1
|
Lee NJ, Jung M, Yang HY, Shim H. A single-domain antibody library based on a stability-engineered human VH3 scaffold. Sci Rep 2024; 14:17747. [PMID: 39085444 PMCID: PMC11291719 DOI: 10.1038/s41598-024-68680-5] [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: 05/14/2024] [Accepted: 07/25/2024] [Indexed: 08/02/2024] Open
Abstract
Using conventional immunoglobulin G (IgG) molecules as therapeutic agents presents several well-known disadvantages owing to their large size and structural complexity, negatively impacting development and production efficiency. Single-domain antibodies (sdAbs) are the smallest functional antibody format (~ 15 kDa) and represent a viable alternative to IgG in many applications. However, unlike natural single-domain antibodies, such as camelid VHH, the variable domains of conventional antibodies show poor physicochemical properties when expressed as sdAbs. This report identified stable sdAb variants of human VH3-23 from a framework region 2-randomized human VH library by phage display selection under thermal challenge. Synthetic complementarity determining region diversity was introduced to one of the selected variants with high thermal stability, expression level, and monomeric content to construct a human VH sdAb library. The library was validated by panning against a panel of antigens, and target-specific binders were identified and characterized for their affinity and biophysical properties. The results of this study suggest that a synthetic sdAb library based on a stability-engineered human VH scaffold could be a facile source of high-quality sdAb for many practical applications.
Collapse
Affiliation(s)
- Nam Ju Lee
- Department of Bioinspired Sciences, Ewha Womans University, Seoul, Korea
| | - Mooyoung Jung
- Department of Bioinspired Sciences, Ewha Womans University, Seoul, Korea
| | - Hye Young Yang
- Department of Life Sciences, Ewha Womans University, Seoul, Korea
| | - Hyunbo Shim
- Department of Bioinspired Sciences, Ewha Womans University, Seoul, Korea.
- Department of Life Sciences, Ewha Womans University, Seoul, Korea.
| |
Collapse
|
2
|
Edwards JP, Swers JS, Buonato JM, Zaritskaya L, Mu CJ, Gupta A, Shachar S, LaFleur DW, Richman LK, Tice DA, Hilbert DM. Controlling CAR-T cell activity and specificity with synthetic SparX adapters. Mol Ther 2024; 32:1835-1848. [PMID: 38659225 PMCID: PMC11184337 DOI: 10.1016/j.ymthe.2024.04.027] [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: 10/30/2023] [Revised: 03/18/2024] [Accepted: 04/18/2024] [Indexed: 04/26/2024] Open
Abstract
While conventional chimeric antigen-receptor (CAR)-T therapies have shown remarkable clinical activity in some settings, they can induce severe toxicities and are rarely curative. To address these challenges, we developed a controllable cell therapy where synthetic D-domain-containing proteins (soluble protein antigen-receptor X-linker [SparX]) bind one or more tumor antigens and mark those cells for elimination by genetically modified T cells (antigen-receptor complex [ARC]-T). The chimeric antigen receptor was engineered with a D-domain that specifically binds to the SparX protein via a unique TAG, derived from human alpha-fetoprotein. The interaction is mediated through an epitope on the TAG that is occluded in the native alpha-fetoprotein molecule. In vitro and in vivo data demonstrate that the activation and cytolytic activity of ARC-T cells is dependent on the dose of SparX protein and only occurs when ARC-T cells are engaged with SparX proteins bound to antigen-positive cells. ARC-T cell specificity was also redirected in vivo by changing SparX proteins that recognized different tumor antigens to combat inherent or acquired tumor heterogeneity. The ARC-SparX platform is designed to expand patient and physician access to cell therapy by controlling potential toxicities through SparX dosing regimens and enhancing tumor elimination through sequential or simultaneous administration of SparX proteins engineered to bind different tumor antigens.
Collapse
MESH Headings
- Humans
- Receptors, Chimeric Antigen/metabolism
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/genetics
- Animals
- Mice
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Immunotherapy, Adoptive/methods
- Antigens, Neoplasm/immunology
- Antigens, Neoplasm/metabolism
- Cell Line, Tumor
- Xenograft Model Antitumor Assays
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Antigen, T-Cell/immunology
- Neoplasms/therapy
- Neoplasms/immunology
- Neoplasms/metabolism
- Protein Binding
Collapse
|
3
|
Early-phenotype CAR-T cells for the treatment of pediatric cancers. Ann Oncol 2021; 32:1366-1380. [PMID: 34375680 DOI: 10.1016/j.annonc.2021.07.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 06/19/2021] [Accepted: 07/30/2021] [Indexed: 01/19/2023] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy is a promising approach for the treatment of childhood cancers, particularly high-risk tumors that fail to respond to standard therapies. CAR-T cells have been highly successful in treating some types of hematological malignancies. However, CAR-T cells targeting solid cancers have had limited success so far for multiple reasons, including their poor long-term persistence and proliferation. Evidence is emerging to show that maintaining CAR-T cells in an early, less differentiated state in vitro results in superior persistence, proliferation, and anti-tumor effects in vivo. Children are ideal candidates for receiving less-differentiated CAR-T cells, because their peripheral T cell pool primarily comprises naïve cells that could readily be harvested in large numbers to generate early-phenotype CAR-T cells. Although several studies have reported different approaches to successfully generate early CAR-T cells, there are only a few clinical trials testing these in adult patients. No trials are currently testing early CAR-T cells in children. Here, we summarize the different strategies used to maintain CAR-T cells in an early phenotypic stage, and present evidence suggesting that this approach may be particularly relevant to treating childhood cancers.
