1
|
Dorogin J, Benz MA, Moore CJ, Benoit DSW, Hettiaratchi MH. Recombinant and Synthetic Affibodies Function Comparably for Modulating Protein Release. Cell Mol Bioeng 2024; 17:305-312. [PMID: 39372554 PMCID: PMC11450113 DOI: 10.1007/s12195-024-00815-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 08/16/2024] [Indexed: 10/08/2024] Open
Abstract
Purpose Affibodies are a class of versatile affinity proteins with a wide variety of therapeutic applications, ranging from contrast agents for imaging to cell-targeting therapeutics. We have identified several affibodies specific to bone morphogenetic protein-2 (BMP-2) with a range of binding affinities and demonstrated the ability to tune release rate of BMP-2 from affibody-conjugated poly(ethylene glycol) maleimide (PEG-mal) hydrogels based on affibody affinity strength. In this work, we compare the purity, structure, and activity of recombinant, bacterially-expressed BMP-2-specific affibodies with affibodies synthesized via solid-phase peptide synthesis. Methods High- and low-affinity BMP-2-specific affibodies were recombinantly expressed using BL21(DE3) E. coli and chemically synthesized using microwave-assisted solid-phase peptide synthesis with Fmoc-Gly-Wang resin. The secondary structures of the affibodies and dissociation constants of affibody-BMP-2 binding were characterized by circular dichroism and biolayer interferometry, respectively. Endotoxin levels were measured using chromogenic limulus amebocyte lysate (LAL) assays. Affibody-conjugated PEG-mal hydrogels were fabricated and loaded with BMP-2 to evaluate hydrogel capacity for controlled release, quantified by enzyme-linked immunosorbent assays (ELISA). Results Synthetic and recombinant affibodies were determined to be α-helical by circular dichroism. The synthetic high- and low-affinity BMP-2-specific affibodies demonstrated comparable BMP-2 binding dissociation constants to their recombinant counterparts. Recombinant affibodies retained some endotoxins after purification, while endotoxins were not detected in the synthetic affibodies above FDA permissible limits. High-affinity affibody-conjugated hydrogels reduced cumulative BMP-2 release compared to the low-affinity affibody-conjugated hydrogels and hydrogels without affibodies. Conclusions Synthetic affibodies demonstrate comparable structure and function to recombinant affibodies while reducing endotoxin contamination and increasing product yield, indicating that solid-phase peptide synthesis is a viable method of producing affibodies for controlled protein release and other applications.
Collapse
Affiliation(s)
- Jonathan Dorogin
- Department of Bioengineering, University of Oregon, Knight Campus, Eugene, Oregon USA
| | - Morrhyssey A. Benz
- Department of Bioengineering, University of Oregon, Knight Campus, Eugene, Oregon USA
- Department of Chemistry and Biochemistry, University of Oregon, Eugene, Oregon USA
| | - Cameron J. Moore
- Department of Bioengineering, University of Oregon, Knight Campus, Eugene, Oregon USA
| | - Danielle S. W. Benoit
- Department of Bioengineering, University of Oregon, Knight Campus, Eugene, Oregon USA
| | - Marian H. Hettiaratchi
- Department of Bioengineering, University of Oregon, Knight Campus, Eugene, Oregon USA
- Department of Chemistry and Biochemistry, University of Oregon, Eugene, Oregon USA
| |
Collapse
|
2
|
Badenhorst M, Windhorst AD, Beaino W. Navigating the landscape of PD-1/PD-L1 imaging tracers: from challenges to opportunities. Front Med (Lausanne) 2024; 11:1401515. [PMID: 38915766 PMCID: PMC11195831 DOI: 10.3389/fmed.2024.1401515] [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: 03/15/2024] [Accepted: 05/20/2024] [Indexed: 06/26/2024] Open
Abstract
Immunotherapy targeted to immune checkpoint inhibitors, such as the program cell death receptor (PD-1) and its ligand (PD-L1), has revolutionized cancer treatment. However, it is now well-known that PD-1/PD-L1 immunotherapy response is inconsistent among patients. The current challenge is to customize treatment regimens per patient, which could be possible if the PD-1/PD-L1 expression and dynamic landscape are known. With positron emission tomography (PET) imaging, it is possible to image these immune targets non-invasively and system-wide during therapy. A successful PET imaging tracer should meet specific criteria concerning target affinity, specificity, clearance rate and target-specific uptake, to name a few. The structural profile of such a tracer will define its properties and can be used to optimize tracers in development and design new ones. Currently, a range of PD-1/PD-L1-targeting PET tracers are available from different molecular categories that have shown impressive preclinical and clinical results, each with its own advantages and disadvantages. This review will provide an overview of current PET tracers targeting the PD-1/PD-L1 axis. Antibody, peptide, and antibody fragment tracers will be discussed with respect to their molecular characteristics and binding properties and ways to optimize them.
Collapse
Affiliation(s)
- Melinda Badenhorst
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Radiology and Nuclear Medicine, De Boelelaan, Amsterdam, Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, Netherlands
| | - Albert D. Windhorst
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Radiology and Nuclear Medicine, De Boelelaan, Amsterdam, Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, Netherlands
| | - Wissam Beaino
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Radiology and Nuclear Medicine, De Boelelaan, Amsterdam, Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, Netherlands
| |
Collapse
|
3
|
Blanchard PL, Knick BJ, Whelan SA, Hackel BJ. Hyperstable Synthetic Mini-Proteins as Effective Ligand Scaffolds. ACS Synth Biol 2023; 12:3608-3622. [PMID: 38010428 PMCID: PMC10822706 DOI: 10.1021/acssynbio.3c00409] [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] [Indexed: 11/29/2023]
Abstract
Small, single-domain protein scaffolds are compelling sources of molecular binding ligands with the potential for efficient physiological transport, modularity, and manufacturing. Yet, mini-proteins require a balance between biophysical robustness and diversity to enable new functions. We tested the developability and evolvability of millions of variants of 43 designed libraries of synthetic 40-amino acid βαββ proteins with diversified sheet, loop, or helix paratopes. We discovered a scaffold library that yielded hundreds of binders to seven targets while exhibiting high stability and soluble expression. Binder discovery yielded 6-122 nM affinities without affinity maturation and Tms averaging ≥78 °C. Broader βαββ libraries exhibited varied developability and evolvability. Sheet paratopes were the most consistently developable, and framework 1 was the most evolvable. Paratope evolvability was dependent on target, though several libraries were evolvable across many targets while exhibiting high stability and soluble expression. Select βαββ proteins are strong starting points for engineering performant binders.
Collapse
Affiliation(s)
- Paul L. Blanchard
- Department of Chemical Engineering and Materials Science, University of Minnesota – Twin Cities, 421 Washington Avenue SE, Minneapolis, MN 55455
| | - Brandon J. Knick
- Department of Chemical Engineering and Materials Science, University of Minnesota – Twin Cities, 421 Washington Avenue SE, Minneapolis, MN 55455
| | - Sarah A. Whelan
- Department of Chemical Engineering and Materials Science, University of Minnesota – Twin Cities, 421 Washington Avenue SE, Minneapolis, MN 55455
| | - Benjamin J. Hackel
- Department of Chemical Engineering and Materials Science, University of Minnesota – Twin Cities, 421 Washington Avenue SE, Minneapolis, MN 55455
| |
Collapse
|
4
|
Hjelm LC, Lindberg H, Ståhl S, Löfblom J. Affibody Molecules Intended for Receptor-Mediated Transcytosis via the Transferrin Receptor. Pharmaceuticals (Basel) 2023; 16:956. [PMID: 37513868 PMCID: PMC10383291 DOI: 10.3390/ph16070956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 06/27/2023] [Accepted: 06/30/2023] [Indexed: 07/30/2023] Open
Abstract
The development of biologics for diseases affecting the central nervous system has been less successful compared to other disease areas, in part due to the challenge of delivering drugs to the brain. The most well-investigated and successful strategy for increasing brain uptake of biological drugs is using receptor-mediated transcytosis over the blood-brain barrier and, in particular, targeting the transferrin receptor-1 (TfR). Here, affibody molecules are selected for TfR using phage display technology. The two most interesting candidates demonstrated binding to human TfR, cross-reactivity to the murine orthologue, non-competitive binding with human transferrin, and binding to TfR-expressing brain endothelial cell lines. Single amino acid mutagenesis of the affibody molecules revealed the binding contribution of individual residues and was used to develop second-generation variants with improved properties. The second-generation variants were further analyzed and showed an ability for transcytosis in an in vitro transwell assay. The new TfR-specific affibody molecules have the potential for the development of small brain shuttles for increasing the uptake of various compounds to the central nervous system and thus warrant further investigations.
Collapse
Affiliation(s)
- Linnea Charlotta Hjelm
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, 106 91 Stockholm, Sweden
| | - Hanna Lindberg
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, 106 91 Stockholm, Sweden
| | - Stefan Ståhl
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, 106 91 Stockholm, Sweden
| | - John Löfblom
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, 106 91 Stockholm, Sweden
| |
Collapse
|
5
|
Kamara S, Guo Y, Wen H, Liu Y, Liu L, Zheng M, Zhang J, Zhou L, Chen J, Zhu S, Zhang L. Novel Bifunctional Affibody Molecules with Specific Binding to Both EBV LMP1 and LMP2 for Targeted Therapy of Nasopharyngeal Carcinoma. Int J Mol Sci 2023; 24:10126. [PMID: 37373272 DOI: 10.3390/ijms241210126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/30/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
Antibodies are considered highly specific therapeutic agents in cancer medicines, and numerous formats have been developed. Among them, bispecific antibodies (BsAbs) have gained a lot of attention as a next-generation strategy for cancer therapy. However, poor tumor penetration is a major challenge because of their large size and thus contributes to suboptimal responses within cancer cells. On the other hand, affibody molecules are a new class of engineered affinity proteins and have achieved several promising results with their applications in molecular imaging diagnostics and targeted tumor therapy. In this study, an alternative format for bispecific molecules was constructed and investigated, named ZLMP110-277 and ZLMP277-110, that targets Epstein-Barr virus latent membrane protein 1 (LMP1) and latent membrane protein 2 (LMP2). Surface plasmon resonance (SPR), indirect immunofluorescence assay, co-immunoprecipitation, and near-infrared (NIR) imaging clearly demonstrated that ZLMP110-277 and ZLMP277-110 have good binding affinity and specificity for both LMP1 and LMP2 in vitro and in vivo. Moreover, ZLMP110-277 and ZLMP277-110, especially ZLMP277-110, significantly reduced the cell viability of C666-1 and CNE-2Z as compared to their monospecific counterparts. ZLMP110-277 and ZLMP277-110 could inhibit phosphorylation of proteins modulated by the MEK/ERK/p90RSK signaling pathway, ultimately leading to suppression of oncogene nuclear translocations. Furthermore, ZLMP110-277 and ZLMP277-110 showed significant antitumor efficacy in nasopharyngeal carcinoma-bearing nude mice. Overall, our results demonstrated that ZLMP110-277 and ZLMP277-110, especially ZLMP277-110, are promising novel prognostic indicators for molecular imaging and targeted tumor therapy of EBV-associated nasopharyngeal carcinoma.
