throbber
610
`
`Phage display in pharmaceutical biotechnology
`Sachdev S Sidhu
`
`Over the past year, methods for the construction of M13
`phage-display libraries have been significantly improved and
`new display formats have been developed. Phage-displayed
`peptide libraries have been used to isolate specific ligands for
`numerous protein targets. New phage antibody libraries have
`further expanded the practical applications of the technology
`and phage cDNA libraries have proven useful in defining
`natural binding interactions. In addition, phage-display methods
`have been developed for the rapid determination of binding
`energetics at protein–protein interfaces.
`
`Developments in phage-display technology
`Library construction
`The success of any selection experiment ultimately
`depends on the diversity and quality of the initial library.
`Over the years, methods have been refined to the point
`where extremely large libraries can now be rapidly and
`reliably constructed. Sidhu et al. [6•,7] have described opti-
`mized methods that enable the construction of libraries
`with diversities greater than 1012, almost 100-fold greater
`than previously thought practical.
`
`Addresses
`Department of Protein Engineering, Genentech, Inc., 1 DNA Way,
`South San Francisco, CA 94080, USA; e-mail: sidhu@gene.com
`
`Current Opinion in Biotechnology 2000, 11:610–616
`
`0958-1669/00/$ — see front matter
`© 2000 Elsevier Science Ltd. All rights reserved.
`
`Abbreviation
`VEGF
`vascular endothelial growth factor
`
`Introduction
`Phage display is a powerful technology for selecting and
`engineering polypeptides with novel functions. If DNA
`fragments encoding polypeptides are fused to certain bac-
`teriophage coat protein genes, the fusion genes can be
`encapsulated within phage particles that also display the
`encoded polypeptides on their surfaces. This establishes a
`physical linkage between phenotype and genotype.
`Highly diverse libraries can be constructed by fusing
`degenerate DNA to a coat protein gene, and library mem-
`bers with desired binding specificities can be isolated by
`binding to an immobilized receptor in vitro. The
`sequences of selected polypeptides can be determined
`from the sequence of the encapsulated, encoding DNA.
`
`Phage display was first developed with the Escherichia coli-
`specific bacteriophage M13 [1], and the success of M13
`phage display has prompted the development of numerous
`alternative display systems. These include systems that uti-
`lize other E. coli-specific phage, such as λ-phage [2] and T4
`phage [3], and also systems that use eukaryotic viruses [4].
`In addition, polypeptides have been displayed on the sur-
`faces of bacteria and yeast [5]. Although these alternative
`systems have proven advantageous in special applications,
`M13 phage display remains the dominant technology.
`
`This review covers developments in M13 phage display
`made over the past year. I discuss technological improve-
`ments that enable the construction of larger libraries and
`new display formats that extend the technology to new
`applications. I also highlight some important applications
`of peptide and protein phage display, with particular
`emphasis on pharmaceutical biotechnology.
`
`Vectors and display formats
`The M13 phage particle consists of a single-stranded DNA
`core surrounded by a coat composed of five different pro-
`teins (Figure 1). The length of the filament is covered by
`several thousand copies of the major coat protein, pro-
`tein-8 (P8). Each end of the particle is capped by five
`copies each of two minor coat proteins: protein-3 (P3) and
`protein-6 (P6) at one end, and protein-7 (P7) and protein-
`9 (P9) at the other end [8]. In early examples of phage
`display, polypeptides were fused to the amino-terminus of
`either P3 or P8 in the viral genome [9,10]. These systems
`were severely limited because large polypeptides (>10
`residues for P8 display) compromised coat protein function
`and so could not be efficiently displayed. The develop-
`ment of phagemid display systems solved this problem
`because, in such systems, polypeptides were fused to an
`additional coat protein gene encoded by a phagemid vec-
`tor [11]. Subsequent infection with a helper phage
`produced particles with phagemid DNA encapsulated in a
`coat composed mainly of wild-type coat proteins from the
`helper phage but also containing some fusion coat proteins
`from the phagemid (Figure 1).
