therapeutic potential by AMFA engineering was investigated using two antibodies directed against soluble antigens: infliximab (IFX), directed against tumor necrosis factor α (TNF-α), and bevacizumab (BVZ), directed against the vascular endothelial growth factor (VEGF). AMFA conjugations to the antibodies were performed either on the oligosaccharidic chains of the antibodies or on the lysine residues. Both conjugations were controlled and reproducible and provided a novel affinity for the M6PR without altering the affinity for the antigen. The grafting of AMFA to mAb increased their cellular uptake through an M6PR-dependent mechanism. The antigens were also 2.6 to 5.7 times more internalized by mAb-AMFA and rapidly degraded in the cells. Additional cell culture studies also proved the significantly higher efficacy of IFX-AMFA and BVZ-AMFA compared to their unconjugated counterparts in inhibiting TNF-α and VEGF activities. Finally, studies in a zebrafish embryo model of angiogenesis and in xenografted chick embryos showed that BVZ-AMFA was more effective than BVZ in reducing angiogenesis. These results demonstrate that AMFA grafting induces the degradation of soluble antigens and a significant increase in the therapeutic efficacy. Engineering with mannose 6-phosphate analogues has the potential to develop a new class of antibodies for autoimmune and inflammatory diseases.
Collapse
Affiliation(s)
| | - Corentin Gauthier
- NanoMedSyn, Montpellier, France
- Institut des Biomolécules Max Mousseron (IBMM), University of Montpellier, CNRS, ENSCM, Montpellier, France
| | | | - Lamiaa M. A. Ali
- Institut des Biomolécules Max Mousseron (IBMM), University of Montpellier, CNRS, ENSCM, Montpellier, France
- Department of Biochemistry Medical Research Institute, University of Alexandria, Alexandria, Egypt
| | - Elodie Morère
- NanoMedSyn, Montpellier, France
- Institut des Biomolécules Max Mousseron (IBMM), University of Montpellier, CNRS, ENSCM, Montpellier, France
| | - Nadir Bettache
- Institut des Biomolécules Max Mousseron (IBMM), University of Montpellier, CNRS, ENSCM, Montpellier, France
| | - Magali Gary-Bobo
- Institut des Biomolécules Max Mousseron (IBMM), University of Montpellier, CNRS, ENSCM, Montpellier, France
| | - Alain Morère
- Institut des Biomolécules Max Mousseron (IBMM), University of Montpellier, CNRS, ENSCM, Montpellier, France
| | | | | | | |
Collapse
|
6
|
Wells JA, Kumru K. Extracellular targeted protein degradation: an emerging modality for drug discovery. Nat Rev Drug Discov 2024; 23:126-140. [PMID: 38062152 DOI: 10.1038/s41573-023-00833-z] [Citation(s) in RCA: 60] [Impact Index Per Article: 60.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/23/2023] [Indexed: 02/08/2024]
Abstract
Targeted protein degradation (TPD) has emerged in the past decade as a major new drug modality to remove intracellular proteins with bispecific small molecules that recruit the protein of interest (POI) to an E3 ligase for degradation in the proteasome. Unlike classic occupancy-based drugs, intracellular TPD (iTPD) eliminates the target and works catalytically, and so can be more effective and sustained, with lower dose requirements. Recently, this approach has been expanded to the extracellular proteome, including both secreted and membrane proteins. Extracellular targeted protein degradation (eTPD) uses bispecific antibodies, conjugates or small molecules to degrade extracellular POIs by trafficking them to the lysosome for degradation. Here, we focus on recent advances in eTPD, covering degrader systems, targets, molecular designs and parameters to advance them. Now almost any protein, intracellular or extracellular, is addressable in principle with TPD.
Collapse
Affiliation(s)
- James A Wells
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA.
- Department of Cellular & Molecular Pharmacology, University of California San Francisco, San Francisco, CA, USA.
| | - Kaan Kumru
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
7
|
Wei W, Sulea T. Sequence-based engineering of pH-sensitive antibodies for tumor targeting or endosomal recycling applications. MAbs 2024; 16:2404064. [PMID: 39289783 PMCID: PMC11409498 DOI: 10.1080/19420862.2024.2404064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 08/20/2024] [Accepted: 09/10/2024] [Indexed: 09/19/2024] Open
Abstract
The engineering of pH-sensitive therapeutic antibodies, particularly for improving effectiveness and specificity in acidic solid-tumor microenvironments, has recently gained traction. While there is a justified need for pH-dependent immunotherapies, current engineering techniques are tedious and laborious, requiring repeated rounds of experiments under different pH conditions. Inexpensive computational techniques to predict the effectiveness of His pH-switches require antibody-antigen complex structures, but these are lacking in most cases. To circumvent these requirements, we introduce a sequence-based in silico method for predicting His mutations in the variable region of antibodies, which could lead to pH-biased antigen binding. This method, called Sequence-based Identification of pH-sensitive Antibody Binding (SIpHAB), was trained on 3D-structure-based calculations of 3,490 antibody-antigen complexes with solved experimental structures. SIpHAB was parametrized to enhance preferential binding either toward or against the acidic pH, for selective targeting of solid tumors or for antigen release in the endosome, respectively. Applications to nine antibody-antigen systems with previously reported binding preferences at different pHs demonstrated the utility and enrichment capabilities of this high-throughput computational tool. SIpHAB, which only requires knowledge of the antibody primary amino-acid sequence, could enable a more efficient triage of pH-sensitive antibody candidates than could be achieved conventionally. An online webserver for running SipHAB is available freely at https://mm.nrc-cnrc.gc.ca/software/siphab/runner/.
Collapse
Affiliation(s)
- Wanlei Wei
- Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, Quebec, Canada
| | - Traian Sulea
- Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, Quebec, Canada
| |
Collapse
|
8
|
Devanaboyina SC, Li P, LaGory EL, Poon-Andersen C, Cook KD, Soto M, Wang Z, Dang K, Uyeda C, Case RB, Thomas VA, Primack R, Ponce M, Di M, Ouyang B, Kaner J, Lam SK, Mostafavi M. Rapid depletion of "catch-and-release" anti-ASGR1 antibody in vivo. MAbs 2024; 16:2383013. [PMID: 39051531 PMCID: PMC11275528 DOI: 10.1080/19420862.2024.2383013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 07/16/2024] [Accepted: 07/17/2024] [Indexed: 07/27/2024] Open
Abstract
Targeting antigens with antibodies exhibiting pH/Ca2+-dependent binding against an antigen is an attractive strategy to mitigate target-mediated disposition and antigen buffering. Studies have reported improved serum exposure of antibodies exhibiting pH/Ca2+-binding against membrane-bound receptors. Asialoglycoprotein receptor 1 (ASGR1) is a membrane-bound receptor primarily localized in hepatocytes. With a high expression level of approximately one million receptors per cell, high turnover, and rapid recycling, targeting this receptor with a conventional antibody is a challenge. In this study, we identified an antibody exhibiting pH/Ca2+-dependent binding to ASGR1 and generated antibody variants with increased binding to neonatal crystallizable fragment receptor (FcRn). Serum exposures of the generated anti-ASGR1 antibodies were analyzed in transgenic mice expressing human FcRn. Contrary to published reports of increased serum exposure of pH/Ca2+-dependent antibodies, the pH/Ca2+-dependent anti-ASGR1 antibody had rapid serum clearance in comparison to a conventional anti-ASGR1 antibody. We conducted sub-cellular trafficking studies of the anti-ASGR1 antibodies along with receptor quantification analysis for mechanistic understanding of the rapid serum clearance of pH/Ca2+-dependent anti-ASGR1 antibody. The findings from our study provide valuable insights in identifying the antigens, especially membrane bound, that may benefit from targeting with pH/Ca2+-dependent antibodies to obtain increased serum exposure.
Collapse
Affiliation(s)
- Siva Charan Devanaboyina
- Department of Pharmacokinetics and Drug Metabolism, Amgen Research, Amgen Inc, South San Francisco, CA, USA
| | - Peng Li
- Department of Biologics, Amgen Research, Amgen Inc, South San Francisco, CA, USA
| | - Edward L. LaGory
- Department of Pharmacokinetics and Drug Metabolism, Amgen Research, Amgen Inc, South San Francisco, CA, USA
| | - Carrie Poon-Andersen
- Department of Pharmacokinetics and Drug Metabolism, Amgen Research, Amgen Inc, South San Francisco, CA, USA
| | - Kevin D. Cook
- Department of Pharmacokinetics and Drug Metabolism, Amgen Research, Amgen Inc, South San Francisco, CA, USA
| | - Marcus Soto
- Department of Pharmacokinetics and Drug Metabolism, Amgen Research, Amgen Inc, Thousand Oaks, CA, USA
| | - Zhe Wang
- Department of Pharmacokinetics and Drug Metabolism, Amgen Research, Amgen Inc, South San Francisco, CA, USA
| | - Khue Dang
- Department of Biologics, Amgen Research, Amgen Inc, South San Francisco, CA, USA
| | - Craig Uyeda
- Department of Pharmacokinetics and Drug Metabolism, Amgen Research, Amgen Inc, South San Francisco, CA, USA
| | - Ryan B. Case
- Department of Lead Discovery and Characterization, Amgen Research, Amgen Inc, South San Francisco, CA, USA
| | - Veena A. Thomas
- Department of Pharmacokinetics and Drug Metabolism, Amgen Research, Amgen Inc, South San Francisco, CA, USA
| | - Ronya Primack
- Department of Pharmacokinetics and Drug Metabolism, Amgen Research, Amgen Inc, Thousand Oaks, CA, USA
| | - Manuel Ponce
- Department of Pharmacokinetics and Drug Metabolism, Amgen Research, Amgen Inc, Thousand Oaks, CA, USA
| | - Mei Di
- Department of Cardiometabolic disorders, Amgen Research, Amgen Inc, South San Francisco, CA, USA
| | - Brian Ouyang
- Department of Biologics, Amgen Research, Amgen Inc, South San Francisco, CA, USA
| | - Joelle Kaner
- Department of Biologics, Amgen Research, Amgen Inc, South San Francisco, CA, USA
| | - Sheung Kwan Lam
- Department of Biologics, Amgen Research, Amgen Inc, South San Francisco, CA, USA
| | - Mina Mostafavi
- Department of Biologics, Amgen Research, Amgen Inc, South San Francisco, CA, USA
| |
Collapse
|
9
|
Tulika T, Pedersen RW, Rimbault C, Ahmadi S, Rivera‐de‐Torre E, Fernández‐Quintero ML, Loeffler JR, Bohn M, Ljungars A, Ledsgaard L, Voldborg BG, Ruso‐Julve F, Andersen JT, Laustsen AH. Phage display assisted discovery of a pH-dependent anti-α-cobratoxin antibody from a natural variable domain library. Protein Sci 2023; 32:e4821. [PMID: 37897425 PMCID: PMC10659949 DOI: 10.1002/pro.4821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 09/28/2023] [Accepted: 10/24/2023] [Indexed: 10/30/2023]
Abstract
Recycling IgG antibodies bind to their target antigen at physiological pH in the blood stream and release them upon endocytosis when pH levels drop, allowing the IgG antibodies to be recycled into circulation via FcRn-mediated cellular pathways, while the antigens undergo lysosomal degradation. This enables recycling antibodies to achieve comparable therapeutic effect at lower doses than their non-recycling counterparts. The development of such antibodies is typically achieved by histidine doping of their variable regions or by performing in vitro antibody selection campaigns utilizing histidine doped libraries. Both are strategies that may introduce sequence liabilities. Here, we present a methodology that employs a naïve antibody phage display library, consisting of natural variable domains, to discover antibodies that bind α-cobratoxin from the venom of Naja kaouthia in a pH-dependent manner. As a result, an antibody was discovered that exhibits a 7-fold higher off-rate at pH 5.5 than pH 7.4 in bio-layer interferometry experiments. Interestingly, no histidine residues were found in its variable domains, and in addition, the antibody showed pH-dependent binding to a histidine-devoid antigen mutant. As such, the results demonstrate that pH-dependent antigen-antibody binding may not always be driven by histidine residues. By employing molecular dynamics simulations, different protonation states of titratable residues were found, which potentially could be responsible for the observed pH-dependent antigen binding properties of the antibody. Finally, given the typically high diversity of naïve antibody libraries, the methodology presented here can likely be applied to discover recycling antibodies against different targets ab initio without the need for histidine doping.
