51
|
Grimm S, Salahshour S, Nygren PÅ. Monitored whole gene in vitro evolution of an anti-hRaf-1 affibody molecule towards increased binding affinity. N Biotechnol 2011; 29:534-42. [PMID: 22027369 DOI: 10.1016/j.nbt.2011.10.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2011] [Revised: 10/11/2011] [Accepted: 10/12/2011] [Indexed: 01/02/2023]
Abstract
The use of library technologies for the generation of affinity proteins often includes an affinity maturation step, based on the construction of secondary libraries from which second generation variants with improved affinities are selected. Here, we describe for the first time the affinity maturation of affibody molecules based on step-wise in vitro molecular evolution, involving cycles of error-prone PCR (epPCR) amplification for the introduction of diversity over the entire 58-residue three-helix bundle structure and ribosome display (RD) for the selection of improved variants. The model affibody molecule for the process was Z(RAF322), binding with a 1.9μm equilibrium dissociation constant (K(D)) to human Raf-1 (hRaf-1), a protein kinase of central importance in the MAPK/ERK proliferation pathway. The molecular evolution process was followed on both gene and protein levels via DNA sequencing and a biosensor-based binding analysis of pools of selected variants. After two cycles of diversification and selection, a significant increase in binding response of selected pools was seen. DNA sequencing showed that a dominant alanine to valine substitution had been effectively enriched, and was found in 83% of all selected clones, either alone or in combination with other enriched substitutions. The evolution procedure resulted in variants showing up to 26-fold increases in affinity to the hRaf-1 target. Noteworthy, for the two variants showing the highest affinities, substitutions were also found in affibody framework positions, corresponding to regions of the protein domain not addressed by traditional affibody molecule affinity maturation strategies. Interestingly, thermal melting point (T(m)) analyses showed that an increased affinity could be associated with both higher and lower T(m) values. All investigated variants showed excellent refolding properties and selective binding to hRaf-1, as analysed using a multiplexed bead-based binding assay, making them potentially valuable affinity reagents for cell biology studies.
Collapse
Affiliation(s)
- Sebastian Grimm
- Division of Molecular Biotechnology, Royal Institute of Technology, SE-106 91 Stockholm, Sweden
| | | | | |
Collapse
|
52
|
Ribosome display selection of a murine IgG₁ Fab binding affibody molecule allowing species selective recovery of monoclonal antibodies. Mol Biotechnol 2011; 48:263-76. [PMID: 21197589 PMCID: PMC3115053 DOI: 10.1007/s12033-010-9367-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Affinity reagents recognizing constant parts of antibody molecules are invaluable tools in immunotechnology applications, including purification, immobilization, and detection of immunoglobulins. In this article, murine IgG1, the primary isotype of monoclonal antibodies (mAbs) was used as target for selection of novel binders from a combinatorial ribosome display (RD) library of 1011 affibody molecules. Four rounds of selection using three different mouse IgG1 mAbs as alternating targets resulted in the identification of binders with broad mIgG1 recognition and dissociation constants (KD) in the low nanomolar to low micromolar range. For one of the binders, denoted Zmab25, competition in binding to full length mIgG1 by a streptococcal protein G (SPG) fragment and selective affinity capture of mouse IgG1 Fab fragments after papain cleavage of a full mAb suggest that an epitope functionally overlapping with the SPG-binding site in the CH1 domain of mouse IgG1 had been addressed. Interestingly, biosensor-based binding experiments showed that neither human IgG1 nor bovine Ig, the latter present in fetal bovine serum (FBS) was recognized by Zmab25. This selective binding profile towards murine IgG1 was successfully exploited in species selective recovery of two different mouse mAbs from complex samples containing FBS, resembling a hybridoma culture supernatant.
Collapse
|
53
|
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]
|
54
|
Abstract
Peptides within the knottin family have been shown to possess inherent stability, making them attractive scaffolds for the development of therapeutic and diagnostic agents. Given its remarkable stability to proteases, the cyclic peptide kalata B1 was employed as a scaffold to create a large knottin library displayed on the surface of E. coli. A library exceeding 10(9) variants was constructed by randomizing seven amino acids within a loop of the kalata B1 scaffold and screened using fluorescence-activated cell sorting to identify peptide ligands specific for the active site of human thrombin. Refolded thrombin binders exhibited high nanomolar affinities in solution and slow dissociation rates and were able to inhibit thrombin's enzymatic activity. Importantly, 80% of a knottin-based thrombin inhibitor remained intact after a 2 h incubation both with trypsin and with chymotrypsin, demonstrating that modifying the kalata B1 sequence did not compromise its stability properties. In addition, the knottin variant mediated 20-fold enhanced affinity for thrombin, when compared to the same seven residue binding epitope constrained by a single disulfide bond. Our results indicate that peptide libraries derived from the kalata B1 scaffold can yield high-affinity protein ligands that retain the remarkable protease resistance associated with the parent scaffold. More generally, this strategy may prove useful in the development of stable peptide ligands suitable for in vivo applications.
Collapse
Affiliation(s)
- Jennifer A. Getz
- Department of Chemical Engineering, University of California, Santa Barbara, CA 93106
- Institute for Collaborative Biotechnologies, University of California, Santa Barbara, CA 93106
| | - Jeffrey J. Rice
- Department of Chemical Engineering, University of California, Santa Barbara, CA 93106
- Institute for Collaborative Biotechnologies, University of California, Santa Barbara, CA 93106
| | - Patrick S. Daugherty
- Department of Chemical Engineering, University of California, Santa Barbara, CA 93106
- Institute for Collaborative Biotechnologies, University of California, Santa Barbara, CA 93106
- Biomolecular Science and Engineering, University of California, Santa Barbara, CA 93106
| |
Collapse
|
55
|
Gong R, Wang Y, Feng Y, Zhao Q, Dimitrov DS. Shortened engineered human antibody CH2 domains: increased stability and binding to the human neonatal Fc receptor. J Biol Chem 2011; 286:27288-93. [PMID: 21669873 PMCID: PMC3149322 DOI: 10.1074/jbc.m111.254219] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2011] [Revised: 06/02/2011] [Indexed: 01/20/2023] Open
Abstract
The immunoglobulin (Ig) constant CH2 domain is critical for antibody effector functions. Isolated CH2 domains are promising scaffolds for construction of libraries containing diverse binders that could also confer some effector functions. We have shown previously that an isolated human CH2 domain is relatively unstable to thermally induced unfolding, but its stability can be improved by engineering an additional disulfide bond (Gong, R., Vu, B. K., Feng, Y., Prieto, D. A., Dyba, M. A., Walsh, J. D., Prabakaran, P., Veenstra, T. D., Tarasov, S. G., Ishima, R., and Dimitrov, D. S. (2009) J. Biol. Chem. 284, 14203-14210). We have hypothesized that the stability of this engineered antibody domain could be further increased by removing unstructured residues. To test our hypothesis, we removed the seven N-terminal residues that are in a random coil as suggested by our analysis of the isolated CH2 crystal structure and NMR data. The resulting shortened engineered CH2 (m01s) was highly soluble, monomeric, and remarkably stable, with a melting temperature (T(m)) of 82.6 °C, which is about 10 and 30 °C higher than those of the original stabilized CH2 (m01) and CH2, respectively. m01s and m01 were more resistant to protease digestion than CH2. A newly identified anti-CH2 antibody that recognizes a conformational epitope bound to m01s significantly better (>10-fold higher affinity) than to CH2 and slightly better than to m01. m01s bound to a recombinant soluble human neonatal Fc receptor at pH 6.0 more strongly than CH2. These data suggest that shortening the m01 N terminus significantly increases stability without disrupting its conformation and that our approach for increasing stability and decreasing size by removing unstructured regions may also apply to other proteins.
Collapse
Affiliation(s)
- Rui Gong
- From the Protein Interactions Group, Center for Cancer Research Nanobiology Program, Center for Cancer Research, and
| | - Yanping Wang
- From the Protein Interactions Group, Center for Cancer Research Nanobiology Program, Center for Cancer Research, and
- SAIC-Frederick, Inc., NCI-Frederick, National Institutes of Health, Frederick, Maryland 21702
| | - Yang Feng
- From the Protein Interactions Group, Center for Cancer Research Nanobiology Program, Center for Cancer Research, and
| | - Qi Zhao
- From the Protein Interactions Group, Center for Cancer Research Nanobiology Program, Center for Cancer Research, and
| | - Dimiter S. Dimitrov
- From the Protein Interactions Group, Center for Cancer Research Nanobiology Program, Center for Cancer Research, and
| |
Collapse
|
56
|
Naimuddin M, Kubo T. Display of disulfide-rich proteins by complementary DNA display and disulfide shuffling assisted by protein disulfide isomerase. Anal Biochem 2011; 419:33-9. [PMID: 21867670 DOI: 10.1016/j.ab.2011.07.034] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2011] [Revised: 07/07/2011] [Accepted: 07/27/2011] [Indexed: 10/17/2022]
Abstract
We report an efficient system to produce and display properly folded disulfide-rich proteins facilitated by coupled complementary DNA (cDNA) display and protein disulfide isomerase-assisted folding. The results show that a neurotoxin protein containing four disulfide linkages can be displayed in the folded state. Furthermore, it can be refolded on a solid support that binds efficiently to its natural acetylcholine receptor. Probing the efficiency of the display proteins prepared by these methods provided up to 8-fold higher enrichment by the selective enrichment method compared with cDNA display alone, more than 10-fold higher binding to its receptor by the binding assays, and more than 10-fold higher affinities by affinity measurements. Cotranslational folding was found to have better efficiency than posttranslational refolding between the two investigated methods. We discuss the utilities of efficient display of such proteins in the preparation of superior quality proteins and protein libraries for directed evolution leading to ligand discovery.