`
`Phagemid systems permit the display of polypeptides that
`could not be displayed in simple phage systems, because
`the deleterious effects of the fusion protein are attentuat-
`ed by the presence of wild-type coat proteins from the
`helper phage. Such systems have also enabled the devel-
`opment of new display formats. Jespers et al. [12] showed
`that proteins could be displayed as fusions to the carboxyl-
`terminus of P6. More recently, Gao et al. [13] have
`demonstrated the display of antibody fragments fused to
`the amino-terminus of P7 and P9, whereas Fuh et al. [14••]
`have demonstrated carboxy-terminal P8 display. In addi-
`tion, we have found that polypeptides fused to the
`carboxyl-terminus of P3 are displayed at levels comparable
`to conventional amino-terminal fusions [15]. Thus, in
`phagemid systems, functional polypeptide display has now
`been demonstrated with all five M13 coat proteins.
`
`Although conventional amino-terminal display formats are
`likely to dominate established applications, carboxy-termi-
`nal display enables studies unsuited to amino-terminal
`
`Lassen - Exhibit 1027, p. 1
`
`

`

`display. These include the study of protein–protein inter-
`actions requiring free carboxy-termini and functional
`cDNA cloning (see below). Also, carboxy-terminal display
`may be especially useful for the display of intracellular pro-
`teins. Before phage assembly, P3 and P8 reside in the
`E. coli inner membrane with their carboxy-termini in the
`cytoplasm and their amino-termini in the periplasm [16].
`Amino-terminal display has worked well for the display of
`secreted proteins evolved for folding in the oxidizing
`periplasmic environment, but few intracellular proteins
`have been displayed in this manner. In contrast, carboxy-
`terminal fusions would allow for folding in the reducing
`cytoplasm, and this may be ideal for intracellular proteins.
`
`Engineering the M13 coat for phage display
`In a phagemid system where wild-type P8 from the helper
`phage maintains the integrity of the phage coat, Sidhu et al.
`[17] have shown that an additional phagemid-encoded P8
`can tolerate a surprisingly large number of mutations.
`Furthermore, some of these mutations actually promote
`incorporation into the phage coat and thus increase the dis-
`play of fusion proteins. Subsequent mutational analyses
`demonstrated that only a small subset of the first 30 P8
`residues is critical for coat incorporation [18]. Weiss and
`Sidhu [19••] have further demonstrated the malleability of
`the phage coat by evolving completely artificial coat pro-
`teins that resemble P8 in an inverted orientation. These
`artificial coat proteins incorporate into the phage coat and
`permit the display of carboxy-terminal-fused polypeptides.
`Together, these reports show that the phage coat is
`extremely tolerant to the addition of new proteins, and
`these proteins can be specifically engineered for improved
`phage display.
`
`Applications for phage-displayed peptide
`libraries
`Phage-displayed peptide libraries can be used to isolate
`peptides that bind with high specificity and affinity to vir-
`tually any target protein. These binding peptides can be
`used as reagents to understand molecular recognition, as
`minimized mimics for receptors, or as lead molecules in
`drug design.
`
`Mimics of extracellular protein–protein interactions
`Extracellular protein–protein interactions often involve
`large, flat contact surfaces. Conventional small-molecule
`screening efforts that work well against concave surfaces
`have largely failed in targeting these interactions [20], but
`in contrast, phage-displayed peptide libraries have proven
`remarkably successful. Numerous phage-derived peptides
`that bind extracellular protein surfaces have been report-
`ed (reviewed in [6]), and several three-dimensional
`structures have also been solved (Figure 2). Ferrer and
`Harrison [21••] isolated peptides that bind to HIV gp120
`and inhibit its interaction with CD4. Although this inter-
`action has been a prime target for antiviral drug
`development, previous efforts had been largely unsuc-
`cessful because of the extended nature of the binding
`
`Phage display in pharmaceutical biotechnology Sidhu 611
`
`Figure 1
`
`Promoter
`
`Secretion
`signal
`
`Displayed
`protein
`
`M13 coat protein
`
`Ampr
`
`Phagemid
`vector
`
`322 ori
`
`
`
`f1 ori
`
`Escherichia coli
`
`
`
`Recycle
`
`Add helper phage
`
`P7 and P9
`
`P3 and P6
`
`P8
`
`Bind to receptor
`
` Binders
`
`Current Opinion in Biotechnology
`
`An M13 phagemid vector designed for phage display. A phagemid
`vector contains origins of single-stranded (f1 ori) and double-stranded
`(322 ori) DNA replication and a selective marker, such as the
`β-lactamase gene (Ampr) which confers resistance to ampicillin. For
`phage display, the phagemid also contains a cassette consisting of a
`promoter that drives transcription of an open reading frame encoding a
`secretion signal and the displayed protein fused to an M13 coat
`protein. The vector replicates in E. coli as a double-stranded plasmid,
`but coinfection with a helper phage results in the production of single-
`stranded DNA that is packaged into phage particles. The phage coat
`contains five different proteins, and polypeptides can be displayed as
`either amino-terminal fusions (with P3, P8, P7, or P9) or carboxy-
`terminal fusions (with P6, P8, or P3). The phage particles can be used
`in binding selections, and binding clones can be amplified by recycling
`through an E. coli host.