Collapse
Affiliation(s)
- Tulika Tulika
- Department of Biotechnology and BiomedicineTechnical University of DenmarkLyngbyDenmark
| | - Rasmus W. Pedersen
- Department of Biotechnology and BiomedicineTechnical University of DenmarkLyngbyDenmark
| | - Charlotte Rimbault
- Department of Biotechnology and BiomedicineTechnical University of DenmarkLyngbyDenmark
| | - Shirin Ahmadi
- Department of Biotechnology and BiomedicineTechnical University of DenmarkLyngbyDenmark
| | | | - Monica L. Fernández‐Quintero
- Center for Molecular Biosciences Innsbruck, Department of GeneralInorganic and Theoretical Chemistry, University of InnsbruckInnsbruckAustria
| | - Johannes R. Loeffler
- Center for Molecular Biosciences Innsbruck, Department of GeneralInorganic and Theoretical Chemistry, University of InnsbruckInnsbruckAustria
| | - Markus‐Frederik Bohn
- Department of Biotechnology and BiomedicineTechnical University of DenmarkLyngbyDenmark
| | - Anne Ljungars
- Department of Biotechnology and BiomedicineTechnical University of DenmarkLyngbyDenmark
| | - Line Ledsgaard
- Department of Biotechnology and BiomedicineTechnical University of DenmarkLyngbyDenmark
| | - Bjørn G. Voldborg
- Department of Biotechnology and BiomedicineTechnical University of DenmarkLyngbyDenmark
| | - Fulgencio Ruso‐Julve
- Department of PharmacologyUniversity of OsloOsloNorway
- Department of ImmunologyOslo University Hospital RikshospitaletOsloNorway
- Precision Immunotherapy AllianceUniversity of OsloOsloNorway
| | - Jan Terje Andersen
- Department of PharmacologyUniversity of OsloOsloNorway
- Department of ImmunologyOslo University Hospital RikshospitaletOsloNorway
- Precision Immunotherapy AllianceUniversity of OsloOsloNorway
| | - Andreas H. Laustsen
- Department of Biotechnology and BiomedicineTechnical University of DenmarkLyngbyDenmark
| |
Collapse
|
10
|
Dervisevic M, Dervisevic E, Esser L, Easton CD, Cadarso VJ, Voelcker NH. Wearable microneedle array-based sensor for transdermal monitoring of pH levels in interstitial fluid. Biosens Bioelectron 2023; 222:114955. [PMID: 36462430 DOI: 10.1016/j.bios.2022.114955] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/19/2022] [Accepted: 11/24/2022] [Indexed: 11/27/2022]
Abstract
Microneedle-based wearable sensors offer an alternative approach to traditional invasive blood-based health monitoring and disease diagnostics techniques. Instead of blood, microneedle-based sensors target the skin interstitial fluid (ISF), in which the biomarker type and concentration profile resemble the one found in the blood. However, unlike blood, interstitial fluid does not have the same pH-buffering capacity causing deviation of pH levels from the physiological range. Information about the skin ISF pH levels can be used as a biomarker for a wide range of pathophysiological conditions and as a marker for the calibration of a wearable sensor. The ISF pH can significantly affect the detection accuracy of other biomarkers as it influences enzyme activity, aptamer affinity, and antibody-antigen interaction. Herein, we report the fabrication of a high-density polymeric microneedle array-based (PMNA) sensing patch and its optimization for the potentiometric transdermal monitoring of pH levels in ISF. The wearable sensor utilizes a polyaniline-coated PMNA having a density of ∼10,000 microneedles per cm2, containing individual microneedles with a height of ∼250 μm, and a tip diameter of ∼2 μm. To prevent interference from other body fluids like sweat, an insulating layer is deposited at the base of the PMNA. The wearable pH sensor operates from pH 4.0 to 8.6 with a sensitivity of 62.9 mV per pH unit and an accuracy of ±0.036 pH units. Furthermore, testing on a mouse demonstrates the ability of the PMNA to provide a real-time reading of the transdermal pH values. This microneedle-based system will significantly contribute to advancing transdermal wearable sensors technology, simplifying the fabrication process, and improving the cost-effectiveness of such devices.
Collapse
Affiliation(s)
- Muamer Dervisevic
- Drug Delivery, Disposition, and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia
| | - Esma Dervisevic
- Department of Mechanical and Aerospace Engineering, Monash University, Room 227, New Horizons Building, 20 Research Way, Clayton, Victoria, 3800, Australia
| | - Lars Esser
- Drug Delivery, Disposition, and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia; Commonwealth Scientific and Industrial Research Organisation (CSIRO), Clayton, Victoria, 3168, Australia
| | - Christopher D Easton
- Commonwealth Scientific and Industrial Research Organisation (CSIRO), Clayton, Victoria, 3168, Australia
| | - Victor J Cadarso
- Department of Mechanical and Aerospace Engineering, Monash University, Room 227, New Horizons Building, 20 Research Way, Clayton, Victoria, 3800, Australia; Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Clayton, Victoria, 3168, Australia
| | - Nicolas H Voelcker
- Drug Delivery, Disposition, and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia; Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Clayton, Victoria, 3168, Australia; Materials Science and Engineering, Monash University, Clayton, Victoria, 3168, Australia.
| |
Collapse
|
11
|
Jalalizadeh M, Yadollahvandmiandoab R, Reis LO. Immune Checkpoint Glycoproteins Have Polymorphism: Are Monoclonal Antibodies Too Specific? Curr Oncol 2023; 30:1267-1274. [PMID: 36661747 PMCID: PMC9857673 DOI: 10.3390/curroncol30010098] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 01/11/2023] [Accepted: 01/13/2023] [Indexed: 01/19/2023] Open
Abstract
Since the 2018 Nobel prize in medicine was granted to the discovery of immune escape by cancer cells, billions of dollars have been spent on a new form of cancer immunotherapy called immune checkpoint inhibition (ICI). In this treatment modality, monoclonal antibodies (mAbs) are used to block cell-surface glycoproteins responsible for cancer immune escape. However, only a subset of patients benefit from this treatment. In this commentary, we focus on the polymorphism in the target molecules of these mAbs, namely PD-1, PD-L1 and CTLA4; we explain that using a single mAb from one clone is unlikely to succeed in treating all humans because humans have a genotype and phenotype polymorphism in these molecules. Monoclonal antibodies are highly specific and are capable of recognizing only one epitope ("monospecific"), which makes them ideal for use in laboratory animals because these animals are generationally inbred and genetically identical (isogenic). In humans, however, the encoding genes for PD-1, PD-L1 and CTLA4 have variations (alleles), and the final protein products have phenotype polymorphism. This means that small differences exist in these proteins among individual humans, rendering one mAb too specific to cover all patients. Our suggestion for the next step in advancing this oncotherapy is to focus on methods to tailor the mAb treatment individually for each patient or replace a single clone of mAb with less specific alternatives, e.g., a "cocktail of mAbs", oligoclonal antibodies or recombinant polyclonal antibodies. Fortunately, there are ongoing clinical trials on oligoclonal antibodies at the moment.
Collapse
Affiliation(s)
- Mehrsa Jalalizadeh
- UroScience, School of Medical Sciences, University of Campinas, UNICAMP, Campinas 13083-970, SP, Brazil
| | - Reza Yadollahvandmiandoab
- UroScience, School of Medical Sciences, University of Campinas, UNICAMP, Campinas 13083-970, SP, Brazil
| | - Leonardo Oliveira Reis
- UroScience, School of Medical Sciences, University of Campinas, UNICAMP, Campinas 13083-970, SP, Brazil
- Center for Life Sciences, Pontifical Catholic University of Campinas, PUC-Campinas, Campinas 13034-685, SP, Brazil
| |
Collapse
|
12
|
Yadollahvandmiandoab R, Jalalizadeh M, Buosi K, Garcia-Perdomo HA, Reis LO. Immunogenic Cell Death Role in Urothelial Cancer Therapy. Curr Oncol 2022; 29:6700-6713. [PMID: 36135095 PMCID: PMC9498148 DOI: 10.3390/curroncol29090526] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/07/2022] [Accepted: 09/16/2022] [Indexed: 12/05/2022] Open
Abstract
PURPOSE Bladder cancer is the 13th most common cause of cancer death with the highest lifetime cost for treatment of all cancers. This scoping review clarifies the available evidence on the role of a novel therapeutic approach called immunogenic cell death (ICD) in urothelial cancer of the bladder. METHODS In accordance with the recommendations of the Joanna Briggs Institute, we searched MEDLINE (Ovid), EMBASE, CENTRAL databases, and supplemented with manual searches through the conferences, Google scholar, and clinicaltrials.gov for published studies up to April 2022. We included literature that studied molecular mechanisms of ICD and the role of certain danger-associated molecular patterns (DAMPs) in generating ICD, safety and efficacy of different ICD inducers, and their contributions in combination with other urothelial cancer treatments. RESULTS Oncolytic viruses, radiotherapy, certain chemo/chemo radiation therapy combinations, photodynamic therapy, and novel agents were studied as ICD-inducing treatment modalities in the included studies. ICD was observed in vitro (murine or human urothelial carcinoma) in ten studies, eight studies were performed on mouse models (orthotopic or subcutaneous), and five clinical trials assessed patient response to ICD inducing agents. The most common studied DAMPs were Calreticulin, HMGB1, ATP, and Heat Shock Proteins (HSP) 70 and 90, which were either expressed on the cancer cells or released. CONCLUSION ICD inducers were able to generate lasting antitumor immune responses with memory formation in animal studies (vaccination effect). In clinical trials these agents generally had low side effects, except for one trial, and could be used alone or in combination with other cancer treatment strategies in urothelial cancer patients.
Collapse
Affiliation(s)
- Reza Yadollahvandmiandoab
- UroScience, School of Medical Sciences, University of Campinas, UNICAMP, Campinas, Sao Paulo 13083-970, Brazil
| | - Mehrsa Jalalizadeh
- UroScience, School of Medical Sciences, University of Campinas, UNICAMP, Campinas, Sao Paulo 13083-970, Brazil
| | - Keini Buosi
- UroScience, School of Medical Sciences, University of Campinas, UNICAMP, Campinas, Sao Paulo 13083-970, Brazil
| | - Herney Andrés Garcia-Perdomo
- Division of Urology/Urooncology, Department of Surgery, School of Medicine, Universidad del Valle, Cali 72824, Colombia
| | - Leonardo Oliveira Reis
- UroScience, School of Medical Sciences, University of Campinas, UNICAMP, Campinas, Sao Paulo 13083-970, Brazil
- Center for Life Sciences, Pontifical Catholic University of Campinas, PUC-Campinas, Sao Paulo 13087-571, Brazil
| |
Collapse
|
13
|
Monoclonal Antibody Engineering and Design to Modulate FcRn Activities: A Comprehensive Review. Int J Mol Sci 2022; 23:ijms23179604. [PMID: 36077002 PMCID: PMC9455995 DOI: 10.3390/ijms23179604] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 08/19/2022] [Accepted: 08/22/2022] [Indexed: 01/03/2023] Open
Abstract
Understanding the biological mechanisms underlying the pH-dependent nature of FcRn binding, as well as the various factors influencing the affinity to FcRn, was concurrent with the arrival of the first recombinant IgG monoclonal antibodies (mAbs) and IgG Fc-fusion proteins in clinical practice. IgG Fc–FcRn became a central subject of interest for the development of these drugs for the comfort of patients and good clinical responses. In this review, we describe (i) mAb mutations close to and outside the FcRn binding site, increasing the affinity for FcRn at acidic pH and leading to enhanced mAb half-life and biodistribution, and (ii) mAb mutations increasing the affinity for FcRn at acidic and neutral pH, blocking FcRn binding and resulting, in vivo, in endogenous IgG degradation. Mutations modifying FcRn binding are discussed in association with pH-dependent modulation of antigen binding and (iii) anti-FcRn mAbs, two of the latest innovations in anti-FcRn mAbs leading to endogenous IgG depletion. We discuss the pharmacological effects, the biological consequences, and advantages of targeting IgG–FcRn interactions and their application in human therapeutics.