Collapse
|
4
|
Zhang C, Codina N, Tang J, Yu H, Chakroun N, Kozielski F, Dalby PA. Comparison of the pH- and thermally-induced fluctuations of a therapeutic antibody Fab fragment by molecular dynamics simulation. Comput Struct Biotechnol J 2021; 19:2726-2741. [PMID: 34093988 PMCID: PMC8131956 DOI: 10.1016/j.csbj.2021.05.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 04/15/2021] [Accepted: 05/01/2021] [Indexed: 11/27/2022] Open
Abstract
Successful development of protein therapeutics depends critically on achieving stability under a range of conditions. A deeper understanding of the drivers of instability across different stress conditions, will enable the engineering of more robust protein scaffolds. We compared the impacts of low pH and high temperature stresses on the structure of a humanized antibody fragment (Fab) A33, using atomistic molecular dynamics simulations, using a recent 2.5 Å crystal structure. This revealed that low-pH induced the loss of native contacts in the domain CL. By contrast, thermal stress led to 5-7% loss of native contacts in all four domains, and simultaneous loss of >30% of native contacts in the VL-VH and CL-CH interfaces. This revealed divergent destabilising pathways under the two different stresses. The underlying cause of instability was probed using FoldX and Rosetta mutation analysis, and packing density calculations. These agreed that mutations in the CL domain, and CL-CH1 interface have the greatest potential for stabilisation of Fab A33. Several key salt bridge losses underpinned the conformational change in CL at low pH, whereas at high temperature, salt bridges became more dynamic, thus contributing to an overall destabilization. Lastly, the unfolding events at the two stress conditions exposed different predicted aggregation-prone regions (APR) to solvent, which would potentially lead to different aggregation mechanisms. Overall, our results identified the early stages of unfolding and stability-limiting regions of Fab A33, and the VH and CL domains as interesting future targets for engineering stability to both pH- and thermal-stresses simultaneously.
Collapse
Affiliation(s)
- Cheng Zhang
- Department of Biochemical Engineering, University College London, Gordon Street, London WC1E 7JE, United Kingdom
| | - Nuria Codina
- Department of Biochemical Engineering, University College London, Gordon Street, London WC1E 7JE, United Kingdom
| | - Jiazhi Tang
- Department of Pharmaceutical and Biological Chemistry, School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, United Kingdom
| | - Haoran Yu
- Department of Chemistry, University College London, 20 Gordon Street, London, WC1H 0AJ, United Kingdom
| | - Nesrine Chakroun
- Department of Biochemical Engineering, University College London, Gordon Street, London WC1E 7JE, United Kingdom
| | - Frank Kozielski
- Department of Pharmaceutical and Biological Chemistry, School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, United Kingdom
| | - Paul A Dalby
- Department of Biochemical Engineering, University College London, Gordon Street, London WC1E 7JE, United Kingdom
| |
Collapse
|
5
|
Ajina A, Maher J. Strategies to Address Chimeric Antigen Receptor Tonic Signaling. Mol Cancer Ther 2018; 17:1795-1815. [PMID: 30181329 PMCID: PMC6130819 DOI: 10.1158/1535-7163.mct-17-1097] [Citation(s) in RCA: 146] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 03/19/2018] [Accepted: 06/18/2018] [Indexed: 12/18/2022]
Abstract
Adoptive cell transfer using chimeric antigen receptors (CAR) has emerged as one of the most promising new therapeutic modalities for patients with relapsed or refractory B-cell malignancies. Thus far, results in patients with advanced solid tumors have proven disappointing. Constitutive tonic signaling in the absence of ligand is an increasingly recognized complication when deploying these synthetic fusion receptors and can be a cause of poor antitumor efficacy, impaired survival, and reduced persistence in vivo In parallel, ligand-dependent tonic signaling can mediate toxicity and promote T-cell anergy, exhaustion, and activation-induced cell death. Here, we review the mechanisms underpinning CAR tonic signaling and highlight the wide variety of effects that can emerge after making subtle structural changes or altering the methodology of CAR transduction. We highlight strategies to prevent unconstrained tonic signaling and address its deleterious consequences. We also frame this phenomenon in the context of endogenous TCR tonic signaling, which has been shown to regulate peripheral tolerance, facilitate the targeting of foreign antigens, and suggest opportunities to coopt ligand-dependent CAR tonic signaling to facilitate in vivo persistence and efficacy. Mol Cancer Ther; 17(9); 1795-815. ©2018 AACR.
Collapse
MESH Headings
- Cell Differentiation/genetics
- Cell Differentiation/immunology
- Humans
- Immunotherapy, Adoptive/methods
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/therapy
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/metabolism
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/immunology
- Recombinant Fusion Proteins/metabolism
- Signal Transduction/genetics
- Signal Transduction/immunology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
Collapse
Affiliation(s)
- Adam Ajina
- CAR Mechanics Group, King's College London, London, United Kingdom.
