Вы находитесь на странице: 1из 33

Biotechnology Advances 19 (2001) 1 33

Research review paper

Biotechnological applications of phage and cell display


Itai Benhar*
Department of Molecular Microbiology and Biotechnology, The George S. Wise Faculty of Life Sciences, Green Building, Room 202, Tel-Aviv University, Ramat Aviv 69978, Israel

Abstract In recent years, the use of surface-display vectors for displaying polypeptides on the surface of bacteriophage and bacteria, combined with in vitro selection technologies, has transformed the way in which we generate and manipulate ligands, such as enzymes, antibodies and peptides. Phage display is based on expressing recombinant proteins or peptides fused to a phage coat protein. Bacterial display is based on expressing recombinant proteins fused to sorting signals that direct their incorporation on the cell surface. In both systems, the genetic information encoding for the displayed molecule is physically linked to its product via the displaying particle. Using these two complementary technologies, we are now able to design repertoires of ligands from scratch and use the power of affinity selection to select those ligands having the desired (biological) properties from a large excess of irrelevant ones. With phage display, tailor-made proteins (fused peptides, antibodies, enzymes, DNA-binding proteins) may be synthesized and selected to acquire the desired catalytic properties or affinity of binding and specificity for in vitro and in vivo diagnosis, for immunotherapy of human disease or for biocatalysis. Bacterial surface display has found a range of applications in the expression of various antigenic determinants, heterologous enzymes, single-chain antibodies, and combinatorial peptide libraries. This review explains the basis of phage and bacterial surface display and discusses the contributions made by these two leading technologies to biotechnological applications. This review focuses mainly on three areas where phage and cell display have had the greatest impact, namely, antibody engineering, enzyme technology and vaccine development. D 2001 Elsevier Science Inc. All rights reserved.
Keywords: Antibody engineering; Bacterial surface display; Enzyme evolution; Immobilized enzymes; Phage display; Vaccine development

* Tel.: +972-3-6407511; fax: +972-3-6409407. E-mail address: benhar@post.tau.ac.il (I. Benhar). 0734-9750/01/$ see front matter D 2001 Elsevier Science Inc. All rights reserved. PII: S 0 7 3 4 - 9 7 5 0 ( 0 0 ) 0 0 0 5 4 - 9

I. Benhar / Biotechnology Advances 19 (2001) 133

1. Introduction As a result of ongoing genome projects, a large-scale effort to measure and analyze protein protein interactions using experimental methods is under way. These include biochemical techniques such as co-immunoprecipitation or crosslinking (Silver and Hunt, 1993), molecular biological techniques such as the two-hybrid system (Fashena et al., 2000) or the phage- and cell-display systems (reviewed herein), and genetic techniques such as mutant isolation and analysis (Gargus, 1987). Therefore, determining functions of novel gene products identified in genome sequencing efforts has become an important scientific challenge. Identifying proteins that interact with novel gene products can help in deducing their activity, regulation, and potential role in disease. Traditional biochemical analyses of enzyme structure and function and elucidation of receptor or antibody recognition processes are based typically on the utilization of efficient model systems. These are characterized by practical considerations, such as: enrichment of the molecules being studied, a source of tissue or cells that can be obtained in abundance with reasonable ease and the stability of the components to biochemical manipulations. The reductionist's approach is to systematically weed out the molecules of interest until purity. The advent of molecular genetics has enhanced the study of proteinligand interactions both by providing means to produce rare molecules in large quantities via gene-expression systems and by focusing on specific regions or residues of the proteins using site-directed mutagenesis. However, the most commonly used technologies for identifying proteinligand interactions, such as the yeast two-hybrid system (Fashena et al., 2000) and expression library screening (Maser and Calvet, 1995; McCarrey and Williams, 1994) are labor-intensive and often not amenable to high-throughput evaluation of multiple gene products. Two leading methodologies have been added to those mentioned above that provide new horizons for the study of proteinprotein interactions. These technologies are based on the production and presentation of enormous collections of peptides or proteins to be screened by corresponding `binders.' This enables the identification of binding pairs or the selection of molecules by virtue of their function. The approaches are based on the concept of generating a complete set of all possible combinations of the peptides or proteins of interest, collectively referred to as `combinatorial display.' The respective approaches involve the display of (poly)peptides on the surface of bacteria and the combinatorial display of synthetic peptides, organic structures and recombinant proteins on bacteriophage. 2. Phage display The generation of new drugs has long involved the search amongst hundreds of thousands of components using well-defined in vitro screening tests, the output of which was chosen to mimic as closely as possible the desired in vivo activity of the new drug. Today, new library methodologies offer many alternative and at least as powerful routes, by combining the generation of billions of components with a fast screening or selection procedure to identify the most interesting lead candidates.

I. Benhar / Biotechnology Advances 19 (2001) 133

Phage display has proven to be a very powerful technique for obtaining libraries containing millions or even billions of different peptides or proteins. Phage display (Smith, 1985) has been used for affinity screening of combinatorial peptide libraries to identify ligands for peptide receptors, define epitopes for monoclonal antibodies, select enzyme substrates (Kay et al., 1996), and screen cloned antibody repertoires (Griffiths and Duncan, 1998). Regulated associations between select proteins are the basis of cellular signal transduction pathways, and defining a signal transduction pathway can, in many cases, be reduced to tracing a chain of proteinprotein interactions. One of the most successful applications of phage display has been the isolation of monoclonal antibodies, and fragments thereof, using large phage antibody libraries (Winter et al., 1994). Indeed, in the last few years, very efficient techniques have been developed to design and build large libraries of antibody fragments, and ingenious selection procedures have been established to derive antibodies with the desired characteristics. Much progress has been made in this rapidly developing field and many possible applications of phage technology have been developed (Hoogenboom et al., 1998). These include the creation and screening of libraries to discover novel therapeutic targets and methods for selection of biologically active ligands. The most widely used library methodology is based on the use of filamentous phage (Smith, 1985), a bacteriophage that infects male Escherichia coli. Display systems based on other phage were also developed, including display on phage l heads (Sternberg and Hoess, 1995; Mikawa et al., 1996; Santini et al., 1998) and tail (Maruyama et al., 1994; Kuwabara et al., 1997), P4 capsid (Lindqvist and Naderi, 1995), T7 capsid (Houshmand et al., 1999; Yamamoto et al., 1999), T4 capsid (Jiang et al., 1997; Ren and Black, 1998; Mullaney and Black, 1996, 1998), and MS2 coat (Mastico et al., 1993; Heal et al., 1999). The phage-display systems and the molecules that were displayed using them are listed in Table 1. Filamentous phage display is based on cloning DNA fragments encoding millions of variants of certain ligands (e.g. peptides, proteins or fragments thereof) into the phage genome, fused to the gene encoding one of the phage coat proteins (usually pIII, but also pIV, PVI or pVIII, Table 1). Upon expression, the coat protein fusion is incorporated into new phage particles that are assembled in the periplasmic space of the bacterium. Expression of the gene fusion product and its subsequent incorporation into the mature phage coat results in the ligand being presented on the phage surface, while its genetic material resides within the phage particle. This connection between genotype and phenotype allows the enrichment of specific phage, e.g. using selection on an immobilized target. Phage that display a relevant ligand are retained by virtue of their binding to the target, while nonadherent phages are washed away. Bound phage can be recovered from the surface, used to reinfect bacteria and reproduced for further enrichment, and eventually for analysis of binding. The success of ligand phage display hinges on the combination of this display and enrichment method, with the synthesis of large combinatorial repertoires on phage. Phage selection is not limited to the isolation of antibodies or short peptides as described above. As listed in Table 1, this approach has also been instrumental in studies and manipulation of a variety of other biologically active molecules, e.g. cytokines (Gram et al., 1993; Saggio et al., 1995; Buchli et al., 1997), receptors (Lowman et al., 1991; Scarselli et al., 1993), enzymes (McCafferty et al., 1991; Soumillion et al., 1994; Pedersen et al., 1998;