Collapse
|
14
|
Biophysical differences in IgG1 Fc-based therapeutics relate to their cellular handling, interaction with FcRn and plasma half-life. Commun Biol 2022; 5:832. [PMID: 35982144 PMCID: PMC9388496 DOI: 10.1038/s42003-022-03787-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 08/01/2022] [Indexed: 01/07/2023] Open
Abstract
Antibody-based therapeutics (ABTs) are used to treat a range of diseases. Most ABTs are either full-length IgG1 antibodies or fusions between for instance antigen (Ag)-binding receptor domains and the IgG1 Fc fragment. Interestingly, their plasma half-life varies considerably, which may relate to how they engage the neonatal Fc receptor (FcRn). As such, there is a need for an in-depth understanding of how different features of ABTs affect FcRn-binding and transport behavior. Here, we report on how FcRn-engagement of the IgG1 Fc fragment compare to clinically relevant IgGs and receptor domain Fc fusions, binding to VEGF or TNF-α. The results reveal FcRn-dependent intracellular accumulation of the Fc, which is in line with shorter plasma half-life than that of full-length IgG1 in human FcRn-expressing mice. Receptor domain fusion to the Fc increases its half-life, but not to the extent of IgG1. This is mirrored by a reduced cellular recycling capacity of the Fc-fusions. In addition, binding of cognate Ag to ABTs show that complexes of similar size undergo cellular transport at different rates, which could be explained by the biophysical properties of each ABT. Thus, the study provides knowledge that should guide tailoring of ABTs regarding optimal cellular sorting and plasma half-life. Analysis of clinically approved antibody-based therapeutics reveals different structural designs, such as full-length IgG1 or Fc-fusions, entail distinct biophysical properties that affect FcRn binding, intracellular transport and plasma half-life.
Collapse
|
15
|
Watkins JM, Watkins JD. An Engineered Monovalent Anti-TNF-α Antibody with pH-Sensitive Binding Abrogates Immunogenicity in Mice following a Single Intravenous Dose. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:829-839. [PMID: 35896334 PMCID: PMC10580234 DOI: 10.4049/jimmunol.2101180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 06/07/2022] [Indexed: 10/19/2023]
Abstract
Therapeutic Abs directed toward TNF-α display significant immunogenicity in humans, frequently leading to lower serum concentrations of the Ab that are associated with lower treatment efficacy. The enhanced incidence of immunogenicity observed with this class of therapeutics may be mediated by the expression of TNF-α as a homotrimer, both as a soluble serum protein and as a membrane-associated protein (mTNF-α) on the surface of dendritic cells. The TNF-α homotrimer enables the formation of polyvalent Ab-TNF-α immune complexes (ICs) that enhance binding to FcR and neonatal FcR. Polyvalent ICs and Ab bound to mTNF-α on the surface of dendritic cells can internalize, traffic to the lysosomes, and be processed for presentation by MHC molecules. To diminish immunogenicity caused by trafficking of ICs and mTNF-α to the lysosomes, we engineered a monovalent format of adalimumab with pH-sensitive binding to TNF-α. The engineered variant, termed AF-M2637, did not cross-link TNF-α trimers and consequently formed small, nonprecipitating ICs only. AF-M2637 bound TNF-α with high affinity at pH 7.4 (EC50 = 1.1 nM) and displayed a significantly faster dissociation rate than adalimumab at pH 6.0. No immune response to AF-M2637 was detected in mice following a single i.v. dose. In contrast, rapid immunization was detected following the injection of a single i.v. dose of adalimumab, monovalent adalimumab, or the bivalent form of the pH-sensitive variant. These data suggest that ICs and mTNF-α both contribute to the immunogenicity of adalimumab in mice and provide a general strategy for engineering less immunogenic therapeutic TNF-α Abs.
Collapse
|
16
|
Ledsgaard L, Ljungars A, Rimbault C, Sørensen CV, Tulika T, Wade J, Wouters Y, McCafferty J, Laustsen AH. Advances in antibody phage display technology. Drug Discov Today 2022; 27:2151-2169. [PMID: 35550436 DOI: 10.1016/j.drudis.2022.05.002] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 03/24/2022] [Accepted: 05/04/2022] [Indexed: 01/06/2023]
Abstract
Phage display technology can be used for the discovery of antibodies for research, diagnostic, and therapeutic purposes. In this review, we present and discuss key parameters that can be optimized when performing phage display selection campaigns, including the use of different antibody formats and advanced strategies for antigen presentation, such as immobilization, liposomes, nanodiscs, virus-like particles, and whole cells. Furthermore, we provide insights into selection strategies that can be used for the discovery of antibodies with complex binding requirements, such as targeting a specific epitope, cross-reactivity, or pH-dependent binding. Lastly, we provide a description of specialized phage display libraries for the discovery of bispecific antibodies and pH-sensitive antibodies. Together, these methods can be used to improve antibody discovery campaigns against all types of antigen. Teaser: This review provides an overview of the different strategies that can be exploited to improve the success rate of antibody phage display discovery campaigns, addressing key parameters, such as antigen presentation, selection methodologies, and specialized libraries.
Collapse
Affiliation(s)
- Line Ledsgaard
- Department of Biotechnology and Biomedicine, Technical University of Denmark, DK-2800 Kongens Lyngby, Denmark.
| | - Anne Ljungars
- Department of Biotechnology and Biomedicine, Technical University of Denmark, DK-2800 Kongens Lyngby, Denmark
| | - Charlotte Rimbault
- Department of Biotechnology and Biomedicine, Technical University of Denmark, DK-2800 Kongens Lyngby, Denmark
| | - Christoffer V Sørensen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, DK-2800 Kongens Lyngby, Denmark
| | - Tulika Tulika
- Department of Biotechnology and Biomedicine, Technical University of Denmark, DK-2800 Kongens Lyngby, Denmark
| | - Jack Wade
- Department of Biotechnology and Biomedicine, Technical University of Denmark, DK-2800 Kongens Lyngby, Denmark
| | - Yessica Wouters
- Department of Biotechnology and Biomedicine, Technical University of Denmark, DK-2800 Kongens Lyngby, Denmark
| | - John McCafferty
- Department of Medicine, Addenbrookes Hospital, Box 157, Hills Road, Cambridge, CB2 0QQ, UK; Department of Medicine, Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 0QQ, UK
| | - Andreas H Laustsen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, DK-2800 Kongens Lyngby, Denmark.
| |
Collapse
|
17
|
Lee PS, MacDonald KG, Massi E, Chew PV, Bee C, Perkins P, Chau B, Thudium K, Lohre J, Nandi P, Deyanova EG, Barman I, Gudmundsson O, Dollinger G, Sproul T, Engelhardt JJ, Strop P, Rajpal A. Improved therapeutic index of an acidic pH-selective antibody. MAbs 2022; 14:2024642. [PMID: 35192429 PMCID: PMC8865267 DOI: 10.1080/19420862.2021.2024642] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Although therapeutically efficacious, ipilimumab can exhibit dose-limiting toxicity that prevents maximal efficacious clinical outcomes and can lead to discontinuation of treatment. We hypothesized that an acidic pH-selective ipilimumab (pH Ipi), which preferentially and reversibly targets the acidic tumor microenvironment over the neutral periphery, may have a more favorable therapeutic index. While ipilimumab has pH-independent CTLA-4 affinity, pH Ipi variants have been engineered to have up to 50-fold enhanced affinity to CTLA-4 at pH 6.0 compared to pH 7.4. In hCTLA-4 knock-in mice, these variants have maintained anti-tumor activity and reduced peripheral activation, a surrogate marker for toxicity. pH-sensitive therapeutic antibodies may be a differentiating paradigm and a novel modality for enhanced tumor targeting and improved safety profiles.
Collapse
Affiliation(s)
- Peter S Lee
- Discovery Biotherapeutics, Bristol Myers Squibb, Redwood City, CA, USA
| | | | - Evan Massi
- Immuno-Oncology Research, Bristol Myers Squibb, Redwood City, CA, USA
| | - Pamela V Chew
- Oncology Biology, Gilead Sciences, Foster City, CA, USA
| | - Christine Bee
- Discovery Biology, Frontier Medicines, South San Francisco, CA, USA
| | - Padma Perkins
- Immuno-Oncology Research, Bristol Myers Squibb, Redwood City, CA, USA
| | - Bryant Chau
- Kyverna, Synthetic Biology, Emeryville, CA, USA
| | - Kent Thudium
- Immuno-Oncology Research, Bristol Myers Squibb, Redwood City, CA, USA
| | - Jack Lohre
- In Vivo Pharmacology, Bristol Myers Squibb, Redwood City, CA, USA
| | - Pradyot Nandi
- Pharmaceutical Candidate Optimization, Bristol Myers Squibb, Lawrenceville, NJ, USA
| | - Ekaterina G Deyanova
- Pharmaceutical Candidate Optimization, Bristol Myers Squibb, Lawrenceville, NJ, USA
| | - Ishita Barman
- Therapeutic Discovery, 3T Biosciences, South San Francisco, CA, USA
| | - Olafur Gudmundsson
- Pharmaceutical Candidate Optimization, Bristol Myers Squibb, Lawrenceville, NJ, USA
| | - Gavin Dollinger
- Discovery Biotherapeutics, Bristol Myers Squibb, Redwood City, CA, USA
| | - Tim Sproul
- In Vivo Pharmacology, UNITY Biotechnology, South San Francisco, CA, USA
| | | | - Pavel Strop
- Biologics Discovery, Tallac Therapeutics, Burlingame, CA, USA
| | - Arvind Rajpal
- Large Molecule Drug Discovery, Genentech Research and Early Development, South San Francisco, CA, USA
| |
Collapse
|
18
|
Hori Y, Ohmine K, Katada H, Noguchi Y, Sato K, Nambu T, Adeline LR, Wan GS, Haraya K, Ozeki K, Nanami M, Tachibana T, Sampei Z, Kuramochi T, Nezu J, Hattori K, Igawa T. Elimination of plasma soluble antigen in cynomolgus monkeys by combining pH-dependent antigen binding and novel Fc engineering. MAbs 2022; 14:2068213. [PMID: 35482905 PMCID: PMC9067469 DOI: 10.1080/19420862.2022.2068213] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
A conventional antibody targeting a soluble antigen in circulation typically requires a huge dosage and frequent intravenous administration to neutralize the antigen. This is because antigen degradation is reduced by the formation of antigen–antibody immune complexes, which escape from lysosomal degradation using neonatal Fc receptor (FcRn)-mediated recycling. To address this, we developed an antigen-sweeping antibody that combines pH-dependent antigen binding and Fc engineering to enhance Fc receptor binding. The sweeping antibody actively eliminates the plasma antigens by increasing the cellular uptake of the immune complex and dissociating the antigens in the acidic endosome for degradation. Strong antigen sweeping can reduce the dosage, potentially achieve higher efficacy, and expand the scope of antigen space available for targeting by antibodies. In this study, to further improve the sweeping efficacy, we developed a novel antibody Fc variant by enhancing Fcγ receptor IIb (FcγRIIb) binding and modulating charge characteristics for increased cellular uptake of the immune complex, together with enhancing FcRn binding for efficient salvage of the antigen-free antibodies. Our Fc variant achieved strong antigen sweeping in cynomolgus monkeys with antibody pharmacokinetics comparable to a wild-type human IgG1 antibody. The positive-charge substitutions enhanced uptake of the immune complex by FcγRIIb-expressing cells in vitro, which was completely inhibited by an anti-FcγRIIb antibody. This suggests that the strong in vivo sweeping efficacy improved by the charge engineering is more likely achieved by FcγRIIb-dependent uptake of the immune complex rather than nonspecific uptake. We expect this novel Fc engineering can maximize the antigen sweeping efficacy even in humans and create novel therapeutic antibodies that meet unmet medical needs for patients.