- School of Cancer and Pharmaceutical Studies, Guy's Hospital, London, United Kingdom
| | - John Maher
- CAR Mechanics Group, King's College London, London, United Kingdom
- School of Cancer and Pharmaceutical Studies, Guy's Hospital, London, United Kingdom
- Department of Clinical Immunology and Allergy, King's College Hospital NHS Foundation Trust, London, United Kingdom
- Department of Immunology, Eastbourne Hospital, East Sussex, United Kingdom
| |
Collapse
|
6
|
Li J, Wang J, Grewal YS, Howard CB, Raftery LJ, Mahler S, Wang Y, Trau M. Multiplexed SERS Detection of Soluble Cancer Protein Biomarkers with Gold–Silver Alloy Nanoboxes and Nanoyeast Single-Chain Variable Fragments. Anal Chem 2018; 90:10377-10384. [DOI: 10.1021/acs.analchem.8b02216] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Junrong Li
- Centre for Personalized Nanomedicine, Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Jing Wang
- Centre for Personalized Nanomedicine, Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Yadveer S. Grewal
- Centre for Personalized Nanomedicine, Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Christopher B. Howard
- Centre for Advanced Imaging, Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Lyndon J. Raftery
- Centre for Advanced Imaging, Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Stephen Mahler
- Centre for Advanced Imaging, Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Yuling Wang
- Department of Molecular Sciences, ARC Centre of Excellence for Nanoscale BioPhotonics, Faculty of Science and Engineering, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Matt Trau
- Centre for Personalized Nanomedicine, Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland 4072, Australia
| |
Collapse
|
7
|
Herold EM, John C, Weber B, Kremser S, Eras J, Berner C, Deubler S, Zacharias M, Buchner J. Determinants of the assembly and function of antibody variable domains. Sci Rep 2017; 7:12276. [PMID: 28947772 PMCID: PMC5613017 DOI: 10.1038/s41598-017-12519-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 09/12/2017] [Indexed: 01/17/2023] Open
Abstract
The antibody Fv module which binds antigen consists of the variable domains VL and VH. These exhibit a conserved ß-sheet structure and comprise highly variable loops (CDRs). Little is known about the contributions of the framework residues and CDRs to their association. We exchanged conserved interface residues as well as CDR loops and tested the effects on two Fvs interacting with moderate affinities (KDs of ~2.5 µM and ~6 µM). While for the rather instable domains, almost all mutations had a negative effect, the more stable domains tolerated a number of mutations of conserved interface residues. Of particular importance for Fv association are VLP44 and VHL45. In general, the exchange of conserved residues in the VL/VH interface did not have uniform effects on domain stability. Furthermore, the effects on association and antigen binding do not strictly correlate. In addition to the interface, the CDRs modulate the variable domain framework to a significant extent as shown by swap experiments. Our study reveals a complex interplay of domain stability, association and antigen binding including an unexpected strong mutual influence of the domain framework and the CDRs on stability/association on the one side and antigen binding on the other side.
Collapse
Affiliation(s)
- Eva Maria Herold
- Center for Integrated Protein Science Munich (CIPSM) at the Department Chemie, Technische Universität München, 85747, Garching, Germany.,Sanofi-Aventis GmbH, Industriepark Höchst, 65926, Frankfurt am Main, Germany
| | - Christine John
- Center for Integrated Protein Science Munich (CIPSM) at the Department Chemie, Technische Universität München, 85747, Garching, Germany
| | - Benedikt Weber
- Center for Integrated Protein Science Munich (CIPSM) at the Department Chemie, Technische Universität München, 85747, Garching, Germany
| | - Stephan Kremser
- Center for Integrated Protein Science Munich (CIPSM) at the Physics Department, Technische Universität München, 85747, Garching, Germany
| | - Jonathan Eras
- ETH Zürich, Otto-Stern-Weg 5, 8093, Zuerich, Switzerland
| | - Carolin Berner
- Center for Integrated Protein Science Munich (CIPSM) at the Department Chemie, Technische Universität München, 85747, Garching, Germany
| | - Sabrina Deubler
- Center for Integrated Protein Science Munich (CIPSM) at the Department Chemie, Technische Universität München, 85747, Garching, Germany
| | - Martin Zacharias
- Center for Integrated Protein Science Munich (CIPSM) at the Physics Department, Technische Universität München, 85747, Garching, Germany
| | - Johannes Buchner
- Center for Integrated Protein Science Munich (CIPSM) at the Department Chemie, Technische Universität München, 85747, Garching, Germany.
| |
Collapse
|
8
|
Knapp B, Dunbar J, Alcala M, Deane CM. Variable Regions of Antibodies and T-Cell Receptors May Not Be Sufficient in Molecular Simulations Investigating Binding. J Chem Theory Comput 2017; 13:3097-3105. [PMID: 28617587 DOI: 10.1021/acs.jctc.7b00080] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Antibodies and T-cell receptors are important proteins of the immune system that share similar structures. Both contain variable and constant regions. Insight into the dynamics of their binding can be provided by computational simulations. For these simulations the constant regions are often removed to save runtime as binding occurs in the variable regions. Here we present the first study to investigate the effect of removing the constant regions from antibodies and T-cell receptors on such simulations. We performed simulations of an antibody/antigen and T-cell receptor/MHC system with and without constant regions using 10 replicas of 100 ns of each of the four setups. We found that simulations without constant regions show significantly different behavior compared to simulations with constant regions. If the constant regions are not included in the simulations alterations in the binding interface hydrogen bonds and even partial unbinding can occur. These results indicate that constant regions should be included in antibody and T-cell receptor simulations for reliable conclusions to be drawn.