Collapse
Affiliation(s)
- Saidu Kamara
- Institute of Molecular Virology and Immunology, Department of Microbiology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Yanru Guo
- Institute of Molecular Virology and Immunology, Department of Microbiology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - He Wen
- Institute of Molecular Virology and Immunology, Department of Microbiology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Ying Liu
- Institute of Molecular Virology and Immunology, Department of Microbiology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Lei Liu
- Institute of Molecular Virology and Immunology, Department of Microbiology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Maolin Zheng
- Institute of Molecular Virology and Immunology, Department of Microbiology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Jing Zhang
- Institute of Molecular Virology and Immunology, Department of Microbiology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Luqi Zhou
- Institute of Molecular Virology and Immunology, Department of Microbiology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Jun Chen
- Institute of Molecular Virology and Immunology, Department of Microbiology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Shanli Zhu
- Institute of Molecular Virology and Immunology, Department of Microbiology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Lifang Zhang
- Institute of Molecular Virology and Immunology, Department of Microbiology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| |
Collapse
|
6
|
Giang KA, Nygren PÅ, Nilvebrant J. Selection of Affibody Affinity Proteins from Phagemid Libraries. Methods Mol Biol 2023; 2702:373-392. [PMID: 37679630 DOI: 10.1007/978-1-0716-3381-6_19] [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] [Indexed: 09/09/2023]
Abstract
Herein, we describe a general protocol for the selection of target-binding affinity protein molecules from a phagemid-encoded library. The protocol is based on our experience with phage display selections of non-immunoglobulin affibody affinity proteins but can in principle be applied to perform biopanning experiments from any phage-displayed affinity protein library available in a similar phagemid vector. The procedure begins with an amplification of the library from frozen bacterial glycerol stocks via cultivation and helper phage superinfection, followed by a step-by-step instruction of target protein preparation, selection cycles, and post-selection analyses. The described procedures in this standard protocol are relatively conservative and rely on ordinary reagents and equipment available in most molecular biology laboratories.
Collapse
Affiliation(s)
- Kim Anh Giang
- Division of Protein Engineering, School of Chemistry, Biotechnology and Health, Royal Institute of Technology, Stockholm, Sweden
| | - Per-Åke Nygren
- Division of Protein Engineering, School of Chemistry, Biotechnology and Health, Royal Institute of Technology, Stockholm, Sweden
- Science for Life Laboratory, Solna, Sweden
| | - Johan Nilvebrant
- Division of Protein Engineering, School of Chemistry, Biotechnology and Health, Royal Institute of Technology, Stockholm, Sweden.
| |
Collapse
|
7
|
Shibasaki S, Karasaki M, Matsui K, Iwasaki T. Functional Evaluation of Anti-TNF-α Affibody Molecules in Biochemical Detection and Inhibition to Signalling Pathways of a Synovial Cell. Curr Pharm Biotechnol 2021; 22:1228-1234. [PMID: 33069194 DOI: 10.2174/1389201021666201016143730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 09/15/2020] [Accepted: 09/21/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND An affibody molecule obtained from a bioengineered staphylococcal protein was previously shown to act as an affinity binder for a wide range of targets and develop Tumour Necrosis Factor α (TNF-α)-binding clones. METHODS In this study, we demonstrated that affibody molecules against TNF-α could bind to recombinant TNF-α on the membrane for biochemical detection. In addition, we examined whether the affibody molecules could block binding between recombinant TNF-α and its receptor on MH7A synovial cells. RESULTS When a TNF-α-binding affibody was added, the production level of inflammatory mediators IL-6 and MMP-3 in MH7A were found to decrease up to 44%. Additionally, proliferation of synovial cells was also inhibited by the addition of TNF-α to cultivation media. CONCLUSION These results suggest that affibody molecules against TNF-α could be candidate molecules for the detection of TNF-α during biochemical analysis and pharmacotherapy for rheumatoid arthritis.
Collapse
Affiliation(s)
- Seiji Shibasaki
- Department of Internal Medicine, Hyogo College of Medicine, Mukogawa-cho, Nishinomiya, 663-8501, Japan
| | - Miki Karasaki
- General Education Center, Hyogo University of Health Sciences, Minatojima 1-3-6, Kobe, 650-8530, Japan
| | - Kiyoshi Matsui
- Department of Internal Medicine, Hyogo College of Medicine, Mukogawa-cho, Nishinomiya, 663-8501, Japan
| | - Tsuyoshi Iwasaki
- Department of Internal Medicine, Hyogo College of Medicine, Mukogawa-cho, Nishinomiya, 663-8501, Japan
| |
Collapse
|
8
|
Grindel B, Engel BJ, Hall CG, Kelderhouse LE, Lucci A, Zacharias NM, Takahashi TT, Millward SW. Mammalian Expression and In Situ Biotinylation of Extracellular Protein Targets for Directed Evolution. ACS OMEGA 2020; 5:25440-25455. [PMID: 33043224 PMCID: PMC7542843 DOI: 10.1021/acsomega.0c03990] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 09/08/2020] [Indexed: 05/17/2023]
Abstract
Directed evolution is a powerful tool for the selection of functional ligands from molecular libraries. Extracellular domains (ECDs) of cell surface receptors are common selection targets for therapeutic and imaging agent development. Unfortunately, these proteins are often post-translationally modified and are therefore unsuitable for expression in bacterial systems. Directional immobilization of these targets is further hampered by the absence of biorthogonal groups for site-specific chemical conjugation. We have developed a nonadherent mammalian expression system for rapid, high-yield expression of biotinylated ECDs. ECDs from EGFR, HER2, and HER3 were site-specifically biotinylated in situ and recovered from the cell culture supernatant with yields of up to 10 mg/L at >90% purity. Biotinylated ECDs also contained a protease cleavage site for rapid and selective release of the ECD after immobilization on avidin/streptavidin resins and library binding. A model mRNA display selection round was carried out against the HER2 ECD with the HER2 affibody expressed as an mRNA-protein fusion. HER2 affibody-mRNA fusions were selectively released by thrombin and quantitative PCR revealed substantial improvements in the enrichment of functional affibody-mRNA fusions relative to direct PCR amplification of the resin-bound target. This methodology allows rapid purification of high-quality targets for directed evolution and selective elution of functional sequences at the conclusion of each selection round.
Collapse
Affiliation(s)
- Brian
J. Grindel
- Department
of Cancer Systems Imaging, MD Anderson Cancer
Center, Houston, Texas 77030, United States
| | - Brian J. Engel
- Department
of Cancer Systems Imaging, MD Anderson Cancer
Center, Houston, Texas 77030, United States
| | - Carolyn G. Hall
- Department
of Breast Surgical Oncology, MD Anderson
Cancer Center, Houston, Texas 77030, United States
| | - Lindsay E. Kelderhouse
- Department
of Cancer Systems Imaging, MD Anderson Cancer
Center, Houston, Texas 77030, United States
| | - Anthony Lucci
- Department
of Breast Surgical Oncology, MD Anderson
Cancer Center, Houston, Texas 77030, United States
| | - Niki M. Zacharias
- Department
of Urology, MD Anderson Cancer Center, Houston, Texas 77030, United States
| | - Terry T. Takahashi
- Department
of Chemistry, University of Southern California, Los Angeles, California 90089, United States
| | - Steven W. Millward
- Department
of Cancer Systems Imaging, MD Anderson Cancer
Center, Houston, Texas 77030, United States
| |
Collapse
|
9
|
Barozzi A, Lavoie RA, Day KN, Prodromou R, Menegatti S. Affibody-Binding Ligands. Int J Mol Sci 2020; 21:ijms21113769. [PMID: 32471034 PMCID: PMC7312911 DOI: 10.3390/ijms21113769] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 05/21/2020] [Accepted: 05/24/2020] [Indexed: 02/03/2023] Open
Abstract
While antibodies remain established therapeutic and diagnostic tools, other protein scaffolds are emerging as effective and safer alternatives. Affibodies in particular are a new class of small proteins marketed as bio-analytic reagents. They feature tailorable binding affinity, low immunogenicity, high tissue permeation, and high expression titer in bacterial hosts. This work presents the development of affibody-binding peptides to be utilized as ligands for their purification from bacterial lysates. Affibody-binding candidates were identified by screening a peptide library simultaneously against two model affibodies (anti-immunoglobulin G (IgG) and anti-albumin) with the aim of selecting peptides targeting the conserved domain of affibodies. An ensemble of homologous sequences identified from screening was synthesized on Toyopearl® resin and evaluated via binding studies to select sequences that afford high product binding and recovery. The affibody-peptide interaction was also evaluated by in silico docking, which corroborated the targeting of the conserved domain. Ligand IGKQRI was validated through purification of an anti-ErbB2 affibody from an Escherichia coli lysate. The values of binding capacity (~5 mg affibody per mL of resin), affinity (KD ~1 μM), recovery and purity (64-71% and 86-91%), and resin lifetime (100 cycles) demonstrate that IGKQRI can be employed as ligand in affibody purification processes.