Collapse
Affiliation(s)
- Mohammed Naimuddin
- Janusys Corporation, Saitama Industrial Technology Center, Skip City, Kawaguchi, Saitama 333-0844, Japan.
| | | |
Collapse
|
57
|
Pazos E, Mosquera J, Vázquez ME, Mascareñas JL. DNA Recognition by Synthetic Constructs. Chembiochem 2011; 12:1958-73. [DOI: 10.1002/cbic.201100247] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2011] [Indexed: 12/29/2022]
|
58
|
Löfblom J, Frejd FY, Ståhl S. Non-immunoglobulin based protein scaffolds. Curr Opin Biotechnol 2011; 22:843-8. [PMID: 21726995 DOI: 10.1016/j.copbio.2011.06.002] [Citation(s) in RCA: 110] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2011] [Revised: 04/27/2011] [Accepted: 06/01/2011] [Indexed: 10/18/2022]
Abstract
Non-immunoglobulin based protein scaffolds have been reported as promising alternatives to traditional monoclonal antibodies for over a decade and are often mentioned as part of the next-generation immunotherapeutics. Today, this class of biologics is beginning to demonstrate its potential for therapeutic applications and several are currently in preclinical or clinical development. A common denominator for most of these new scaffolds is the attractive properties that differentiate them from monoclonal antibodies including small size, cysteine-free sequence, flexible pharmacokinetic properties, and ease of generating multispecific molecules. In addition to therapeutic applications, substantial evidence point to superior performance of several of these scaffolds in molecular imaging compared to full-length antibodies. Here we review the most recent progress using alternative protein scaffolds for therapy and medical imaging.
Collapse
Affiliation(s)
- John Löfblom
- Division of Molecular Biotechnology, School of Biotechnology, Royal Institute of Technology (KTH), AlbaNova University Center, SE-106 91 Stockholm, Sweden
| | | | | |
Collapse
|
59
|
Riihimäki TA, Hiltunen S, Rangl M, Nordlund HR, Määttä JAE, Ebner A, Hinterdorfer P, Kulomaa MS, Takkinen K, Hytönen VP. Modification of the loops in the ligand-binding site turns avidin into a steroid-binding protein. BMC Biotechnol 2011; 11:64. [PMID: 21658230 PMCID: PMC3201017 DOI: 10.1186/1472-6750-11-64] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2011] [Accepted: 06/09/2011] [Indexed: 01/20/2023] Open
Abstract
Background Engineered proteins, with non-immunoglobulin scaffolds, have become an important alternative to antibodies in many biotechnical and therapeutic applications. When compared to antibodies, tailored proteins may provide advantageous properties such as a smaller size or a more stable structure. Results Avidin is a widely used protein in biomedicine and biotechnology. To tailor the binding properties of avidin, we have designed a sequence-randomized avidin library with mutagenesis focused at the loop area of the binding site. Selection from the generated library led to the isolation of a steroid-binding avidin mutant (sbAvd-1) showing micromolar affinity towards testosterone (Kd ~ 9 μM). Furthermore, a gene library based on the sbAvd-1 gene was created by randomizing the loop area between β-strands 3 and 4. Phage display selection from this library led to the isolation of a steroid-binding protein with significantly decreased biotin binding affinity compared to sbAvd-1. Importantly, differential scanning calorimetry and analytical gel-filtration revealed that the high stability and the tetrameric structure were preserved in these engineered avidins. Conclusions The high stability and structural properties of avidin make it an attractive molecule for the engineering of novel receptors. This methodology may allow the use of avidin as a universal scaffold in the development of novel receptors for small molecules.
Collapse
Affiliation(s)
- Tiina A Riihimäki
- Institute of Biomedical Technology, University of Tampere and Tampere University Hospital, FI-33520 Tampere, Finland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
60
|
Molecular imprinting of fructose using a polymerizable benzoboroxole: Effective complexation at pH 7.4. POLYMER 2011. [DOI: 10.1016/j.polymer.2011.04.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
61
|
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: 2.1] [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
|
62
|
Peptide phage display as a tool for drug discovery: targeting membrane receptors. Molecules 2011; 16:857-87. [PMID: 21258295 PMCID: PMC6259427 DOI: 10.3390/molecules16010857] [Citation(s) in RCA: 135] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2010] [Revised: 01/14/2011] [Accepted: 01/19/2011] [Indexed: 12/14/2022] Open
Abstract
Ligands selected from phage-displayed random peptide libraries tend to be directed to biologically relevant sites on the surface of the target protein. Consequently, peptides derived from library screenings often modulate the target protein’s activity in vitro and in vivo and can be used as lead compounds in drug design and as alternatives to antibodies for target validation in both genomics and drug discovery. This review discusses the use of phage display to identify membrane receptor modulators with agonistic or antagonistic activities. Because isolating or producing recombinant membrane proteins for use as target molecules in library screening is often impossible, innovative selection strategies such as panning against whole cells or tissues, recombinant receptor ectodomains, or neutralizing antibodies to endogenous binding partners were devised. Prominent examples from a two-decade history of peptide phage display will be presented, focusing on the design of affinity selection experiments, methods for improving the initial hits, and applications of the identified peptides.
Collapse
|
63
|
Naimuddin M, Kobayashi S, Tsutsui C, Machida M, Nemoto N, Sakai T, Kubo T. Directed evolution of a three-finger neurotoxin by using cDNA display yields antagonists as well as agonists of interleukin-6 receptor signaling. Mol Brain 2011; 4:2. [PMID: 21214917 PMCID: PMC3024951 DOI: 10.1186/1756-6606-4-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2010] [Accepted: 01/07/2011] [Indexed: 12/16/2022] Open
Abstract
Background Directed evolution of biomolecules such as DNA, RNA and proteins containing high diversity has emerged as an effective method to obtain molecules for various purposes. In the recent past, proteins from non-immunoglobulins have attracted attention as they mimic antibodies with respect to binding potential and provide further potential advantages. In this regard, we have attempted to explore a three-finger neurotoxin protein (3F). 3F proteins are small (~7 kDa), structurally well defined, thermally stable and resistant to proteolysis that presents them as promising candidates for directed evolution. Results We have engineered a snake α-neurotoxin that belongs to the 3F family by randomizing the residues in the loops involved in binding with acetylcholine receptors and employing cDNA display to obtain modulators of interleukin-6 receptor (IL-6R). Selected candidates were highly specific for IL-6R with dissociation constants and IC50s in the nanomolar range. Antagonists as well as agonists were identified in an IL-6 dependent cell proliferation assay. Size minimization yielded peptides of about one-third the molecular mass of the original proteins, without significant loss of activities and, additionally, lead to the identification of the loops responsible for function. Conclusions This study shows 3F protein is amenable to introduce amino acid changes in the loops that enable preparation of a high diversity library that can be utilized to obtain ligands against macromolecules. We believe this is the first report of protein engineering to convert a neurotoxin to receptor ligands other than the parent receptor, the identification of an agonist from non-immunoglobulin proteins, the construction of peptide mimic of IL-6, and the successful size reduction of a single-chain protein.