`
`surface [21••]. Phage display probably suceeded because
`peptidic ligands can adopt extended conformations that
`effectively compliment such extended surfaces.
`
`In theory, peptides could bind to a protein anywhere on its
`solvent-exposed surface. Thus, it is notable that most select-
`ed peptides bind at sites that coincide with natural
`ligand-binding sites, and consequently act as antagonists or
`agonists of natural protein–protein interactions. It seems that
`natural binding sites possess features that predispose them for
`ligand binding. Delano et al. [22••] isolated peptides that
`bound to the constant fragment of immunoglobulin G (IgG-
`Fc) and found that a ‘consensus’ binding site that interacts
`with at least four natural proteins was also the preferred site
`
`Lassen - Exhibit 1027, p. 2
`
`

`

`612 Pharmaceutical biotechnology
`
`Figure 2
`
`(a)
`
`(b)
`
`(c)
`
`(d)
`
`(e)
`
`Structures of binding peptides isolated from
`phage-displayed peptide libraries. Mainchains
`are shown as dark grey ribbons and sidechains
`are shown in light grey. Each peptide contains
`a single disulfide bond. Structures (a) and
`(b) were determined by NMR with the free
`peptides in solution. The other structures were
`determined by X-ray crystallography with the
`peptides in complex with their cognate ligands.
`(a) A turn-helix conformation is adopted by a
`peptide that binds to the insulin-like growth
`factor binding protein 1 [49]. (b) Within the
`disulfide-bonded loop, a FVIIa-binding peptide
`consists of a type I reverse turn and an
`irregular turn of a helix that extends to the
`carboxyl-terminus [29••]. (c) A peptide that
`binds to the IgG-Fc is a β-hairpin [22••].
`(d) Another β-hairpin peptide forms a
`non-covalent, symmetric dimer that acts as an
`agonist by dimerizing two erythropoietin
`receptors (the monomer is shown) [50]. (e) A
`peptide that antagonizes the activity of VEGF
`consists of a disulfide-bonded loop and an
`extended amino-terminus [51].
`
`Current Opinion in Biotechnology
`
`for peptide binding. These results bode well for the use of
`peptide libraries in drug development because they demon-
`strate that a large proportion of binding peptides is likely to
`target biologically relevant sites.
`
`Intracellular protein-binding domains
`There are several families of intracellular protein domains
`that bind other proteins and thus regulate cellular function,
`and many of these interactions involve the recognition of
`
`Lassen - Exhibit 1027, p. 3
`
`

`

`small continuous stretches of amino acids in the binding
`partner. Phage-displayed peptide libraries have been used to
`study the binding specificity of SH3 domains and WW
`domains that recognize proline-rich sequences [23,24]. It has
`also been shown that phage-displayed peptides can be phos-
`phorylated in vitro and these modified libraries can be used
`to isolate ligands for SH2 and phosphotyrosine-binding
`(PTB) domains that bind to phosphotyrosine-containing
`peptides [25,26]. Recently, Chen and Sigler [27•] deter-
`mined the crystal structure of the GroEL chaperonin alone
`and in complex with a high-affinity peptide isolated from a
`random library. Their studies shed light on the structural
`basis for GroEL’s diverse substrate specificity and the mech-
`anism of GroEL-mediated protein folding. In another study,
`peptide libraries fused to the carboxy-terminus of P8 were
`used to select ligands for PDZ domains, a recently identified
`domain family whose members generally bind the extreme
`carboxy-termini of other proteins and, in so doing, regulate
`subcellular protein localization [14••].