Collapse
Affiliation(s)
- Yuji Hori
- Research Division, Chugai Pharmaceutical Co, Ltd, Gotemba, Japan
| | - Ken Ohmine
- Research Division, Chugai Pharmaceutical Co, Ltd, Gotemba, Japan
| | | | - Yuki Noguchi
- Research Division, Chugai Pharmaceutical Co, Ltd, Gotemba, Japan
| | - Kazuki Sato
- Research Division, Chugai Pharmaceutical Co, Ltd, Gotemba, Japan
| | | | | | | | - Kenta Haraya
- Research Division, Chugai Pharmaceutical Co, Ltd, Gotemba, Japan
| | - Kazuhisa Ozeki
- Research Division, Chugai Pharmaceutical Co, Ltd, Gotemba, Japan
| | - Masahiko Nanami
- Research Division, Chugai Pharmaceutical Co, Ltd, Gotemba, Japan
| | | | - Zenjiro Sampei
- Research Division, Chugai Pharmaceutical Co, Ltd, Gotemba, Japan
| | - Taichi Kuramochi
- Research Division, Chugai Pharmaceutical Co, Ltd, Gotemba, Japan
| | - Junichi Nezu
- Research Division, Chugai Pharmaceutical Co, Ltd, Chuo-ku, Tokyo, Japan
| | - Kunihiro Hattori
- Research Division, Chugai Pharmaceutical Co, Ltd, Kamakura, Japan
| | - Tomoyuki Igawa
- Research Division, Chugai Pharmaceutical Co, Ltd, Chuo-ku, Tokyo, Japan
| |
Collapse
|
19
|
Grevys A, Frick R, Mester S, Flem-Karlsen K, Nilsen J, Foss S, Sand KMK, Emrich T, Fischer JAA, Greiff V, Sandlie I, Schlothauer T, Andersen JT. Antibody variable sequences have a pronounced effect on cellular transport and plasma half-life. iScience 2022; 25:103746. [PMID: 35118359 PMCID: PMC8800109 DOI: 10.1016/j.isci.2022.103746] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 11/11/2021] [Accepted: 01/05/2022] [Indexed: 11/15/2022] Open
Abstract
Monoclonal IgG antibodies are the fastest growing class of biologics, but large differences exist in their plasma half-life in humans. Thus, to design IgG antibodies with favorable pharmacokinetics, it is crucial to identify the determinants of such differences. Here, we demonstrate that the variable region sequences of IgG antibodies greatly affect cellular uptake and subsequent recycling and rescue from intracellular degradation by endothelial cells. When the variable sequences are masked by the cognate antigen, it influences both their transport behavior and binding to the neonatal Fc receptor (FcRn), a key regulator of IgG plasma half-life. Furthermore, we show how charge patch differences in the variable domains modulate both binding and transport properties and that a short plasma half-life, due to unfavorable charge patches, may partly be overcome by Fc-engineering for improved FcRn binding. IgG variable region sequences greatly affect cellular uptake and recycling Variable region charge patches affect FcRn binding and transport The presence of cognate antigen modulates cellular transport and FcRn binding Fc-engineering for improved FcRn binding can overcome unfavorable charge patches
Collapse
Affiliation(s)
- Algirdas Grevys
- Centre for Immune Regulation (CIR) and Department of Biosciences, University of Oslo, 0371 Oslo, Norway
- CIR and Department of Immunology, Oslo University Hospital Rikshospitalet, 0372 Oslo, Norway
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, 82377 Penzberg, Germany
- Corresponding author
| | - Rahel Frick
- CIR and Department of Immunology, Oslo University Hospital Rikshospitalet, 0372 Oslo, Norway
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway
| | - Simone Mester
- Centre for Immune Regulation (CIR) and Department of Biosciences, University of Oslo, 0371 Oslo, Norway
- CIR and Department of Immunology, Oslo University Hospital Rikshospitalet, 0372 Oslo, Norway
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway
| | - Karine Flem-Karlsen
- CIR and Department of Immunology, Oslo University Hospital Rikshospitalet, 0372 Oslo, Norway
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway
| | - Jeannette Nilsen
- CIR and Department of Immunology, Oslo University Hospital Rikshospitalet, 0372 Oslo, Norway
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway
| | - Stian Foss
- CIR and Department of Immunology, Oslo University Hospital Rikshospitalet, 0372 Oslo, Norway
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway
| | - Kine Marita Knudsen Sand
- Centre for Immune Regulation (CIR) and Department of Biosciences, University of Oslo, 0371 Oslo, Norway
- CIR and Department of Immunology, Oslo University Hospital Rikshospitalet, 0372 Oslo, Norway
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway
| | - Thomas Emrich
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, 82377 Penzberg, Germany
| | | | - Victor Greiff
- Department of Immunology, Institute of Clinical Medicine, University of Oslo, 0424 Oslo, Norway
| | - Inger Sandlie
- Centre for Immune Regulation (CIR) and Department of Biosciences, University of Oslo, 0371 Oslo, Norway
- CIR and Department of Immunology, Oslo University Hospital Rikshospitalet, 0372 Oslo, Norway
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway
| | - Tilman Schlothauer
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, 82377 Penzberg, Germany
| | - Jan Terje Andersen
- CIR and Department of Immunology, Oslo University Hospital Rikshospitalet, 0372 Oslo, Norway
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway
- Corresponding author
| |
Collapse
|
20
|
Veissi M, Maktabi S, Ramezani Z, Khosravi M. Highly Sensitive Fluorescence Assay of Enterotoxin A in Milk Using Carbon Quantum Dots as a Fluorophore. FOOD ANAL METHOD 2021. [DOI: 10.1007/s12161-021-02009-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
21
|
Sun Y, Estevez A, Schlothauer T, Wecksler AT. Antigen physiochemical properties allosterically effect the IgG Fc-region and Fc neonatal receptor affinity. MAbs 2021; 12:1802135. [PMID: 32795110 PMCID: PMC7531492 DOI: 10.1080/19420862.2020.1802135] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The neonatal Fc receptor (FcRn) is a key membrane protein that plays an integral role in serum immunoglobulin (IgG) recycling, which extends the half-life of antibody. In addition, FcRn is known to traffic antigen-bound immunoglobulins (Ag-IgGs), and to interact with immune complexes to facilitate the antigen cross-presentation of peptides derived from the immune complexes in antigen-presenting cells (APCs). Studies on the IgG-FcRn molecular interactions have primarily focused on the Fc region, and only recently have shown the potential impact of the antigen-binding fragment physiochemical properties on FcRn binding. However, the effect of the antigen physiochemical properties on IgG structure as it relates to Ag-IgG-FcRn binding is not well understood. Here we used an IgG-peptide antigen complex as a model system to investigate the structural effects of the antigen's physiochemical properties on the IgG structure, and the subsequent effects of Ag-IgG-FcRn interactions. We used hydroxyl radical footprinting-mass spectrometry to investigate the structural impact on an IgG upon antigen binding, and observed that the physicochemical properties of the antigen differentially induce conformational changes in the IgG FcRn binding region. The extent of these structural changes directly correlates to the magnitude of the affinity differences between the Ag-IgG complexes and FcRn. Moreover, the antigen's physicochemical properties differentially induce structural differences within the Ag-IgG-FcRn ternary complex. We also provide electron microscopy data that shows corroborating Fab-FcRn interactions, and confirms the hypothesis of potential 2:1 FcRn:IgG binding stoichiometry. These data demonstrate antigen-induced Fc structural rearrangements affect both the affinity toward FcRn and the trimeric antigen-IgG-FcRn complex, providing novel molecular insights in the first steps toward understanding interactions of FcRn-containing large(r)-sized immune complex.
Collapse
Affiliation(s)
- Yue Sun
- Protein Analytical Chemistry, Genentech Inc ., South San Francisco, CA, USA
| | - Alberto Estevez
- Structural Biology, Genentech Inc ., South San Francisco, CA, USA
| | - Tilman Schlothauer
- Roche Pharma Research & Early Development, Roche Innovation Center Munich , Penzberg, Germany.,Biological Technologies, Genentech Inc ., South San Francisco, CA, USA
| | - Aaron T Wecksler
- Protein Analytical Chemistry, Genentech Inc ., South San Francisco, CA, USA
| |
Collapse
|
22
|
Qi T, Cao Y. In Translation: FcRn across the Therapeutic Spectrum. Int J Mol Sci 2021; 22:3048. [PMID: 33802650 PMCID: PMC8002405 DOI: 10.3390/ijms22063048] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/11/2021] [Accepted: 03/15/2021] [Indexed: 12/14/2022] Open
Abstract
As an essential modulator of IgG disposition, the neonatal Fc receptor (FcRn) governs the pharmacokinetics and functions many therapeutic modalities. In this review, we thoroughly reexamine the hitherto elucidated biological and thermodynamic properties of FcRn to provide context for our assessment of more recent advances, which covers antigen-binding fragment (Fab) determinants of FcRn affinity, transgenic preclinical models, and FcRn targeting as an immune-complex (IC)-clearing strategy. We further comment on therapeutic antibodies authorized for treating SARS-CoV-2 (bamlanivimab, casirivimab, and imdevimab) and evaluate their potential to saturate FcRn-mediated recycling. Finally, we discuss modeling and simulation studies that probe the quantitative relationship between in vivo IgG persistence and in vitro FcRn binding, emphasizing the importance of endosomal transit parameters.
Collapse
Affiliation(s)
| | - Yanguang Cao
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA;
| |
Collapse
|
23
|
Sulea T, Rohani N, Baardsnes J, Corbeil CR, Deprez C, Cepero-Donates Y, Robert A, Schrag JD, Parat M, Duchesne M, Jaramillo ML, Purisima EO, Zwaagstra JC. Structure-based engineering of pH-dependent antibody binding for selective targeting of solid-tumor microenvironment. MAbs 2021; 12:1682866. [PMID: 31777319 PMCID: PMC6927761 DOI: 10.1080/19420862.2019.1682866] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Recent development of monoclonal antibodies as mainstream anticancer agents demands further optimization of their safety for use in humans. Potent targeting and/or effector activities on normal tissues is an obvious toxicity concern. Optimization of specific tumor targeting could be achieved by taking advantage of the extracellular acidity of solid tumors relative to normal tissues. Here, we applied a structure-based computational approach to engineer anti-human epidermal growth factor receptor 2 (Her2) antibodies with selective binding in the acidic tumor microenvironment. We used an affinity maturation platform in which dual-pH histidine-scanning mutagenesis was implemented for pH selectivity optimization. Testing of a small set of designs for binding to the recombinant Her2 ectodomain led to the identification of antigen-binding fragment (Fab) variants with the desired pH-dependent binding behavior. Binding selectivity toward acidic pH was improved by as much as 25-fold relative to the parental bH1-Fab. In vitro experiments on cells expressing intact Her2 confirmed that designed variants formatted as IgG1/k full-size antibodies have high affinity and inhibit the growth of tumor spheroids at a level comparable to that of the benchmark anti-Her2 antibody trastuzumab (Herceptin®) at acidic pH, whereas these effects were significantly reduced at physiological pH. In contrast, both Herceptin and the parental bH1 antibody exhibited strong cell binding and growth inhibition irrespective of pH. This work demonstrates the feasibility of computational optimization of antibodies for selective targeting of the acidic environment such as that found in many solid tumors.