4 Table 1 Phage display systems Phage Coat protein M13 pIII (minor)

I. Benhar / Biotechnology Advances 19 (2001) 133

Displayed molecules Peptides

References

M13 M13

M13 l

P4 T7 T4

MS2

Numerous reports. Recent reviews: Daniels and Lane, 1996; Lowman, 1997; Rodi and Makowski, 1999; Cabilly, 1999 Antibodies Numerous reports. Recent reviews: Winter et al., 1994; Burton and Barbas, 1994; Rader and Barbas, 1997; Hoogenboom, 1997; Hoogenboom et al., 1998; Griffiths and Duncan, 1998 Cytokines Gram et al., 1993; Saggio et al., 1995; Buchli et al., 1997 Receptors Lowman et al., 1991; Scarselli et al., 1993 Enzymes McCafferty et al., 1991; Soumillion et al., 1994; Pedersen et al., 1998; Demartis et al., 1999; Forrer et al., 1999 Enzyme inhibitors Roberts et al., 1992; Pannekoek et al., 1993; van Meijer et al., 1996; Huang et al., 1998 Catalytic antibodies Janda et al., 1994; Sastry et al., 1995; Baca et al., 1997; Fujii et al., 1998 DNA-binding Rebar and Pabo, 1994; Jamieson et al., 1994; proteins Choo and Klug, 1994; Choo et al., 1997 Cellulose-binding Smith et al., 1998; Berdichevsky et al., 1999 proteins pVI Enzyme inhibitors Jespers et al., 1995 Enzymes Hufton et al., 1999 cDNA libraries Hufton et al., 1999 pVIII (major) Peptides Numerous reports. Recent reviews: Felici et al., 1995; Ladner, 1995; Cortese et al., 1996; Lowman, 1997; Wilson and Finlay, 1998 Antibodies Kang et al., 1991; Wang et al., 1997 Enzymes Corey et al., 1993 Enzyme inhibitors Markland et al., 1991, 1996 pVII/pIX Antibodies Gao et al., 1999 D (Head protein) Peptides Sternberg and Hoess, 1995 IgG-binding protein Sternberg and Hoess, 1995 Enzymes Mikawa et al., 1996 Protein A Mikawa et al., 1996 cDNA libraries Santini et al., 1998 pV (Tail protein) Peptides Maruyama et al., 1994; Kuwabara et al., 1997 Enzymes Maruyama et al., 1994 Psu capsid protein Peptides Lindqvist and Naderi, 1995 10B capsid protein Peptides Houshmand et al., 1999 cDNA libraries Yamamoto et al., 1999 Hoc capsid protein Peptides Jiang et al., 1997 Antibodies Ren and Black, 1998 Soc capsid protein Peptides Jiang et al., 1997 Antibodies Ren and Black, 1998 Internal protein III Enzymes Mullaney and Black, 1996, 1998 Green fluorescent Mullaney and Black, 1996 protein Coat protein Peptides Mastico et al., 1993; Heal et al., 1999

I. Benhar / Biotechnology Advances 19 (2001) 133

Demartis et al., 1999; Forrer et al., 1999), substrates (Matthews and Wells, 1993; Ohkubo et al., 1999) enzyme inhibitors (Roberts et al., 1992; Pannekoek et al., 1993; van Meijer et al., 1996; Huang et al., 1998), catalytic antibodies (Sastry et al., 1995; Janda et al., 1994; Baca et al., 1997; Fujii et al., 1998) DNA-binding proteins (Rebar and Pabo, 1994; Jamieson et al., 1994; Choo and Klug, 1994; Choo et al., 1997), receptor domains for constructing binding molecules with novel properties (reviewed in Nygren and Uhlen, 1997), and cellulosebinding domains (Smith et al., 1998; Berdichevsky et al., 1999). Scaffolds different from antibodies have been reported to form suitable binding ligands for many types of molecules. There are ample examples of host scaffolds that contain sufficient permissive regions to accommodate a reasonable numbers of substitutions, which may be used to generate a library of localized variability. Alternative scaffolds reported to date include b-sheet proteins (Tramontano et al., 1994), a-helical bundle proteins (Ku and Schultz, 1995), combinations of these two (Tramontano et al., 1994), a separate group of highly constrained protease inhibitors (Markland et al., 1996; Tramontano et al., 1994; Rottgen and Collins, 1995), and, most recently, Green fluorescent protein (GFP) (Abedi et al., 1998) and cellulose-binding domains (Smith et al., 1998). Since secreted as well as cytoplasmic and nuclear proteins have been displayed on phage, the use of phage display is often the first strategy to define permissive sites for randomization and to generate ligand-binding variants (Choo et al., 1997). Alternatively, the use of lambda phage (Mikawa et al., 1996), bacterial cell display (Georgiou et al., 1997), and eukaryotic cell-display methods have been reported. With regard to functional selection methods, choosing other types of molecules besides antibodies is validated by the fact that antibody expression and folding may be impaired in the subcellular location where the desired functional activity is required. It would be advantageous to engineer the antibody for intracellular expression, for example by building stable disulfide-free antibodies (Proba et al., 1997), or to use libraries of scaffolds that are naturally produced in the targeted cell organelle, provided that effective and sufficient structural diversity may be obtained. 3. Bacterial surface display The targeting and anchoring of heterologous proteins to the outer surface of yeast and mammalian cells (Schreuder et al., 1991) have been utilized for various applications. However, this review is focused on bacterial surface-display systems that are becoming an increasingly important research area (Georgiou et al., 1997; Hofnung, 1991; Little et al., 1993; Francisco and Georgiou, 1994; Fischetti et al., 1996). Surface display of heterologous proteins in bacteria has been employed as a tool for basic and applied research in microbiology, molecular biology, vaccinology and biotechnology. The first examples of heterologous surface display were reported a decade ago, when short gene fragments were inserted into the genes for the E. coli outer membrane proteins LamB, OmpA and PhoE, and the gene fusion products were found to be accessible on the outer surface of the recombinant bacteria (Charbit et al., 1988; Agterberg et al., 1990a,b; Tommassen et al., 1994; Janssen and Tommassen, 1994). Those display systems, however, were not suitable in most cases for the display of large proteins (Tommassen et al., 1994).