Collapse
Affiliation(s)
- Annalisa Barozzi
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC 27695-7905, USA; (A.B.); (R.A.L.); (K.N.D.); (R.P.)
| | - R. Ashton Lavoie
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC 27695-7905, USA; (A.B.); (R.A.L.); (K.N.D.); (R.P.)
| | - Kevin N. Day
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC 27695-7905, USA; (A.B.); (R.A.L.); (K.N.D.); (R.P.)
| | - Raphael Prodromou
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC 27695-7905, USA; (A.B.); (R.A.L.); (K.N.D.); (R.P.)
| | - Stefano Menegatti
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC 27695-7905, USA; (A.B.); (R.A.L.); (K.N.D.); (R.P.)
- Biomanufacturing Training and Education Center (BTEC), North Carolina State University, Raleigh, NC 27695-7905, USA
- Correspondence: ; Tel.: +1-919-753-3276
| |
Collapse
|
10
|
Myrhammar A, Rosik D, Karlström AE. Photocontrolled Reversible Binding between the Protein A-Derived Z Domain and Immunoglobulin G. Bioconjug Chem 2020; 31:622-630. [DOI: 10.1021/acs.bioconjchem.9b00786] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Anders Myrhammar
- Department of Protein Science School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology AlbaNova University Center, S−106 91 Stockholm, Sweden
| | - Daniel Rosik
- Department of Protein Science School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology AlbaNova University Center, S−106 91 Stockholm, Sweden
| | - Amelie Eriksson Karlström
- Department of Protein Science School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology AlbaNova University Center, S−106 91 Stockholm, Sweden
| |
Collapse
|
11
|
Richards DA. Exploring alternative antibody scaffolds: Antibody fragments and antibody mimics for targeted drug delivery. DRUG DISCOVERY TODAY. TECHNOLOGIES 2018; 30:35-46. [PMID: 30553519 DOI: 10.1016/j.ddtec.2018.10.005] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 10/12/2018] [Accepted: 10/15/2018] [Indexed: 05/20/2023]
Abstract
The field of targeted therapeutics has benefitted immeasurably from the development of high-affinity antibodies. These important ligands have facilitated the development of effective therapies, particularly when conjugated to potent cytotoxic payloads i.e. in antibody-drug conjugates (ADCs). The success of ADCs is evidenced by rapid adoption within the pharmaceuticals community; many major companies have dedicated ADC research programmes. However, despite the advantages, the field of ADCs has failed to live up to its full potential. Studies have emerged suggesting that traditional IgG scaffolds may not be the optimal format for targeted payload delivery. In response, the protein engineering community has begun to explore alternative high-binding protein scaffolds as antibody mimics. In this short review I will summarise the generation, modification, and application of emerging antibody fragments and synthetic antibody mimics, with a focus on their use as drug carriers. The review aims to highlight the advantages of antibody mimics, and how they could be employed to overcome the issues and limitations of traditional ADCs.
Collapse
Affiliation(s)
- Daniel A Richards
- Department of Chemistry, University College London, 20 Gordon Street, London, WC1H 0AJ, UK.
| |
Collapse
|
12
|
Nilsson A, Lindgren J, Eriksson Karlström A. Intramolecular Thioether Crosslinking to Increase the Proteolytic Stability of Affibody Molecules. Chembiochem 2017; 18:2056-2062. [PMID: 28836374 DOI: 10.1002/cbic.201700350] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Indexed: 11/12/2022]
Abstract
Protein therapeutics suffer from low oral bioavailability, mainly due to poor membrane permeability and digestion by gastrointestinal proteases. To improve proteolytic stability, intramolecular thioether crosslinks were introduced into a three-helix affibody molecule binding the human epidermal growth factor receptor (EGFR). Solid-phase peptide synthesis was used to produce an unmodified control protein domain and three different crosslinked protein domain variants: one with a thioether crosslink between the N-terminal lysine residue and a cysteine residue in the second loop region (denoted K4), a second with a crosslink between the C-terminal lysine residue and a cysteine residue in the first loop region (denoted K58), and a third with crosslinks in both positions (denoted K4K58). Circular dichroism (CD) and surface-plasmon-resonance-based (SPR-based) biosensor studies of the protein domains showed that the three-helix structure and high-affinity binding to EGFR were preserved in the crosslinked protein domains. In vitro digestion by gastrointestinal proteases demonstrated that the crosslinked protein domains showed increased stability towards pepsin and towards a combination of trypsin and chymotrypsin.
Collapse
Affiliation(s)
- Anders Nilsson
- KTH Royal Institute of Technology, School of Biotechnology, Division of Protein Technology, AlbaNova University Center, 106 91, Stockholm, Sweden
| | - Joel Lindgren
- KTH Royal Institute of Technology, School of Biotechnology, Division of Protein Technology, AlbaNova University Center, 106 91, Stockholm, Sweden
| | - Amelie Eriksson Karlström
- KTH Royal Institute of Technology, School of Biotechnology, Division of Protein Technology, AlbaNova University Center, 106 91, Stockholm, Sweden
| |
Collapse
|
13
|
Woldring DR, Holec PV, Stern LA, Du Y, Hackel BJ. A Gradient of Sitewise Diversity Promotes Evolutionary Fitness for Binder Discovery in a Three-Helix Bundle Protein Scaffold. Biochemistry 2017; 56:1656-1671. [PMID: 28248518 DOI: 10.1021/acs.biochem.6b01142] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Engineered proteins provide clinically and industrially impactful molecules and utility within fundamental research, yet inefficiencies in discovering lead variants with new desired functionality, while maintaining stability, hinder progress. Improved function, which can result from a few strategic mutations, is fundamentally separate from discovering novel function, which often requires large leaps in sequence space. While a highly diverse combinatorial library covering immense sequence space would empower protein discovery, the ability to sample only a minor subset of sequence space and the typical destabilization of random mutations preclude this strategy. A balance must be reached. At library scale, compounding several destabilizing mutations renders many variants unable to properly fold and devoid of function. Broadly searching sequence space while reducing the level of destabilization may enhance evolution. We exemplify this balance with affibody, a three-helix bundle protein scaffold. Using natural ligand data sets, stability and structural computations, and deep sequencing of thousands of binding variants, a protein library was designed on a sitewise basis with a gradient of mutational levels across 29% of the protein. In direct competition of biased and uniform libraries, both with 1 × 109 variants, for discovery of 6 × 104 ligands (5 × 103 clusters) toward seven targets, biased amino acid frequency increased ligand discovery 13 ± 3-fold. Evolutionarily favorable amino acids, both globally and site-specifically, are further elucidated. The sitewise amino acid bias aids evolutionary discovery by reducing the level of mutant destabilization as evidenced by a midpoint of denaturation (62 ± 4 °C) 15 °C higher than that of unbiased mutants (47 ± 11 °C; p < 0.001). Sitewise diversification, identified by high-throughput evolution and rational library design, improves discovery efficiency.
Collapse
Affiliation(s)
- Daniel R Woldring
- Department of Chemical Engineering and Materials Science, University of Minnesota-Twin Cities , 421 Washington Avenue Southeast, Minneapolis, Minnesota 55455, United States
| | - Patrick V Holec
- Department of Chemical Engineering and Materials Science, University of Minnesota-Twin Cities , 421 Washington Avenue Southeast, Minneapolis, Minnesota 55455, United States
| | - Lawrence A Stern
- Department of Chemical Engineering and Materials Science, University of Minnesota-Twin Cities , 421 Washington Avenue Southeast, Minneapolis, Minnesota 55455, United States
| | - Yang Du
- Molecular and Cellular Physiology, Stanford University , 279 Campus Drive, Stanford, California 94305, United States
| | - Benjamin J Hackel
- Department of Chemical Engineering and Materials Science, University of Minnesota-Twin Cities , 421 Washington Avenue Southeast, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
14
|
Leenheer D, ten Dijke P, Hipolito CJ. A current perspective on applications of macrocyclic-peptide-based high-affinity ligands. Biopolymers 2016; 106:889-900. [PMID: 27352774 PMCID: PMC5132055 DOI: 10.1002/bip.22900] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Revised: 05/24/2016] [Accepted: 05/31/2016] [Indexed: 01/05/2023]
Abstract
Monoclonal antibodies can bind with high affinity and high selectivity to their targets. As a tool in therapeutics or diagnostics, however, their large size (∼150 kDa) reduces penetration into tissue and prevents passive cellular uptake. To overcome these and other problems, minimized protein scaffolds have been chosen or engineered, with care taken to not compromise binding affinity or specificity. An alternate approach is to begin with a minimal non-antibody scaffold and select functional ligands from a de novo library. We will discuss the structure, production, applications, strengths, and weaknesses of several classes of antibody-derived ligands, that is, antibodies, intrabodies, and nanobodies, and nonantibody-derived ligands, that is, monobodies, affibodies, and macrocyclic peptides. In particular, this review is focussed on macrocyclic peptides produced by the Random non-standard Peptides Integrated Discovery (RaPID) system that are small in size (typically ∼2 kDa), but are able to perform tasks typically handled by larger proteinaceous ligands.