Collapse
Affiliation(s)
- Traian Sulea
- Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, Quebec, Canada
| | - Nazanin Rohani
- Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, Quebec, Canada
| | - Jason Baardsnes
- Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, Quebec, Canada
| | - Christopher R Corbeil
- Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, Quebec, Canada
| | - Christophe Deprez
- Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, Quebec, Canada
| | - Yuneivy Cepero-Donates
- Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, Quebec, Canada
| | - Alma Robert
- Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, Quebec, Canada
| | - Joseph D Schrag
- Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, Quebec, Canada
| | - Marie Parat
- Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, Quebec, Canada
| | - Mélanie Duchesne
- Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, Quebec, Canada
| | - Maria L Jaramillo
- Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, Quebec, Canada
| | - Enrico O Purisima
- Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, Quebec, Canada
| | - John C Zwaagstra
- Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, Quebec, Canada
| |
Collapse
|
24
|
The covalent SNAP tag for protein display quantification and low-pH protein engineering. J Biotechnol 2020; 320:50-56. [PMID: 32561362 DOI: 10.1016/j.jbiotec.2020.06.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 06/14/2020] [Accepted: 06/15/2020] [Indexed: 11/22/2022]
Abstract
Yeast display has become an important tool for modern biotechnology with many advantages for eukaryotic protein engineering. Antibody-based peptide interactions are often used to quantify yeast surface expression (e.g., by fusing a target protein to a FLAG, Myc, polyhistidine, or other peptide tag). However, antibody-antigen interactions require high stability for accurate quantification, and conventional tag systems based on such interactions may not be compatible with a low pH environment. In this study, a SNAP tag was introduced to a yeast display platform to circumvent disadvantages of conventional antibody display tags at low pH. SNAP forms a covalent bond with its small-molecule substrate, enabling precise and pH-independent protein display tagging. We compared the SNAP tag to conventional antibody-based peptide fusion and to direct fluorescent domain fusion using antibody fragment crystallizable (Fc) gene libraries as a case study in low pH protein engineering. Our results demonstrated that covalent SNAP tags can effectively quantify protein-surface expression at low pH, enabling the enrichment of Fc variants with increased affinity at pH 6.0 to the neonatal Fc receptor (FcRn). Incorporation of a covalent SNAP tag thus overcomes disadvantages of conventional antibody-based expression tags and enables protein-engineering applications outside of physiological pH.
Collapse
|
25
|
Conner KP, Devanaboyina SC, Thomas VA, Rock DA. The biodistribution of therapeutic proteins: Mechanism, implications for pharmacokinetics, and methods of evaluation. Pharmacol Ther 2020; 212:107574. [PMID: 32433985 DOI: 10.1016/j.pharmthera.2020.107574] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Accepted: 04/30/2020] [Indexed: 02/08/2023]
Abstract
Therapeutic proteins (TPs) are a diverse drug class that include monoclonal antibodies (mAbs), recombinantly expressed enzymes, hormones and growth factors, cytokines (e.g. chemokines, interleukins, interferons), as well as a wide range of engineered fusion scaffolds containing IgG1 Fc domain for half-life extension. As the pharmaceutical industry advances more potent and selective protein-based medicines through discovery and into the clinical stages of development, it has become widely appreciated that a comprehensive understanding of the mechanisms of TP biodistribution can aid this endeavor. This review aims to highlight the literature that has advanced our understanding of the determinants of TP biodistribution. A particular emphasis is placed on the multi-faceted role of the neonatal Fc receptor (FcRn) in mAb and Fc-fusion protein disposition. In addition, characterization of the TP-target interaction at the cell-level is discussed as an essential strategy to establish pharmacokinetic-pharmacodynamic (PK/PD) relationships that may lead to more informed human dose projections during clinical development. Methods for incorporation of tissue and cell-level parameters defining these characteristics into higher-order mechanistic and semi-mechanistic PK models will also be presented.
Collapse
Affiliation(s)
- Kip P Conner
- Dept. of Pharmacokinetics and Drug Metabolism, Amgen Inc, 1120 Veterans Blvd, South San Francisco, CA 94080, USA.
| | - Siva Charan Devanaboyina
- Dept. of Pharmacokinetics and Drug Metabolism, Amgen Inc, 1120 Veterans Blvd, South San Francisco, CA 94080, USA.
| | - Veena A Thomas
- Dept. of Pharmacokinetics and Drug Metabolism, Amgen Inc, 1120 Veterans Blvd, South San Francisco, CA 94080, USA.
| | - Dan A Rock
- Dept. of Pharmacokinetics and Drug Metabolism, Amgen Inc, 1120 Veterans Blvd, South San Francisco, CA 94080, USA.
| |
Collapse
|
26
|
Yang F, Lin S, Ye F, Yang J, Qi J, Chen Z, Lin X, Wang J, Yue D, Cheng Y, Chen Z, Chen H, You Y, Zhang Z, Yang Y, Yang M, Sun H, Li Y, Cao Y, Yang S, Wei Y, Gao GF, Lu G. Structural Analysis of Rabies Virus Glycoprotein Reveals pH-Dependent Conformational Changes and Interactions with a Neutralizing Antibody. Cell Host Microbe 2020; 27:441-453.e7. [DOI: 10.1016/j.chom.2019.12.012] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 12/06/2019] [Accepted: 12/30/2019] [Indexed: 12/21/2022]
|
27
|
Klyukin K, Alexandrov V. Kinetics of pH-dependent interactions between PD-1 and PD-L1 immune checkpoint proteins from molecular dynamics. Proteins 2020; 88:1162-1168. [PMID: 32105362 DOI: 10.1002/prot.25885] [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: 11/05/2019] [Revised: 02/05/2020] [Accepted: 02/23/2020] [Indexed: 12/17/2022]
Abstract
Immune checkpoint blockade of signaling pathways such as PD-1/PD-L1 has recently opened up a new avenue for highly efficient immunotherapeutic strategies to treat cancer. Since tumor microenvironments are characterized by lower pH (5.5-7.0), pH-dependent protein-ligand interactions can be exploited as efficient means to regulate drug affinity and specificity for a variety of malignancies. In this article, we investigate the mechanism and kinetics of pH-dependent binding and unbinding processes for the PD-1/PD-L1 checkpoint pair employing classical molecular dynamics simulations. Two representative pH levels corresponding to circumneutral physiological conditions of blood (pH 7.4) and acidic tumor microenvironment (pH 5.5) are considered. Our calculations demonstrate that pH plays a key role in protein-ligand interactions with small pH changes leading to several orders of magnitude increase in binding affinity. By identifying the binding pocket in the PD-1/PD-L1 complex, we show a pivotal role of the His68 protonation state of PD-1in the complex stabilization at low pH. The results on the reaction rate constants are in qualitative agreement with available experimental data. The obtained molecular details are important for further engineering of binding/unbinding kinetics to formulate more efficient immune checkpoint blockade strategies.
Collapse
Affiliation(s)
- Konstantin Klyukin
- Department of Chemical and Biomolecular Engineering, University of Nebraska-Lincoln, Lincoln, Nebraska
| | - Vitaly Alexandrov
- Department of Chemical and Biomolecular Engineering, University of Nebraska-Lincoln, Lincoln, Nebraska.,Nebraska Center for Materials and Nanoscience, University of Nebraska-Lincoln, Lincoln, Nebraska
| |
Collapse
|
28
|
Application of interpretable artificial neural networks to early monoclonal antibodies development. Eur J Pharm Biopharm 2019; 141:81-89. [DOI: 10.1016/j.ejpb.2019.05.017] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 05/17/2019] [Accepted: 05/17/2019] [Indexed: 11/20/2022]
|
29
|
A two-stage method for automated detection of ring-like endosomes in fluorescent microscopy images. PLoS One 2019; 14:e0218931. [PMID: 31246999 PMCID: PMC6597078 DOI: 10.1371/journal.pone.0218931] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 06/12/2019] [Indexed: 01/21/2023] Open
Abstract
Endosomes are subcellular organelles which serve as important transport compartments in eukaryotic cells. Fluorescence microscopy is a widely applied technology to study endosomes at the subcellular level. In general, a microscopy image can contain a large number of organelles and endosomes in particular. Detecting and annotating endosomes in fluorescence microscopy images is a critical part in the study of subcellular trafficking processes. Such annotation is usually performed by human inspection, which is time-consuming and prone to inaccuracy if carried out by inexperienced analysts. This paper proposes a two-stage method for automated detection of ring-like endosomes. The method consists of a localization stage cascaded by an identification stage. Given a test microscopy image, the localization stage generates a voting-map by locally comparing the query endosome patches and the test image based on a bag-of-words model. Using the voting-map, a number of candidate patches of endosomes are determined. Subsequently, in the identification stage, a support vector machine (SVM) is trained using the endosome patches and the background pattern patches. Each of the candidate patches is classified by the SVM to rule out those patches of endosome-like background patterns. The performance of the proposed method is evaluated with real microscopy images of human myeloid endothelial cells. It is shown that the proposed method significantly outperforms several state-of-the-art competing methods using multiple performance metrics.
Collapse
|
30
|
de Souza AR, Yamin M, Gava D, Zanella JRC, Gatti MSV, Bonafe CFS, de Lima Neto DF. Porcine parvovirus VP1/VP2 on a time series epitope mapping: exploring the effects of high hydrostatic pressure on the immune recognition of antigens. Virol J 2019; 16:75. [PMID: 31159841 PMCID: PMC6547530 DOI: 10.1186/s12985-019-1165-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 04/17/2019] [Indexed: 11/24/2022] Open
Abstract
Porcine parvovirus (PPV) is a DNA virus that causes reproductive failure in gilts and sows, resulting in embryonic and fetal losses worldwide. Epitope mapping of PPV is important for developing new vaccines. In this study, we used spot synthesis analysis for epitope mapping of the capsid proteins of PPV (NADL-2 strain) and correlated the findings with predictive data from immunoinformatics. The virus was exposed to three conditions prior to inoculation in pigs: native (untreated), high hydrostatic pressure (350 MPa for 1 h) at room temperature and high hydrostatic pressure (350 MPa for 1 h) at − 18 °C, and was compared with a commercial vaccine produced using inactivated PPV. The screening of serum samples detected 44 positive spots corresponding to 20 antigenic sites. Each type of inoculated antigen elicited a distinct epitope set. In silico prediction located linear and discontinuous epitopes in B cells that coincided with several epitopes detected in spot synthesis of sera from pigs that received different preparations of inoculum. The conditions tested elicited antibodies against the VP1/VP2 antigen that differed in relation to the response time and the profile of structurally available regions that were recognized.
Collapse
Affiliation(s)
- Ancelmo Rabelo de Souza
- Departamento de Bioquímica e Biologia Tecidual, Universidade Estadual de Campimas (UNICAMP), Rua Monteiro Lobato, 255, Cidade Universitária Zeferino Vaz, Campinas, SP, 13083-862, Brazil
| | - Marriam Yamin
- Departamento de Bioquímica e Biologia Tecidual, Universidade Estadual de Campimas (UNICAMP), Rua Monteiro Lobato, 255, Cidade Universitária Zeferino Vaz, Campinas, SP, 13083-862, Brazil
| | - Danielle Gava
- Embrapa Suínos e Aves, Laboratório de Virologia de Suínos, Concórdia, SC, 89715-899, Brazil
| | | | - Maria Sílvia Viccari Gatti
- Departamento de Bioquímica e Biologia Tecidual, Universidade Estadual de Campimas (UNICAMP), Rua Monteiro Lobato, 255, Cidade Universitária Zeferino Vaz, Campinas, SP, 13083-862, Brazil
| | - Carlos Francisco Sampaio Bonafe
- Departamento de Bioquímica e Biologia Tecidual, Universidade Estadual de Campimas (UNICAMP), Rua Monteiro Lobato, 255, Cidade Universitária Zeferino Vaz, Campinas, SP, 13083-862, Brazil
| | - Daniel Ferreira de Lima Neto
- Departamento de Bioquímica e Biologia Tecidual, Universidade Estadual de Campimas (UNICAMP), Rua Monteiro Lobato, 255, Cidade Universitária Zeferino Vaz, Campinas, SP, 13083-862, Brazil. .,Departamento de Genética, Evolução e Bioagentes, Instituto de Biologia, Universidade Estadual de Campinas (UNICAMP), Rua Monteiro Lobato, 255, Cidade Universitária Zeferino Vaz, Campinas, SP, 13083-862, Brazil.
| |
Collapse
|
31
|
Kang JC, Sun W, Khare P, Karimi M, Wang X, Shen Y, Ober RJ, Ward ES. Engineering a HER2-specific antibody-drug conjugate to increase lysosomal delivery and therapeutic efficacy. Nat Biotechnol 2019; 37:523-526. [PMID: 30936563 PMCID: PMC6668989 DOI: 10.1038/s41587-019-0073-7] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 02/20/2019] [Indexed: 12/11/2022]
Abstract
We improve the potency of antibody-drug conjugates (ADCs) containing the
HER2-specific antibody pertuzumab by reducing their affinity for HER2 by
>250-fold at acidic endosomal pH relative to near neutral pH. These
engineered pertuzumab variants show increased lysosomal delivery and
cytotoxicity towards tumor cells expressing intermediate HER2 levels. In
HER2int xenograft tumor models in mice, the variants show higher
therapeutic efficacy than the parent ADC and a clinically-approved HER2-specific
ADC.