Collapse
Affiliation(s)
- Mohammed Naimuddin
- National Institute of Advanced Industrial Science and Technology, Tsukuba, Ibaraki, Japan.
| | | | | | | | | | | | | |
Collapse
|
64
|
Lim TS, Schütze T, Lehrach H, Glökler J, Konthur Z. Diversity visualization by endonuclease: a rapid assay to monitor diverse nucleotide libraries. Anal Biochem 2010; 411:16-21. [PMID: 21185254 DOI: 10.1016/j.ab.2010.12.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2010] [Revised: 12/15/2010] [Accepted: 12/16/2010] [Indexed: 11/19/2022]
Abstract
Many experiments require a fast and cost-effective method to monitor nucleic acid sequence diversity. Here we describe a method called diversity visualization by endonuclease (DiVE) that allows rapid visualization of sequence diversity of polymerase chain reaction (PCR) products based on DNA hybridization kinetics coupled with the activity of a single-strand specific nuclease. The assay involves only a limited number of steps and can be performed in less than 4h, including the initial PCR. After PCR, the homoduplex double-stranded DNA (dsDNA) is denatured and reannealed under stringent conditions. During the reannealing process, incubation with S1 nuclease removes single-stranded loops of formed heteroduplexes and the resulting digest is visualized on agarose gel. The sequence diversity is inversely proportional to the band intensities of S1 nuclease surviving dsDNA molecules of expected size. As an example, we employed DiVE to monitor the diversity of panning rounds from a single-framework, semisynthetic single-chain antibody fragment (scFv) phage display library. The results are in good agreement with the observed decrease in diversity in phage display panning rounds toward the selection of monoclonal scFv. We conclude that the DiVE assay allows rapid and cost-effective monitoring of diversities of various nucleotide libraries and proves to be particularly suitable for scaffold-based randomized libraries.
Collapse
Affiliation(s)
- Theam Soon Lim
- Department of Vertebrate Genomics, Max Planck Institute for Molecular Genetics, D-14195 Berlin, Germany
| | | | | | | | | |
Collapse
|
65
|
Kronqvist N, Malm M, Göstring L, Gunneriusson E, Nilsson M, Höidén Guthenberg I, Gedda L, Frejd FY, Ståhl S, Löfblom J. Combining phage and staphylococcal surface display for generation of ErbB3-specific Affibody molecules. Protein Eng Des Sel 2010; 24:385-96. [PMID: 21177282 DOI: 10.1093/protein/gzq118] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Emerging evidence suggests that the catalytically inactive ErbB3 (HER3) protein plays a fundamental role in normal tyrosine kinase receptor signaling as well as in aberrant functioning of these signaling pathways, resulting in several forms of human cancers. ErbB3 has recently also been implicated in resistance to ErbB2-targeting therapies. Here we report the generation of high-affinity ErbB3-specific Affibody molecules intended for future molecular imaging and biotherapeutic applications. Using a high-complexity phage-displayed Affibody library, a number of ErbB3 binders were isolated and specific cell-binding activity was demonstrated in immunofluorescence microscopic studies. Subsequently, a second-generation library was constructed based on sequences of the candidates from the phage display selection. By exploiting the sensitive affinity discrimination capacity of a novel bacterial surface display technology, the affinity of candidate Affibody molecules was further increased down to subnanomolar affinity. In summary, the demonstrated specific targeting of native ErbB3 receptor on human cancer cell lines as well as competition with the heregulin/ErbB3 interaction indicates that these novel biological agents may become useful tools for diagnostic and therapeutic targeting of ErbB3-expressing cancers. Our studies also highlight the powerful approach of combining the advantages of different display technologies for generation of functional high-affinity protein-based binders. Potential future applications, such as radionuclide-based diagnosis and treatment of human cancers are discussed.
Collapse
Affiliation(s)
- Nina Kronqvist
- Department of Molecular Biotechnology, School of Biotechnology, Royal Institute of Technology (KTH), AlbaNova University Center, SE-106 91 Stockholm
| | | | | | | | | | | | | | | | | | | |
Collapse
|
66
|
A Novel Unstructured Scaffold Based on 4EBP1 Enables the Functional Display of a Wide Range of Bioactive Peptides. J Mol Biol 2010; 404:819-31. [DOI: 10.1016/j.jmb.2010.09.063] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2010] [Revised: 09/22/2010] [Accepted: 09/29/2010] [Indexed: 01/11/2023]
|
67
|
Sharkey RM, Rossi EA, Chang CH, Goldenberg DM. Improved cancer therapy and molecular imaging with multivalent, multispecific antibodies. Cancer Biother Radiopharm 2010; 25:1-12. [PMID: 20187791 DOI: 10.1089/cbr.2009.0690] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Antibodies are highly versatile proteins with the ability to be used to target diverse compounds, such as radionuclides for imaging and therapy, or drugs and toxins for therapy, but also can be used unconjugated to elicit therapeutically beneficial responses, usually with minimal toxicity. This update describes a new procedure for forming multivalent and/or multispecific proteins, known as the dock-and-lock (DNL) technique. Developed as a procedure for preparing bispecific antibodies capable of binding divalently to a tumor antigen and monovalently to a radiolabeled hapten-peptide for pretargeted imaging and therapy, this methodology has the flexibility to create a number of other biologic agents of therapeutic interest. A variety of constructs, based on anti-CD20 and CD22 antibodies, have been made, with results showing that multispecific antibodies have very different properties from the respective parental monospecific antibodies. The technique is not restricted to antibody combination, but other biologics, such as interferon-alpha2b, have been prepared. These types of constructs not only allow small biologics to be sustained in the blood longer, but also to be selectively targeted. Thus, DNL technology is a highly flexible platform that can be used to prepare many different types of agents that could further improve cancer detection and therapy.
Collapse
Affiliation(s)
- Robert M Sharkey
- Center for Molecular Medicine and Immunology (CMMI), Belleville, New Jersey, USA
| | | | | | | |
Collapse
|
68
|
Miao Z, Ren G, Liu H, Jiang L, Cheng Z. Small-animal PET imaging of human epidermal growth factor receptor positive tumor with a 64Cu labeled affibody protein. Bioconjug Chem 2010; 21:947-54. [PMID: 20402512 DOI: 10.1021/bc900515p] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Epidermal growth factor receptor (EGFR) has become an attractive target for cancer molecular imaging and therapy. Affibody proteins against EGFR have been reported, and thus, we were interested in evaluating their potential for positron emission tomography (PET) imaging of EGFR positive cancer. An Affibody analogue (Ac-Cys-Z(EGFR:1907)) binding to EGFR was made through conventional solid phase peptide synthesis. The purified protein was site-specifically coupled with the 1,4,7,10-tetraazacyclododecane-1,4,7-tris-aceticacid-10-maleimidethylacetamide (maleimido-mono-amide-DOTA) to produce the bioconjugate, DOTA-Z(EGFR:1907). (64)Cu labeled probe (64)Cu-DOTA-Z(EGFR:1907) displayed a moderate specific activity (5-8 MBq/nmol, 22-35 microCi/microg). Cell uptake assays by pre-incubating without or with 300 times excess unlabeled Ac-Cys-Z(EGFR:1907) showed high EGFR-specific uptake (20% applied activity at 0.5 h) in A431 epidermoid carcinoma cancer cells. The affinity (K(D)) of (64)Cu-DOTA-Z(EGFR:1907) as tested by cell saturation analysis was 20 nM. The serum stability test showed excellent stability of the probe with >95% intact after 4 h of incubation in mouse serum. In vivo small-animal PET imaging showed fast tumor targeting, high tumor accumulation (approximately 10% ID/g at 1 h p.i.), and good tumor-to-normal tissue contrast of (64)Cu-DOTA-Z(EGFR:1907) spiked with a wide dose range of Ac-Cys-Z(EGFR:1907). Bio-distribution studies further demonstrated that the probe had high tumor, blood, liver, and kidney uptakes, while blood radioactivity concentration dropped dramatically at increased spiking doses. Co-injection of the probe with 500 microg of Ac-Cys-Z(EGFR:1907) for blocking significantly reduced the tumor uptake. Thus, (64)Cu-DOTA-Z(EGFR:1907) showed potential as a high tumor contrast EGFR PET imaging reagent. The probe spiked with 50 microg of Ac-Cys-Z(EGFR:1907) improved tumor imaging contrast which may have important clinical applications.
Collapse
Affiliation(s)
- Zheng Miao
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University, Stanford, California 94305-5344, USA
| | | | | | | | | |
Collapse
|
69
|
Grimm S, Lundberg E, Yu F, Shibasaki S, Vernet E, Skogs M, Nygren PÅ, Gräslund T. Selection and characterisation of affibody molecules inhibiting the interaction between Ras and Raf in vitro. N Biotechnol 2010; 27:766-73. [PMID: 20674812 DOI: 10.1016/j.nbt.2010.07.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2010] [Revised: 07/16/2010] [Accepted: 07/23/2010] [Indexed: 10/19/2022]
Abstract
Development of molecules with the ability to selectively inhibit particular protein-protein interactions is important in providing tools for understanding cell biology. In this work, we describe efforts to select small Ras- and Raf-specific three-helix bundle affibody binding proteins capable of inhibiting the interaction between H-Ras and Raf-1, from a combinatorial library displayed on bacteriophage. Target-specific variants with typically high nanomolar or low micromolar affinities (K(D)) could be selected successfully against both proteins, as shown by dot blot, ELISA and real-time biospecific interaction analyses. Affibody molecule variants selected against H-Ras were shown to bind epitopes overlapping each other at a site that differed from that at which H-Ras interacts with Raf-1. In contrast, an affibody molecule isolated during selection against Raf-1 was shown to effectively inhibit the interaction between H-Ras and Raf-1 in a dose-dependent manner. Possible intracellular applications of the selected affibody molecules are discussed.