`
`These studies demonstrate that different types of phage-
`displayed libraries are useful for investigating different
`types of binding interactions. Despite the specific
`demands of some binding interactions, peptide phage dis-
`play is applicable to the majority of intracellular
`protein–protein interactions.
`
`Enzyme inhibitors
`Enzymes typically contain many deep clefts — including
`active sites and allosteric regulatory sites — that are well suit-
`ed for the binding of small ligands. Hyde-DeRuyscher et al.
`[28•] obtained peptide inhibitors for six out of seven distinct
`enzyme classes they tested. In addition, they showed that
`these peptides could be used as reagents for the detection of
`small-molecule inhibitors in high-throughput screens.
`
`Dennis et al. [29••] isolated a peptide that non-competi-
`tively inhibits the activity of the serine protease factor VIIa
`(FVIIa) with exquisite specificity and high potency.
`Extensive mutational and structural analyses showed that
`the peptide binds at a previously unknown ‘exosite’ dis-
`tinct from the active site, and apparently inhibits activity
`by an allosteric mechanism. These results are of substan-
`tial therapeutic relevance; FVIIa is a key regulator of the
`blood coagulation pathway and, in the past, it has been
`extremely difficult to selectively inhibit individual pro-
`teases of the coagulation cascade [29••].
`
`Phage-displayed peptide libraries seem to be ideal sources of
`peptidic enzyme inhibitors. Inhibitory peptides should prove
`useful as drug discovery reagents, or perhaps even as drugs
`themselves. Furthermore, the phage-display process explores
`the entire exposed surface of a target enzyme and, as a result,
`it can sometimes identify new sites for inhibitor design.
`
`Applications for phage-displayed proteins
`The ability to display large proteins on M13 phage has led
`to numerous applications. I focus on areas that have
`
`Phage display in pharmaceutical biotechnology Sidhu 613
`
`advanced significantly in the past year and are likely to
`have a major impact on biotechnology.
`
`Antibody phage
`Over the past decade, phage-displayed antibody fragments
`have been the subject of intensive reasearch (reviewed in
`[30,31]). As a result, antibody phage libraries have become
`practical tools for drug discovery and several phage-derived
`antibodies are in advanced clinical trials. Although most
`libraries have been constructed by cloning the natural
`immune repertoire into phage-display vectors, an important
`advance has been the development of high-quality libraries
`with completely synthetic complementarity-determining
`regions. Knappik et al. [32] have constructed a library in
`which a limited number of human frameworks are used and
`diversity is introduced by means of synthetic cassettes.
`Such a system is very amenable to the generation of thera-
`peutic antibodies because preferred frameworks can be
`used and affinity maturation is aided by the use of defined
`mutagenic cassettes. The construction of large, high-quali-
`ty libraries should also be aided by general improvements
`in library construction methodologies [6,7] and by the intro-
`duction of improved in vivo recombination systems [33].
`
`Although it is possible to obtain specific antibodies direct-
`ly from naïve phage-displayed repertoires, another
`important application for phage-display technology has
`been the humanization and affinity maturation of conven-
`tional murine antibodies. Such antibodies have been
`proven effective in a number of therapeutic applications.
`For example, a humanized antibody to vascular endothe-
`lial growth factor (VEGF) is in clinical trials as a cancer
`treatment, and Chen et al. [34] have used phage display to
`produce an affinity matured variant that is ~100-fold
`improved in both affinity and potency.
`
`Phage-displayed cDNA libraries
`Phage-displayed cDNA libraries can be used to identify
`natural binding partners for orphan receptors and ligands.