6 Table 2 Bacterial display systems Bacterium

I. Benhar / Biotechnology Advances 19 (2001) 133

Membrane protein

Displayed molecules Peptides Peptides Metal binding proteins Antibodies Protein A Peptides Peptides Peptides Peptides Peptides Enzymes Antibodies Receptors Toxin domain Cellulose binding proteins Peptides Toxin domain Peptides Enzymes HIV GP120 Peptides Peptides Peptides Toxin domain Peptides Peptides Peptides Peptides Protein A Enzymes Peptides Receptors Antibodies Enzymes Peptides

References Charbit et al., 1988; Leclerc et al., 1991; Bradbury et al., 1993; Sousa et al., 1996; Persic et al., 1999 Pistor and Hobom, 1988; Agterberg et al., 1990a,b Kotrba et al., 1999 Fuchs et al., 1991, 1996; Dhillon et al., 1999 Steidler et al., 1993 Tommassen et al., 1994; Janssen and Tommassen, 1994 Chang and Lo, 2000; Chang et al., 1999 Hedegaard and Klemm, 1989 Lu et al., 1995 Pallesen et al., 1995 Francisco et al., 1992 Francisco and Georgiou, 1994; Francisco et al., 1993b; Chen et al., 1996 Benhar et al., 2000 Benhar et al., 2000 Francisco et al., 1993a Maurer et al., 1997 Maurer et al., 1997 Malmborg et al., 1997 Jung et al., 1998a,b Kwak et al., 1999 Sewani et al., 1998 Xu and Lee, 1999 Wu et al., 1989; Newton et al., 1989 Klauser et al., 1990 Schorr et al., 1991 Bingle et al., 1997 Pozzi et al., 1992 Oggioni et al., 1999 Schneewind et al., 1992, 1993 Schneewind et al., 1992, 1993 Hansson et al., 1992 Hansson et al., 1992 Gunneriusson et al., 1996, 1999 Strauss and Gotz, 1996 Mesnage et al., 1999

A. Gram-negative bacteria E. coli LamB OmpA

Pal Pap PhoE TraT Type 1 fimbriae FliC FimH Lpp-OmpA

AIDA-I F pilin Pseudomonas Inp ExtB OmpC Flagellin Neisseria IgAb E. coli OmpA RsaA

Salmonella Caulobacter

B. Gram-positive bacteria Streptococcus M1 protein M6 protein Staphylococcus Synthetic anchor Protein A FnBPB S-layer protein EA1

B. anthracis

I. Benhar / Biotechnology Advances 19 (2001) 133

Since then, not only outer membrane proteins but also lipoproteins, fimbria proteins and flagellar proteins, as well as dedicated systems with coupled translocation and surface anchoring, have been employed to achieve heterologous surface display on gram-negative bacteria (Hofnung, 1991; Little et al., 1993; Francisco and Georgiou, 1994), for example E. coli and Salmonella spp. In parallel, systems have been described for heterologous surface display on gram-positive bacteria such as staphylococci (Hansson et al., 1992), streptococci (Pozzi et al., 1992; Hanski et al., 1992) and mycobacteria (Stover et al., 1993). The bacterial display systems and the molecules that were displayed using them are listed in Table 2. The most common application of bacterial surface display has been in the development of live bacterial vaccine-delivery systems. In this context, the cell-surface display of heterologous antigenic determinants has been considered advantageous for the induction of antigenspecific antibody responses when using live recombinant cells for immunization (Stover et al., 1993; Georgiou et al., 1997; Nguyen et al., 1995). Bacterial surface display has also been applied in generating whole-cell bioadsorbents for environmental purposes (Sousa et al., 1996), novel microbial biocatalysts [surface display of active enzymes on E. coli (Francisco et al., 1992, 1993a,b) and staphylococci (Strauss and Gotz, 1996)], diagnostic tools [bacteria with surface-displayed antibody fragments (Fuchs et al., 1991; Francisco et al., 1992)] and for the display of entire peptide libraries (Lu et al., 1995) as an alternative to the rapidly developing phage-display technology. It should, however, be specified that combinatorial display is commonly applied using phage systems, and rarely using bacterial display systems. The latter are usually dedicated to the display of functionally defined molecules. 4. Surface display in gram-negative bacteria Display systems have also been developed for the expression of a number of heterologous proteins on the surface of gram-negative bacteria. Most often, foreign gene products have been fused to outer membrane proteins, such as the maltoporin LamB (Charbit et al., 1988), the phosphate-inducible porin PhoE (Tommassen et al., 1994), the outer membrane protein OmpA (Agterberg et al., 1990a,b), and to lipoproteins such as the major lipoprotein Lpp, the TraT lipoprotein and the peptidoglycan-associated lipoprotein PAL (Fuchs et al., 1991, 1996; Dhillon et al., 1999; Chang and Lo, 2000; Chang et al., 1999). Proteins from the filamentous structures present on gram-negative bacteria have also been employed for surface-expression purposes. These include fimbria proteins such as the FimA protein (Hedegaard and Klemm, 1989) and the FimH adhesin of type I fimbriae (Pallesen et al., 1995), the flagellar protein flagellin (Wu et al., 1989; Newton et al., 1989) and pili proteins such as the PapA pilus subunit (Steidler et al., 1993) and F Pilin (Malmborg et al., 1997). Certain display systems have been based on proteins that have specific mechanisms for translocation and surface anchoring. The lipoprotein pullulanase from Klebsiella pneumoniae has been employed for surface-display purposes in E. coli (Kornacker and Pugsley, 1990). Using this system, the target protein becomes transiently anchored to the outer membrane by its N-terminal fatty acid moiety and is subsequently released into the culture medium. The b-domain of the Neisseria gonorrhoeae IgA protease precursor (Igab) and the E. coli AIDA-I have also been employed for surface exposure of various heterologous proteins. When used as a C-terminal-