Collapse
Affiliation(s)
- Sebastian Grimm
- Division of Molecular Biotechnology, Royal Institute of Technology, Stockholm, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
70
|
|
71
|
Hohlbaum AM, Skerra A. Anticalins: the lipocalin family as a novel protein scaffold for the development of next-generation immunotherapies. Expert Rev Clin Immunol 2010; 3:491-501. [PMID: 20477155 DOI: 10.1586/1744666x.3.4.491] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Anticalins are engineered ligand-binding proteins based on the human lipocalin scaffold. Their architecture is characterized by a rigid beta-barrel that supports four structurally hypervariable loops. Similar to antibodies, these loops form the natural ligand-binding site, usually for vitamins, hormones or secondary metabolites. Anticalins with novel specificities can be engineered by reshaping this loop region, using targeted random mutagenesis in combination with functional display and guided selection. Several drug candidates with specificities for exogenous low-molecular-weight substances, peptides and even protein targets (e.g., several disease-related cell surface receptors) have been obtained in this way. Owing to their exquisite specificity and high affinity, Anticalins are particularly attractive as antagonists for the manipulation of immune mechanisms, leading to either inhibitory or stimulatory effects. Compared with antibodies, Anticalins offer several practical advantages as they are much smaller, consist of a single polypeptide chain and can be produced easily in microbial expression systems.
Collapse
|
72
|
Dübel S, Stoevesandt O, Taussig MJ, Hust M. Generating recombinant antibodies to the complete human proteome. Trends Biotechnol 2010; 28:333-9. [PMID: 20538360 DOI: 10.1016/j.tibtech.2010.05.001] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2010] [Revised: 04/29/2010] [Accepted: 05/03/2010] [Indexed: 10/19/2022]
Abstract
In vitro antibody generation technologies have now been available for two decades. Research reagents prepared via phage display are becoming available and several recent studies have demonstrated that these technologies are now sufficiently advanced to facilitate generation of a comprehensive renewable resource of antibodies for any protein encoded by the approximately 22,500 human protein-coding genes. Antibody selection in vitro offers properties not available in animal-based antibody generation methods. By adjusting the biochemical milieu during selection, it is possible to control the antigen conformation recognized, the antibody affinity or unwanted cross-reactivity. For larger-scale antibody generation projects, the handling, transport and storage logistics and bacterial production offer cost benefits. Because the DNA sequence encoding the antibody is available, modifications, such as site-specific in vivo biotinylation and multimerization, are only a cloning step away. This opinion article summarizes opportunities for the generation of antibodies for proteome research using in vitro technologies.
Collapse
Affiliation(s)
- Stefan Dübel
- Technische Universität Braunschweig, Institute of Biochemistry and Biotechnology, D-38106 Braunschweig, Germany.
| | | | | | | |
Collapse
|
73
|
Molecular basis of the structural stability of a Top7-based scaffold at extreme pH and temperature conditions. J Mol Graph Model 2010; 28:755-65. [DOI: 10.1016/j.jmgm.2010.01.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2009] [Revised: 12/29/2009] [Accepted: 01/31/2010] [Indexed: 11/22/2022]
|
74
|
Miao Z, Ren G, Liu H, Kimura RH, Jiang L, Cochran JR, Gambhir SS, Cheng Z. An engineered knottin peptide labeled with 18F for PET imaging of integrin expression. Bioconjug Chem 2010; 20:2342-7. [PMID: 19908826 DOI: 10.1021/bc900361g] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Knottins are small constrained polypeptides that share a common disulfide-bonded framework and a triple-stranded beta-sheet fold. Previously, directed evolution of the Ecballium elaterium trypsin inhibitor (EETI-II) knottin led to the identification of a mutant that bound to tumor-specific alpha(v)beta(3) and alpha(v)beta(5) integrin receptors with low nanomolar affinity. The objective of this study was to prepare and evaluate a radiofluorinated version of this knottin (termed 2.5D) for microPET imaging of integrin positive tumors in living subjects. Knottin peptide 2.5D was prepared by solid-phase synthesis and folded in vitro, and its free N-terminal amine was reacted with N-succinimidyl-4-18/19F-fluorobenzoate (18/19F-SFB) to produce the fluorinated peptide 18/19F-FB-2.5D. The binding affinities of unlabeled knottin peptide 2.5D and 19F-FB-2.5D to U87MG glioblastoma cells were measured by competition binding assay using 125I-labeled echistatin. It was found that unlabeled 2.5D and 19F-FB-2.5D competed with 125I-echistatin for binding to cell surface integrins with IC(50) values of 20.3 +/- 7.3 and 13.2 +/- 5.4 nM, respectively. Radiosynthesis of 18F-FB-2.5D resulted in a product with high specific activity (ca. 100 GBq/micromol). Next, biodistribution and positron emission tomography (PET) imaging studies were performed to evaluate the in vivo behavior of 18F-FB-2.5D. Approximately 3.7 MBq 18F-FB-2.5D was injected into U87MG tumor-bearing mice via the tail vein. Biodistribution studies demonstrated that 18F-FB-2.5D had moderate tumor uptake at 0.5 h post injection, and coinjection of a large excess of the unlabeled peptidomimetic c(RGDyK) as a blocking agent significantly reduced tumor uptake (1.90 +/- 1.15 vs 0.57 +/- 0.14%ID/g, 70% inhibition, P < 0.05). In vivo microPET imaging showed that 18F-FB-2.5D rapidly accumulated in the tumor and quickly cleared from the blood through the kidneys, allowing excellent tumor-to-normal tissue contrast to be obtained. Collectively, 18F-FB-2.5D allows integrin-specific PET imaging of U87MG tumors with good contrast and further demonstrates that knottins are excellent peptide scaffolds for development of PET probes with potential for clinical translation.
Collapse
Affiliation(s)
- Zheng Miao
- Molecular Imaging Program at Stanford (MIPS), Bio-X Program, Stanford University, Stanford, California 94305, USA
| | | | | | | | | | | | | | | |
Collapse
|
75
|
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: 494] [Impact Index Per Article: 32.9] [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]
|
76
|
Dimitrov DS. Engineered CH2 domains (nanoantibodies). MAbs 2010; 1:26-8. [PMID: 20046570 DOI: 10.4161/mabs.1.1.7480] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2008] [Accepted: 11/21/2008] [Indexed: 11/19/2022] Open
Abstract
Currently, almost all FDA approved therapeutic antibodies (except ReoPro, Lucentis and Cimzia which are Fabs), and the vast majority of those in clinical trials are full-size antibodies mostly in IgG1 format of about 150 kDa size. A fundamental problem for such large molecules is their poor penetration into tissues (e.g., solid tumors) and poor or absent binding to regions on the surface of some molecules (e.g., on the HIV envelope glycoprotein) which are fully accessible only by molecules of smaller size. Therefore, much work especially during the last decade has been aimed at developing novel scaffolds of much smaller size and high stability. Here I briefly describe a proposition to use the immunoglobulin (Ig) constant CH2 domain (CH3 for IgE and IgM) as a scaffold. CH2 is critical for the Ig effector functions. Isolated CH2 is stable monomer in contrast to all other constant domains and most of the variable domains. CH2 and engineered CH2 domains with improved stability can be used as scaffolds for construction of libraries containing diverse binders to various antigens. Such binders based on a CH2 scaffold could also confer some effector functions. Because the CH2 domains are the smallest independently folded antibody domains that can be engineered to contain simultaneously antigen-binding sites and binding sites mediating effector and stability functions, and to distinguish them from domain antibodies which are used to denote engineered VH or VL domains or nanobodies which are used to denote camelid VHH, I termed them nanoantibodies (nAbs).