`cDNA libraries have been displayed with several different
`formats using either M13 phage [35–38] or other E. coli
`phage [2,39,40]. Although these systems show consider-
`able promise, no single display format has proven
`universally applicable. This is probably because of the fact
`that only a subset of eukaryotic proteins can be efficiently
`expressed in E. coli, and only a fraction of these can be effi-
`ciently displayed with any given system. Hopefully, new
`M13 display formats may also prove useful in these
`applications (see above).
`
`Mapping protein functional epitopes
`A major goal of modern biology is to obtain a detailed
`understanding of the protein–protein interactions that
`underly cellular processes. Alanine-scanning is a system-
`atic method that has been particularly effective in
`mapping ‘functional binding epitopes’: the contact
`residues at a protein–protein interface that make ener-
`getic contributions to binding [41]. Because alanine
`
`Lassen - Exhibit 1027, p. 4
`
`

`

`614 Pharmaceutical biotechnology
`
`mutations remove sidechain atoms past the β-carbon, they
`can be used to infer the roles of individual sidechains. A
`comprehensive alanine scan is laborious, however,
`because many single alanine mutants must be construct-
`ed, purified, and then analyzed for structural integrity and
`binding affinity. The use of phage-displayed alanine
`mutants can greatly expedite the process because proteins
`can be easily purified in association with phage particles
`and binding affinities can be realiably determined with
`simple assays that use anti-phage antibodies to quantify
`bound proteins. Using this approach, Dubaquié and
`Lowman [42] completed an impressive total alanine-scan
`of insulin-like growth factor-1 and mapped its binding
`interactions with two natural ligands.
`
`The mapping of protein functional epitopes may be fur-
`ther expedited with a combinatorial alanine-scanning
`strategy [43•]. In this method, a special phage-displayed
`protein library is constructed in which sidechains are pref-
`erentially allowed to vary only as the wild type or alanine.
`After binding selections to isolate functional clones, DNA
`sequencing is used to determine the alanine/wild-type
`ratio at each varied position. This ratio can be used to cal-
`culate binding energy contributions of
`individual
`sidechains with accuracies close to those obtained with
`conventional alanine-scanning mutagenesis. The method
`is very rapid because many sidechains are analyzed simul-
`taneously and binding energetics are derived from
`statistical analysis of DNA sequences, thus circumventing
`the need for protein purification and biophysical analysis.
`
`Engineering binding affinity and specificity
`Phage display can be used to improve or alter the binding
`properties of displayed proteins. Several groups have used
`phage display to engineer zinc-finger domains with
`designed DNA-binding specificities that can be used to
`control gene expression [44–46]. In another application,
`Hiipakka et al. [47] selected SH3 domains with improved
`or altered binding properties, thus demonstrating that
`phage display can be used to engineer signaling protein
`interaction domains. Phage display can also be used to sim-
`plify complex signaling pathways. For example, VEGF is a
`pleiotropic factor that binds two different receptors (Flt-1
`and KDR) and thus exerts a multitude of biological effects.
`Using rational design and phage display, Li et al. [48] have
`generated VEGF variants that selectively bind only to
`either Flt-1 or KDR [48]. These receptor-selective variants
`can be used to elucidate the specific role of each receptor.
`
`Conclusions
`Intensive efforts from many researchers have made phage
`display an invaluable component of biotechnology.
`Improved library construction methods — in combination
`with numerous vectors and display formats — will extend
`the technology even further. Phage antibodies are likely to
`play an even greater role in the generation of analytical
`reagents and therapeutic drugs. Highly diverse peptide
`libraries can be used to isolate specific ligands for virtually
`
`any target of interest, and these ligands should be useful in
`therapeutic target validation and as leads in drug design.
`cDNA phage display can be used to identify natural pro-
`tein–protein interactions, which can be mapped in detail
`with methods that use phage display as a tool for measur-
`ing binding energetics. Further research will continue to
`improve the reliability of phage-display methods and many
`aspects of the technology may eventually be automated. It
`seems probable that extremely diverse phage-display
`libraries will contain multiple solutions to most binding
`problems, and perhaps the most effective applications will
`be those that exhaustively explore these solutions by com-
`bining phage-display selections with high-throughput
`screening and DNA sequencing. These technologies will
`generate vast databases that explore the links between
`protein structure and function, and this information will in
`turn expedite the process of drug development.