I. Benhar / Biotechnology Advances 19 (2001) 133

fusion partner, the Igab and AIDA-l mediate attachment of hybrid proteins on the outer surface of Salmonella typhimurium and E. coli strains without the OmpT protease (Maurer et al., 1997; Klauser et al., 1990). One particularly interesting system is based on the combined features of an Lpp-OmpAH chimera (Francisco and Georgiou, 1994; Francisco et al., 1992, 1993a,b; Stathopoulos et al., 1996; Georgiou et al., 1997; Daugherty et al., 1998). The Lpp-OmpAH chimera is comprised of the signal sequence and first nine amino acids of Lpp, fused to a region encompassing either three or five transmembrane helices of OmpA. This Lpp-OmpAH display system has been shown to give efficient translocation and surface anchoring of the fused gene products, resulting in a high number of chimeric surface proteins present in an accessible form on E. coli cells. 5. Surface display in gram-positive bacteria Gram-positive bacteria have also been considered suitable for bacterial surface-display purposes (Hansson et al., 1992; Pozzi et al., 1992; Fischetti et al., 1996; Georgiou et al., 1997). A range of proteins, including antigenic determinants, heterologous enzymes and single-chain Fv (scFv) antibodies, have been targeted and anchored to the cell surface of gram-positive bacteria. Several gram-positive bacterial hosts have been investigated in this context, but until recently the research has focused on nonpathogenic staphylococci used in food-fermentation processes (Hansson et al., 1992; Pozzi et al., 1992; Hanski et al., 1992; Samuelson et al., 1995). Schneewind et al. (1992, 1993, 1995) have elucidated the mechanisms of cell-surface targeting and subsequent anchoring of surface proteins on staphylococcal cells several years after heterologous surface display had been achieved on staphylococci and streptococci. They found that Staphylococcus aureus protein A was sorted to the cell surface by its C-terminal surface-anchoring region. That region consists of a charged repetitive region, postulated to interact with the peptidoglycan cell wall. The Ctermini of numerous gram-positive bacterial surface receptors are highly homologous, suggesting that this or a similar mechanism is utilized for their targeting to the cell surface (Pozzi et al., 1992; Schneewind et al., 1993, 1995). The surface-display systems developed for Staphylococcus xylosus (Hansson et al., 1992) and Staphylococcus carnosus (Samuelson et al., 1995) both take advantage of the cell-surface-anchoring regions of protein A. The Staphylococcus gordinii surface-display system (Pozzi et al., 1992) uses the similar C-terminal region of the M6 protein of Streptococcus pyogenes to achieve surface exposure of various chimeric surface proteins. While the staphylococcal systems utilize gene expression from plasmid vectors, the streptococcal system is based on incorporation of the target genes into the streptococcal chromosome by homologous recombination (Hansson et al., 1992). Recently, heterologous surface expression was achieved in Bacillus anthracis by integrating into the chromosome a translational fusion harboring the DNA fragments encoding the cell wall-targeting domain of the S-layer protein EA1 tetanus toxin fragment C. This construct was expressed under the control of the promoter of the S-layer component gene (Mesnage et al., 1999).

I. Benhar / Biotechnology Advances 19 (2001) 133

The major applications of bacterial surface display (see below) are antibody and enzyme display, and vaccine development. In addition, other less common applications include peptide display for mapping epitopes of monoclonal antibodies. In one example, a random peptide library in a conformationally constrained thioredoxin region was introduced into the flagellin gene of E. coli and thus surface-exposed on the E. coli flagellum (Lu et al., 1995). The epitopes for three different immobilized monoclonal antibodies were mapped by the selection of bacterial clones and the identified consensus epitopes had several amino acids in common with motifs found in the original antigens used to generate the monoclonal antibodies. A very different application of bacterial surface display was recently suggested by Sousa et al. (1996), who displayed polyhistidyl peptides on the surface of E. coli using the LamB system. E. coli cells with surface-exposed histidine tags adsorbed approximately 11 times more Cd2 + ions than control cells. Such bacteria could perhaps be used for bioadsorption of heavy metal ions, potentially valuable for environmental (bioremediation) applications. Powerful techniques to monitor and characterize the exposed target proteins being displayed are crucial for the full exploitation of any display technology. Traditional techniques for studying (recombinant) surface-displayed proteins include immunofluorescence and immunogold methods. Although these methods reveal whether a heterologous protein is produced in an accessible form or not, they fail to give reliable quantitative information about the number of exposed chimeric surface proteins per bacterial cell. Immunofluorescent staining can give misleading results by positive staining of partially lysed cells and should thus be accompanied by additional assays. Flow cytometry, in the form of fluorescence-activated cell sorting (FACS), has been used as the method of choice to characterize surface display on bacteria (Fuchs et al., 1991; Stover et al., 1993; Francisco et al., 1993b; Nguyen et al., 1995; Samuelson et al., 1995). 6. Surface display of antibodies Phage display is by far the major tool for the isolation and engineering of recombinant antibodies, while bacterial display plays a minor role in that field. Antibodies in the form of recombinant antibody fragments were the first proteins to be successfully displayed on the surface of phage (McCafferty et al., 1990). This was achieved by fusing the coding sequence of the antibody variable (V) regions encoding for a single-chain Fv (scFv) fragment to the amino terminus of the phage gene III, coding for the phage minor coat protein pIII. Initial attempts to display FabH fragments fused to pVIII, the phage major coat protein, were also successful (Gram et al., 1992). However, the pVIII site, although very popular for peptide phage display, is not suitable for the efficient display of large polypeptides such as antibodies. For this reason, most antibody phage-display systems utilize the pIII site. Antibodies were first displayed using a phage vector, based on the genome of fd-tet (Zacher et al., 1980) and its gene III as fusion partner. In this vector, the genes coding for antibody scFv fragments were cloned in-frame with gene III and downstream of the gene III signal sequence, which normally directs the export of the phage-coat protein to the periplasm. Here, the antibody VH and VL domains may fold correctly, both stabilized by an