Collapse
Affiliation(s)
- Jeffrey C Kang
- Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, College Station, TX, USA
| | - Wei Sun
- Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, College Station, TX, USA
| | - Priyanka Khare
- Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, College Station, TX, USA
| | - Mostafa Karimi
- Department of Electrical & Computer Engineering, Texas A&M University, College Station, TX, USA
| | - Xiaoli Wang
- Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, College Station, TX, USA
| | - Yang Shen
- Department of Electrical & Computer Engineering, Texas A&M University, College Station, TX, USA
| | - Raimund J Ober
- Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, College Station, TX, USA. .,Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA. .,Cancer Sciences Unit, Centre for Cancer Immunology, Faculty of Medicine, University of Southampton, Southampton, UK.
| | - E Sally Ward
- Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, College Station, TX, USA. .,Cancer Sciences Unit, Centre for Cancer Immunology, Faculty of Medicine, University of Southampton, Southampton, UK. .,Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX, USA.
| |
Collapse
|
32
|
Sampei Z, Haraya K, Tachibana T, Fukuzawa T, Shida-Kawazoe M, Gan SW, Shimizu Y, Ruike Y, Feng S, Kuramochi T, Muraoka M, Kitazawa T, Kawabe Y, Igawa T, Hattori K, Nezu J. Antibody engineering to generate SKY59, a long-acting anti-C5 recycling antibody. PLoS One 2018; 13:e0209509. [PMID: 30592762 PMCID: PMC6310256 DOI: 10.1371/journal.pone.0209509] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 12/06/2018] [Indexed: 01/08/2023] Open
Abstract
Modulating the complement system is a promising strategy in drug discovery for disorders with uncontrolled complement activation. Although some of these disorders can be effectively treated with an antibody that inhibits complement C5, the high plasma concentration of C5 requires a huge dosage and frequent intravenous administration. Moreover, a conventional anti-C5 antibody can cause C5 to accumulate in plasma by reducing C5 clearance when C5 forms an immune complex (IC) with the antibody, which can be salvaged from endosomal vesicles by neonatal Fc receptor (FcRn)-mediated recycling. In order to neutralize the increased C5, an even higher dosage of the antibody would be required. This antigen accumulation can be suppressed by giving the antibody a pH-dependent C5-binding property so that C5 is released from the antibody in the acidic endosome and then trafficked to the lysosome for degradation, while the C5-free antibody returns back to plasma. We recently demonstrated that a pH-dependent C5-binding antibody, SKY59, exhibited long-lasting neutralization of C5 in cynomolgus monkeys, showing potential for subcutaneous delivery or less frequent administration. Here we report the details of the antibody engineering involved in generating SKY59, from humanizing a rabbit antibody to improving the C5-binding property. Moreover, because the pH-dependent C5-binding antibodies that we first generated still accumulated C5, we hypothesized that the surface charges of the ICs partially contributed to a slow uptake rate of the C5–antibody ICs. This idea motivated us to engineer the surface charges of the antibody. Our surface-charge engineered antibody consequently exhibited a high capacity to sweep C5 and suppressed the C5 accumulation in vivo by accelerating the cycle of sweeping: uptake of ICs into cells, release of C5 from the antibody in endosomes, and salvage of the antigen-free antibody. Thus, our engineered anti-C5 antibody, SKY59, is expected to provide significant benefits for patients with complement-mediated disorders.
Collapse
Affiliation(s)
- Zenjiro Sampei
- Research Division, Chugai Pharmaceutical Co., Ltd., Gotemba, Shizuoka, Japan
- * E-mail:
| | - Kenta Haraya
- Research Division, Chugai Pharmaceutical Co., Ltd., Gotemba, Shizuoka, Japan
| | - Tatsuhiko Tachibana
- Research Division, Chugai Pharmaceutical Co., Ltd., Kamakura, Kanagawa, Japan
| | - Taku Fukuzawa
- Research Division, Chugai Pharmaceutical Co., Ltd., Gotemba, Shizuoka, Japan
| | - Meiri Shida-Kawazoe
- Research Division, Chugai Pharmaceutical Co., Ltd., Gotemba, Shizuoka, Japan
| | - Siok Wan Gan
- Chugai Pharmabody Research Pte. Ltd., Singapore, Singapore
| | | | - Yoshinao Ruike
- Research Division, Chugai Pharmaceutical Co., Ltd., Kamakura, Kanagawa, Japan
| | - Shu Feng
- Chugai Pharmabody Research Pte. Ltd., Singapore, Singapore
| | | | - Masaru Muraoka
- Chugai Pharmabody Research Pte. Ltd., Singapore, Singapore
| | - Takehisa Kitazawa
- Research Division, Chugai Pharmaceutical Co., Ltd., Gotemba, Shizuoka, Japan
| | - Yoshiki Kawabe
- Research Division, Chugai Pharmaceutical Co., Ltd., Gotemba, Shizuoka, Japan
| | - Tomoyuki Igawa
- Chugai Pharmabody Research Pte. Ltd., Singapore, Singapore
| | - Kunihiro Hattori
- Research Division, Chugai Pharmaceutical Co., Ltd., Kamakura, Kanagawa, Japan
| | - Junichi Nezu
- Research Division, Chugai Pharmaceutical Co., Ltd., Kamakura, Kanagawa, Japan
| |
Collapse
|
33
|
Kroetsch A, Qiao C, Heavey M, Guo L, Shah DK, Park S. Engineered pH-dependent recycling antibodies enhance elimination of Staphylococcal enterotoxin B superantigen in mice. MAbs 2018; 11:411-421. [PMID: 30526311 DOI: 10.1080/19420862.2018.1545510] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
A new modality in antibody engineering has emerged in which the antigen affinity is designed to be pH dependent (PHD). In particular, combining high affinity binding at neutral pH with low affinity binding at acidic pH leads to a novel antibody that can more effectively neutralize the target antigen while avoiding antibody-mediated antigen accumulation. Here, we studied how the in vivo pharmacokinetics of the superantigen, Staphylococcal enterotoxin B (SEB), is affected by an engineered antibody with pH-dependent binding. PHD anti-SEB antibodies were engineered by introducing mutations into a high affinity anti-SEB antibody, 3E2, by rational design and directed evolution. Three antibody mutants engineered in the study have an affinity at pH 6.0 that is up to 68-fold weaker than the control antibody. The pH dependency of each mutant, measured as the pH-dependent affinity ratio (PAR - ratio of affinity at pH 7.4 and pH 6.0), ranged from 6.7-11.5 compared to 1.5 for the control antibody. The antibodies were characterized in mice by measuring their effects on the pharmacodynamics and pharmacokinetics (PK) of SEB after co-administration. All antibodies were effective in neutralizing the toxin and reducing the toxin-induced cytokine production. However, engineered PHD antibodies led to significantly faster elimination of the toxin from the circulation than wild type 3E2. The area under the curve computed from the SEB PK profile correlated well with the PAR value of antibody, indicating the importance of fine tuning the pH dependency of binding. These results suggest that a PHD recycling antibody may be useful to treat intoxication from a bacterial toxin by accelerating its clearance.
Collapse
Affiliation(s)
- Andrew Kroetsch
- a Department of Chemical and Biological Engineering , University at Buffalo , Buffalo , New York , USA
| | - Chunxia Qiao
- b Department of Pharmaceutical Sciences , University at Buffalo , Buffalo , New York , USA
| | - Mairead Heavey
- b Department of Pharmaceutical Sciences , University at Buffalo , Buffalo , New York , USA
| | - Leiming Guo
- b Department of Pharmaceutical Sciences , University at Buffalo , Buffalo , New York , USA
| | - Dhaval K Shah
- b Department of Pharmaceutical Sciences , University at Buffalo , Buffalo , New York , USA
| | - Sheldon Park
- a Department of Chemical and Biological Engineering , University at Buffalo , Buffalo , New York , USA
| |
Collapse
|
34
|
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: 24] [Impact Index Per Article: 3.4] [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
|
35
|
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: 83] [Impact Index Per Article: 11.9] [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
|
36
|
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
|
37
|
Srisa-Art M, Boehle KE, Geiss BJ, Henry CS. Highly Sensitive Detection of Salmonella typhimurium Using a Colorimetric Paper-Based Analytical Device Coupled with Immunomagnetic Separation. Anal Chem 2017; 90:1035-1043. [PMID: 29211962 DOI: 10.1021/acs.analchem.7b04628] [Citation(s) in RCA: 147] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Salmonella causes over a million foodborne illnesses per year in the United States resulting in more hospitalizations and deaths than any other foodborne bacterial pathogen. To help prevent outbreaks, a rapid, portable, sensitive, and reliable method for onsite detection of bacteria that can be used in different sample matrices would be beneficial. Herein, we present a colorimetric paper-based analytical device (PAD) combined with immunomagnetic separation (IMS) for detecting Salmonella typhimurium. IMS anti-Salmonella coated magnetic beads were applied to capture and separate bacteria from the sample matrix and preconcentrate it into small volumes before testing on paper. To directly detect S. typhimurium after IMS, a sandwich immunoassay was implemented into the procedure with β-galactosidase (β-gal) as the detection enzyme. Using the antibody/enzyme complex, we performed a colorimetric assay with chlorophenol red-β-d-galactopyranoside (CPRG) for bacteria quantification. The method was confirmed to be highly specific to S. typhimurium without interference from other pathogenic bacteria like Escherichia coli. Using this system, the limit of detection of S. typhimurium was found to be 102 CFU mL-1 in culturing solution without any pre-enrichment. In addition, distance-based detection where the concentration is read as the length of colored band formed on the reaction was also demonstrated. This assay had a detection limit of 102 CFU mL-1 for S. typhimurium, providing an instrument-free quantitative analysis alternative to spot tests, which require image analysis. Finally, the proposed platform was applied for detection of S. typhimurium in inoculated Starling bird fecal samples and whole milk with detection limits of 105 CFU g-1 and 103 CFU mL-1, respectively, and this is the first published paper-based detection method for S. typhimurium in bird feces and whole milk.
Collapse
Affiliation(s)
- Monpichar Srisa-Art
- Department of Chemistry, Faculty of Science, Chulalongkorn University , Bangkok, 10330, Thailand
| | | | | | | |
Collapse
|
38
|
Lin D, Lin Z, Velmurugan R, Ober RJ. Automatic Endosomal Structure Detection And Localization in Fluorescence Microscopic Images. IEEE INTERNATIONAL SYMPOSIUM ON CIRCUITS AND SYSTEMS PROCEEDINGS. IEEE INTERNATIONAL SYMPOSIUM ON CIRCUITS AND SYSTEMS 2017; 2017. [PMID: 30906101 DOI: 10.1109/iscas.2017.8050242] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
This paper proposes a modified spatially-constrained similarity measure (mSCSM) method for endosomal structure detection and localization under the bag-of-words (BoW) framework. To our best knowledge, the proposed mSCSM is the first method for fully automatic detection and localization of complex subcellular compartments like endosomes. Essentially, a new similarity score and a novel two-stage output control scheme are proposed for localization by extracting discriminative information within a group of query images. Compared with the original SCSM which is formulated for instance localization, the proposed mSCSM can address category based localization problems. The preliminary experimental results show the proposed mSCSM can correctly detect and localize 79.17% of the existing endosomal structures in the microscopic images of human myeloid endothelial cells.