Collapse
Affiliation(s)
- Bernhard Knapp
- Department of Statistics, Protein Informatics Group, University of Oxford , Oxford OX1 3BD, U.K.,Department of Basic Sciences, Faculty of Medicine and Health Sciences, International University of Catalonia , 08195 Sant Cugat del Vallès, Barcelona, Spain
| | - James Dunbar
- Department of Statistics, Protein Informatics Group, University of Oxford , Oxford OX1 3BD, U.K.,Pharma Research and Early Development, Large Molecule Research, Roche Innovation Center Munich 82377 Penzberg, Germany
| | - Marta Alcala
- Department of Basic Sciences, Faculty of Medicine and Health Sciences, International University of Catalonia , 08195 Sant Cugat del Vallès, Barcelona, Spain
| | - Charlotte M Deane
- Department of Statistics, Protein Informatics Group, University of Oxford , Oxford OX1 3BD, U.K
| |
Collapse
|
9
|
Grewal Y, Shiddiky MJA, Mahler SM, Cangelosi GA, Trau M. Nanoyeast and Other Cell Envelope Compositions for Protein Studies and Biosensor Applications. ACS APPLIED MATERIALS & INTERFACES 2016; 8:30649-30664. [PMID: 27762541 PMCID: PMC5114700 DOI: 10.1021/acsami.6b09263] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 10/20/2016] [Indexed: 05/06/2023]
Abstract
Rapid progress in disease biomarker discovery has increased the need for robust detection technologies. In the past several years, the designs of many immunoaffinity reagents have focused on lowering costs and improving specificity while also promoting stability. Antibody fragments (scFvs) have long been displayed on the surface of yeast and phage libraries for selection; however, the stable production of such fragments presents challenges that hamper their widespread use in diagnostics. Membrane and cell wall proteins similarly suffer from stability problems when solubilized from their native environment. Recently, cell envelope compositions that maintain membrane proteins in native or native-like lipid environment to improve their stability have been developed. This cell envelope composition approach has now been adapted toward stabilizing antibody fragments by retaining their native cell wall environment. A new class of immunoaffinity reagents has been developed that maintains antibody fragment attachment to yeast cell wall. Herein, we review recent strategies that incorporate cell wall fragments with functional scFvs, which are designed for easy production while maintaining specificity and stability when in use with simple detection platforms. These cell wall based antibody fragments are globular in structure, and heterogeneous in size, with fragments ranging from tens to hundreds of nanometers in size. These fragments appear to retain activity once immobilized onto biosensor surfaces for the specific and sensitive detection of pathogen antigens. They can be quickly and economically generated from a yeast display library and stored lyophilized, at room temperature, for up to a year with little effect on stability. This new format of scFvs provides stability, in a simple and low-cost manner toward the use of scFvs in biosensor applications. The production and "panning" of such antibody cell wall composites are also extremely facile, enabling the rapid adoption of stable and inexpensive affinity reagents for emerging infectious threats.
Collapse
Affiliation(s)
- Yadveer
S. Grewal
- Centre
for Personalised Nanomedicine, Australian Institute for Bioengineering
and Nanotechnology (AIBN), University of
Queensland, Brisbane, Queensland 4072, Australia
| | - Muhammad J. A. Shiddiky
- Centre
for Personalised Nanomedicine, Australian Institute for Bioengineering
and Nanotechnology (AIBN), University of
Queensland, Brisbane, Queensland 4072, Australia
| | - Stephen M. Mahler
- ARC Training Centre for Biopharmaceutical Innovation, Australian Institute for Bioengineering and Nanotechnology
(AIBN), University of Queensland, Brisbane, Queensland 4072, Australia
- School
of Chemical Engineering, University of Queensland, Brisbane, Queensland 4072, Australia
| | - Gerard A. Cangelosi
- School
of Public Health, University of Washington, Seattle, Washington 98195, United States
| | - Matt Trau
- Centre
for Personalised Nanomedicine, Australian Institute for Bioengineering
and Nanotechnology (AIBN), University of
Queensland, Brisbane, Queensland 4072, Australia
- School
of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, Queensland 4072, Australia
| |
Collapse
|
10
|
Single-domain antibody based thermally stable electrochemical immunosensor. Biosens Bioelectron 2016; 83:162-8. [PMID: 27125838 DOI: 10.1016/j.bios.2016.04.054] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 04/15/2016] [Accepted: 04/18/2016] [Indexed: 02/07/2023]
Abstract
Conventional monoclonal and polyclonal antibodies are sensitive to changes in environmental factors such as temperature, pH, humidity, etc. This limits the current cost-effective and portable electrochemical immunosensors in harsh environments. Using Ricin Chain-A, a naturally occurring toxin, as a model analyte we report fabrication of a thermally stable electrochemical immunosensor. Single-domain antibodies (sdAb) or nanobodies have been employed as recognition elements for direct detection of Ricin at temperatures great than 4°C. Immunosensor fabricated using the conventional Ricin monoclonal and polyclonal antibodies have also been demonstrated for comparison. In the case of sdAb immunosensor, Ricin was detected in a linear range of 1log(fg/mL)-1log(μg/mL) with a sensitivity of 0.07μA/log(g/mL)/cm(2) using cyclic voltammetry. The fabricated miniaturized sensors have demonstrated higher shelf life and stability at temperatures up to 40°C. Therefore these electrochemical sensors can be integrated as a part of a portable device for point-of-care immunosensing.