10

I. Benhar / Biotechnology Advances 19 (2001) 133

intramolecular disulfide bridge, and pair to form a functional scFv (Skerra and Pluckthun, 1988; Better et al., 1988). Initially, phage vectors that carried all the genetic information required for the phage life cycle were used (McCafferty et al., 1990; Clackson et al., 1991). Later, phagemids became the popular type of vector for display. Phagemids are small plasmid vectors that carry gene III with appropriate cloning sites (for the insertion of cloned antibody genes) and the phage intergenic region (for rolling-circle replication and packaging) (Bass et al., 1990; Hoogenboom et al., 1991; Garrard et al., 1991). In phagemids, the scFv may be fused at the Nterminus of an intact gene III (McCafferty et al., 1990; Hoogenboom et al., 1991) or at the Nterminus of a truncated gene III, lacking the first two N-terminal domains (Bass et al., 1990; Barbas et al., 1991). Phagemids have high transformation efficiencies and are therefore ideally suited for generating very large repertoires. They may also be formatted for direct secretion of the unfused antibody fragment, without subcloning (Hoogenboom et al., 1991). Many phagemids utilize the lac promoter to control the expression of the antibodypIII fusion product (Hoogenboom et al., 1991; Barbas et al., 1991). For display of the antibody pIII product, glucose, acting in catabolite repression of the lac promoter, is removed or depleted, leading to the expression of sufficient fusion product to generate monovalent phage particles. Whenever expression-mediated toxicity is an issue [which is the case for some, mostly hybridoma-derived, antibody fragments (Thompson et al., 1996)], tighter regulation of expression levels may be required. The use of a lac promoter with an additional transcriptional terminator (Krebber et al., 1996), the tightly regulated tet promoter (Zahn et al., 1999) or of the phage shock promoter (psp) (Rakonjac et al., 1997) may allow display of relatively toxic products and reduce expression-mediated library biases. The phagemid DNA encoding the antibodypIII fusion are preferentially packaged into phage particles using a helper phage such as M13KO7 or VCS-M13, which supplies all structural proteins. Since the helper phage genome encodes wild-type pIII, typically over 90% of rescued phage display no antibody at all and the vast majority of the rescued phage particles that do display the fusion product will only contain a single copy. More efficient, multivalent display may be preferable when selecting very large antibody libraries, to guarantee selection when a limited number of phage particles per clone are available. Monovalent display, on the other hand, may be essential when selecting antibodies for higher affinity. Therefore, the use of inducible promoters (Lutz and Bujard, 1997) or the use of a helper phage with gene III deleted (Griffiths et al., 1993), may allow the modulation of the valency of displayed antibodies. Phage antibody selection involves the sequential enrichment of specific binding phage from a large excess of non-binding ones. This, as shown in Fig. 1, is achieved by multiple rounds of phage binding to the target, washing to remove nonspecific phage and elution to retrieve specific binding phage. Any method that separates phage that bind from those that do not, can be used for phage selection, and indeed, many different selection methods have been used. The most popular selection methods include affinity selection (also called biopanning) on immobilized antigen coated onto solid supports, columns or BIAcore sensor chips (Clackson et al., 1991; Marks et al., 1991; Griffiths et al., 1994; Malmborg et al., 1996), selection using biotinylated antigen (Hawkins et al., 1992), panning on fixed prokaryotic cells (Bradbury et

I. Benhar / Biotechnology Advances 19 (2001) 133

11

Fig. 1. The phage affinity-selection (biopanning cycle). Recombinant DNA techniques are used to generate a library consisting of millions of different antibodies, or of variants (mutants) of an existing antibody. The resulting phage library is subjected to several cycles of affinity selection including capture of phage with antigen, washing to remove unbound phage, elution to release antigen-bound phage, and amplification in E. coli. When adequate enrichment has been obtained, individual antigen-specific clones are isolated and characterized.

al., 1993) and on mammalian cells (Cai and Garen, 1996), subtractive selection using sorting procedures (de Kruif et al., 1995), enrichment on tissue sections or pieces of tissue (van Ewijk et al., 1997), and, finally, selections using living animals, as reported for peptide phage libraries (Pasqualini and Ruoslahti, 1996). Phage antibodies bound to antigen may be eluted in different ways: with (one step or gradients of) acidic solutions such as HCl or glycine buffers, with basic solutions like triethylamine, with chaotropic agents, with DTT when biotin is linked to antigen by a disulfide bridge (Marks et al., 1991), by enzymatic cleavage of a protease site engineered between the antibody and gene III, or by competition with excess antigen (Marks et al., 1991; Bradbury et al., 1993; Griffiths et al., 1994; de Kruif et al., 1995). Antibody phage display has been the leading tool in antibody engineering during the last decade. Initially applied to the isolation of antibodies against various target molecules, the