`
`Acknowledgements
`I thank Nicholas Skelton and David Wood for help with figures.
`
`References and recommended reading
`Papers of particular interest, published within the annual period of review,
`have been highlighted as:
`• of special interest
`•• of outstanding interest
`Smith GP: Filamentous fusion phage: novel expression vectors
`that display cloned antigens on the virion surface. Science 1985,
`228:315-1317.
`
`1.
`
`2.
`
`3.
`
`4.
`
`Santini C, Brennan D, Mennuni C, Hoess RH, Nicosia A, Cortese R,
`Luzzago A: Efficient display of an HCV cDNA expression library as
`C-terminal fusion to the capsid protein D of bacteriophage
`lambda. J Mol Biol 1998, 282:125-135.
`
`Ren Z, Black LW: Phage T4 SOC and HOC display of biologically
`active, full-length proteins on the viral capsid. Gene 1998,
`215:439-444.
`
`Possee RD: Baculoviruses as expression vectors. Curr Opin
`Biotechnol 1997, 8:569-572.
`
`5. Georgiou G, Stathopoulos C, Daugherty PS, Nayak AR,
`Iverson BL, Curtiss R III: Display of heterologous proteins on
`the surface of microorganisms: from the screening of
`combinatorial libraries to live recombinant vaccines. Nat
`Biotechnol 1997, 15:29-34.
`
`6.
`•
`
`Sidhu SS, Lowman HB, Cunningham BC, Wells JA: Phage display
`for selection of novel binding peptides. Methods Enzymol 2000,
`328:333-363.
`This chapter provides detailed descriptions of optimized methods for the
`construction of extremely diverse phage-displayed libraries. With highly con-
`centrated E. coli and DNA, a single electroporation reaction produces
`libraries with >1010 unique members. Multiple reactions can be combined to
`produce diversities >1012.
`
`7.
`
`Sidhu SS, Weiss GA: Constructing phage display libraries by
`oligonucleotide-directed mutagenesis. In Phage Display: A
`Practical Approach. Edited by Clackson T, Lowman HB. Oxford:
`Oxford University Press; in press.
`
`8. Marvin DA: Filamentous phage structure, infection and assembly.
`Curr Opin Struct Biol 1998, 8:150-158.
`
`9.
`
`Scott JK, Smith GP: Searching for peptide ligands with an epitope
`library. Science 1990, 249:386-390.
`
`10. Greenwood J, Willis AE, Perham RN: Multiple display of foreign
`peptides on a filamentous bacteriophage. J Mol Biol 1991,
`220:821-827.
`
`11. Bass S, Green R, Wells JA: Hormone phage: an enrichment
`method for variant proteins with altered binding properties.
`Proteins: Struct Funct Genet 1990, 8:309-314.
`
`Lassen - Exhibit 1027, p. 5
`
`

`

`12.
`
`Jespers LS, Messens JH, De Keyser A, Eeckhout D, Van Den Brande I,
`Gansemans YG, Lauwereys MJ, Vlasuk GP, Stanssens PE: Surface
`expression and ligand-based selection of cDNAs fused to
`filamentous phage gene VI. Bio/Technology 1995, 13:378-382.
`
`13. Gao C, Mao S, Lo CH, Wirsching P, Lerner RA, Janda KD: Making
`artificial antibodies: a format for phage display of combinatorial
`heterodimeric arrays. Proc Natl Acad Sci USA 1999,
`96:6025-6030.
`
`14. Fuh G, Pisabarro MT, Li Y, Quan C, Lasky LA, Sidhu SS: Analysis of
`PDZ domain-ligand interactions using carboxyl-terminal phage
`••
`display. J Biol Chem 2000, 275:21486-21491.
`The authors report a new display format in which peptids are fused to the
`carboxyl-terminus of P8. A random peptide library displayed in this format is
`used to identify high-affinity ligands for PDZ domains that bind peptides with
`free carboxy-termini. The binding interactions are rationalized using synthet-
`ic peptides and homology modeling.
`
`15. Fuh G, Sidhu SS: Efficient phage display of polypeptides fused to
`the carboxy-terminus of the M13 gene-3 minor coat protein.