Collapse
Affiliation(s)
- Daniël Leenheer
- Ph.D. Program in Human Biology, School of Integrative and Global MajorsUniversity of TsukubaTsukubaIbarakiJapan
| | - Peter ten Dijke
- Leiden University Medical Center, Department of Molecular Cell BiologyLeidenSouth HollandThe Netherlands
- Cancer Signaling, Graduate School of Comprehensive Human Sciences and Faculty of Medicine, University of TsukubaTsukubaIbarakiJapan
| | - Christopher John Hipolito
- Cancer Signaling, Graduate School of Comprehensive Human Sciences and Faculty of Medicine, University of TsukubaTsukubaIbarakiJapan
| |
Collapse
|
15
|
Zhang T, Niu X, Yuan T, Tessari M, de Vries MP, Permentier HP, Bischoff R. Efficient and Selective Chemical Labeling of Electrochemically Generated Peptides Based on Spirolactone Chemistry. Anal Chem 2016; 88:6465-71. [PMID: 27247048 DOI: 10.1021/acs.analchem.6b01154] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Specific digestion of proteins is an essential step for mass spectrometry-based proteomics, and the chemical labeling of the resulting peptides is often used for peptide enrichment or the introduction of desirable tags. Cleavage of the peptide bond following electrochemical oxidation of Tyr or Trp results in a spirolactone moiety at the newly formed C-terminus offering a handle for chemical labeling. In this work, we developed a highly efficient and selective chemical labeling approach based on spirolactone chemistry. Electrochemically generated peptide-spirolactones readily undergo an intramolecular rearrangement yielding isomeric diketopiperazines precluding further chemical labeling. A strategy was established to prevent intramolecular arrangement by acetylating the N-terminal amino group prior to electrochemical oxidation and cleavage allowing the complete and selective chemical labeling of the tripeptide LWL and the decapeptide ACTH 1-10 with amine-containing reagents. As examples, we show the successful introduction of a fluorescent label and biotin for detection or affinity enrichment. Electrochemical digestion of peptides and proteins followed by efficient chemical labeling constitutes a new, powerful tool in protein chemistry and protein analysis.
Collapse
Affiliation(s)
- Tao Zhang
- Analytical Biochemistry and Interfaculty Mass Spectrometry Center, Department of Pharmacy, University of Groningen , A. Deusinglaan 1, 9713 AV, Groningen, The Netherlands
| | - Xiaoyu Niu
- Analytical Biochemistry and Interfaculty Mass Spectrometry Center, Department of Pharmacy, University of Groningen , A. Deusinglaan 1, 9713 AV, Groningen, The Netherlands
| | - Tao Yuan
- Analytical Biochemistry and Interfaculty Mass Spectrometry Center, Department of Pharmacy, University of Groningen , A. Deusinglaan 1, 9713 AV, Groningen, The Netherlands
| | - Marco Tessari
- Institute for Molecules and Materials, Radboud University , Heyendaalseweg 135, 6525 AJ, Nijmegen, The Netherlands
| | - Marcel P de Vries
- Department of Pediatrics, University Medical Center Groningen, University of Groningen , Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Hjalmar P Permentier
- Analytical Biochemistry and Interfaculty Mass Spectrometry Center, Department of Pharmacy, University of Groningen , A. Deusinglaan 1, 9713 AV, Groningen, The Netherlands
| | - Rainer Bischoff
- Analytical Biochemistry and Interfaculty Mass Spectrometry Center, Department of Pharmacy, University of Groningen , A. Deusinglaan 1, 9713 AV, Groningen, The Netherlands
| |
Collapse
|
16
|
Westerlund K, Honarvar H, Norrström E, Strand J, Mitran B, Orlova A, Eriksson Karlström A, Tolmachev V. Increasing the Net Negative Charge by Replacement of DOTA Chelator with DOTAGA Improves the Biodistribution of Radiolabeled Second-Generation Synthetic Affibody Molecules. Mol Pharm 2016; 13:1668-78. [PMID: 27010700 DOI: 10.1021/acs.molpharmaceut.6b00089] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
A promising strategy to enable patient stratification for targeted therapies is to monitor the target expression in a tumor by radionuclide molecular imaging. Affibody molecules (7 kDa) are nonimmunoglobulin scaffold proteins with a 25-fold smaller size than intact antibodies. They have shown an apparent potential as molecular imaging probes both in preclinical and clinical studies. Earlier, we found that hepatic uptake can be reduced by the incorporation of negatively charged purification tags at the N-terminus of Affibody molecules. We hypothesized that liver uptake might similarly be reduced by positioning the chelator at the N-terminus, where the chelator-radionuclide complex will provide negative charges. To test this hypothesis, a second generation synthetic anti-HER2 ZHER2:2891 Affibody molecule was synthesized and labeled with (111)In and (68)Ga using DOTAGA and DOTA chelators. The chelators were manually coupled to the N-terminus of ZHER2:2891 forming an amide bond. Labeling DOTAGA-ZHER2:2891 and DOTA-ZHER2:2891 with (68)Ga and (111)In resulted in stable radioconjugates. The tumor-targeting and biodistribution properties of the (111)In- and (68)Ga-labeled conjugates were compared in SKOV-3 tumor-bearing nude mice at 2 h postinjection. The HER2-specific binding of the radioconjugates was verified both in vitro and in vivo. Using the DOTAGA chelator gave significantly lower radioactivity in liver and blood for both radionuclides. The (111)In-labeled conjugates showed more rapid blood clearance than the (68)Ga-labeled conjugates. The most pronounced influence of the chelators was found when they were labeled with (68)Ga. The DOTAGA chelator gave significantly higher tumor-to-blood (61 ± 6 vs 23 ± 5, p < 0.05) and tumor-to-liver (10.4 ± 0.6 vs 4.5 ± 0.5, p < 0.05) ratios than the DOTA chelator. This study demonstrated that chelators may be used to alter the uptake of Affibody molecules, and most likely other scaffold-based imaging probes, for improvement of imaging contrast.
Collapse
Affiliation(s)
- Kristina Westerlund
- Division of Protein Technology, School of Biotechnology, KTH Royal Institute of Technology, AlbaNova University Center , 106 91 Stockholm, Sweden
| | - Hadis Honarvar
- Institute for Immunology, Genetics and Pathology, Uppsala University , 751 05 Uppsala, Sweden
| | - Emily Norrström
- Division of Protein Technology, School of Biotechnology, KTH Royal Institute of Technology, AlbaNova University Center , 106 91 Stockholm, Sweden
| | - Joanna Strand
- Institute for Immunology, Genetics and Pathology, Uppsala University , 751 05 Uppsala, Sweden
| | - Bogdan Mitran
- Preclinical PET Platform, Department of Medicinal Chemistry, Uppsala University , 751 05 Uppsala, Sweden
| | - Anna Orlova
- Preclinical PET Platform, Department of Medicinal Chemistry, Uppsala University , 751 05 Uppsala, Sweden
| | - Amelie Eriksson Karlström
- Division of Protein Technology, School of Biotechnology, KTH Royal Institute of Technology, AlbaNova University Center , 106 91 Stockholm, Sweden
| | - Vladimir Tolmachev
- Institute for Immunology, Genetics and Pathology, Uppsala University , 751 05 Uppsala, Sweden
| |
Collapse
|
17
|
Non-immunoglobulin scaffolds: a focus on their targets. Trends Biotechnol 2015; 33:408-18. [DOI: 10.1016/j.tibtech.2015.03.012] [Citation(s) in RCA: 137] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Revised: 03/28/2015] [Accepted: 03/30/2015] [Indexed: 12/21/2022]
|
18
|
|
19
|
Wållberg H, Ståhl S. Design and evaluation of radiolabeled tracers for tumor imaging. Biotechnol Appl Biochem 2014; 60:365-83. [PMID: 24033592 DOI: 10.1002/bab.1111] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Accepted: 02/20/2013] [Indexed: 12/22/2022]
Abstract
The growing understanding of tumor biology and the identification of tumor-specific genetic and molecular alterations, such as the overexpression of membrane receptors and other proteins, allows for personalization of patient management using targeted therapies. However, this puts stringent demands on the diagnostic tools used to identify patients who are likely to respond to a particular treatment. Radionuclide molecular imaging is a promising noninvasive method to visualize and characterize the expression of such targets. A number of different proteins, from full-length antibodies and their derivatives to small scaffold proteins and peptide receptor-ligands, have been applied to molecular imaging, each demonstrating strengths and weaknesses. Here, we discuss the concept of molecular targeting and, in particular, molecular imaging of cancer-associated targets. Additionally, we describe important biotechnological considerations and desired features when designing and developing tracers for radionuclide molecular imaging.
Collapse
Affiliation(s)
- Helena Wållberg
- Division of Molecular Biotechnology, School of Biotechnology, AlbaNova University Center, KTH Royal Institute of Technology, Stockholm, Sweden
| | | |
Collapse
|
20
|
Perols A, Honarvar H, Strand J, Selvaraju R, Orlova A, Karlström AE, Tolmachev V. Influence of DOTA chelator position on biodistribution and targeting properties of (111)In-labeled synthetic anti-HER2 affibody molecules. Bioconjug Chem 2012; 23:1661-70. [PMID: 22768790 DOI: 10.1021/bc3002369] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Affibody molecules are a class of affinity proteins. Their small size (7 kDa) in combination with the high (subnanomolar) affinity for a number of cancer-associated molecular targets makes them suitable for molecular imaging. Earlier studies demonstrated that the selection of radionuclide and chelator may substantially influence the tumor-targeting properties of affibody molecules. Moreover, the placement of chelators for labeling of affibody molecules with (99m)Tc at different positions in affibody molecules influenced both blood clearance rate and uptake in healthy tissues. This introduces an opportunity to improve the contrast of affibody-mediated imaging. In this comparative study, 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA) was conjugated to the synthetic affibody molecule Z(HER2:S1) at three different positions: DOTA-A1-Z(HER2:S1) (N-terminus), DOTA-K58-Z(HER2:S1) (C-terminus), and DOTA-K50-Z(HER2:S1) (middle of helix 3). The affinity for HER2 differed slightly among the variants and the K(D) values were determined to be 133 pM, 107 pM and 94 pM for DOTA-A1-Z(HER2:S1), DOTA-K50-Z(HER2:S1), and DOTA-K58-Z(HER2:S1), respectively. Z(HER2:S1)-K50-DOTA showed a slightly lower melting point (57 °C) compared to DOTA-A1-Z(HER2:S1) (64 °C) and DOTA-K58-Z(HER2:S1) (62 °C), but all variants showed good refolding properties after heat treatment. All conjugates were successfully labeled with (111)In resulting in a radiochemical yield of 99% with preserved binding capacity. In vitro specificity studies using SKOV-3 and LS174T cell lines showed that the binding of the radiolabeled compounds was HER2 receptor-mediated, which also was verified in vivo using BALB/C nu/nu mice with LS174T and Ramos lymphoma xenografts. The three conjugates all showed specific uptake in LS174T xenografts in nude mice, where DOTA-A1-Z(HER2:S1)and DOTA-K58-Z(HER2:S1) showed the highest uptake. Overall, DOTA-K58-Z(HER2:S1) provided the highest tumor-to-blood ratio, which is important for a high-contrast imaging. In conclusion, the positioning of the DOTA chelator influences the cellular processing and the biodistribution pattern of radiolabeled affibody molecules, creating preconditions for imaging optimization.