12

I. Benhar / Biotechnology Advances 19 (2001) 133

technology has evolved into an efficient tool for the in vitro manipulation of antibody affinity, specificity and stability. The most successful application of antibody phage display has been the isolation of antibodies from large libraries. For that purpose, three types of libraries were used. First, immune libraries that were generated by cloning antibody genes from B cells (usually from spleens) of immunized donors. This approach takes advantage of the fact that antibodies directed against the immunogen are enriched in such donors, and that they have been subjected to in vivo affinity maturation by the host immune system (Clackson et al., 1991; Burton et al., 1991). A further advantage of immune libraries is the ability to isolate many antibodies that are immunogen-specific from a single library. Immune libraries have been produced from a variety of species, including mouse (Clackson et al., 1991; Chester et al., 1994; Kettleborough et al., 1994; Berdichevsky et al., 1999), human (Barbas et al., 1993; Cai and Garen, 1995), rabbit (Lang et al., 1996; Li et al., 2000a), camelids (Arbabi Ghahroudi et al., 1997; Muyldermans and Lauwereys, 1999), sheep (Li et al., 2000b), and chicken (Davies et al., 1995; Yamanaka et al., 1996). Immunization, however, is not always feasible, due to ethical considerations, possible toxicity or lack of immunogenicity of the antigen. Consequently, non-immune libraries were developed. Non-immune libraries are further divided into naive and synthetic antibody libraries. Naive libraries were constructed using the same methodology that was applied in the construction of immune libraries. The difference was that here B cells from non-immunized donors were used as sources for antibody genes. Several naive libraries have been constructed, and because they are not biased towards any specific antigen, were used to isolate antibodies against multiple antigens (Marks et al., 1991; Gram et al., 1992; Vaughan et al., 1996). With naive libraries, the affinities of isolated antibodies correlated with the library size. Hence it was necessary to construct very large libraries to obtain high-affinity antibodies (Vaughan et al., 1996; de Haard et al., 1999). Synthetic or semisynthetic libraries were the next generation of non-immune libraries. These were constructed artificially by assembling in vitro antibody V genes with synthetic D and J segments. The DJ segments contained information for coding CDR3 loops of random sequence and varying length. The resulting libraries were sometimes referred to as `singlepot' libraries, because each such library was a source of antibodies directed against multiple antigens (Hoogenboom and Winter, 1992; Garrard and Henner, 1993; Nissim et al., 1994; de Kruif et al., 1995). Antibodies selected from phage libraries are not always suitable for direct application as therapeutics or biotechnological reagents. In some cases, manipulation of the antibody affinity, valency, specificity or stability was required. For such cases, phage display was applied, in a manner similar to the process of producing synthetic libraries and selecting the best binders from them. Such secondary libraries contained variants of the initially isolated antibodies, where mutations were introduced either randomly, or following rational design. Mutations were introduced into the antibody genes using one of several methods, including site-directed mutagenesis (Schier et al., 1996; Chowdhury and Pastan, 1999) error-prone PCR (Hawkins et al., 1992), Chain shuffling (Clackson et al., 1991; Marks et al., 1992), DNA shuffling (Crameri et al., 1996), and mutator E. coli strains (Low et al., 1996; Irving et al., 1996). Using such approaches it was possible to obtain antibodies having extremely high affinities, well below 100 pM (Schier et al., 1996; Yang et al., 1995). Advanced phage-

I. Benhar / Biotechnology Advances 19 (2001) 133

13

selection schemes in combination with secondary antibody libraries were useful in the isolation of antibodies having improved stability and folding properties (Spada et al., 1998; Forrer et al., 1999; Jung et al., 1999). Finally, phage display enabled the isolation of catalytic antibodies, where catalysis or binding to transition-state analogs were used as the selective criteria (Sastry et al., 1995; Janda et al., 1994; Baca et al., 1997; Fujii et al., 1998). The expression of functional antibodies on the surface of E. coli (Fuchs et al., 1991; Little et al., 1993; Francisco et al., 1993b; Maurer et al., 1997) and staphylococci (Gunneriusson et al., 1996, 1999) has led to discussion whether this strategy would be a way to create inexpensive diagnostic tools or alternatives to the rapidly developing phage technology for the selection of peptides or recombinant antibody fragments from large libraries (Little et al., 1993; Georgiou et al., 1997). Most studies of bacterial display of antibodies involved the Lpp-OmpAH system developed by Georgiou and coworkers (Francisco and Georgiou, 1994; Francisco et al., 1993b; Georgiou et al., 1997; Chen et al., 1996) (schematically drawn in Fig. 2). The Lpp-OmpAH system was applied for the development of a quantitative immunoassay that utilizes E. coli bacteria expressing scFv antibody fragments attached to the cell surface (Chen et al., 1996). Scatchard analysis demonstrated that the antibodies on the surface of the cells retained full binding activity and that about 60 000 scFv molecules were displayed per cell. The bacteria were used as the antibody reagent in that assay, and, following incubation with analyte simple centrifugation could separate the antibody-bound from unbound analyte. The Lpp-OmpAH system was also applied for antibody affinity maturation by bacterial surface display of scFv libraries utilizing FACS (Daugherty et al., 1998). This system was employed to isolate clones with high affinity to a fluorescently labeled hapten from libraries

Fig. 2. Schematic representation of the Lpp-OmpAH display system. Surface display on E. coli is facilitated by the Lpp signal peptide (residues 1 9) followed by residues 46 159 of OmpA. The OmpA fragment is composed of five membrane-spanning b-strands, with the C-terminus located outside the cell. A protein linked to the Cterminus of the construct is thus located at the cell surface.

14

I. Benhar / Biotechnology Advances 19 (2001) 133

constructed by randomizing heavy and light-chain residues in the anti-digoxin 26-10 derived scFv-antibody. The use of flow cytometry enabled the detection of rare library members directly in heterogeneous populations and the optimization of selection conditions prior to sorting. A similar approach was used for in vitro scanning saturation mutagenesis of the antidigoxin 26-10 antibody's binding site that allows the high throughput production and characterization of antibody mutants with improved binding affinity (Chen et al., 1999), and to investigate the effects of mutation frequency on the affinity maturation of the displayed antibody (Daugherty et al., 2000). A possible advantage of the bacterial-display systems over the phage-display techniques lies in the fact that bacterial selection can be accomplished through FACS technology, using fluorescently labelled antigens. This avoids crucial steps in phage-display selection procedures such as immobilization of the antigen, elution of bound phage and reinfection of bacteria with eluted phage. Filamentous phages are too small to be compatible with current FACS technology (Francisco and Georgiou, 1994). It has, however, been shown that E. coli cells expressing functional cell-surface-anchored antibody fragments can he separated by FACS (Francisco and Georgiou, 1994; Francisco et al., 1993b; Fuchs et al., 1996; Chen et al., 1999; Daugherty et al., 2000). Moreover, by manipulating the concentration of the fluorescent ligand it is possible to obtain higher and lower affinity antibodies in a single experiment based on signal intensity. Using this approach it was possible to analyze quantitatively the effect of mutation frequency (inserted by error-prone PCR) on the frequency of clones that remain functional and on the likelihood of isolating gain-of-function mutants of a high-affinity single-chain antibodies (Daugherty et al., 2000). The results demonstrated that large protein libraries can be surface displayed on bacteria, that scFv fragments can be functionally expressed on bacterial cells, and that such cells can be efficiently enriched by FACS technology. This approach should promote future use of bacterial display as an alternative or at least a complement to the phage systems. 7. Surface display of enzymes, substrates and inhibitors Both phage and cell display have contributed to the field of enzyme technology. A number of enzymes have been displayed on phage (McCafferly et al., 1991; Corey et al., 1993; Soumillion et al., 1994; Maruyama et al., 1994; Mikawa et al., 1996; Mullaney and Black, 1996; Pedersen et al., 1998; Demartis et al., 1999; Forrer et al., 1999; Hufton et al., 1999; Legendre et al., 2000) and bacteria (Francisco et al., 1992; Schneewind et al., 1992, 1993; Strauss and Gotz, 1996; Jung et al., 1998a, b). Initially attempts were made to apply phage display for the isolation of `improved' enzymes, or for enzymes with altered substrate specificity by selecting for improved binding to transition-state analogs. This approach resulted in modest improvements for catalytic antibodies displayed on phage (Fujii et al., 1998; Baca et al., 1997; Hansson et al., 1997; Arkin and Wells, 1998). However, binding of transition-state analogs does not correlate well with improved catalysis (Fujii et al., 1998; Arkin and Wells, 1998). Specialized selection schemes using reactive substrates (Janda et al., 1994), inhibitors (Light and Lerner, 1995), active-site ligands (Widersten and Mannervik, 1995), and reactive products (Janda et al., 1997) have also been used in attempts to select for