`FEBS Lett 2000, 480:231-234.
`
`16. Rodi DJ, Makowski L: Phage-display technology — finding a needle
`in a vast molecular haystack. Curr Opin Biotechnol 1999, 10:87-93.
`
`17. Sidhu SS, Weiss GA, Wells JA: High copy display of large proteins
`on phage for functional selections. J Mol Biol 2000, 296:487-495.
`
`18. Weiss GA, Wells JA, Sidhu SS: Mutational anlaysis of the major
`coat protein of M13 identifies residues that control protein
`display. Protein Sci 2000, 9:647-654.
`
`19. Weiss GA, Sidhu SS: Design and evolution of artificial M13 coat
`proteins. J Mol Biol 2000, 300:213-219.
`••
`Completely artificial M13 coat proteins are selected from large libraries of
`sequences designed to mimic P8 in a reverse orientation. These results
`demonstrate that the M13 phage coat is very tolerant to the incorporation of
`new proteins that need not be related to natural coat proteins. Thus, it will
`be possible to engineer artificial coat proteins specifically for phage display.
`
`20. Cochran AG: Antagonists of protein–protein interactions. Chem
`Biol 2000, 7:R85-R94.
`
`21. Ferrer M, Harrison SC: Peptide ligands to human
`immunodeficiency virus type 1 gp120 identified from phage
`••
`display libraries. J Virol 1999, 73:5795-5802.
`The authors use a naïve library to isolate a 12-residue peptide that binds
`gp120 from three different HIV strains and inhibits the interaction between
`gp120 and CD4. Although the peptide did not show inhibitory activity in a
`cell–cell fusion assay, it may still be a useful lead for the development of anti-
`HIV compounds.
`
`22. Delano WL, Ultsch MH, de Vos AM, Wells JA: Convergent solutions
`to binding at a protein–protein interface. Science 2000,
`••
`287:1279-1283.
`The crystal structure of a disulfide-constrained peptide complexed with
`IgG-Fc reveals a β-hairpin structure. The peptide binds to a consensus bind-
`ing site that also interacts with four structurally distinct natural proteins.
`A detailed analysis reveals that the different ligands use similar binding con-
`tacts and the consensus binding site is highly accessible, hydrophobic, and
`adaptive. These properties may be common attributes in natural binding sites.
`
`23. Sparks AB, Rider JE, Hoffman NG, Fowlkes DM, Quilliam LA, Kay BK:
`Distinct ligand preferences of Src homology 3 domains from Src,
`Yes, Abl, Cortactin, p53bp2, PLCγ, Crk, and Grb2. Proc Natl Acad
`USA 1996, 93:1540-1544.
`
`24. Lin H, Ermekova KS, Rentschler S, Sparks AB, Kay BK, Sudol M:
`Using molecular repertoires to identify high-affinity peptide
`ligands of the WW domain of human and mouse YAP. Biol Chem
`1997, 378:531-537.
`
`25. Dente L, Vetriani C, Zucconi A, Pelicci G, Lanfrancone L, Pelicci PG,
`Cesareni G: Modified phage peptide libraries as a tool to study
`specificity of phosphorylation and recognition of tyrosine
`containing peptides. J Mol Biol 1997, 269:694-703.
`
`26. Gram H, Schmitz R, Zuber JF, Baumann G: Identification of
`phosphopeptide ligands for the Src-homology 2 (SH2) domain of
`Grb2 by phage display. Eur J Biochem 1997, 246:633-637.
`
`27. Chen L, Sigler PB: The crystal structure of a GroEL/peptide
`complex: plasticity as a basis for substrate diversity. Cell 1999,
`•
`99:757-768.
`X-ray crystallography is used to investigate the binding interaction between
`GroEL chaperonin and a phage-derived peptide. The authors use this infor-
`mation to investigate the structural basis for GroEL’s diverse substrate
`
`Phage display in pharmaceutical biotechnology Sidhu 615
`
`specificity, and they conclude that various modes of molecular plasticity are
`responsible for tight but promiscuous binding.