Collapse
Affiliation(s)
- Dimiter S Dimitrov
- Protein Interactions Group, Center for Cancer Research Nanobiology Program, National Cancer Institute, National Institutes of Health, Frederick, Maryland 21702, USA.
| |
Collapse
|
77
|
Cinier M, Petit M, Williams MN, Fabre RM, Pecorari F, Talham DR, Bujoli B, Tellier C. Bisphosphonate Adaptors for Specific Protein Binding on Zirconium Phosphonate-based Microarrays. Bioconjug Chem 2009; 20:2270-7. [DOI: 10.1021/bc9002597] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Mathieu Cinier
- Laboratoire de Biotechnologie, Biocatalyse et Biorégulation, UFR Sciences et Techniques, Université de Nantes, CNRS, UMR 6204, 2, rue de la Houssinière, BP 92208, 44322 NANTES Cedex 3, France, Chimie Et Interdisciplinarité: Synthèse Analyse Modélisation (CEISAM), Université de Nantes, CNRS, UMR 6230, 2 Rue de la Houssinière, BP92208, 44322 Nantes Cedex 03, France, and Department of Chemistry, University of Florida, Gainesville, Florida 32611-7200
| | - Marc Petit
- Laboratoire de Biotechnologie, Biocatalyse et Biorégulation, UFR Sciences et Techniques, Université de Nantes, CNRS, UMR 6204, 2, rue de la Houssinière, BP 92208, 44322 NANTES Cedex 3, France, Chimie Et Interdisciplinarité: Synthèse Analyse Modélisation (CEISAM), Université de Nantes, CNRS, UMR 6230, 2 Rue de la Houssinière, BP92208, 44322 Nantes Cedex 03, France, and Department of Chemistry, University of Florida, Gainesville, Florida 32611-7200
| | - Monique N. Williams
- Laboratoire de Biotechnologie, Biocatalyse et Biorégulation, UFR Sciences et Techniques, Université de Nantes, CNRS, UMR 6204, 2, rue de la Houssinière, BP 92208, 44322 NANTES Cedex 3, France, Chimie Et Interdisciplinarité: Synthèse Analyse Modélisation (CEISAM), Université de Nantes, CNRS, UMR 6230, 2 Rue de la Houssinière, BP92208, 44322 Nantes Cedex 03, France, and Department of Chemistry, University of Florida, Gainesville, Florida 32611-7200
| | - Roxane M. Fabre
- Laboratoire de Biotechnologie, Biocatalyse et Biorégulation, UFR Sciences et Techniques, Université de Nantes, CNRS, UMR 6204, 2, rue de la Houssinière, BP 92208, 44322 NANTES Cedex 3, France, Chimie Et Interdisciplinarité: Synthèse Analyse Modélisation (CEISAM), Université de Nantes, CNRS, UMR 6230, 2 Rue de la Houssinière, BP92208, 44322 Nantes Cedex 03, France, and Department of Chemistry, University of Florida, Gainesville, Florida 32611-7200
| | - Frédéric Pecorari
- Laboratoire de Biotechnologie, Biocatalyse et Biorégulation, UFR Sciences et Techniques, Université de Nantes, CNRS, UMR 6204, 2, rue de la Houssinière, BP 92208, 44322 NANTES Cedex 3, France, Chimie Et Interdisciplinarité: Synthèse Analyse Modélisation (CEISAM), Université de Nantes, CNRS, UMR 6230, 2 Rue de la Houssinière, BP92208, 44322 Nantes Cedex 03, France, and Department of Chemistry, University of Florida, Gainesville, Florida 32611-7200
| | - Daniel R. Talham
- Laboratoire de Biotechnologie, Biocatalyse et Biorégulation, UFR Sciences et Techniques, Université de Nantes, CNRS, UMR 6204, 2, rue de la Houssinière, BP 92208, 44322 NANTES Cedex 3, France, Chimie Et Interdisciplinarité: Synthèse Analyse Modélisation (CEISAM), Université de Nantes, CNRS, UMR 6230, 2 Rue de la Houssinière, BP92208, 44322 Nantes Cedex 03, France, and Department of Chemistry, University of Florida, Gainesville, Florida 32611-7200
| | - Bruno Bujoli
- Laboratoire de Biotechnologie, Biocatalyse et Biorégulation, UFR Sciences et Techniques, Université de Nantes, CNRS, UMR 6204, 2, rue de la Houssinière, BP 92208, 44322 NANTES Cedex 3, France, Chimie Et Interdisciplinarité: Synthèse Analyse Modélisation (CEISAM), Université de Nantes, CNRS, UMR 6230, 2 Rue de la Houssinière, BP92208, 44322 Nantes Cedex 03, France, and Department of Chemistry, University of Florida, Gainesville, Florida 32611-7200
| | - Charles Tellier
- Laboratoire de Biotechnologie, Biocatalyse et Biorégulation, UFR Sciences et Techniques, Université de Nantes, CNRS, UMR 6204, 2, rue de la Houssinière, BP 92208, 44322 NANTES Cedex 3, France, Chimie Et Interdisciplinarité: Synthèse Analyse Modélisation (CEISAM), Université de Nantes, CNRS, UMR 6230, 2 Rue de la Houssinière, BP92208, 44322 Nantes Cedex 03, France, and Department of Chemistry, University of Florida, Gainesville, Florida 32611-7200
| |
Collapse
|
78
|
Chen W, Zhu Z, Feng Y, Dimitrov DS. A large human domain antibody library combining heavy and light chain CDR3 diversity. Mol Immunol 2009; 47:912-21. [PMID: 19883941 DOI: 10.1016/j.molimm.2009.09.039] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2009] [Revised: 09/23/2009] [Accepted: 09/30/2009] [Indexed: 10/20/2022]
Abstract
Domain antibodies (dAbs) are promising candidate therapeutics and diagnostics. Efficient selection of novel potent dAbs with potential for clinical utility is critically dependent on the library diversity and size, and the scaffold stability. We have previously constructed a large (size approximately 2.5 x 10(10)) dAb library by grafting human antibody heavy chain complementarity determining regions (CDRs) 2 and 3 (H2s, H3s) into their cognate positions in a human heavy chain variable domain (VH) scaffold and mutagenizing the CDR1 (H1). High-affinity binders against some antigens were selected from this library but panning against others was not very successful likely due to limited diversity. We have hypothesized that by grafting highly variable, both in length and composition, human CDRs into non-cognate positions, the dAb library diversity could be significantly increased and the library would allow for more efficient selection of high-affinity antibodies against some targets. To test this hypothesis we designed a novel type of dAb library containing CDRs in non-cognate positions. It is based on our previous library where H1 was replaced by a library of human light chain CDR3s (L3s) thus combining three most diversified fragments (L3, H3 and H2) in one VH scaffold. This large (size approximately 10(10)) phage-displayed library was highly diversified as determined by analyzing the sequences of 126 randomly selected clones. Novel high-affinity dAbs against components of the human insulin-like growth factor (IGF) system were selected from the new library that could not be selected from the previously constructed one. Most of the newly identified dAbs were highly soluble, expressible, monomeric and may have potential as candidate cancer therapeutics. The new library could be used not only for the selection of such dAbs thus complementing existing libraries but also as a research tool for the exploration of the mechanisms determining folding and stability of human antibody domains.
Collapse
Affiliation(s)
- Weizao Chen
- Protein Interactions Group, Center for Cancer Research Nanobiology Program, National Cancer Institute-Frederick, National Institutes of Health, Frederick, MD 21702-1201, USA
| | | | | | | |
Collapse
|
79
|
Zoller F, Eisenhut M, Haberkorn U, Mier W. Endoradiotherapy in cancer treatment--basic concepts and future trends. Eur J Pharmacol 2009; 625:55-62. [PMID: 19836381 DOI: 10.1016/j.ejphar.2009.05.035] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2009] [Revised: 05/07/2009] [Accepted: 05/18/2009] [Indexed: 11/24/2022]
Abstract
Endoradiotherapy represents an alternative therapeutic method in cancer treatment with advantageous features compared to chemotherapy and radiation therapy. Intelligent dose delivery concepts using small drugs, peptides or antibodies as radionuclide carriers enable the verification of a selective accumulation in the tumour lesion and to reduce radiation toxicity for the peripheral organs. The development of endoradiotherapeutic agents, especially chelator-conjugated biomolecules, for example ibritumomab tiuxetan or DOTATOC, gains importance due to the stable complexation of versatile radiometals, such as (90)Y or (177)Lu. The rational design of novel target binding sides and their grafting into a drug scaffold is a highly promising strategy, which may promote further implication in endoradiotherapy. This review highlights the basic concepts of endoradiotherapy and discusses the potential of targeted therapy and the properties of energy-rich particles emitted by radionuclides for tumour therapy.