I. Benhar / Biotechnology Advances 19 (2001) 133

15

improved catalysts. The selection from libraries of phage-displayed enzymes based on catalytic activity of the mutants is more complex than selection based on affinity for a ligand, which is how phage display is routinely applied. Several strategies have been proposed to alleviate this problem (Soumillion et al., 1994; Hansson et al., 1997; Atwell and Wells, 1999; Demartis et al., 1999; Jestin et al., 1999). These novel approaches are based on linking the enzymatic reaction product to the surface of the phage that displays the active enzyme. It is thus possible to identify the reaction products (and the phage they are linked to) with an appropriate antibody. Recently, a technique based on phage display for the directed evolution of enzymes of industrial interest was proposed. This technique is based on the use of relevant suicide inhibitors specifically designed to irreversibly bind the active pocket of the enzyme following their cleavage (Janda et al., 1997). By applying phage display for enzyme evolution, it should also be possible to focus selection on particular properties besides catalysis. The robustness of the phage themselves (their tolerance to a wide pH range and buffer conditions) makes it is possible to improve the stability and the catalytic properties of the displayed enzymes by altering library construction and selection conditions. In addition to the display of enzymes, phage were used to display enzyme substrates and enzyme inhibitors. The term `substrate phage' was coined by Matthews and Wells (1993) to describe a prototype approach for identifying novel substrates for enzymes (mostly proteases). A library of fusion proteins was constructed containing an amino-terminal domain used to bind to an affinity support, followed by a randomized protease substrate sequence and the carboxylterminal domain of M13 gene III. Each fusion protein was displayed as a single copy on filamentous phagemid particles (substrate phage). Phage were then bound to an affinity support and treated with the protease of interest. Phage that displayed good protease substrates were released, whereas phage with substrates that resisted proteolysis remained bound. This approach was valuable in identifying novel substrates or optimizing existing ones (Matthews and Wells, 1993; O'Boyle et al., 1997; Ke et al., 1997a,b; Ohkubo et al., 1999). A direct approach similar to that used for the isolation of antibodies from phage libraries was undertaken for the isolation of enzyme inhibitors. In these studies, engineered variants, based on small proteins derived from natural inhibitors (Roberts et al., 1992; Pannekoek et al., 1993; van Meijer et al., 1996; Huang et al., 1998) or peptide libraries (Ploug et al., 1998), were displayed on phage. Inhibitors were isolated by selecting phage on immobilized enzymes. Potent inhibitors of trypsin (Markland et al., 1991; Roberts et al., 1992), tPA (Pannekoek et al., 1993; van Meijer et al., 1996; Ploug et al., 1998), beta-lactamase (Huang et al., 1998), serine proteases (Jespers et al., 1995), human plasma kallikrein and human thrombin (Markland et al., 1996) were readily isolated using this approach. Enzyme inhibitors that are based on novel protein scaffolds were isolated by Sollazzo and coworkers (Martin et al., 1997; Dimasi et al., 1997). They isolated inhibitors of hepatitis C virus NS3 protease from an antibody `camelized VH' library (Martin et al., 1997) and from a `Minibodi' library (Dimasi et al., 1997). While phage display was used primarily for the molecular evolution of enzymes, substrates and inhibitors, with cell display the emphasis was different. Certain enzymes have been expressed with retained activity on the surface of gram-negative and grampositive bacteria (Table 2), and the potential use of such recombinant bacteria as novel

16

I. Benhar / Biotechnology Advances 19 (2001) 133

microbial biocatalysts has been investigated. E. coli b-lactamase, which is normally a periplasmic enzyme, has been successfully exposed on the outer surface of E. coli using the pullulanase system (Kornacker and Pugsley, 1990) or the Lpp-OmpAH system (Francisco et al., 1992) as has been the Cex exoglucanase of Cellulomonas fimi (Francisco et al., 1993a). Bacteria displaying heterologous enzymes on their surface hold great potential as wholecell biocatalysts. For industrial applications, such surface-engineered cells need to be killed and chemically fixed to prevent disintegration and leakage of the displayed proteins under process conditions. It is also highly desirable to couple the chemically stabilized cells onto a solid support matrix for additional mechanical stability, flexibility in reactor choice, and easy separation from processed medium. Using such an approach, a method designed to prepare an immobilized whole cell biocatalyst from beta-lactamase-displaying E. coli cells was recently described (Freeman et al., 1999). The method is based on crosslinking carried out in the presence of beta-lactamase inhibitors, that protects the active site from chemical modification resulting in up to threefold higher specific activities without affecting the cellstabilizing effect of the glutaraldehyde treatment. However, not all enzymes can be efficiently exposed on the bacterial surface. Alkaline phosphatase, which is normally periplasmic, was found to be retained in the periplasm of E. coli when expressed via the Lpp-OmpAH system (Stathopoulos et al., 1996). The efficacy of using E. coli with surface-displayed heterologous enzymes as novel microbial biocatalysts remains to be seen. Surface expression is undoubtedly an inexpensive way to produce immobilized enzymes, but gram-negative bacteria might suffer from practical drawbacks such as inhibition of growth or cell lysis. The E. coli display systems seem to be limited in the size of proteins they are capable of exporting to the surface (Tommassen et al., 1985; Stathopoulos et al., 1996). Recently, the ice-nucleation protein (Inp), a glycosyl phosphatidylinositolanchored outer membrane protein found in some gram-negative bacteria was used for enzyme display in E. coli. By using Pseudomonas syringae Inp as an anchoring motif, the functional display of Zymomonas mobilis levansucrase (LevU) on the cell surface was obtained (Jung et al., 1998a,b). The cells expressing Inp-LevU were found to retain both the ice-nucleation and whole-cell levansucrase enzyme activities. Viability of the cells was also maintained over 48 h in the stationary phase. When the levansucrase-displayed cells were used as the enzyme source, levan was efficiently synthesized from sucrose with 34% conversion yield, generating glucose as a by-product. Regarding enzyme display on gram-positive bacteria, a lipase from Staphylococcus hyicus and E. coli b-lactamase were expressed on the outer cell surface of S. carnosus with retained activity (Strauss and Gotz, 1996). Approximately 10 000 enzyme molecules were found to be present on each cell and it was suggested that the rigid structure of gram-positive bacteria would make them particularly appropriate as microbial catalysts. More recently, bacterial surface display was applied for enzyme evolution. The E. coli surface-display system based on Inp (Jung et al., 1998a,b) was used to selectively screen for improved variants of carboxymethyl cellulase (CMCase) (Kim et al., 2000). A library of mutated CMCase genes generated by DNA shuffling was fused to the ice nucleation protein gene so that the resulting fusion proteins were displayed on the bacterial cell surface. Some cells displaying mutant proteins grew more rapidly on carboxymethyl cellulose plates than controls, forming heterogeneous colonies. In contrast, cells displaying the parent CMCase