Collapse
Affiliation(s)
- Anna Perols
- Division of Molecular Biotechnology, School of Biotechnology, KTH Royal Institute of Technology, Stockholm, Sweden
| | | | | | | | | | | | | |
Collapse
|
21
|
A general route to xyloglucan-peptide conjugates for the activation of cellulose surfaces. Carbohydr Res 2012; 354:116-20. [DOI: 10.1016/j.carres.2012.03.038] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Revised: 03/29/2012] [Accepted: 03/30/2012] [Indexed: 11/18/2022]
|
22
|
Lindgren J, Ekblad C, Abrahmsén L, Eriksson Karlström A. A Native Chemical Ligation Approach for Combinatorial Assembly of Affibody Molecules. Chembiochem 2012; 13:1024-31. [DOI: 10.1002/cbic.201200052] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
23
|
Mascini M, Palchetti I, Tombelli S. Nucleic acid and peptide aptamers: fundamentals and bioanalytical aspects. Angew Chem Int Ed Engl 2011; 51:1316-32. [PMID: 22213382 DOI: 10.1002/anie.201006630] [Citation(s) in RCA: 244] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2010] [Indexed: 12/11/2022]
Abstract
In recent years new nucleic acid and protein-based combinatorial molecules have attracted the attention of researchers working in various areas of science, ranging from medicine to analytical chemistry. These molecules, called aptamers, have been proposed as alternatives to antibodies in many different applications. The aim of this Review is to illustrate the peculiarities of these combinatorial molecules which have initially been explored for their importance in molecular medicine, but have enormous potential in other biotechnological fields historically dominated by antibodies, such as bioassays. A description of these molecules is given, and the methods for their selection and production are also summarized. Moreover, critical aspects related to these molecules are discussed.
Collapse
Affiliation(s)
- Marco Mascini
- Dipartimento di Chimica Ugo Schiff, Università degli Studi di Firenze, Via della Lastruccia 3, 50019 Sesto Fiorentino, Italy.
| | | | | |
Collapse
|
24
|
Mascini M, Palchetti I, Tombelli S. Nucleinsäure- und Peptidaptamere: Grundlagen und bioanalytische Aspekte. Angew Chem Int Ed Engl 2011. [DOI: 10.1002/ange.201006630] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
25
|
Malmberg J, Perols A, Varasteh Z, Altai M, Braun A, Sandström M, Garske U, Tolmachev V, Orlova A, Eriksson Karlström A. Comparative evaluation of synthetic anti-HER2 Affibody molecules site-specifically labelled with 111In using N-terminal DOTA, NOTA and NODAGA chelators in mice bearing prostate cancer xenografts. Eur J Nucl Med Mol Imaging 2011; 39:481-92. [DOI: 10.1007/s00259-011-1992-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2011] [Accepted: 11/02/2011] [Indexed: 11/29/2022]
|
26
|
Konrad A, Karlström AE, Hober S. Covalent immunoglobulin labeling through a photoactivable synthetic Z domain. Bioconjug Chem 2011; 22:2395-403. [PMID: 22026370 DOI: 10.1021/bc200052h] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Traditionally, labeling of antibodies has been performed by covalent conjugation to amine or carboxyl groups. These methods are efficient but suffer from nonspecificity, since all free and available amine/carboxyl groups have the possibility to react. This drawback may lead to uncontrolled levels and locations of the labeling. Hence, the labeled molecules might behave differently and, possibly, the binding site of the antibody will also be affected. In this project, we have developed a highly stringent method for labeling of antibodies by utilizing an immunoglobulin-binding domain from protein A, the Z domain. Domain Z has been synthesized with an amino acid analogue, benzoylphenylalanine, capable of forming covalent attachment to other amino acids upon UV-exposure. This feature has been used for directed labeling of immunoglobulins and subsequent use of these in different assays.
Collapse
Affiliation(s)
- Anna Konrad
- Division of Proteomics, AlbaNova University Center, Stockholm, Sweden
| | | | | |
Collapse
|
27
|
Lin X, Xie J, Chen X. Protein-based tumor molecular imaging probes. Amino Acids 2011; 41:1013-36. [PMID: 20232092 PMCID: PMC3617487 DOI: 10.1007/s00726-010-0545-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2010] [Accepted: 02/24/2010] [Indexed: 12/30/2022]
Abstract
Molecular imaging is an emerging discipline which plays critical roles in diagnosis and therapeutics. It visualizes and quantifies markers that are aberrantly expressed during the disease origin and development. Protein molecules remain to be one major class of imaging probes, and the option has been widely diversified due to the recent advances in protein engineering techniques. Antibodies are part of the immunosystem which interact with target antigens with high specificity and affinity. They have long been investigated as imaging probes and were coupled with imaging motifs such as radioisotopes for that purpose. However, the relatively large size of antibodies leads to a half-life that is too long for common imaging purposes. Besides, it may also cause a poor tissue penetration rate and thus compromise some medical applications. It is under this context that various engineered protein probes, essentially antibody fragments, protein scaffolds, and natural ligands have been developed. Compared to intact antibodies, they possess more compact size, shorter clearance time, and better tumor penetration. One major challenge of using protein probes in molecular imaging is the affected biological activity resulted from random labeling. Site-specific modification, however, allows conjugation happening in a stoichiometric fashion with little perturbation of protein activity. The present review will discuss protein-based probes with focus on their application and related site-specific conjugation strategies in tumor imaging.
Collapse
Affiliation(s)
- Xin Lin
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | |
Collapse
|
28
|
Miao Z, Levi J, Cheng Z. Protein scaffold-based molecular probes for cancer molecular imaging. Amino Acids 2011; 41:1037-47. [PMID: 20174842 PMCID: PMC2914822 DOI: 10.1007/s00726-010-0503-9] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2009] [Accepted: 01/25/2010] [Indexed: 01/18/2023]
Abstract
Protein scaffold molecules are powerful reagents for targeting various cell signal receptors, enzymes, cytokines and other cancer-related molecules. They belong to the peptide and small protein platform with distinct properties. For the purpose of development of new generation molecular probes, various protein scaffold molecules have been labeled with imaging moieties and evaluated both in vitro and in vivo. Among the evaluated probes Affibody molecules and analogs, cystine knot peptides, and nanobodies have shown especially good characteristics as protein scaffold platforms for development of in vivo molecular probes. Quantitative data obtained from positron emission tomography, single photon emission computed tomography/CT, and optical imaging together with biodistribution studies have shown high tumor uptakes and high tumor-to-blood ratios for these probes. High tumor contrast imaging has been obtained within 1 h after injection. The success of those molecular probes demonstrates the adequacy of protein scaffold strategy as a general approach in molecular probe development.
Collapse
Affiliation(s)
- Zheng Miao
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, CA 94305-5344, USA
| | | | | |
Collapse
|
29
|
Affiliation(s)
- Per-Ake Nygren
- Division of Molecular Biotechnology, School of Biotechnology, Royal Institute of Technology (KTH), SE-106 91 Stockholm, Sweden
| |
Collapse
|
30
|
Nilvebrant J, Alm T, Hober S, Löfblom J. Engineering bispecificity into a single albumin-binding domain. PLoS One 2011; 6:e25791. [PMID: 21991353 PMCID: PMC3185003 DOI: 10.1371/journal.pone.0025791] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2011] [Accepted: 09/11/2011] [Indexed: 01/13/2023] Open
Abstract
Bispecific antibodies as well as non-immunoglobulin based bispecific affinity proteins are considered to have a very high potential in future biotherapeutic applications. In this study, we report on a novel approach for generation of extremely small bispecific proteins comprised of only a single structural domain. Binding to tumor necrosis factor-α (TNF-α) was engineered into an albumin-binding domain while still retaining the original affinity for albumin, resulting in a bispecific protein composed of merely 46 amino acids. By diversification of the non albumin-binding side of the three-helix bundle domain, followed by display of the resulting library on phage particles, bispecific single-domain proteins were isolated using selections with TNF-α as target. Moreover, based on the obtained sequences from the phage selection, a second-generation library was designed in order to further increase the affinity of the bispecific candidates. Staphylococcal surface display was employed for the affinity maturation, enabling efficient isolation of improved binders as well as multiparameter-based sortings with both TNF-α and albumin as targets in the same selection cycle. Isolated variants were sequenced and the binding to albumin and TNF-α was analyzed. This analysis revealed an affinity for TNF-α below 5 nM for the strongest binders. From the multiparameter sorting that simultaneously targeted TNF-α and albumin, several bispecific candidates were isolated with high affinity to both antigens, suggesting that cell display in combination with fluorescence activated cell sorting is a suitable technology for engineering of bispecificity. To our knowledge, the new binders represent the smallest engineered bispecific proteins reported so far. Possibilities and challenges as well as potential future applications of this novel strategy are discussed.
Collapse
Affiliation(s)
- Johan Nilvebrant
- Department of Proteomics, School of Biotechnology, Royal Institute of Technology, AlbaNova University Center, Stockholm, Sweden
| | | | | | | |
Collapse
|
31
|
Petrovskaya LE, Shingarova LN, Dolgikh DA, Kirpichnikov MP. Alternative scaffold proteins. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2011; 37:581-91. [DOI: 10.1134/s1068162011050141] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
32
|
Application of the logic of cysteine-free native chemical ligation to the synthesis of Human Parathyroid Hormone (hPTH). Proc Natl Acad Sci U S A 2011; 108:5986-9. [PMID: 21444787 DOI: 10.1073/pnas.1103118108] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The power of chemical synthesis of large cysteine-free polypeptides has been significantly enhanced through the use of nonproteogenic constructs which bear strategically placed thiol groups, enabling native chemical ligation. Central to these much expanded capabilities is the specific, radical-induced, metal-free dethiolation, which can be accomplished in aqueous medium.