Collapse
|
11
|
Vaidyanathan R, Rauf S, Grewal YS, Spadafora LJ, Shiddiky MJA, Cangelosi GA, Trau M. Enhancing Protein Capture Using a Combination of Nanoyeast Single-Chain Fragment Affinity Reagents and Alternating Current Electrohydrodynamic Forces. Anal Chem 2015; 87:11673-81. [DOI: 10.1021/acs.analchem.5b02490] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Ramanathan Vaidyanathan
- Centre
for Personalized NanoMedicine, Australian Institute for Bioengineering
and Nanotechnology (AIBN), Corner College and Cooper Roads (Bldg 75), The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Sakandar Rauf
- Centre
for Personalized NanoMedicine, Australian Institute for Bioengineering
and Nanotechnology (AIBN), Corner College and Cooper Roads (Bldg 75), The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Yadveer S. Grewal
- Centre
for Personalized NanoMedicine, Australian Institute for Bioengineering
and Nanotechnology (AIBN), Corner College and Cooper Roads (Bldg 75), The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Lauren J. Spadafora
- School
of Public Health, University of Washington, Seattle, Washington 98195, United States
| | - Muhammad J. A. Shiddiky
- Centre
for Personalized NanoMedicine, Australian Institute for Bioengineering
and Nanotechnology (AIBN), Corner College and Cooper Roads (Bldg 75), The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Gerard A. Cangelosi
- School
of Public Health, University of Washington, Seattle, Washington 98195, United States
| | - Matt Trau
- Centre
for Personalized NanoMedicine, Australian Institute for Bioengineering
and Nanotechnology (AIBN), Corner College and Cooper Roads (Bldg 75), The University of Queensland, Brisbane, Queensland 4072, Australia
- School
of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland 4072, Australia
| |
Collapse
|
12
|
Zhang K, Geddie ML, Kohli N, Kornaga T, Kirpotin DB, Jiao Y, Rennard R, Drummond DC, Nielsen UB, Xu L, Lugovskoy AA. Comprehensive optimization of a single-chain variable domain antibody fragment as a targeting ligand for a cytotoxic nanoparticle. MAbs 2015; 7:42-52. [PMID: 25484041 DOI: 10.4161/19420862.2014.985933] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Antibody-targeted nanoparticles have the potential to significantly increase the therapeutic index of cytotoxic anti-cancer therapies by directing them to tumor cells. Using antibodies or their fragments requires careful engineering because multiple parameters, including affinity, internalization rate and stability, all need to be optimized. Here, we present a case study of the iterative engineering of a single chain variable fragment (scFv) for use as a targeting arm of a liposomal cytotoxic nanoparticle. We describe the effect of the orientation of variable domains, the length and composition of the interdomain protein linker that connects VH and VL, and stabilizing mutations in both the framework and complementarity-determining regions (CDRs) on the molecular properties of the scFv. We show that variable domain orientation can alter cross-reactivity to murine antigen while maintaining affinity to the human antigen. We demonstrate that tyrosine residues in the CDRs make diverse contributions to the binding affinity and biophysical properties, and that replacement of non-essential tyrosines can improve the stability and bioactivity of the scFv. Our studies demonstrate that a comprehensive engineering strategy may be required to identify a scFv with optimal characteristics for nanoparticle targeting.
Collapse
Affiliation(s)
- Kathy Zhang
- a Merrimack Pharmaceuticals, Inc. ; Cambridge , MA USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Abstract
The use of monoclonal antibodies as therapeutics requires optimizing several of their key attributes. These include binding affinity and specificity, folding stability, solubility, pharmacokinetics, effector functions, and compatibility with the attachment of additional antibody domains (bispecific antibodies) and cytotoxic drugs (antibody-drug conjugates). Addressing these and other challenges requires the use of systematic design methods that complement powerful immunization and in vitro screening methods. We review advances in designing the binding loops, scaffolds, domain interfaces, constant regions, post-translational and chemical modifications, and bispecific architectures of antibodies and fragments thereof to improve their bioactivity. We also highlight unmet challenges in antibody design that must be overcome to generate potent antibody therapeutics.