Collapse
Affiliation(s)
- Dongyun Lin
- School of Electrical & Electronic Engineering, Nanyang Technological University, Singapore
| | - Zhiping Lin
- School of Electrical & Electronic Engineering, Nanyang Technological University, Singapore
| | - Ramraj Velmurugan
- Department of Biomedical Engineering, Texas A&M University, Texas, US
| | - Raimund J Ober
- Department of Biomedical Engineering, Texas A&M University, Texas, US
| |
Collapse
|
39
|
Li R, Chiguru S, Li L, Kim D, Velmurugan R, Kim D, Devanaboyina SC, Tian H, Schroit A, Mason RP, Ober RJ, Ward ES. Targeting Phosphatidylserine with Calcium-Dependent Protein-Drug Conjugates for the Treatment of Cancer. Mol Cancer Ther 2017; 17:169-182. [PMID: 28939556 DOI: 10.1158/1535-7163.mct-17-0092] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 05/04/2017] [Accepted: 08/29/2017] [Indexed: 12/18/2022]
Abstract
In response to cellular stress, phosphatidylserine is exposed on the outer membrane leaflet of tumor blood vessels and cancer cells, motivating the development of phosphatidylserine-specific therapies. The generation of drug-conjugated phosphatidylserine-targeting agents represents an unexplored therapeutic approach, for which antitumor effects are critically dependent on efficient internalization and lysosomal delivery of the cytotoxic drug. In the current study, we have generated phosphatidylserine-targeting agents by fusing phosphatidylserine-binding domains to a human IgG1-derived Fc fragment. The tumor localization and pharmacokinetics of several phosphatidylserine-specific Fc fusions have been analyzed in mice and demonstrate that Fc-Syt1, a fusion containing the synaptotagmin 1 C2A domain, effectively targets tumor tissue. Conjugation of Fc-Syt1 to the cytotoxic drug monomethyl auristatin E results in a protein-drug conjugate (PDC) that is internalized into target cells and, due to the Ca2+ dependence of phosphatidylserine binding, dissociates from phosphatidylserine in early endosomes. The released PDC is efficiently delivered to lysosomes and has potent antitumor effects in mouse xenograft tumor models. Interestingly, although an engineered, tetravalent Fc-Syt1 fusion shows increased binding to target cells, this higher avidity variant demonstrates reduced persistence and therapeutic effects compared with bivalent Fc-Syt1. Collectively, these studies show that finely tuned, Ca2+-switched phosphatidylserine-targeting agents can be therapeutically efficacious. Mol Cancer Ther; 17(1); 169-82. ©2017 AACR.
Collapse
Affiliation(s)
- Ran Li
- Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, College Station, Texas
| | - Srinivas Chiguru
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Li Li
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Dongyoung Kim
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas
| | - Ramraj Velmurugan
- Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, College Station, Texas.,Biomedical Engineering Graduate Program, University of Texas Southwestern Medical Center, Dallas, Texas
| | - David Kim
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas
| | - Siva Charan Devanaboyina
- Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, College Station, Texas
| | - Hong Tian
- China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Alan Schroit
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Ralph P Mason
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Raimund J Ober
- Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, College Station, Texas.,Department of Biomedical Engineering, Texas A&M University, College Station, Texas
| | - E Sally Ward
- Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, College Station, Texas. .,Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, College Station, Texas
| |
Collapse
|
40
|
Yang D, Giragossian C, Castellano S, Lasaro M, Xiao H, Saraf H, Hess Kenny C, Rybina I, Huang ZF, Ahlberg J, Bigwarfe T, Myzithras M, Waltz E, Roberts S, Kroe-Barrett R, Singh S. Maximizing in vivo target clearance by design of pH-dependent target binding antibodies with altered affinity to FcRn. MAbs 2017; 9:1105-1117. [PMID: 28786732 PMCID: PMC5627591 DOI: 10.1080/19420862.2017.1359455] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Antibodies with pH-dependent binding to both target antigens and neonatal Fc receptor (FcRn) provide an alternative tool to conventional neutralizing antibodies, particularly for therapies where reduction in antigen level is challenging due to high target burden. However, the requirements for optimal binding kinetic framework and extent of pH dependence for these antibodies to maximize target clearance from circulation are not well understood. We have identified a series of naturally-occurring high affinity antibodies with pH-dependent target binding properties. By in vivo studies in cynomolgus monkeys, we show that pH-dependent binding to the target alone is not sufficient for effective target removal from circulation, but requires Fc mutations that increase antibody binding to FcRn. Affinity-enhanced pH-dependent FcRn binding that is double-digit nM at pH 7.4 and single-digit nM at pH 6 achieved maximal target reduction when combined with similar target binding affinities in reverse pH directions. Sustained target clearance below the baseline level was achieved 3 weeks after single-dose administration at 1.5 mg/kg. Using the experimentally derived mechanistic model, we demonstrate the essential kinetic interplay between target turnover and antibody pH-dependent binding during the FcRn recycling, and identify the key components for achieving maximal target clearance. These results bridge the demand for improved patient dosing convenience with the “know-how” of therapeutic modality by design.
Collapse
Affiliation(s)
- Danlin Yang
- a Biotherapeutics Discovery Research , Boehringer Ingelheim Pharmaceuticals, Inc. , Ridgefield , Connecticut , USA
| | - Craig Giragossian
- a Biotherapeutics Discovery Research , Boehringer Ingelheim Pharmaceuticals, Inc. , Ridgefield , Connecticut , USA
| | - Steven Castellano
- b Duke University School of Medicine , Marilee Glen Court, Durham , North Carolina , USA
| | - Marcio Lasaro
- a Biotherapeutics Discovery Research , Boehringer Ingelheim Pharmaceuticals, Inc. , Ridgefield , Connecticut , USA
| | - Haiguang Xiao
- a Biotherapeutics Discovery Research , Boehringer Ingelheim Pharmaceuticals, Inc. , Ridgefield , Connecticut , USA
| | - Himanshu Saraf
- a Biotherapeutics Discovery Research , Boehringer Ingelheim Pharmaceuticals, Inc. , Ridgefield , Connecticut , USA
| | - Cynthia Hess Kenny
- a Biotherapeutics Discovery Research , Boehringer Ingelheim Pharmaceuticals, Inc. , Ridgefield , Connecticut , USA
| | - Irina Rybina
- a Biotherapeutics Discovery Research , Boehringer Ingelheim Pharmaceuticals, Inc. , Ridgefield , Connecticut , USA
| | - Zhong-Fu Huang
- a Biotherapeutics Discovery Research , Boehringer Ingelheim Pharmaceuticals, Inc. , Ridgefield , Connecticut , USA
| | - Jennifer Ahlberg
- a Biotherapeutics Discovery Research , Boehringer Ingelheim Pharmaceuticals, Inc. , Ridgefield , Connecticut , USA
| | - Tammy Bigwarfe
- a Biotherapeutics Discovery Research , Boehringer Ingelheim Pharmaceuticals, Inc. , Ridgefield , Connecticut , USA
| | - Maria Myzithras
- a Biotherapeutics Discovery Research , Boehringer Ingelheim Pharmaceuticals, Inc. , Ridgefield , Connecticut , USA
| | - Erica Waltz
- a Biotherapeutics Discovery Research , Boehringer Ingelheim Pharmaceuticals, Inc. , Ridgefield , Connecticut , USA
| | - Simon Roberts
- a Biotherapeutics Discovery Research , Boehringer Ingelheim Pharmaceuticals, Inc. , Ridgefield , Connecticut , USA
| | - Rachel Kroe-Barrett
- a Biotherapeutics Discovery Research , Boehringer Ingelheim Pharmaceuticals, Inc. , Ridgefield , Connecticut , USA
| | - Sanjaya Singh
- c Janssen BioTherapeutics , Janssen Research & Development , LLC, Spring House, Pennsylvania , USA
| |
Collapse
|
41
|
Adler LN, Jiang W, Bhamidipati K, Millican M, Macaubas C, Hung SC, Mellins ED. The Other Function: Class II-Restricted Antigen Presentation by B Cells. Front Immunol 2017; 8:319. [PMID: 28386257 PMCID: PMC5362600 DOI: 10.3389/fimmu.2017.00319] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 03/07/2017] [Indexed: 12/31/2022] Open
Abstract
Mature B lymphocytes (B cells) recognize antigens using their B cell receptor (BCR) and are activated to become antibody-producing cells. In addition, and integral to the development of a high-affinity antibodies, B cells utilize the specialized major histocompatibility complex class II (MHCII) antigen presentation pathway to process BCR-bound and internalized protein antigens and present selected peptides in complex with MHCII to CD4+ T cells. This interaction influences the fate of both types of lymphocytes and shapes immune outcomes. Specific, effective, and optimally timed antigen presentation by B cells requires well-controlled intracellular machinery, often regulated by the combined effects of several molecular events. Here, we delineate and summarize these events in four steps along the antigen presentation pathway: (1) antigen capture and uptake by B cells; (2) intersection of internalized antigen/BCRs complexes with MHCII in peptide-loading compartments; (3) generation and regulation of MHCII/peptide complexes; and (4) exocytic transport for presentation of MHCII/peptide complexes at the surface of B cells. Finally, we discuss modulation of the MHCII presentation pathway across B cell development and maturation to effector cells, with an emphasis on the shaping of the MHCII/peptide repertoire by two key antigen presentation regulators in B cells: HLA-DM and HLA-DO.
Collapse
Affiliation(s)
- Lital N Adler
- Department of Pediatrics, Stanford University, Stanford, CA, USA; Program in Immunology, Stanford University, Stanford, CA, USA
| | - Wei Jiang
- Department of Pediatrics, Stanford University, Stanford, CA, USA; Program in Immunology, Stanford University, Stanford, CA, USA
| | | | | | - Claudia Macaubas
- Department of Pediatrics, Stanford University, Stanford, CA, USA; Program in Immunology, Stanford University, Stanford, CA, USA
| | - Shu-Chen Hung
- Department of Pediatrics, Stanford University, Stanford, CA, USA; Program in Immunology, Stanford University, Stanford, CA, USA
| | - Elizabeth D Mellins
- Department of Pediatrics, Stanford University, Stanford, CA, USA; Program in Immunology, Stanford University, Stanford, CA, USA
| |
Collapse
|
42
|
Martins JP, Kennedy PJ, Santos HA, Barrias C, Sarmento B. A comprehensive review of the neonatal Fc receptor and its application in drug delivery. Pharmacol Ther 2016; 161:22-39. [PMID: 27016466 DOI: 10.1016/j.pharmthera.2016.03.007] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Advances in the understanding of neonatal Fc receptor (FcRn) biology and function have demonstrated that this receptor, primarily identified for the transfer of passive immunity from mother infant, is involved in several biological and immunological processes. In fact, FcRn is responsible for the long half-life of IgG and albumin in the serum, by creating an intracellular protein reservoir, which is protected from lysosomal degradation and, importantly, trafficked across the cell. Such discovery has led researchers to hypothesize the role for this unique receptor in the controlled delivery of therapeutic agents. A great amount of FcRn-based strategies are already under extensive investigation, in which FcRn reveals to have profound impact on the biodistribution and half-life extension of therapeutic agents. This review summarizes the main findings on FcRn biology, function and distribution throughout different tissues, together with the main advances on the FcRn-based therapeutic opportunities and model systems, which indicate that this receptor is a potential target for therapeutic regimen modification.