Collapse
Affiliation(s)
- Frederic Zoller
- Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Centre, INF 280, 69120 Heidelberg, Germany
| | | | | | | |
Collapse
|
80
|
Cheng Z, Campbell RE. An engineered tryptophan zipper-type peptide as a molecular recognition scaffold. J Pept Sci 2009; 15:523-32. [PMID: 19551843 DOI: 10.1002/psc.1153] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
In an effort to develop a structured peptide scaffold that lacks a disulfide bond and is thus suitable for molecular recognition applications in the reducing environment of the cytosol, we investigated engineered versions of the trpzip class of beta-hairpin peptides. We have previously shown that even most highly folded members of the trpzip class (i.e. the 16mer peptide HP5W4) are substantially destabilized by the introduction of mutations in the turn region and therefore not an ideal peptide scaffold. To address this issue, we used a FRET-based live cell screening system to identify extended trpzip-type peptides with additional stabilizing interactions. One of the most promising of these extended trpzip-type variants is the 24mer xxtz1-peptide with the sequence KAWTHDWTWNPATGKWTWLWRKNK. A phage display library of this peptide with randomization of six residues with side chains directed towards one face of the hairpin was constructed and panned against immobilized streptavidin. We have also explored the use of xxtz1-peptide for the presentation of an unstructured peptide 'loop' inserted into the turn region. Although NMR analysis provided no direct evidence for structure in the xxtz1-peptide with the loop insertion, we did attempt to use this construct as a scaffold for phage display of randomized peptide libraries. Panning of the resulting libraries against streptavidin resulted in the identification of peptide sequences with submicromolar affinities. Interestingly, substitution of key residues in the hairpin-derived portion of the peptide resulted in a 400-fold decrease in K(d), suggesting that the hairpin-derived portion plays an important role in preorganization of the loop region for molecular recognition.
Collapse
Affiliation(s)
- Zihao Cheng
- Department of Chemistry, University of Alberta, Edmonton, Alberta T6G2G2, Canada
| | | |
Collapse
|
81
|
Cheng Z, De Jesus OP, Kramer DJ, De A, Webster JM, Gheysens O, Levi J, Namavari M, Wang S, Park JM, Zhang R, Liu H, Lee B, Syud FA, Gambhir SS. 64Cu-labeled affibody molecules for imaging of HER2 expressing tumors. Mol Imaging Biol 2009; 12:316-24. [PMID: 19779897 DOI: 10.1007/s11307-009-0256-6] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2009] [Revised: 06/14/2009] [Accepted: 07/09/2009] [Indexed: 11/29/2022]
Abstract
INTRODUCTION The development of molecular probes based on novel engineered protein constructs is under active investigation due to the great potential of this generalizable strategy for imaging a variety of tumor targets. DISCUSSION In this report, human epidermal growth factor receptor type 2 (HER2)-binding Affibody molecules were radiolabeled with (64)Cu and their imaging ability was further evaluated in tumor mice models to understand the promise and limitations of such probes. The anti-HER2 Affibody molecules in monomeric (Z(HER2:477)) and dimeric [(Z(HER2:477))(2)] forms were site specifically modified with the maleimide-functionalized chelator, 1,4,7,10-tetraazacyclododecane-1,4,7-tris(acetic acid)-10-acetate mono (N-ethylmaleimide amide) (Mal-DOTA). The resulting DOTA-Affibody conjugates were radiolabeled with (64)Cu and evaluated in nude mice bearing subcutaneous SKOV3 tumors. Biodistribution experiments showed that tumor uptake values of (64)Cu-DOTA-Z(HER2:477) and (64)Cu-DOTA-(Z(HER2:477))(2) were 6.12 +/- 1.44% and 1.46 +/- 0.50% ID/g, respectively, in nude mice (n = 3 each) at 4 h postinjection. Moreover, (64)Cu-labeled monomer exhibited significantly higher tumor/blood ratio than that of radiolabeled dimeric counterpart at all time points examined in this study. MicroPET imaging of (64)Cu-DOTA-Z(HER2:477) in SKOV3 tumor mice clearly showed good and specific tumor localization. This study demonstrates that (64)Cu-labeled Z(HER2:477) is a promising targeted molecular probe for imaging HER2 receptor expression in living mice. Further work is needed to improve the excretion properties, hence dosimetry and imaging efficacy, of the radiometal-based probe.
Collapse
Affiliation(s)
- Zhen Cheng
- Molecular Imaging Program at Stanford, Departments of Radiology and Bioengineering, Bio-X Program, Stanford University, California, CA 94305-5344, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
82
|
Engineering and characterization of a bispecific HER2 x EGFR-binding affibody molecule. Biotechnol Appl Biochem 2009; 54:121-31. [PMID: 19492986 DOI: 10.1042/ba20090096] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
HER2 (human epidermal-growth-factor receptor-2; ErbB2) and EGFR (epidermal-growth-factor receptor) are overexpressed in various forms of cancer, and the co-expression of both HER2 and EGFR has been reported in a number of studies. The simultaneous targeting of HER2 and EGFR has been discussed as a strategy with which to potentially increase efficiency and selectivity in molecular imaging and therapy of certain cancers. In an effort to generate a molecule capable of bispecifically targeting HER2 and EGFR, a gene fragment encoding a bivalent HER2-binding affibody molecule was genetically fused in-frame with a bivalent EGFR-binding affibody molecule via a (G4S)3 [(Gly4-Ser)3]-encoding gene fragment. The encoded 30 kDa affibody construct (ZHER2)2-(G4S)3-(ZEGFR)2, with potential for bs (bispecific) binding to HER2 and EGFR, was expressed in Escherichia coli and characterized in terms of its binding capabilities. The retained ability to bind HER2 and EGFR separately was demonstrated using both biosensor technology and flow-cytometric analysis, the latter using HER2- and EGFR-overexpressing cells. Furthermore, simultaneous binding to HER2 and EGFR was demonstrated in: (i) a sandwich format employing real-time biospecific interaction analysis where the bs affibody molecule bound immobilized EGFR and soluble HER2; (ii) immunofluorescence microscopy, where the bs affibody molecule bound EGFR-overexpressing cells and soluble HER2; and (iii) a cell-cell interaction analysis where the bs affibody molecule bound HER2-overexpressing SKBR-3 cells and EGFR-overexpressing A-431 cells. This is, to our knowledge, the first reported bs affinity protein with potential ability for the simultaneous targeting of HER2 and EGFR. The potential future use of this and similar constructs, capable of bs targeting of receptors to increase the efficacy and selectivity in imaging and therapy, is discussed.
Collapse
|
83
|
Ren G, Zhang R, Liu Z, Webster JM, Miao Z, Gambhir SS, Syud FA, Cheng Z. A 2-helix small protein labeled with 68Ga for PET imaging of HER2 expression. J Nucl Med 2009; 50:1492-9. [PMID: 19690041 DOI: 10.2967/jnumed.109.064287] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
UNLABELLED Affibody molecules are a class of scaffold proteins being developed into a generalizable approach to targeting tumors. Many 3-helix-based Affibody proteins have shown excellent in vivo properties for tumor imaging and therapy. By truncating one alpha-helix that is not responsible for receptor recognition in the Affibody and maturating the protein affinity through synthetic strategies, we have successfully identified in our previous research several small 2-helix proteins with excellent binding affinities to human epidermal growth factor receptor type 2 (HER2). With preferential properties such as faster blood clearance and tumor accumulation, lower immunogenic potential, and facile and economically viable synthetic schemes, we hypothesized that these 2-helix protein binders could become excellent molecular imaging probes for monitoring HER2 expression and modulation. METHODS In this study, a 2-helix small protein, MUT-DS, was chemically modified with a metal chelator, 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA). DOTA-MUT-DS was then site-specifically radiolabeled with an important PET radionuclide, (68)Ga. The resulting radiolabeled anti-HER2 2-helix molecule was further evaluated as a potential molecular probe for small-animal PET HER2 imaging in a SKOV3 tumor mouse model. RESULTS The 2-helix DOTA-MUT-DS showed high HER2-binding affinity (dissociation constant, 4.76 nM). The radiolabeled probe displayed high stability in mouse serum and specificity toward HER2 in cell cultures. Biodistribution and small-animal PET studies further showed that (68)Ga-DOTA-MUT-DS had rapid and high SKOV3 tumor accumulation and quick clearance from normal organs. The specificity of (68)Ga-DOTA-MUT-DS for SKOV3 tumors was confirmed by monitoring modulation of HER2 protein on treatment of tumor mice with heat shock protein 90 inhibitor 17-N,N-dimethyl ethylene diamine-geldanamycin in vivo. CONCLUSION This proof-of-concept research clearly demonstrated that synthetic 2-helix (68)Ga-DOTA-MUT-DS is a promising PET probe for imaging HER2 expression in vivo. The Affibody-derived small 2-helix protein scaffold has great potential for developing targeting agents for a variety of tumor-associated biomarkers.
Collapse
Affiliation(s)
- Gang Ren
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology and Bio-X Program, Stanford University, Stanford, California, USA
| | | | | | | | | | | | | | | |
Collapse
|
84
|
Quirk S, Zhong S, Hernandez R. De novoidentification of binding sequences for antibody replacement molecules. Proteins 2009; 76:693-705. [DOI: 10.1002/prot.22382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
85
|
Cicortas Gunnarsson L, Nordberg Karlsson E, Andersson M, Holst O, Ohlin M. Molecular engineering of a thermostable carbohydrate-binding module. BIOCATAL BIOTRANSFOR 2009. [DOI: 10.1080/10242420500518516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
86
|
Heinis C, Rutherford T, Freund S, Winter G. Phage-encoded combinatorial chemical libraries based on bicyclic peptides. Nat Chem Biol 2009; 5:502-7. [PMID: 19483697 DOI: 10.1038/nchembio.184] [Citation(s) in RCA: 567] [Impact Index Per Article: 35.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2008] [Accepted: 04/29/2009] [Indexed: 02/07/2023]
Abstract
Here we describe a phage strategy for the selection of ligands based on bicyclic or linear peptides attached covalently to an organic core. We designed peptide repertoires with three reactive cysteine residues, each spaced apart by several random amino acid residues, and we fused the repertoires to the phage gene-3-protein. Conjugation with tris-(bromomethyl)benzene via the reactive cysteines generated repertoires of peptide conjugates with two peptide loops anchored to a mesitylene core. Iterative affinity selections yielded several enzyme inhibitors; after further mutagenesis and selection, we were able to chemically synthesize a lead inhibitor (PK15; Ki =1.5 nM) specific to human plasma kallikrein that efficiently interrupted the intrinsic coagulation pathway in human plasma tested ex vivo. This approach offers a powerful means of generating and selecting bicyclic macrocycles (or if cleaved, linear derivatives thereof) as ligands poised at the interface of small-molecule drugs and biologics.
Collapse
Affiliation(s)
- Christian Heinis
- Laboratory of Molecular Biology, Medical Research Council, Cambridge, UK
| | | | | | | |
Collapse
|
87
|
Mirecka EA, Hey T, Fiedler U, Rudolph R, Hatzfeld M. Affilin Molecules Selected against the Human Papillomavirus E7 Protein Inhibit the Proliferation of Target Cells. J Mol Biol 2009; 390:710-21. [DOI: 10.1016/j.jmb.2009.05.027] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2009] [Revised: 05/10/2009] [Accepted: 05/15/2009] [Indexed: 01/18/2023]
|
88
|
Gong R, Vu BK, Feng Y, Prieto DA, Dyba MA, Walsh JD, Prabakaran P, Veenstra TD, Tarasov SG, Ishima R, Dimitrov DS. Engineered human antibody constant domains with increased stability. J Biol Chem 2009; 284:14203-10. [PMID: 19307178 PMCID: PMC2682868 DOI: 10.1074/jbc.m900769200] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2009] [Revised: 03/06/2009] [Indexed: 11/06/2022] Open
Abstract
The immunoglobulin (Ig) constant CH2 domain is critical for antibody effector functions. Isolated CH2 domains are promising as scaffolds for construction of libraries containing diverse binders that could also confer some effector functions. However, previous work has shown that an isolated murine CH2 domain is relatively unstable to thermally induced unfolding. To explore unfolding mechanisms of isolated human CH2 and increase its stability gamma1 CH2 was cloned and a panel of cysteine mutants was constructed. Human gamma1 CH2 unfolded at a higher temperature (T(m) = 54.1 degrees C, as measured by circular dichroism) than that previously reported for a mouse CH2 (41 degrees C). One mutant (m01) was remarkably stable (T(m) = 73.8 degrees C). Similar results were obtained by differential scanning calorimetry. This mutant was also significantly more stable than the wild-type CH2 against urea induced unfolding (50% unfolding at urea concentration of 6.8 m versus 4.2 m). The m01 was highly soluble and monomeric. The existence of the second disulfide bond in m01 and its correct position were demonstrated by mass spectrometry and nuclear magnetic resonance spectroscopy, respectively. The loops were on average more flexible than the framework in both CH2 and m01, and the overall secondary structure was not affected by the additional disulfide bond. These data suggest that a human CH2 domain is relatively stable to unfolding at physiological temperature, and that both CH2 and the highly stable mutant m01 are promising new scaffolds for the development of therapeutics against human diseases.
Collapse
Affiliation(s)
- Rui Gong
- Protein Interactions Group, CCRNP, CCR, National Institutes of Health, Frederick, Maryland 21702, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
89
|
Boschek CB, Apiyo DO, Soares TA, Engelmann HE, Pefaur NB, Straatsma TP, Baird CL. Engineering an ultra-stable affinity reagent based on Top7. Protein Eng Des Sel 2009; 22:325-32. [DOI: 10.1093/protein/gzp007] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
|
90
|
Uhlén M, Hober S. Generation and validation of affinity reagents on a proteome-wide level. J Mol Recognit 2009; 22:57-64. [PMID: 18546091 DOI: 10.1002/jmr.891] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
There is a need for protein-specific affinity reagents to explore the gene products encoded by the genome. Recently, systematic efforts to generate validated affinity reagents on a whole human proteome level have been initiated. There are several issues for such efforts, including choice of antigen, type of affinity reagent, and the subsequent validation of the generated protein-specific binders. The advantages and disadvantages with the different approaches are discussed and the problems related to quality assessment of antibodies to be used in multi-platform applications are addressed. This review also describes the efforts to create a virtual resource of validated antibodies using a community-based portal and summarizes the status and visions for the publicly available human protein atlas (http://www.proteinatlas.org) showing the human protein profiles in a large number of normal and cancer tissues as well as a large set of human cell lines.
Collapse
Affiliation(s)
- Mathias Uhlén
- Department of Proteomics, School of Biotechnology, Royal Institute of Technology (KTH), AlbaNova University Center, Stockholm, Sweden.
| | | |
Collapse
|
91
|
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: 76] [Impact Index Per Article: 4.8] [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]
|
92
|
Liang S, Li L, Hsu WL, Pilcher MN, Uversky V, Zhou Y, Dunker AK, Meroueh SO. Exploring the molecular design of protein interaction sites with molecular dynamics simulations and free energy calculations. Biochemistry 2009; 48:399-414. [PMID: 19113835 PMCID: PMC2754190 DOI: 10.1021/bi8017043] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The significant work that has been invested toward understanding protein-protein interaction has not translated into significant advances in structure-based predictions. In particular redesigning protein surfaces to bind to unrelated receptors remains a challenge, partly due to receptor flexibility, which is often neglected in these efforts. In this work, we computationally graft the binding epitope of various small proteins obtained from the RCSB database to bind to barnase, lysozyme, and trypsin using a previously derived and validated algorithm. In an effort to probe the protein complexes in a realistic environment, all native and designer complexes were subjected to a total of nearly 400 ns of explicit-solvent molecular dynamics (MD) simulation. The MD data led to an unexpected observation: some of the designer complexes were highly unstable and decomposed during the trajectories. In contrast, the native and a number of designer complexes remained consistently stable. The unstable conformers provided us with a unique opportunity to define the structural and energetic factors that lead to unproductive protein-protein complexes. To that end we used free energy calculations following the MM-PBSA approach to determine the role of nonpolar effects, electrostatics and entropy in binding. Remarkably, we found that a majority of unstable complexes exhibited more favorable electrostatics than native or stable designer complexes, suggesting that favorable electrostatic interactions are not prerequisite for complex formation between proteins. However, nonpolar effects remained consistently more favorable in native and stable designer complexes reinforcing the importance of hydrophobic effects in protein-protein binding. While entropy systematically opposed binding in all cases, there was no observed trend in the entropy difference between native and designer complexes. A series of alanine scanning mutations of hot-spot residues at the interface of native and designer complexes showed less than optimal contacts of hot-spot residues with their surroundings in the unstable conformers, resulting in more favorable entropy for these complexes. Finally, disorder predictions revealed that secondary structures at the interface of unstable complexes exhibited greater disorder than the stable complexes.