Collapse
Affiliation(s)
- Kathryn E Tiller
- Center for Biotechnology and Interdisciplinary Studies, Isermann Department of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, Troy, New York 12180;
| | - Peter M Tessier
- Center for Biotechnology and Interdisciplinary Studies, Isermann Department of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, Troy, New York 12180;
| |
Collapse
|
14
|
Kim YP, Park D, Kim JJ, Choi WJ, Lee SH, Lee SY, Kim S, Chung JM, Jeon J, Lee BD, Shin JH, Lee YI, Cho H, Lee JM, Kang HC. Effective therapeutic approach for head and neck cancer by an engineered minibody targeting the EGFR receptor. PLoS One 2014; 9:e113442. [PMID: 25438047 PMCID: PMC4249956 DOI: 10.1371/journal.pone.0113442] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Accepted: 10/23/2014] [Indexed: 11/19/2022] Open
Abstract
Cetuximab, a chimeric monoclonal antibody developed for targeting the Epidermal Growth Factor Receptor (EGFR), has been intensively used to treat cancer patients with metastatic colorectal cancer and head and neck cancer. Intact immunoglobulin G (IgG) antibody like cetuximab, however, has some limitations such as high production cost and low penetration rate from vasculature into solid tumor mass due to its large size. In attempt to overcome these limitations, we engineered cetuximab to create single chain variable fragments (scFv-CH3; Minibody) that were expressed in bacterial system. Among three engineered minibodies, we found that MI061 minibody, which is composed of the variable heavy (VH) and light (VL) region joined by an 18-residue peptide linker, displays higher solubility and better extraction properties from bacterial lysate. In addition, we validated that purified MI061 significantly interferes ligand binding to EGFR and blocks EGFR's phosphorylation. By using a protein microarray composed of 16,368 unique human proteins covering around 2,400 plasma membrane associated proteins such as receptors and channels, we also demonstrated that MI061 only recognizes the EGFR but not other proteins as compared with cetuximab. These results indicated that engineered MI061 retains both binding specificity and affinity of cetuximab for EGFR. Although it had relatively short half-life in serum, it was shown to be highly significant anti-tumor effect by inhibiting ERK pathway in A431 xenograft model. Taken together, our present study provides compelling evidence that engineered minibody is more effective and promising agent for in vivo targeting of solid tumors.
Collapse
Affiliation(s)
- Young Pil Kim
- Department of Bio-Engineering, Life Science RD Center, Sinil Pharmaceutical Co., Seongnam, South Korea
| | - Dongsun Park
- Department of Physiology, Ajou University School of Medicine, Suwon, South Korea
| | - Jae Jin Kim
- Department of Physiology, Ajou University School of Medicine, Suwon, South Korea
- Department of Biomedical Sciences, The Graduate School, Ajou University, Suwon, South Korea
| | - Won-Jae Choi
- Department of Bio-Engineering, Life Science RD Center, Sinil Pharmaceutical Co., Seongnam, South Korea
| | - Sun Hee Lee
- Department of Bio-Engineering, Life Science RD Center, Sinil Pharmaceutical Co., Seongnam, South Korea
| | - Seo Yun Lee
- Department of Physiology, Ajou University School of Medicine, Suwon, South Korea
- Department of Biomedical Sciences, The Graduate School, Ajou University, Suwon, South Korea
| | - Soyeon Kim
- Department of Physiology, Ajou University School of Medicine, Suwon, South Korea
- Department of Biomedical Sciences, The Graduate School, Ajou University, Suwon, South Korea
| | - Jee Min Chung
- Department of Physiology, Ajou University School of Medicine, Suwon, South Korea
- Department of Biomedical Sciences, The Graduate School, Ajou University, Suwon, South Korea
| | - Jinseon Jeon
- Department of Physiology, Ajou University School of Medicine, Suwon, South Korea
| | - Byoung Dae Lee
- Department of Neuroscience, Kyung Hee University School of Medicine, Seoul, South Korea
| | - Joo-Ho Shin
- Division of Pharmacology, Department of Molecular Cell Biology, Samsung Biomedical Research Institute, SungKyunKwan University School of Medicine, Suwon, South Korea
| | - Yun-il Lee
- Well Aging Research Center, Samsung Advanced Institute of Technology (SAIT), Suwon, South Korea
| | - Hyeseong Cho
- Department of Biomedical Sciences, The Graduate School, Ajou University, Suwon, South Korea
- Department of Biochemistry, Ajou University School of Medicine, Suwon, South Korea
- * E-mail: (HC); (JL); (HCK)
| | - Jeong-Min Lee
- Department of Bio-Engineering, Life Science RD Center, Sinil Pharmaceutical Co., Seongnam, South Korea
- * E-mail: (HC); (JL); (HCK)
| | - Ho Chul Kang
- Department of Physiology, Ajou University School of Medicine, Suwon, South Korea
- Department of Biomedical Sciences, The Graduate School, Ajou University, Suwon, South Korea
- * E-mail: (HC); (JL); (HCK)
| |
Collapse
|
15
|
Zhang S, Wang Y, Song X, Hong J, Zhang Y, Yao L. Improving Trichoderma reesei Cel7B Thermostability by Targeting the Weak Spots. J Chem Inf Model 2014; 54:2826-33. [DOI: 10.1021/ci500339v] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Affiliation(s)
- Shujun Zhang
- Laboratory
of Biofuels, Qingdao
Institute of Bioenergy and Bioprocess Technology Chinese Academy of Sciences, Qingdao, 266061, China
| | - Yefei Wang
- Laboratory
of Biofuels, Qingdao
Institute of Bioenergy and Bioprocess Technology Chinese Academy of Sciences, Qingdao, 266061, China
| | - Xiangfei Song
- Laboratory
of Biofuels, Qingdao
Institute of Bioenergy and Bioprocess Technology Chinese Academy of Sciences, Qingdao, 266061, China
| | - Jingbo Hong
- Laboratory
of Biofuels, Qingdao
Institute of Bioenergy and Bioprocess Technology Chinese Academy of Sciences, Qingdao, 266061, China
| | - Yu Zhang
- Laboratory
of Biofuels, Qingdao
Institute of Bioenergy and Bioprocess Technology Chinese Academy of Sciences, Qingdao, 266061, China