Collapse
|
33
|
Zoller F, Haberkorn U, Mier W. Miniproteins as phage display-scaffolds for clinical applications. Molecules 2011; 16:2467-85. [PMID: 21407148 PMCID: PMC6259850 DOI: 10.3390/molecules16032467] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2010] [Revised: 03/04/2011] [Accepted: 03/07/2011] [Indexed: 11/16/2022] Open
Abstract
Miniproteins are currently developed as alternative, non-immunoglobin proteins for the generation of novel binding motifs. Miniproteins are rigid scaffolds that are stabilised by alpha-helices, beta-sheets and disulfide-constrained secondary structural elements. They are tolerant to multiple amino acid substitutions, which allow for the integration of a randomised affinity function into the stably folded framework. These properties classify miniprotein scaffolds as promising tools for lead structure generation using phage display technologies. Owing to their high enzymatic resistance and structural stability, miniproteins are ideal templates to display binding epitopes for medical applications in vivo. This review summarises the characteristics and the engineering of miniproteins as a novel class of scaffolds to generate of alternative binding agents using phage display screening. Moreover, recent developments for therapeutic and especially diagnostic applications of miniproteins are reviewed.
Collapse
Affiliation(s)
- Frederic Zoller
- Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center, INF 280, 69120 Heidelberg, Germany; E-Mails: (F.Z.); (U.H.)
- Department of Nuclear Medicine, University Hospital Heidelberg, INF 400, 69120 Heidelberg, Germany
| | - Uwe Haberkorn
- Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center, INF 280, 69120 Heidelberg, Germany; E-Mails: (F.Z.); (U.H.)
- Department of Nuclear Medicine, University Hospital Heidelberg, INF 400, 69120 Heidelberg, Germany
| | - Walter Mier
- Department of Nuclear Medicine, University Hospital Heidelberg, INF 400, 69120 Heidelberg, Germany
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +49-6221-56-7720; Fax: +49-6221-56-5473
| |
Collapse
|
34
|
Lindgren J, Wahlström A, Danielsson J, Markova N, Ekblad C, Gräslund A, Abrahmsén L, Karlström AE, Wärmländer SKTS. N-terminal engineering of amyloid-β-binding Affibody molecules yields improved chemical synthesis and higher binding affinity. Protein Sci 2011; 19:2319-29. [PMID: 20886513 DOI: 10.1002/pro.511] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The aggregation of amyloid-β (Aβ) peptides is believed to be a major factor in the onset and progression of Alzheimer's disease. Molecules binding with high affinity and selectivity to Aβ-peptides are important tools for investigating the aggregation process. An Aβ-binding Affibody molecule, ZAβ3 , has earlier been selected by phage display and shown to bind Aβ(1-40) with nanomolar affinity and to inhibit Aβ-peptide aggregation. In this study, we create truncated functional versions of the ZAβ3 Affibody molecule better suited for chemical synthesis production. Engineered Affibody molecules of different length were produced by solid phase peptide synthesis and allowed to form covalently linked homodimers by S-S-bridges. The N-terminally truncated Affibody molecules ZAβ3 (12-58), ZAβ3 (15-58), and ZAβ3 (18-58) were produced in considerably higher synthetic yield than the corresponding full-length molecule ZAβ3 (1-58). Circular dichroism spectroscopy and surface plasmon resonance-based biosensor analysis showed that the shortest Affibody molecule, ZAβ3 (18-58), exhibited complete loss of binding to the Aβ(1-40)-peptide, while the ZAβ3 (12-58) and ZAβ3 (15-58) Affibody molecules both displayed approximately one order of magnitude higher binding affinity to the Aβ(1-40)-peptide compared to the full-length Affibody molecule. Nuclear magnetic resonance spectroscopy showed that the structure of Aβ(1-40) in complex with the truncated Affibody dimers is very similar to the previously published solution structure of the Aβ(1-40)-peptide in complex with the full-length ZAβ3 Affibody molecule. This indicates that the N-terminally truncated Affibody molecules ZAβ3 (12-58) and ZAβ3 (15-58) are highly promising for further engineering and future use as binding agents to monomeric Aβ(1-40).
Collapse
Affiliation(s)
- Joel Lindgren
- Division of Molecular Biotechnology, School of Biotechnology, Royal Institute of Technology (KTH), AlbaNova University Centre, 106 91 Stockholm, Sweden
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Miao Z, Ren G, Liu H, Jiang L, Cheng Z. Cy5.5-labeled Affibody molecule for near-infrared fluorescent optical imaging of epidermal growth factor receptor positive tumors. JOURNAL OF BIOMEDICAL OPTICS 2010; 15:036007. [PMID: 20615009 DOI: 10.1117/1.3432738] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
Affibody protein is an engineered protein scaffold with a three-helical bundle structure. Affibody molecules of small size (7 kD) have great potential for targeting overexpressed cancer biomarkers in vivo. To develop an Affibody-based molecular probe for in vivo optical imaging of epidermal growth factor receptor (EGFR) positive tumors, an anti-EGFR Affibody molecule, Ac-Cys-Z(EGFR:1907) (7 kD), is site-specifically conjugated with a near-IR fluorescence dye, Cy5.5-mono-maleimide. Using fluorescent microscopy, the binding specificity of the probe Cy5.5-Z(EGFR:1907) is checked by a high-EGFR-expressing A431 cell and low-EGFR-expressing MCF7 cells. The binding affinity of Cy5.5-Z(EGFR:1907) (K(D)) to EGFR is 43.6+/-8.4 nM, as determined by flow cytometry. For an in vivo imaging study, the probe shows fast tumor targeting and good tumor contrast as early as 0.5 h postinjection (p.i.) for A431 tumors, while MCF7 tumors are barely visible. An ex vivo imaging study also demonstrates that Cy5.5-Z(EGFR:1907) has high tumor, liver, and kidney uptakes at 24 h p.i.. In conclusion, Cy5.5-Z(EGFR:1907) shows good affinity and high specificity to the EGFR. There is rapid achievement of good tumor-to-normal-tissue contrasts of Cy5.5-Z(EGFR:1907), thus demonstrating its potential for EGFR-targeted molecular imaging of cancers.
Collapse
Affiliation(s)
- Zheng Miao
- Stanford University, Stanford Cancer Center, Molecular Imaging Program at Stanford, Department of Radiology and Bio-X Program, 1201 Welch Road, Lucas Expansion, P095, Stanford, California 94305-5344, USA
| | | | | | | | | |
Collapse
|
36
|
Löfblom J, Feldwisch J, Tolmachev V, Carlsson J, Ståhl S, Frejd F. Affibody molecules: Engineered proteins for therapeutic, diagnostic and biotechnological applications. FEBS Lett 2010; 584:2670-80. [DOI: 10.1016/j.febslet.2010.04.014] [Citation(s) in RCA: 406] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2010] [Revised: 04/06/2010] [Accepted: 04/08/2010] [Indexed: 01/28/2023]
|
37
|
Feldwisch J, Tolmachev V, Lendel C, Herne N, Sjöberg A, Larsson B, Rosik D, Lindqvist E, Fant G, Höidén-Guthenberg I, Galli J, Jonasson P, Abrahmsén L. Design of an optimized scaffold for affibody molecules. J Mol Biol 2010; 398:232-47. [PMID: 20226194 DOI: 10.1016/j.jmb.2010.03.002] [Citation(s) in RCA: 118] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2009] [Revised: 02/26/2010] [Accepted: 03/03/2010] [Indexed: 12/15/2022]
Abstract
Affibody molecules are non-immunoglobulin-derived affinity proteins based on a three-helical bundle protein domain. Here, we describe the design process of an optimized Affibody molecule scaffold with improved properties and a surface distinctly different from that of the parental scaffold. The improvement was achieved by applying an iterative process of amino acid substitutions in the context of the human epidermal growth factor receptor 2 (HER2)-specific Affibody molecule Z(HER2:342). Replacements in the N-terminal region, loop 1, helix 2 and helix 3 were guided by extensive structural modeling using the available structures of the parent Z domain and Affibody molecules. The effect of several single substitutions was analyzed followed by combination of up to 11 different substitutions. The two amino acid substitutions N23T and S33K accounted for the most dramatic improvements, including increased thermal stability with elevated melting temperatures of up to +12 degrees C. The optimized scaffold contains 11 amino acid substitutions in the nonbinding surface and is characterized by improved thermal and chemical stability, as well as increased hydrophilicity, and enables generation of identical Affibody molecules both by chemical peptide synthesis and by recombinant bacterial expression. A HER2-specific Affibody tracer, [MMA-DOTA-Cys61]-Z(HER2:2891)-Cys (ABY-025), was produced by conjugating MMA-DOTA (maleimide-monoamide-1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid) to the peptide produced either chemically or in Escherichia coli. ABY-025 showed high affinity and specificity for HER2 (equilibrium dissociation constant, K(D), of 76 pM) and detected HER2 in tissue sections of SKOV-3 xenograft and human breast tumors. The HER2-binding capacity was fully retained after three cycles of heating to 90 degrees C followed by cooling to room temperature. Furthermore, the binding surfaces of five Affibody molecules targeting other proteins (tumor necrosis factor alpha, insulin, Taq polymerase, epidermal growth factor receptor or platelet-derived growth factor receptor beta) were grafted onto the optimized scaffold, resulting in molecules with improved thermal stability and a more hydrophilic nonbinding surface.