Collapse
Affiliation(s)
- João Pedro Martins
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; ICBAS - Instituto Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua Jorge Viterbo 228, 4150-180 Porto, Portugal
| | - Patrick J Kennedy
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; ICBAS - Instituto Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua Jorge Viterbo 228, 4150-180 Porto, Portugal; Ipatimup - Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
| | - Hélder A Santos
- Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E, FI -00014 Helsinki, Finland
| | - Cristina Barrias
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
| | - Bruno Sarmento
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; CESPU - Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde and Instituto Universitário de Ciências da Saúde, 4585-116 Gandra, Portugal.
| |
Collapse
|
43
|
Igawa T, Haraya K, Hattori K. Sweeping antibody as a novel therapeutic antibody modality capable of eliminating soluble antigens from circulation. Immunol Rev 2016; 270:132-51. [DOI: 10.1111/imr.12392] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Tomoyuki Igawa
- Research Division; Chugai Pharmaceutical Co. Ltd.; Shizuoka Japan
| | - Kenta Haraya
- Chugai Pharmabody Research Pte. Ltd.; Synapse Singapore
| | - Kunihiro Hattori
- Research Division; Chugai Pharmaceutical Co. Ltd.; Shizuoka Japan
| |
Collapse
|
44
|
Haraya K, Tachibana T, Iwayanagi Y, Maeda A, Ozeki K, Nezu J, Ishigai M, Igawa T. PK/PD analysis of a novel pH-dependent antigen-binding antibody using a dynamic antibody-antigen binding model. Drug Metab Pharmacokinet 2016; 31:123-32. [PMID: 26944099 DOI: 10.1016/j.dmpk.2015.12.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 12/01/2015] [Accepted: 12/21/2015] [Indexed: 01/26/2023]
Abstract
Previously, we have reported novel engineered antibody with pH-dependent antigen-binding (recycling antibody), and with both pH-dependent antigen-binding and increased FcRn-binding at neutral pH (sweeping antibody). The purpose of this study is to perform PK/PD predictions to better understand the potential applications of the antibodies as therapeutics. To demonstrate the applicability of recycling and sweeping antibodies over conventional antibodies, PK/PD analyses were performed. PK/PD parameters for antibody and antigen dynamics were estimated from the results of a pharmacokinetic study in human FcRn transgenic mice. A simulation study was performed using the estimated PK/PD parameters with various target antigen profiles. In comparison to conventional antibody, recycling antibody enhanced antibody-antigen complex clearance by 3 folds, while sweeping antibody accelerated antigen clearance by 10 folds in a pharmacokinetic study. Simulation results showed that recycling and sweeping antibodies can improve dosage frequency and reduce the required dose for target antigens with various clearances, plasma concentrations or binding kinetics. Moreover, importance of the association rate constant to enhance the beneficial effect of antibodies was shown. These results support the conclusion that recycling and sweeping antibodies can be applied to various target antigens with different profiles, and expand the number of antigens that antibodies can target.
Collapse
Affiliation(s)
- Kenta Haraya
- Chugai Pharmabody Research Pte. Ltd., 3 Biopolis Drive, #04-11 to 17 Synapse, 138623, Singapore.
| | - Tatsuhiko Tachibana
- Chugai Pharmabody Research Pte. Ltd., 3 Biopolis Drive, #04-11 to 17 Synapse, 138623, Singapore
| | - Yuki Iwayanagi
- Research Division, Chugai Pharmaceutical Co., Ltd., Gotemba, Shizuoka, Japan
| | - Atsuhiko Maeda
- Research Division, Chugai Pharmaceutical Co., Ltd., Gotemba, Shizuoka, Japan
| | - Kazuhisa Ozeki
- Research Division, Chugai Pharmaceutical Co., Ltd., Gotemba, Shizuoka, Japan
| | - Junichi Nezu
- Chugai Pharmabody Research Pte. Ltd., 3 Biopolis Drive, #04-11 to 17 Synapse, 138623, Singapore
| | - Masaki Ishigai
- Research Division, Chugai Pharmaceutical Co., Ltd., Gotemba, Shizuoka, Japan
| | - Tomoyuki Igawa
- Research Division, Chugai Pharmaceutical Co., Ltd., Gotemba, Shizuoka, Japan
| |
Collapse
|
45
|
Bonvin P, Venet S, Fontaine G, Ravn U, Gueneau F, Kosco-Vilbois M, Proudfoot AE, Fischer N. De novo isolation of antibodies with pH-dependent binding properties. MAbs 2015; 7:294-302. [PMID: 25608219 PMCID: PMC4623423 DOI: 10.1080/19420862.2015.1006993] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
pH-dependent antibodies are engineered to release their target at a slightly acidic pH, a property making them suitable for clinical as well as biotechnological applications. Such antibodies were previously obtained by histidine scanning of pre-existing antibodies, a labor-intensive strategy resulting in antibodies that displayed residual binding to their target at pH 6.0. We report here the de novo isolation of pH-dependent antibodies selected by phage display from libraries enriched in histidines. Strongly pH-dependent clones with various affinity profiles against CXCL10 were isolated by this method. Our best candidate has nanomolar affinity for CXCL10 at pH 7.2, but no residual binding was detected at pH 6.0. We therefore propose that this new process is an efficient strategy to generate pH-dependent antibodies.
Collapse
Key Words
- BLI, bio-layer interferometry
- CDR, complementary determining region
- CDRH, CDR of the heavy chain
- CDRL, CDR of the light chain
- ELISA, enzyme-linked immunosorbent assay
- GPCR, G protein-coupled receptor
- KB, kinetic buffer
- PBS, phosphate buffered saline
- SPR, surface plasmon resonance
- antibody recycling
- chemokine
- histidine
- mAb, monoclonal antibody
- monoclonal antibody
- pH-dependency
- phage display
- phage libraries
- scFv, single-chain variable fragment
Collapse
|
46
|
Hironiwa N, Ishii S, Kadono S, Iwayanagi Y, Mimoto F, Habu K, Igawa T, Hattori K. Calcium-dependent antigen binding as a novel modality for antibody recycling by endosomal antigen dissociation. MAbs 2015; 8:65-73. [PMID: 26496237 PMCID: PMC4966519 DOI: 10.1080/19420862.2015.1110660] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 10/09/2015] [Accepted: 10/15/2015] [Indexed: 11/18/2022] Open
Abstract
The pH-dependent antigen binding antibody, termed a recycling antibody, has recently been reported as an attractive type of second-generation engineered therapeutic antibody. A recycling antibody can dissociate antigen in the acidic endosome, and thus bind to its antigen multiple times. As a consequence, a recycling antibody can neutralize large amounts of antigen in plasma. Because this approach relies on histidine residues to achieve pH-dependent antigen binding, which could limit the epitopes that can be targeted and affect the rate of antigen dissociation in the endosome, we explored an alternative approach for generating recycling antibodies. Since calcium ion concentration is known to be lower in endosome than in plasma, we hypothesized that an antibody with antigen-binding properties that are calcium-dependent could be used as recycling antibody. Here, we report a novel anti-interleukin-6 receptor (IL-6R) antibody, identified from a phage library that binds to IL-6R only in the presence of a calcium ion. Thermal dynamics and a crystal structure study revealed that the calcium ion binds to the heavy chain CDR3 region (HCDR3), which changes and possibly stabilizes the structure of HCDR3 to make it bind to antigen calcium dependently (PDB 5AZE). In vitro and in vivo studies confirmed that this calcium-dependent antigen-binding antibody can dissociate its antigen in the endosome and accelerate antigen clearance from plasma, making it a novel approach for generating recycling antibody.
Collapse
Affiliation(s)
- N Hironiwa
- Research Division; Chugai Pharmaceutical Co., Ltd.; Tokyo, Japan
| | - S Ishii
- Research Division; Chugai Pharmaceutical Co., Ltd.; Tokyo, Japan
| | - S Kadono
- Research Division; Chugai Pharmaceutical Co., Ltd.; Tokyo, Japan
| | - Y Iwayanagi
- Research Division; Chugai Pharmaceutical Co., Ltd.; Tokyo, Japan
| | - F Mimoto
- Research Division; Chugai Pharmaceutical Co., Ltd.; Tokyo, Japan
| | - K Habu
- Research Division; Chugai Pharmaceutical Co., Ltd.; Tokyo, Japan
| | - T Igawa
- Research Division; Chugai Pharmaceutical Co., Ltd.; Tokyo, Japan
| | - K Hattori
- Research Division; Chugai Pharmaceutical Co., Ltd.; Tokyo, Japan
| |
Collapse
|
47
|
Schröter C, Günther R, Rhiel L, Becker S, Toleikis L, Doerner A, Becker J, Schönemann A, Nasu D, Neuteboom B, Kolmar H, Hock B. A generic approach to engineer antibody pH-switches using combinatorial histidine scanning libraries and yeast display. MAbs 2015; 7:138-51. [PMID: 25523975 PMCID: PMC4622719 DOI: 10.4161/19420862.2014.985993] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
There is growing interest in the fast and robust engineering of protein pH-sensitivity that aims to reduce binding at acidic pH, compared to neutral pH. Here, we describe a novel strategy for the incorporation of pH-sensitive antigen binding functions into antibody variable domains using combinatorial histidine scanning libraries and yeast surface display. The strategy allows simultaneous screening for both, high affinity binding at pH 7.4 and pH-sensitivity, and excludes conventional negative selection steps. As proof of concept, we applied this strategy to incorporate pH-dependent antigen binding into the complementary-determining regions of adalimumab. After 3 consecutive rounds of separate heavy and light chain library screening, pH-sensitive variants could be isolated. Heavy and light chain mutations were combined, resulting in 3 full-length antibody variants that revealed sharp, reversible pH-dependent binding profiles. Dissociation rate constants at pH 6.0 increased 230- to 780-fold, while high affinity binding at pH 7.4 in the sub-nanomolar range was retained. Furthermore, binding to huFcRn and thermal stability were not affected by histidine substitutions. Overall, this study emphasizes a generalizable strategy for engineering pH-switch functions potentially applicable to a variety of antibodies and further proteins-based therapeutics.
Collapse
Affiliation(s)
- Christian Schröter
- a Institute for Organic Chemistry and Biochemistry ; Technische Universität Darmstadt ; Darmstadt , Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Targeting FcRn for the modulation of antibody dynamics. Mol Immunol 2015; 67:131-41. [PMID: 25766596 DOI: 10.1016/j.molimm.2015.02.007] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Revised: 02/05/2015] [Accepted: 02/06/2015] [Indexed: 01/08/2023]
Abstract
The MHC class I-related receptor, FcRn, is a multitasking protein that transports its IgG ligand within and across cells of diverse origins. The role of this receptor as a global regulator of IgG homeostasis and transport, combined with knowledge of the molecular details of FcRn-IgG interactions, has led to opportunities to modulate the in vivo dynamics of antibodies and their antigens through protein engineering. Consequently, the generation of half-life extended antibodies has shown a rapid expansion over the past decade. Further, FcRn itself can be targeted by inhibitors to induce decreased levels of circulating IgGs, which could have applications in multiple clinical settings. The engineering of antibody-antigen interactions to reduce antibody-mediated buffering of soluble ligand has also developed into an active area of investigation, leading to novel antibody platforms designed to result in more effective antigen clearance. Similarly, the target-mediated elimination of antibodies by internalizing, membrane bound antigens (receptors) can be decreased using novel engineering approaches. These strategies, combined with subcellular trafficking analyses of antibody/antigen/FcRn behavior in cells to predict in vivo behavior, have considerable promise for the production of next generation therapeutics and diagnostics.
Collapse
|
49
|
Igawa T, Mimoto F, Hattori K. pH-dependent antigen-binding antibodies as a novel therapeutic modality. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2014; 1844:1943-1950. [DOI: 10.1016/j.bbapap.2014.08.003] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Revised: 07/31/2014] [Accepted: 08/05/2014] [Indexed: 12/20/2022]
|