| | - Lishan Yao
- Laboratory
of Biofuels, Qingdao
Institute of Bioenergy and Bioprocess Technology Chinese Academy of Sciences, Qingdao, 266061, China
| |
Collapse
|
16
|
Ahmed M, Hu J, Cheung NKV. Structure Based Refinement of a Humanized Monoclonal Antibody That Targets Tumor Antigen Disialoganglioside GD2. Front Immunol 2014; 5:372. [PMID: 25177320 PMCID: PMC4132262 DOI: 10.3389/fimmu.2014.00372] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Accepted: 07/21/2014] [Indexed: 12/13/2022] Open
Abstract
Disialoganglioside GD2 is an important target on several pediatric and adult cancer types including neuroblastoma, retinoblastoma, melanoma, small-cell lung cancer, brain tumors, sarcomas, and cancer stem cells. We have utilized structural and computational methods to refine the framework of humanized monoclonal antibody 3F8, the highest affinity anti-GD2 antibody in clinical development. Two constructs (V3 and V5) were designed to enhance stability and minimize potential immunogenicity. Construct V3 contained 12 point mutations and had higher thermal stability and comparable affinity and in vitro tumor cells killing as the parental hu3F8. Construct V5 had nine point mutations to minimize potential immunogenicity, but resulted in weaker thermal stability, weaker antigen binding, and reduced tumor killing potency. When construct V3 was combined with the single point mutation HC:G54I, the resulting V3-Ile construct had enhanced stability, antigen binding, and a nearly sixfold increase in tumor cell killing. The resulting product is a lead candidate for clinical development for the treatment of GD2-positive tumors.
Collapse
Affiliation(s)
- Mahiuddin Ahmed
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jian Hu
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nai-Kong V. Cheung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
17
|
Wang S, Liu M, Zeng D, Qiu W, Ma P, Yu Y, Chang H, Sun Z. Increasing stability of antibody via antibody engineering: stability engineering on an anti-hVEGF. Proteins 2014; 82:2620-30. [PMID: 24916692 DOI: 10.1002/prot.24626] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Revised: 05/28/2014] [Accepted: 06/05/2014] [Indexed: 11/09/2022]
Abstract
Antibody stability is very important for expression, activity, specificity, and storage. This knowledge of antibody structure has made it possible for a computer-aided molecule design to be used to optimize and increase antibody stability. Many computational methods have been built based on knowledge or structure, however, a good integrated engineering system has yet to be developed that combines these methods. In the current study, we designed an integrated computer-aided engineering protocol, which included several successful methods. Mutants were designed considering factors that affected stability and multiwall filter screening was used to improve the design accuracy. Using this protocol, the thermo-stability of an anti-hVEGF antibody was significantly improved. Nearly 40% of the single-point mutants proved to be more stable than the parent antibody and most of the mutations could be stacked effectively. The T₅₀ also improved about 7°C by combinational mutation of seven sites in the light chain and three sites in the heavy chain. Data indicate that the protocol is an effective method for optimization of antibody structure, especially for improving thermo-stability. This protocol could also be used to enhance the stability of other antibodies.
Collapse
Affiliation(s)
- Shuang Wang
- Laboratory of Protein Engineering, Beijing Institute of Biotechnology, Beijing, China
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Feige MJ, Buchner J. Principles and engineering of antibody folding and assembly. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2014; 1844:2024-2031. [PMID: 24931831 DOI: 10.1016/j.bbapap.2014.06.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Revised: 06/04/2014] [Accepted: 06/06/2014] [Indexed: 11/20/2022]
Abstract
Antibodies are uniquely suited to serve essential roles in the human immune defense as they combine several specific functions in one hetero-oligomeric protein. Their constant regions activate effector functions and their variable domains provide a stable framework that allows incorporation of highly diverse loop sequences. The combination of non-germline DNA recombination and mutation together with heavy and light chain assembly allows developing variable regions that specifically recognize essentially any antigen they may encounter. However, this diversity also requires tailor-made mechanisms to guarantee that folding and association of antibodies is carefully this diversity also requires tailor-made mechanisms to guarantee that folding and association of antibodies is carefully controlled before the protein is secreted from a plasma cell. Accordingly, the generic immunoglobulin fold β-barrel structure of antibody domains has been fine-tuned during evolution to fit the different requirements. Work over the past decades has identified important aspects of the folding and assembly of antibody domains and chains revealing domain specific variations of a general scheme. The most striking is the folding of an intrinsically disordered antibody domain in the context of its partner domain as the basis for antibody assembly and its control on the molecular level in the cell. These insights have not only allowed a better understanding of the antibody folding process but also provide a wealth of opportunities for rational optimization of antibody molecules. In this review, we summarize current concepts of antibody folding and assembly and discuss how they can be utilized to engineer antibodies with improved performance for different applications. This article is part of a Special Issue entitled: Recent advances in the molecular engineering of antibodies.
Collapse
Affiliation(s)
- Matthias J Feige
- Department of Tumor Cell Biology, St. Jude Children's Research Hospital, Memphis 38105, TN, USA.
| | - Johannes Buchner
- CIPSM at the Department of Chemistry, Technische Universität München, 85748 Garching, Germany.
| |
Collapse
|
19
|
Pybus LP, James DC, Dean G, Slidel T, Hardman C, Smith A, Daramola O, Field R. Predicting the expression of recombinant monoclonal antibodies in Chinese hamster ovary cells based on sequence features of the CDR3 domain. Biotechnol Prog 2013; 30:188-97. [DOI: 10.1002/btpr.1839] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Revised: 10/27/2013] [Indexed: 11/11/2022]
Affiliation(s)
- Leon P. Pybus
- ChELSI Institute; Dept. of Chemical and Biological Engineering; University of Sheffield; Mappin Street, Sheffield S1 3JD U.K
| | - David C. James
- ChELSI Institute; Dept. of Chemical and Biological Engineering; University of Sheffield; Mappin Street, Sheffield S1 3JD U.K
| | - Greg Dean
- MedImmune Ltd.; Granta Park Cambridge CB21 6GH U.K
| | - Tim Slidel
- MedImmune Ltd.; Granta Park Cambridge CB21 6GH U.K
| | | | - Andrew Smith
- MedImmune Ltd.; Granta Park Cambridge CB21 6GH U.K
| | | | - Ray Field
- MedImmune Ltd.; Granta Park Cambridge CB21 6GH U.K
| |
Collapse
|
20
|
Yates BP, Peck MA, Berget PB. Directed evolution of a fluorogen-activating single chain antibody for function and enhanced brightness in the cytoplasm. Mol Biotechnol 2013; 54:829-41. [PMID: 23242633 DOI: 10.1007/s12033-012-9631-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Directed evolution is an exceptionally powerful tool that uses random mutant library generation and screening techniques to engineer or optimize functions of proteins. One class of proteins for which this process is particularly effective is antibodies, where properties such as antigen specificity and affinity can be selected to yield molecules with improved efficacy as molecular labels or in potential therapeutics. Typical antibody structure includes disulfide bonds that are required for stability and proper folding of the domains. However, these bonds are unable to form in the reducing environment of the cytoplasm, stymieing the effectiveness of optimized antibodies in many research applications. We have removed disulfide-forming cysteine residues in a single chain antibody fluorogen-activating protein (FAP), HL4, and employed directed evolution to select a derivative that is capable of activity in the cytoplasm. A subsequent round of directed evolution was targeted at increasing the overall brightness of the fluoromodule (FAP-fluorogen complex). Ultimately, this approach produced a novel FAP that exhibits strong activation of its cognate fluorogen in the reducing environment of the cytoplasm, significantly expanding the range of applications for which fluoromodule technology can be utilized.
Collapse
Affiliation(s)
- Bradley P Yates
- Department of Biological Sciences & Molecular Biosensor and Imaging Center, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, PA 15213, USA
| | | | | |
Collapse
|
21
|
Rodríguez-Rodríguez ER, Ledezma-Candanoza LM, Contreras-Ferrat LG, Olamendi-Portugal T, Possani LD, Becerril B, Riaño-Umbarila L. A Single Mutation in Framework 2 of the Heavy Variable Domain Improves the Properties of a Diabody and a Related Single-Chain Antibody. J Mol Biol 2012; 423:337-50. [DOI: 10.1016/j.jmb.2012.07.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Revised: 07/02/2012] [Accepted: 07/06/2012] [Indexed: 10/28/2022]
|
22
|
Kuroda D, Shirai H, Jacobson MP, Nakamura H. Computer-aided antibody design. Protein Eng Des Sel 2012; 25:507-21. [PMID: 22661385 PMCID: PMC3449398 DOI: 10.1093/protein/gzs024] [Citation(s) in RCA: 173] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2012] [Revised: 04/14/2012] [Accepted: 04/19/2012] [Indexed: 11/12/2022] Open
Abstract
Recent clinical trials using antibodies with low toxicity and high efficiency have raised expectations for the development of next-generation protein therapeutics. However, the process of obtaining therapeutic antibodies remains time consuming and empirical. This review summarizes recent progresses in the field of computer-aided antibody development mainly focusing on antibody modeling, which is divided essentially into two parts: (i) modeling the antigen-binding site, also called the complementarity determining regions (CDRs), and (ii) predicting the relative orientations of the variable heavy (V(H)) and light (V(L)) chains. Among the six CDR loops, the greatest challenge is predicting the conformation of CDR-H3, which is the most important in antigen recognition. Further computational methods could be used in drug development based on crystal structures or homology models, including antibody-antigen dockings and energy calculations with approximate potential functions. These methods should guide experimental studies to improve the affinities and physicochemical properties of antibodies. Finally, several successful examples of in silico structure-based antibody designs are reviewed. We also briefly review structure-based antigen or immunogen design, with application to rational vaccine development.
Collapse
Affiliation(s)
- Daisuke Kuroda
- Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka, Japan.
| | | | | | | |
Collapse
|