Collapse
|
38
|
Haack M, Beck-Sickinger AG. Towards understanding the free and receptor bound conformation of neuropeptide Y by fluorescence resonance energy transfer studies. Chem Biol Drug Des 2009; 73:573-83. [PMID: 19635049 DOI: 10.1111/j.1747-0285.2009.00823.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Despite a considerable sequence identity of the three mammalian hormones of the neuropeptide Y family, namely neuropeptide Y, peptide YY and pancreatic polypeptide, their structure in solution is described to be different. A so-called pancreatic polypeptide-fold has been identified for pancreatic polypeptide, whereas the structure of the N-terminal segment of neuropeptide Y is unknown. This element is important for the binding of neuropeptide Y to two of its relevant receptors, Y(1) and Y(5), but not to the Y(2) receptor subtype. In this study now, three doubly fluorescent-labeled analogs of neuropeptide Y have been synthesized that still bind to the Y(5) receptor with high affinity to investigate the conformation in solution and, for the first time, to probe the conformational changes upon binding of the ligand to its receptor in cell membrane preparations. The results obtained from the fluorescence resonance energy transfer investigations clearly show considerable differences in transfer efficiency that depend both on the solvent as well as on the peptide concentration. However, the studies do not support a pancreatic polypeptide-like folding of neuropeptide Y in the presence of membranes that express the human Y(5) receptor subtype.
Collapse
Affiliation(s)
- Michael Haack
- Institute of Biochemistry, Faculty of Bioscience, Pharmacy and Psychology, Leipzig University, Germany
| | | |
Collapse
|
39
|
Orlova A, Tran TA, Ekblad T, Karlström AE, Tolmachev V. 186Re-maSGS-ZHER2:342, a potential Affibody conjugate for systemic therapy of HER2-expressing tumours. Eur J Nucl Med Mol Imaging 2009; 37:260-9. [DOI: 10.1007/s00259-009-1268-9] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2009] [Accepted: 08/17/2009] [Indexed: 12/23/2022]
|
40
|
Ekblad T, Orlova A, Feldwisch J, Wennborg A, Karlström AE, Tolmachev V. Positioning of 99mTc-chelators influences radiolabeling, stability and biodistribution of Affibody molecules. Bioorg Med Chem Lett 2009; 19:3912-4. [DOI: 10.1016/j.bmcl.2009.03.083] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2009] [Revised: 03/15/2009] [Accepted: 03/20/2009] [Indexed: 10/21/2022]
|
41
|
Ekblad T, Tolmachev V, Orlova A, Lendel C, Abrahmsén L, Karlström AE. Synthesis and chemoselective intramolecular crosslinking of a HER2-binding affibody. Biopolymers 2009; 92:116-23. [PMID: 19140162 DOI: 10.1002/bip.21142] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The human epidermal growth factor receptor HER2 has emerged as an important target for molecular imaging of breast cancer. This article presents the design and synthesis of a HER2-targeting affibody molecule with improved stability and tumor targeting capacity, and with potential use as an imaging agent. The 58 aa three-helix bundle protein was assembled using solid-phase peptide synthesis, and a chemoselective ligation strategy was used to establish an intramolecular thioether bond between the side chain thiol group of a cysteine residue, positioned in the loop between helices I and II, and a chloroacetyl group on the side chain amino group of the C-terminal lysine residue. The tethered protein offered an increased thermal stability, with a melting temperature of 64 degrees C, compared to 54 degrees C for the linear control. The ligation did not have a major influence on the HER2 binding affinity, which was 320 and 380 pM for the crosslinked and linear molecules, respectively. Biodistribution studies were performed both in normal and tumor-bearing mice to evaluate the impact of the crosslinking on the in vivo behavior and on the tumor targeting performance. The distribution pattern was characterized by a low uptake in all organs except kidney, and rapid clearance from blood and normal tissue. Crosslinking of the protein resulted in a significantly increased tumor accumulation, rendering the tethered HER2-binding affibody molecule a valuable lead in the development of superior HER2 imaging agents.
Collapse
Affiliation(s)
- Torun Ekblad
- School of Biotechnology, Royal Institute of Technology, Stockholm, Sweden
| | | | | | | | | | | |
Collapse
|
42
|
Grönwall C, Ståhl S. Engineered affinity proteins—Generation and applications. J Biotechnol 2009; 140:254-69. [DOI: 10.1016/j.jbiotec.2009.01.014] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2008] [Revised: 12/05/2008] [Accepted: 01/26/2009] [Indexed: 12/11/2022]
|
43
|
Ekblad T, Tran T, Orlova A, Widström C, Feldwisch J, Abrahmsén L, Wennborg A, Karlström AE, Tolmachev V. Development and preclinical characterisation of 99mTc-labelled Affibody molecules with reduced renal uptake. Eur J Nucl Med Mol Imaging 2008; 35:2245-55. [PMID: 18594815 DOI: 10.1007/s00259-008-0845-7] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2008] [Accepted: 05/11/2008] [Indexed: 11/29/2022]
Abstract
PURPOSE Affibody molecules are low molecular weight proteins (7 kDa), which can be selected to bind to tumour-associated target proteins with subnanomolar affinity. Because of rapid tumour localisation and clearance from nonspecific compartments, Affibody molecules are promising tracers for molecular imaging. Earlier, (99m)Tc-labelled Affibody molecules demonstrated specific targeting of tumour xenografts. However, the biodistribution was suboptimal either because of hepatobiliary excretion or high renal uptake of the radioactivity. The goal of this study was to optimise the biodistribution of Affibody molecules by chelator engineering. MATERIALS AND METHODS Anti-HER2 Z(HER2:342) Affibody molecules, carrying the mercaptoacetyl-glutamyl-seryl-glutamyl (maESE), mercaptoacetyl-glutamyl-glutamyl-seryl (maEES) and mercaptoacetyl-seryl-glutamyl-glutamyl (maSEE) chelators, were prepared by peptide synthesis and labelled with (99m)Tc. The tumour-targeting capacity of these conjugates was compared with each other and with the best previously available conjugate, (99m)Tc-maEEE-Z(HER2:342,) in nude mice bearing SKOV-3 xenografts. The tumour-targeting capacity of the most promising conjugate, (99m)Tc-maESE-Z(HER2:342,) was compared with radioiodinated Z(HER2:342). RESULTS All novel conjugates demonstrated successful tumour targeting and a low degree of hepatobiliary excretion. The renal uptakes of serine-containing conjugates, 33 +/- 5, 68 +/- 21 and 71 +/- 10%IA/g, for(99m)Tc-maESE-Z(HER2:342), (99m)Tc-maEES-Z(HER2:342) and (99m)Tc-maSEE-Z(HER2:342), respectively, were significantly reduced in comparison with (99m)Tc-maEEE-Z(HER2:342) (102 +/- 13%IA/g). For (99m)Tc-maESE-Z(HER2:342), a tumour uptake of 9.6 +/- 1.8%IA/g and a tumour-to-blood ratio of 58 +/- 6 were reached at 4 h p.i. CONCLUSIONS A combination of serine and glutamic acid residues in the chelator sequence confers increased renal excretion and relatively low renal uptake of (99m)Tc-labelled Affibody molecules. In combination with preserved targeting capacity, this improved imaging of targets in abdominal area.
Collapse
Affiliation(s)
- Torun Ekblad
- School of Biotechnology, Royal Institute of Technology, Stockholm, Sweden
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Nygren PÅ. Alternative binding proteins: Affibody binding proteins developed from a small three-helix bundle scaffold. FEBS J 2008; 275:2668-76. [DOI: 10.1111/j.1742-4658.2008.06438.x] [Citation(s) in RCA: 209] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
45
|
Katritzky AR, Yoshioka M, Narindoshvili T, Chung A, Johnson JV. Fluorescent labeling of peptides on solid phase. Org Biomol Chem 2008; 6:4582-6. [DOI: 10.1039/b811693h] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
46
|
Ahlgren S, Orlova A, Rosik D, Sandström M, Sjöberg A, Baastrup B, Widmark O, Fant G, Feldwisch J, Tolmachev V. Evaluation of Maleimide Derivative of DOTA for Site-Specific Labeling of Recombinant Affibody Molecules. Bioconjug Chem 2007; 19:235-43. [DOI: 10.1021/bc700307y] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Sara Ahlgren
- Division of Nuclear Medicine, Department of Medical Sciences, Uppsala University, Uppsala, Sweden, Affibody AB, Bromma, Sweden, Division of Biomedical Radiation Sciences, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden, Hospital Physics, Department of Oncology, Uppsala University Hospital, Uppsala, Sweden
| | - Anna Orlova
- Division of Nuclear Medicine, Department of Medical Sciences, Uppsala University, Uppsala, Sweden, Affibody AB, Bromma, Sweden, Division of Biomedical Radiation Sciences, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden, Hospital Physics, Department of Oncology, Uppsala University Hospital, Uppsala, Sweden
| | - Daniel Rosik
- Division of Nuclear Medicine, Department of Medical Sciences, Uppsala University, Uppsala, Sweden, Affibody AB, Bromma, Sweden, Division of Biomedical Radiation Sciences, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden, Hospital Physics, Department of Oncology, Uppsala University Hospital, Uppsala, Sweden
| | - Mattias Sandström
- Division of Nuclear Medicine, Department of Medical Sciences, Uppsala University, Uppsala, Sweden, Affibody AB, Bromma, Sweden, Division of Biomedical Radiation Sciences, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden, Hospital Physics, Department of Oncology, Uppsala University Hospital, Uppsala, Sweden
| | - Anna Sjöberg
- Division of Nuclear Medicine, Department of Medical Sciences, Uppsala University, Uppsala, Sweden, Affibody AB, Bromma, Sweden, Division of Biomedical Radiation Sciences, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden, Hospital Physics, Department of Oncology, Uppsala University Hospital, Uppsala, Sweden
| | - Barbro Baastrup
- Division of Nuclear Medicine, Department of Medical Sciences, Uppsala University, Uppsala, Sweden, Affibody AB, Bromma, Sweden, Division of Biomedical Radiation Sciences, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden, Hospital Physics, Department of Oncology, Uppsala University Hospital, Uppsala, Sweden
| | - Olof Widmark
- Division of Nuclear Medicine, Department of Medical Sciences, Uppsala University, Uppsala, Sweden, Affibody AB, Bromma, Sweden, Division of Biomedical Radiation Sciences, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden, Hospital Physics, Department of Oncology, Uppsala University Hospital, Uppsala, Sweden