`
`28. Hyde-DeRuyscher R, Paige LA, Christensen DJ, Hyde-DeRuyscher N,
`Lim A, Fredericks ZL, Kranz J, Gallant P, Zhang J, Rocklage SM et al.:
`•
`Detection of small-molecule enzyme inhibitors with peptides
`isolated from phage-displayed combinatorial peptide libraries.
`Chem Biol 2000, 7:17-25.
`This encouraging study demonstrates that it should be easy to obtain phage-
`derived inhibitory peptides for any enzyme. Furthermore, these peptides can
`be used as sensitive probes for the detection of small molecules that bind
`the enzyme and displace the bound peptide. These probes enable the devel-
`opment of high-throughput screening assays designed to detect small-mol-
`ecule enzyme inhibitors.
`
`29. Dennis MS, Eigenbrot C, Skelton NJ, Ultsch MH, Santell L,
`•• Dwyer MA, O’Connell MP, Lazarus RA: Peptide exosite inhibitors of
`factor VIIa as anticoagulants. Nature 2000, 404:465-470.
`This paper reports an extremely thorough analysis of a peptide that inhibits
`FVIIa activity. The crystal structure of the peptide–FVIIa complex reveals that
`the peptide binds at an allosteric exosite, consistent with the observed non-
`competitive inhibition mode. The structure of the peptide free in solution was
`determined by NMR and found to be very similar to that found in the bound
`state. Thus, phage display identified a potent FVIIa inhibitor and also led to
`the discovery of a new mode of inhibition.
`
`30. Dall’Acqua W, Carter P: Antibody engineering. Curr Opin Struct Biol
`1998, 8:443-450.
`
`31. Griffiths AD, Duncan AR: Strategies for selection of antibodies by
`phage display. Curr Opin Biotechnol 1998, 9:102-108.
`
`32. Knappik A, Ge L, Honegger A, Pack P, Fischer M, Wellnhofer G,
`Hoess A, Wolle J, Pluckthun A, Virnekas B: Fully synthetic human
`combinatorial antibody libraries (HuCAL) based on modular
`consensus frameworks and CDRs randomized with
`trinucleotides. J Mol Biol 2000, 296:57-86.
`
`33. Sblattero D, Bradbury A: Exploiting recombination in single
`bacteria to make large phage antibody libraries. Nat Biotechnol
`2000, 18:75-80.
`
`34. Chen Y, Weismann C, Fuh G, Li B, Christinger HW, McKay P,
`deVos AM, Lowman HB: Selection and analysis of an optimized
`anti-VEGF antibody: crystal structure of an affinity-matured Fab in
`complex with antigen. J Mol Biol 1999, 293:865-881.
`
`35. Fransen M, Van Veldhoven PP, Subramani S: Identification of
`peroxisomal proteins by using M13 phage protein VI phage
`display: molecular evidence that mammalian peroxisomes
`containg a 2,4-dienoyl-CoA reductase. Biochem J 1999,
`340:561-568.
`
`36. Hufton SE, Moerkerk PT, Meulemans EV, de Bruine A, Arends JW,
`Hoogenboom HR: Phage display of cDNA. J Immunol Methods
`1999, 231:39-51.
`
`37. Shanmugavelu M, Baytan AR, Chesnut JD, Bonning BC: A novel
`protein that binds juvenile hormone esterase in fat body tissue
`and pericardial cells of the tobacco hornworm Manduca sexta L.
`J Biol Chem 2000, 275:1802-1806.
`
`38. Cochrane D, Webster C, Masih G, McCafferty J: Identification of
`natural ligands for SH2 domains from a phage display cDNA
`library. J Mol Biol 2000, 297:89-97.
`
`39. Sche PP, McKenzie KM, White JD, Austin DJ: Display cloning:
`functional identification of natural receptors using cDNA-phage
`display. Chem Biol 1999, 6:707-716.
`
`40. Fakok VA, Bratton DL, Rose DM, Pearson A, Ezekewitz RAB,
`Henson PM: A receptor for phosphatidylserine-specific clearance
`of apoptotic cells. Nature 2000, 405:85-90.
`
`41. Wells JA: Systematic mutational analyses of protein–protein
`interfaces. Methods Enzymol 1991, 202:390-41

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket