Please wait a minute...

中国生物工程杂志

China Biotechnology
China Biotechnology  2013, Vol. 33 Issue (1): 95-103    DOI:
    
Overview of Scaffold Protein Used for Selection of Artificial Binding Proteins
YUAN Li, DAI He-ping
State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
Download: HTML   PDF(701KB) HTML
Export: BibTeX | EndNote (RIS)      

Abstract  As the most well-known native binding proteins, antibodies with binding ability to antigen are extensively used in biotechnological and medical applications for over one hundred years. However, the intrinsic limitations of antibodies restrict its applications in many fields. Artificial binding proteins not only have the properties of antibodies, but also are possess of more advantages: smaller size, higher stability, E. Coli production with high throughput and high solubility, ease of modification, high affinity and high specificity, no IP conflict with antibody, therefore it was also called as ideal "the next generational antibody". Artificial binding proteins were selected from protein scaffolds library which was constructed by gene engineering based on stable protein scaffolds. This review will focus on following points: the concept and design of protein scaffold, classify of protein scaffolds, screening technologies of artificial binding proteins based on protein scaffold and application and development of artificial binding proteins.

Key wordsAntibody      Artificial binding protein      Scaffold protein      Phage display     
Received: 12 September 2012      Published: 25 January 2013
ZTFLH:  Q511  
Cite this article:

YUAN Li, DAI He-ping. Overview of Scaffold Protein Used for Selection of Artificial Binding Proteins. China Biotechnology, 2013, 33(1): 95-103.

URL:

https://manu60.magtech.com.cn/biotech/     OR     https://manu60.magtech.com.cn/biotech/Y2013/V33/I1/95

[1] Leader B, Baca Q J, Golan D E. Protein therapeutics: a summary and pharmacological classification. Nature Reviews Drug discovery, 2008, 7:21-39.
[2] Ku J, Schultz P G. Alternate protein frameworks for molecular recognition. Proc Natl Acad Sci U S A, 1995, 92:6552-6556.
[3] Lofblom J, Frejd F Y, Stahl S. Non-immunoglobulin based protein scaffolds. Curr Opin Biotechnol, 2011, 22:843-848.
[4] Nygren P A, Skerra A. Binding proteins from alternative scaffolds. Journal of Immunological Methods, 2004, 290:3-28.
[5] Hey T, Fiedler E, Rudolph R, et al. Artificial, non-antibody binding proteins for pharmaceutical and industrial applications. Trends Biotechnol, 2005, 23:514-522.
[6] Binz H K, Amstutz P, Pluckthun A. Engineering novel binding proteins from nonimmunoglobulin domains. Nature Biotechnology, 2005, 23:1257-1268.
[7] Gebauer M, Skerra A. Engineered protein scaffolds as next-generation antibody therapeutics. Curr Opin Chem Biol, 2009, 13:2452-2455.
[8] 杜建芳, 王金凤, 孙万军, 等. 以Trx为骨架构建可诱导表达的酵母构象型随机肽库. 军事医学科学院院刊, 2006, 30 (3):201-205. Du J F, Wang J F, Sun W J, et al. Construct ion of an inducible yeast random conformat ion-constrained peptide library based on Trx scaffold. Bull Acad Mil Med Sci, 2006, 30 (3):201-205.
[9] 赵安, 薛沿宁, 冯健男, 等. 昆虫防御素A的分子改造及其在噬菌体表面的展示. 军事医学院院刊, 2002, 26 (4):270-272. Zhao A, Xue Y N, Feng J N, et al. Rational design of the insect defensin A and its displaying on the surface of phage. Bull Acad Mil Med Sci, 2002, 26 (4):270-272.
[10] Reichert J M, Valge-Archer V E. Development trends for monoclonal antibody cancer therapeutics. Nature Reviews Drug Discovery, 2007, 6:349-356.
[11] Kohler G, Milstein C. Continuous cultures of fused cells secreting antibody of predefined specificity. Nature, 1975, 256:495-497.
[12] Smith G P. Filamentous fusion phage: novel expression vectors that display cloned antigens on the virion surface. Science, 1985, 228:1315-1317.
[13] McCafferty J, Griffiths A D, Winter G, et al. Phage antibodies: filamentous phage displaying antibody variable domains. Nature, 1990, 348:552-524.
[14] Ebersbach H, Fiedler E, Scheuermann T, et al. Affilin-novel binding molecules based on human gamma-B-crystallin, an All beta-Sheet Protein. J Mol Biol, 2007, 372:172-185.
[15] Hackel B J, Kapila A, Wittrup K D. Picomolar affinity fibronectin domains engineered utilizing loop length diversity, recursive mutagenesis, and loop shuffling. J Mol Biol, 2008, 381:1238-1252.
[16] Koide A, Abbatiello S, Rothgery L, et al. Probing protein conformational changes in living cells by using designer binding proteins: application to the estrogen receptor. Proc Natl Acad Sci U S A, 2002, 99:1253-1258.
[17] Amstutz P, Binz H K, Parizek P, et al. Intracellular kinase inhibitors selected from combinatorial libraries of designed ankyrin repeat proteins. J Biol Chem, 2005, 280:24715-24722.
[18] Roberts B L, Markland W, Ley A C, et al. Directed evolution of a protein: selection of potent neutrophil elastase inhibitors displayed on M13 fusion phage. Proc Natl Acad Sci U S A, 1992, 89:2429-2433.
[19] Dennis M S, Herzka A, Lazarus R A. Potent and selective Kunitz domain inhibitors of plasma kallikrein designed by phage display. J Biol Chem, 1995, 270:25411-25417.
[20] Williams A, Baird L G. DX-88 and HAE: a developmental perspective. Transfus Apher Sci, 2003, 29:255-258..
[21] Wark P A. DX-890 (Dyax). IDrugs: the investigational drugs journal, 2002, 5:586-589.
[22] Baggio R, Burgstaller P, Hale S P, et al. Identification of epitope-like consensus motifs using mRNA display. J Mol Recognit, 2002, 15:126-134.
[23] Hilpert K, Wessner H, Schneider-Mergener J, et al. Design and characterization of a hybrid miniprotein that specifically inhibits porcine pancreatic elastase. J Biol Chem, 2003, 278:24986-24993.
[24] Christmann A, Walter K, Wentzel A, et al. The cystine knot of a squash-type protease inhibitor as a structural scaffold for Escherichia coli cell surface display of conformationally constrained peptides. Protein Engineering, 1999, 12:797-806.
[25] Skerra A. Alternative binding proteins: anticalins - harnessing the structural plasticity of the lipocalin ligand pocket to engineer novel binding activities. FEBS J, 2008, 275:2677-2683.
[26] Gebauer M, Skerra A. Anticalins small engineered binding proteins based on the lipocalin scaffold. Methods Enzymol, 2012, 503:157-188.
[27] Hohlbaum A M, Skerra A. Anticalins: the lipocalin family as a novel protein scaffold for the development of next-generation immunotherapies. Expert Review of Clinical Immunology, 2007, 3:491-501.
[28] Liu J, Ning B, Liu M, et al. Construction of ribosome display library based on lipocalin scaffold and screening anticalins with specificity for estradiol. The Analyst, 2012, 137:2470-2479.
[29] Mercader J V, Skerra A. Generation of anticalins with specificity for a nonsymmetric phthalic acid ester. Anal Biochem, 2002, 308:269-277.
[30] Schlehuber S, Skerra A. Anticalins as an alternative to antibody technology. Expert Opinion on Biological Therapy, 2005, 5:1453-1462.
[31] Schonfeld D, Matschiner G, Chatwell L, et al. An engineered lipocalin specific for CTLA-4 reveals a combining site with structural and conformational features similar to antibodies. Proc Natl Acad Sci U S A, 2009, 106:8198-8203.
[32] Kim H J, Eichinger A, Skerra A. High-affinity recognition of lanthanide(III) chelate complexes by a reprogrammed human lipocalin 2. J Am Chem Soc, 2009, 131:3565-3576.
[33] Schlehuber S, Skerra A. Anticalins in drug development. BioDrugs: clinical immunotherapeutics. Biopharmaceuticals and Gene Therapy, 2005, 19:279-288.
[34] Hufton S E, van Neer N, van den Beuken T, et al. Development and application of cytotoxic T lymphocyte-associated antigen 4 as a protein scaffold for the generation of novel binding ligands. FEBS letters, 2000, 475:225-231.
[35] Irving R A, Coia G, Roberts A, et al. Ribosome display and affinity maturation: from antibodies to single V-domains and steps towards cancer therapeutics. Journal of Immunological Methods, 2001, 248:31-45.
[36] Koide A, Bailey C W, Huang X, et al. The fibronectin type III domain as a scaffold for novel binding proteins. J Mol Biol, 1998, 284:1141-1151.
[37] Binz H K, Amstutz P, Kohl A, et al. High-affinity binders selected from designed ankyrin repeat protein libraries. Nature Biotechnology, 2004, 22:575-582.
[38] Tamaskovic R, Simon M, Stefan N, et al. Designed ankyrin repeat proteins (DARPins) from research to therapy. Methods Enzymol, 2012, 503:101-134.
[39] Binz H K, Stumpp M T, Forrer P, et al. Designing repeat proteins: well-expressed, soluble and stable proteins from combinatorial libraries of consensus ankyrin repeat proteins. J Mol Biol, 2003, 332:489-503.
[40] Boersma Y L, Pluckthun A. DARPins and other repeat protein scaffolds: advances in engineering and applications. Curr Opin Biotechnol, 2011, 22:849-857.
[41] Seeger M A, Mittal A, Velamakanni S, et al. Tuning the drug efflux activity of an ABC transporter in vivo by in vitro selected DARPin binders. PLoS One, 2012, 7:e37845.
[42] Steiner D, Forrer P, Pluckthun A. Efficient selection of DARPins with sub-nanomolar affinities using SRP phage display. J Mol Biol, 2008, 382:1211-1227.
[43] Stumpp M T, Binz H K, Amstutz P. DARPins: a new generation of protein therapeutics. Drug Discovery Today, 2008, 13:695-701.
[44] Nord K, Nilsson J, Nilsson B, et al. A combinatorial library of an alpha-helical bacterial receptor domain. Protein Engineering, 1995, 8:601-608.
[45] Nord K, Gunneriusson E, Ringdahl J, et al. Binding proteins selected from combinatorial libraries of an alpha-helical bacterial receptor domain. Nature Biotechnology, 1997, 15:772-777.
[46] Wahlberg E, Lendel C, Helgstrand M, et al. An affibody in complex with a target protein: structure and coupled folding. Proc Natl Acad Sci U S A, 2003, 100:3185-190.
[47] Mirecka E A, Hey T, Fiedler U, et al. Affilin molecules selected against the human papillomavirus E7 protein inhibit the proliferation of target cells. J Mol Biol, 2009, 390:710-721.
[48] Hoffmann A, Kovermann M, Lilie H, et al. New binding mode to TNF-alpha revealed by ubiquitin-based artificial binding protein. PLoS One, 2012, 7:e31298.
[49] Nilsson B, Berman-Marks C, Kuntz I D, et al. Secretion incompetence of bovine pancreatic trypsin inhibitor expressed in Escherichia coli. J Biol Chem, 1991, 266:2970-2977.
[50] Teter S A, Klionsky D J. How to get a folded protein across a membrane. Trends in Cell Biology, 1999, 9:428-431.
[51] Valent Q A. Signal recognition particle mediated protein targeting in Escherichia coli. Antonie van Leeuwenhoek, 2001, 79:17-31.
[52] Nangola S, Minard P, Tayapiwatana C. Appraisal of translocation pathways for displaying ankyrin repeat protein on phage particles. Protein Expression and Purification, 2010, 74:156-161.
[53] Steiner D, Forrer P, Stumpp M T, et al. Signal sequences directing cotranslational translocation expand the range of proteins amenable to phage display. Nature Biotechnology, 2006, 24:823-831.
[54] Droge M J, Boersma Y L, Braun P G, et al. Phage display of an intracellular carboxylesterase of Bacillus subtilis: comparison of Sec and Tat pathway export capabilities. Applied and Environmental Microbiology, 2006, 72:4589-4595.
[55] Velappan N, Fisher H E, Pesavento E, et al. A comprehensive analysis of filamentous phage display vectors for cytoplasmic proteins: an analysis with different fluorescent proteins. Nucleic Acids Res, 2010, 38:e22.
[56] Paschke M, Hohne W. A twin-arginine translocation (Tat)-mediated phage display system. Gene, 2005, 350:79-88.
[57] Speck J, Arndt K M, Muller K M. Efficient phage display of intracellularly folded proteins mediated by the TAT pathway. Protein Eng Des Sel, 2011, 24:473-484.
[58] Krumpe L R, Atkinson A J, Smythers G W, et al. T7 lytic phage-displayed peptide libraries exhibit less sequence bias than M13 filamentous phage-displayed peptide libraries. Proteomics, 2006, 6:4210-4222.
[59] Herman R E, Badders D, Fuller M, et al. The Trp cage motif as a scaffold for the display of a randomized peptide library on bacteriophage T7. J Biol Chem, 2007, 282:9813-9824.
[60] Dai M, Temirov J, Pesavento E, et al. Using T7 phage display to select GFP-based binders. Protein Eng Des Sel, 2008, 21:413-424.
[61] Beck A, Wurch T, Bailly C, et al. Strategies and challenges for the next generation of therapeutic antibodies. Nature Reviews Immunology, 2010, 10:345-352.
[62] Tolmachev V. Imaging of HER-2 overexpression in tumors for guiding therapy. Current Pharmaceutical Design, 2008, 14:2999-3019.
[63] Miao Z, Levi J, Cheng Z. Protein scaffold-based molecular probes for cancer molecular imaging. Amino Acids, 2011, 41:1037-1047.
[64] Sennhauser G, Grutter M G. Chaperone-assisted crystallography with DARPins. Structure, 2008, 16:1443-1453.
[65] Huber T, Steiner D, Rothlisberger D, et al. In vitro selection and characterization of DARPins and Fab fragments for the co-crystallization of membrane proteins: The Na(+)-citrate symporter CitS as an example. J Struct Biol, 2007, 159:206-221.
[66] Nord K, Gunneriusson E, Uhlen M, et al. Ligands selected from combinatorial libraries of protein A for use in affinity capture of apolipoprotein A-1M and taq DNA polymerase. Journal of Biotechnology, 2000, 80:45-54.
[67] Reina J, Lacroix E, Hobson S D, et al. Computer-aided design of a PDZ domain to recognize new target sequences. Nature Structural Biology, 2002, 9:621-627.
[1] CHEN Xiu-yue,ZHOU Wen-feng,HE Qing,SU Bing,ZOU Ya-wen. Preparation, Purification and Identification of Bacteriophage Qβ Virus-like Particles[J]. China Biotechnology, 2021, 41(7): 42-49.
[2] CHEN Wen-jie,MIAO Xian-feng. Domestic Research and Development Status of Antibody-drug Conjugates and Strategic Layout of Key Enterprises[J]. China Biotechnology, 2021, 41(6): 105-110.
[3] XU Ye-chun,LIU Hong,LI Jian-feng,SHEN Jing-shan,JIANG Hua-liang. Recent Progress in Drug Development against COVID-19[J]. China Biotechnology, 2021, 41(6): 111-118.
[4] YUAN Bo,WANG Jie-wen,KANG Guang-bo,HUANG He. Research Progress and Application of Bispecific Nanobody[J]. China Biotechnology, 2021, 41(2/3): 78-88.
[5] MAO Kai-yun,LI Rong,LI Dan-dan,ZHAO Ruo-chun,FAN Yue-lei,JIANG Hong-bo. Analysis of the Current Status of Global Bispecific Antibody Development[J]. China Biotechnology, 2021, 41(11): 110-118.
[6] ZHAO Yan-shu,ZHANG Jin-hua,SONG Hao. Advances in Production of Monoclonal Antibody and Antibody Fragments in Engineered Prokaryotes and Yeast[J]. China Biotechnology, 2020, 40(8): 74-83.
[7] YANG Xiao-ying,LI Meng,ZHAO Wei,TANG Min,ZHANG Zhi-qian. Preparation and Preliminary Characterization of Anti-α2δ1/CD3 Bispecific Antibody[J]. China Biotechnology, 2020, 40(7): 9-14.
[8] WU Rui-jun,LI Zhi-fei,ZHANG Xin,PU Run,AO Yi,SUN Yan-rong. Development and Prospect of Antibody Drugs for SARS-CoV-2[J]. China Biotechnology, 2020, 40(5): 1-6.
[9] WANG Meng,SONG Hui-ru,CHENG Yu-jie,WANG Yi,YANG Bo,HU Zheng. Accurate Detection of Streptococcus pneumoniae by Using Ribosomal Protein L7 / L12 as Molecular Marker[J]. China Biotechnology, 2020, 40(4): 34-41.
[10] YANG Li,SHI Xiao-yu,LI Wen-lei,LI Jian,XU Han-mei. Optimization of Electroporation Conditions in Construction of Phage Display Antibody Library[J]. China Biotechnology, 2020, 40(4): 42-48.
[11] LI Tong-tong,SONG Cai-ling,YANG Kai-yue,WANG Wen-jing,CHEN Hui-yu,LIU Ming. Preparation and Neutralization Activity of Anti-Canine Parvovirus VP2 Protein Single-chain Antibody[J]. China Biotechnology, 2020, 40(4): 10-16.
[12] CHEN Qiu-li,YANG Li-chao,LI Hui,WEN Sha,LI Gang,HE Min. Prokaryotic Expression,Purification and Preparation of Polyclonal Antibody of Human Nek2 Protein[J]. China Biotechnology, 2020, 40(3): 31-37.
[13] LI Dong-qiao,LV Lu-cheng,YANG Yan-ping. The Research Status and Development Trend of Global Human Coronavirus Antibody Field[J]. China Biotechnology, 2020, 40(1-2): 65-70.
[14] KONG Jian-tao,ZHUANG Ying-ping,GUO Mei-jin. Enhancement of Anti-CD20 Monoclonal Antibody Expression by CHO based on DOE and Amino Acid Supplemental Strategy[J]. China Biotechnology, 2020, 40(12): 41-48.
[15] HE Meng,ZHANG Guo-lin,LI Yan,HAN Xue-bo,LIU Hong-peng,LI Xin,QIAN Ling-ling,LIU Kun-mei,GUO Le. Soluble Expression of Recombinant Antigen CagL from Helicobacter pylori Pathogenicity Island and Preparation and Analysis of Anti-CagA Polyclonal Antibody[J]. China Biotechnology, 2020, 40(11): 21-27.