| | - Gunilla Fant
- Division of Nuclear Medicine, Department of Medical Sciences, Uppsala University, Uppsala, Sweden, Affibody AB, Bromma, Sweden, Division of Biomedical Radiation Sciences, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden, Hospital Physics, Department of Oncology, Uppsala University Hospital, Uppsala, Sweden
| | - Joachim Feldwisch
- Division of Nuclear Medicine, Department of Medical Sciences, Uppsala University, Uppsala, Sweden, Affibody AB, Bromma, Sweden, Division of Biomedical Radiation Sciences, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden, Hospital Physics, Department of Oncology, Uppsala University Hospital, Uppsala, Sweden
| | - Vladimir Tolmachev
- Division of Nuclear Medicine, Department of Medical Sciences, Uppsala University, Uppsala, Sweden, Affibody AB, Bromma, Sweden, Division of Biomedical Radiation Sciences, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden, Hospital Physics, Department of Oncology, Uppsala University Hospital, Uppsala, Sweden
| |
Collapse
|
47
|
Bodin A, Ahrenstedt L, Fink H, Brumer H, Risberg B, Gatenholm P. Modification of Nanocellulose with a Xyloglucan–RGD Conjugate Enhances Adhesion and Proliferation of Endothelial Cells: Implications for Tissue Engineering. Biomacromolecules 2007; 8:3697-704. [DOI: 10.1021/bm070343q] [Citation(s) in RCA: 169] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Aase Bodin
- Biopolymer Technology, Department of Chemical and Biological Engineering, Chalmers University of Technology, SE-412 96 Gothenburg, Sweden, School of Biotechnology, Royal Institute of Technology, Alba Nova University Centre, SE-106-91 Stockholm, Sweden, and Vascular Engineering Centre, Institution of Clinical Sciences, Department of Surgery, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Lage Ahrenstedt
- Biopolymer Technology, Department of Chemical and Biological Engineering, Chalmers University of Technology, SE-412 96 Gothenburg, Sweden, School of Biotechnology, Royal Institute of Technology, Alba Nova University Centre, SE-106-91 Stockholm, Sweden, and Vascular Engineering Centre, Institution of Clinical Sciences, Department of Surgery, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Helen Fink
- Biopolymer Technology, Department of Chemical and Biological Engineering, Chalmers University of Technology, SE-412 96 Gothenburg, Sweden, School of Biotechnology, Royal Institute of Technology, Alba Nova University Centre, SE-106-91 Stockholm, Sweden, and Vascular Engineering Centre, Institution of Clinical Sciences, Department of Surgery, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Harry Brumer
- Biopolymer Technology, Department of Chemical and Biological Engineering, Chalmers University of Technology, SE-412 96 Gothenburg, Sweden, School of Biotechnology, Royal Institute of Technology, Alba Nova University Centre, SE-106-91 Stockholm, Sweden, and Vascular Engineering Centre, Institution of Clinical Sciences, Department of Surgery, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Bo Risberg
- Biopolymer Technology, Department of Chemical and Biological Engineering, Chalmers University of Technology, SE-412 96 Gothenburg, Sweden, School of Biotechnology, Royal Institute of Technology, Alba Nova University Centre, SE-106-91 Stockholm, Sweden, and Vascular Engineering Centre, Institution of Clinical Sciences, Department of Surgery, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Paul Gatenholm
- Biopolymer Technology, Department of Chemical and Biological Engineering, Chalmers University of Technology, SE-412 96 Gothenburg, Sweden, School of Biotechnology, Royal Institute of Technology, Alba Nova University Centre, SE-106-91 Stockholm, Sweden, and Vascular Engineering Centre, Institution of Clinical Sciences, Department of Surgery, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
48
|
Renberg B, Nordin J, Merca A, Uhlén M, Feldwisch J, Nygren PA, Karlström AE. Affibody molecules in protein capture microarrays: evaluation of multidomain ligands and different detection formats. J Proteome Res 2007; 6:171-9. [PMID: 17203961 DOI: 10.1021/pr060316r] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The importance of the ligand presentation format for the production of protein capture microarrays was evaluated using different Affibody molecules, produced either as single 6 kDa monomers or genetically linked head-to-tail multimers containing up to four domains. The performances in terms of selectivity and sensitivity of the monomeric and the multidomain Affibody molecules were compared by immobilization of the ligands on microarray slides, followed by incubation with fluorescent-labeled target protein. An increase in signal intensities for the multimers was demonstrated, with the most pronounced difference observed between monomers and dimers. A protein microarray containing six different dimeric Affibody ligands with specificity for IgA, IgE, IgG, TNF-alpha, insulin, or Taq DNA polymerase was characterized for direct detection of fluorescent-labeled analytes. No cross-reactivity was observed and the limits of detection were 600 fM for IgA, 20 pM for IgE, 70 fM for IgG, 20 pM for TNF-alpha, 60 pM for insulin, and 10 pM for Taq DNA polymerase. Also, different sandwich formats for detection of unlabeled protein were evaluated and used for selective detection of IgA or TNF-alpha in human serum or plasma samples, respectively. Finally, the presence of IgA was determined using detection of directly Cy5-labeled normal or IgA-deficient serum samples.
Collapse
Affiliation(s)
- Björn Renberg
- School of Biotechnology, Division of Molecular Biotechnology, Royal Institute of Technology, AlbaNova University Center, SE - 106 91 Stockholm, Sweden
| | | | | | | | | | | | | |
Collapse
|
49
|
Engfeldt T, Tran T, Orlova A, Widström C, Feldwisch J, Abrahmsen L, Wennborg A, Karlström AE, Tolmachev V. 99mTc-chelator engineering to improve tumour targeting properties of a HER2-specific Affibody molecule. Eur J Nucl Med Mol Imaging 2007; 34:1843-53. [PMID: 17565496 DOI: 10.1007/s00259-007-0474-6] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2006] [Accepted: 04/08/2007] [Indexed: 01/24/2023]
Abstract
PURPOSE Monitoring HER2 expression is crucial for selection of breast cancer patients amenable to HER2-targeting therapy. The Affibody molecule Z(HER2:342) binds to HER2 with picomolar affinity and enables specific imaging of HER2 expression. Previously, Z(HER2:342) with the additional N-terminal mercaptoacetyl-glycyl-glycyl-glycyl (maGGG) sequence was labelled with (99m)Tc and demonstrated specific targeting of HER2-expressing xenografts. However, hepatobiliary excretion caused high radioactivity accumulation in the abdomen. We investigated whether the biodistribution of Z(HER2:342) can be improved by substituting glycyl residues in the chelating sequence with more hydrophilic seryl residues. METHODS The Affibody molecule Z(HER2:342), carrying the chelators mercaptoacetyl-glycyl-seryl-glycyl (maGSG), mercaptoacetyl-glycyl-D: -seryl-glycyl [maG(D-S)G] and mercaptoacetyl-seryl-seryl-seryl (maSSS), were prepared by peptide synthesis and labelled with (99m)Tc. The differences in the excretion pathways were evaluated in normal mice. The tumour targeting capacity of (99m)Tc-maSSS-Z(HER2:342) was studied in nude mice bearing SKOV-3 xenografts and compared with the capacity of radioiodinated Z(HER2:342). RESULTS A shift towards renal excretion was obtained when glycine was substituted with serine in the chelating sequence. The radioactivity in the gastrointestinal tract was reduced threefold for the maSSS conjugate in comparison with the maGGG conjugate 4 h post injection (p.i.). The tumour uptake of (99m)Tc-maSSS-Z(HER2:342) was 11.5 +/- 0.5% IA/g 4 h p.i., and the tumour-to-blood ratio was 76. The pharmacokinetics and uptake characteristics of technetium-labelled Z(HER2:342) were better than those of radioiodinated Z(HER2:342). CONCLUSION The introduction of serine residues in the chelator results in better tumour imaging properties of the Affibody molecule Z(HER2:342) compared with glycyl-containing chelators and is favourable for imaging of tumours and metastases in the abdominal area.
Collapse
Affiliation(s)
- Torun Engfeldt
- School of Biotechnology, Royal Institute of Technology, Stockholm, Sweden
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Friedman M, Nordberg E, Höidén-Guthenberg I, Brismar H, Adams GP, Nilsson FY, Carlsson J, Ståhl S. Phage display selection of Affibody molecules with specific binding to the extracellular domain of the epidermal growth factor receptor. Protein Eng Des Sel 2007; 20:189-99. [PMID: 17452435 DOI: 10.1093/protein/gzm011] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Affibody molecules specific for the epidermal growth factor receptor (EGFR) have been selected by phage display technology from a combinatorial protein library based on the 58-residue, protein A-derived Z domain. EGFR is overexpressed in various malignancies and is frequently associated with poor patient prognosis, and the information provided by targeting this receptor could facilitate both patient diagnostics and treatment. Three selected Affibody variants were shown to selectively bind to the extracellular domain of EGFR (EGFR-ECD). Kinetic biosensor analysis revealed that the three monomeric Affibody molecules bound with similar affinity, ranging from 130 to 185 nM. Head-to-tail dimers of the Affibody molecules were compared for their binding to recombinant EGFR-ECD in biosensor analysis and in human epithelial cancer A431 cells. Although the dimeric Affibody variants were found to bind in a range of 25-50 nM affinities in biosensor analysis, they were found to be low nanomolar binders in the cellular assays. Competition assays using radiolabeled Affibody dimers confirmed specific EGFR-binding and demonstrated that the three Affibody molecules competed for the same epitope. Immunofluorescence microscopy demonstrated that the selected Affibody dimers were initially binding to EGFR at the cell surface of A431, and confocal microscopy analysis showed that the Affibody dimers could thereafter be internalized. The potential use of the described Affibody molecules as targeting agents for radionuclide based imaging applications in various carcinomas is discussed.
Collapse
Affiliation(s)
- M Friedman
- Department of Molecular Biotechnology, AlbaNova University Center, Kungl Tekniska Högskolan, SE-106 91 Stockholm, Sweden
| | | | | | | | | | | | | | | |
Collapse
|