Collapse
Affiliation(s)
- Shide Liang
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202
- Center for Computational Biology and Bioinformatics, Indiana University Purdue University Indianapolis, Indianapolis, Indiana 46202
| | - Liwei Li
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202
- Center for Computational Biology and Bioinformatics, Indiana University Purdue University Indianapolis, Indianapolis, Indiana 46202
| | - Wei-Lun Hsu
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202
- Center for Computational Biology and Bioinformatics, Indiana University Purdue University Indianapolis, Indianapolis, Indiana 46202
| | - Meaghan N. Pilcher
- Center for Computational Biology and Bioinformatics, Indiana University Purdue University Indianapolis, Indianapolis, Indiana 46202
- Department of Chemistry, Indiana University Purdue University Indianapolis, Indianapolis, Indiana 46202
| | - Vladimir Uversky
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202
- Center for Computational Biology and Bioinformatics, Indiana University Purdue University Indianapolis, Indianapolis, Indiana 46202
| | - Yaoqi Zhou
- Center for Computational Biology and Bioinformatics, Indiana University Purdue University Indianapolis, Indianapolis, Indiana 46202
| | - A. Keith Dunker
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202
- Center for Computational Biology and Bioinformatics, Indiana University Purdue University Indianapolis, Indianapolis, Indiana 46202
| | - Samy O. Meroueh
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202
- Center for Computational Biology and Bioinformatics, Indiana University Purdue University Indianapolis, Indianapolis, Indiana 46202
- Department of Chemistry, Indiana University Purdue University Indianapolis, Indianapolis, Indiana 46202
| |
Collapse
|
93
|
Vernet E, Konrad A, Lundberg E, Nygren PÅ, Gräslund T. Affinity-based entrapment of the HER2 receptor in the endoplasmic reticulum using an affibody molecule. J Immunol Methods 2008; 338:1-6. [DOI: 10.1016/j.jim.2008.06.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2008] [Revised: 05/22/2008] [Accepted: 06/25/2008] [Indexed: 11/26/2022]
|
94
|
Björling E, Uhlén M. Antibodypedia, a portal for sharing antibody and antigen validation data. Mol Cell Proteomics 2008; 7:2028-37. [PMID: 18667413 DOI: 10.1074/mcp.m800264-mcp200] [Citation(s) in RCA: 107] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Antibodies are useful tools to characterize the components of the human proteome and to validate potential protein biomarkers discovered through various clinical proteomics efforts. The lack of validation results across various applications for most antibodies often makes it necessary to perform cumbersome investigations to ensure specificity of a particular antibody in a certain application. A need therefore exists for a standardized system for sharing validation data about publicly available antibodies and to allow antibody providers as well as users to contribute and edit experimental evidence data, including data also on the antigen. Here we describe a new publicly available portal called Antibodypedia, which has been developed to allow sharing of information regarding validation of antibodies in which providers can submit their own validation results and reliability scores. We report standardized validation criteria and submission rules for applications such as Western blots, protein arrays, immunohistochemistry, and immunofluorescence. The contributor is expected to provide experimental evidence and a validation score for each antibody, and the users can subsequently provide feedback and comments on the use of the antibody. The database thus provides a virtual resource of publicly available antibodies toward human proteins with accompanying experimental evidence supporting an individual validation score for each antibody in an application-specific manner.
Collapse
Affiliation(s)
- Erik Björling
- School of Biotechnology, Royal Institute of Technology, AlbaNova University Center, SE-106 91 Stockholm, Sweden
| | | |
Collapse
|
95
|
Abstract
The use of affinity-based tools has become invaluable as a platform for basic research and in the development of drugs and diagnostics. Applications include affinity chromatography and affinity tag fusions for efficient purification of proteins as well as methods to probe the protein network interactions on a whole-proteome level. A variety of selection systems has been described for in vitro evolution of affinity reagents using combinatorial libraries, which make it possible to create high-affinity reagents to virtually all biomolecules, as exemplified by generation of therapeutic antibodies and new protein scaffold binders. The strategies for high-throughput generation of affinity reagents have also opened up the possibility of generating specific protein probes on a whole-proteome level. Recently, such affinity proteomics have allowed the detailed analysis of human protein expression in a comprehensive manner both in normal and disease tissue using tissue microarrays and confocal microscopy.
Collapse
Affiliation(s)
- Mathias Uhlén
- School of Biotechnology, AlbaNova University Center, Royal Institute of Technology, Stockholm, Sweden.
| |
Collapse
|
96
|
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: 200] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
97
|
Borghouts C, Kunz C, Delis N, Groner B. Monomeric Recombinant Peptide Aptamers Are Required for Efficient Intracellular Uptake and Target Inhibition. Mol Cancer Res 2008; 6:267-81. [DOI: 10.1158/1541-7786.mcr-07-0245] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
98
|
Friedman M, Orlova A, Johansson E, Eriksson TLJ, Höidén-Guthenberg I, Tolmachev V, Nilsson FY, Ståhl S. Directed evolution to low nanomolar affinity of a tumor-targeting epidermal growth factor receptor-binding affibody molecule. J Mol Biol 2008; 376:1388-402. [PMID: 18207161 DOI: 10.1016/j.jmb.2007.12.060] [Citation(s) in RCA: 125] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2007] [Revised: 12/20/2007] [Accepted: 12/21/2007] [Indexed: 12/20/2022]
Abstract
The epidermal growth factor receptor 1 (EGFR) is overexpressed in various malignancies and is associated with a poor patient prognosis. A small, receptor-specific, high-affinity imaging agent would be a useful tool in diagnosing malignant tumors and in deciding upon treatment and assessing the response to treatment. We describe here the affinity maturation procedure for the generation of Affibody molecules binding with high affinity and specificity to EGFR. A library for affinity maturation was constructed by rerandomization of selected positions after the alignment of first-generation binding variants. New binders were selected with phage display technology, using a single oligonucleotide in a single-library effort, and the best second-generation binders had an approximately 30-fold improvement in affinity (K(d)=5-10 nM) for the soluble extracellular domain of EGFR in biospecific interaction analysis using Biacore. The dissociation equilibrium constant, K(d), was also determined for the Affibody with highest affinity using EGFR-expressing A431 cells in flow cytometric analysis (K(d)=2.8 nM). A retained high specificity for EGFR was verified by a dot blot assay showing staining only of EGFR proteins among a panel of serum proteins and other EGFR family member proteins (HER2, HER3, and HER4). The EGFR-binding Affibody molecules were radiolabeled with indium-111, showing specific binding to EGFR-expressing A431 cells and successful targeting of the A431 tumor xenografts with 4-6% injected activity per gram accumulated in the tumor 4 h postinjection.
Collapse
Affiliation(s)
- Mikaela Friedman
- Department of Molecular Biotechnology, AlbaNova University Center, Kungl Tekniska Högskolan (KTH), SE-106 91 Stockholm, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
99
|
Lindholm L, Henning P, Magnusson MK. Novel strategies in tailoring human adenoviruses into therapeutic cancer gene therapy vectors. Future Virol 2008. [DOI: 10.2217/17460794.3.1.45] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Gene therapy is a novel approach for the treatment of cancer that has so far not been realized. The scope of this review is to try to define the remaining barriers to the successful use of adenovirus vectors for gene and viral therapy of human tumors and to suggest solutions whereby these barriers can be bypassed. It is the conviction of the authors that too many studies have been performed in animal models that are not sufficiently comprehensive to allow conclusions to be drawn for application in humans. For example, in the case of the murine experimental model, in which most studies have been performed, mice are devoid of circulating antibodies to adenovirus type 5 and adenovirus cannot replicate in mouse cells. While the problems are real enough, as witnessed by the quite limited success in human trials, some of the solutions that will be suggested here are hypothetical and have not as yet been tried, even in animals. The review has no ambition to be exhaustive but is intended as a contribution in order to forward the field of gene therapy vectors for systemic clinical application.
Collapse
Affiliation(s)
- Leif Lindholm
- University of Goteborg, Institute for Biomedicine, Department of Microbiology & Immunology, PO Box 435, SE 40530 Goteborg, Sweden, and, Got-A-Gene AB, Östra Kyviksvägen 18, SE 42930 Kullavik, Sweden
| | - Petra Henning
- University of Goteborg, Institute for Biomedicine, Department of Microbiology & Immunology, PO Box 435, SE 40530 Goteborg, Sweden, and, Got-A-Gene AB, Östra Kyviksvägen 18, SE 42930 Kullavik, Sweden
| | - Maria K Magnusson
- University of Goteborg, Institute for Biomedicine, Department of Microbiology & Immunology, PO Box 435, SE 40530 Goteborg, Sweden, and, Got-A-Gene AB, Östra Kyviksvägen 18, SE 42930 Kullavik, Sweden
| |
Collapse
|
100
|
Stoevesandt O, Taussig MJ. Affinity reagent resources for human proteome detection: initiatives and perspectives. Proteomics 2007; 7:2738-50. [PMID: 17639606 DOI: 10.1002/pmic.200700155] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Essential to the ambition of characterising fully the human proteome are systematic and comprehensive collections of specific affinity reagents directed against all human proteins, including splice variants and modifications. Although a large number of affinity reagents are available commercially, their quality is often questionable and only a fraction of the proteome is covered. In order for more targets to be examined, there is a need for broad availability of panels of affinity reagents, including binders against proteins of unknown functions. The most familiar affinity reagents are antibodies and their fragments, but engineered forms of protein scaffolds and nucleic acid aptamers with similar diversity and binding properties are becoming viable alternatives. Recent initiatives in Europe and the USA have been established to improve both the availability and quality of reagents for affinity proteomics, with the ultimate aim of creating standardised collections of well-validated binding molecules for proteome analysis. As well as coordinating affinity reagent production through existing resources and technology providers, these projects aim to benchmark key molecular entities, tools, and applications, and establish the bioinformatics framework and databases needed. The benefits of such reagent resources will be seen in basic research, medicine and the biotechnology and pharmaceutical industries.
Collapse
Affiliation(s)
- Oda Stoevesandt
- Technology Research Group, The Babraham Institute, Cambridge, UK
| | | |
Collapse
|