I. Benhar / Biotechnology Advances 19 (2001) 133

17

formed uniform tiny colonies. These variations in growth rate were assumed to result from altered availability of glucose caused by differences in the activity of variant CMCases at the cell surface. Staining assays indicate that large, rapidly growing colonies exhibited increased CMCase activity. Increased CMCase activity was confirmed by assaying the specific activities of cell extracts after the expression of unfused forms of the variant genes in the cytoplasm. The best-evolved CMCases showed about a 5- and 2.2-fold increase in activity in the fused and free forms, respectively. Sequencing of evolved CMCase variant genes showed that most amino acid substitutions occurred within the catalytic domain of the enzyme. These results demonstrate that the bacterial surface display of enzyme libraries may provide a direct way to correlate evolved enzyme activity with cell growth rates. This technique may provide a useful technology platform for directed evolution and high-throughput screening of industrial enzymes, including hydrolases. 8. Vaccine development Both phage- and cell-display systems have been used for vaccine development, but with differing approaches. Phage display has been used mostly for the identification of immunogenic epitopes or mimotopes on displayed peptides. These, in turn could become lead molecules for the design of peptide-based vaccines. In only a handful of cases have peptidedisplaying phage themselves been used for immunization. A number of groups used monoclonal of polyclonal antibodies to select peptide libraries. At the dawn of the phage-display era, George Smith, who pioneered the phage-display technology (Smith, 1985), proposed that foreign sequences could be inserted within the minor coat protein, pIII, of filamentous phage. This will result in the creation of a fusion protein that is incorporated into the virion; the virions display the foreign amino acids encoded in the insert on their surface. Phage, bearing specific antigenic determinants from a target gene, can be purified in infectious form from a vast excess of phage that carry other determinants, by virtue of their affinity to the antibody directed against the gene product. Fusion phage can be used as a source of antigen, as a carrierhapten conjugate for obtaining immunological reagents in rabbits, and for B epitope mapping. By generating a fusion-phage library expressing virtually all possible short, amino acid sequences, it may be possible to study epitopes on immunologically important proteins without the use of synthetic peptides and without ever having cloned the genes (Parmley and Smith, 1989). Indeed, de la Cruz et al. (1988) showed that cloning of repeat regions of the circumsporozoite protein gene of Plasmodium falciparum into the pIII gene of a filamentous phage results in the display of the recombinant proteins on the phage surface. The displaying phage were both antigenic and immunogenic when injected to rabbits. Shortly thereafter phage display in the form of combinatorial peptide libraries was used to map linear epitopes of monoclonal antibodies (Scott and Smith, 1990; Cwirla et al., 1990; Devlin et al., 1990; Felici et al., 1991, reviewed in Lane and Stephen, 1993; Daniels and Lane, 1996). The potential of phage display for identification of neutralizing viral epitopes was demonstrated by Perham (di Marzo Veronese et al., 1994) who inserted peptide sequences

18

I. Benhar / Biotechnology Advances 19 (2001) 133

from the V3 loop of gp120 from HIV-1 into the N-terminal region of the major coat protein (pVIII) of filamentous bacteriophage fd. This approach led to their display in multiple copies on the surface of the virion. Peptides displayed in this way were shown to be remarkably effective structural mimics of the natural epitope. They were recognized by human HIV antisera and evoked high titres of antibodies in mice, which cross-reacted with other strains of HIV and were capable of neutralizing the virus. Soon it was realized, however, that peptide sequences selected from phage library do not always correspond to the exact (linear) sequence of the original epitope. It was postulated that such peptides mimic the structural features of the epitope in being recognized by epitopespecific antibodies, and by eliciting upon injection into animals, antibodies capable of recognizing the original epitope. Such peptide mimics were thus named `mimotypes' (Cortese et al., 1994; Folgori et al., 1994; Mennuni et al., 1996; Demangel et al., 1996). Mimotypes were later identified for various nonprotein molecules such as the O-antigen part of the human pathogen Shigella flexneri serotype 5a lypopolysaccharide (Phalipon et al., 1997), the Neisseria meningitidis group B polysaccharide (Moe et al., 1999) and meningococcal group C polysaccharide (Grothaus et al., 2000). While most of the studies were carried out using filamentous phage peptide libraries, other phage systems were later developed for similar purposes. A 36-amino-acid PorA peptide from N. meningitidis was cloned into the display vectors to generate fusions at the N terminus of Hoc or Soc capsid genes of phage T4 (Jiang et al., 1997). The PorA-Hoc and PorA-Soc fusion proteins retained the ability to bind to the capsid surface, and the bound peptide was displayed in an accessible form as shown by its reactivity with specific monoclonal antibodies. Both the PorA-Hoc and PorA-Soc recombinant phages were highly immunogenic in mice and elicited strong antipeptide antibody titers. Once it was established that phage display can be useful for epitope identification, the use of displaying phage as immunogens was considered. A number of groups compared the magnitude of antibody response that was elicited by various immunization protocols. Tested criteria included display valency (monovalent vs. polyvalent display), the presence and nature of added adjuvants, and the route of administration. Willis et al. (1993) displayed the antigenic determinants of the circumsporozoite protein of the malaria parasite, P. falciparum in multiple copies on pVIII of Fd phage. The peptide epitopes in the hybrid phage were found to be strongly immunogenic in four different strains of mice without the use of external adjuvants, and the antibodies were highly specific to the individual epitopes. When tested in nude mice, the immune response was found to be T-cell dependent and to undergo class-switching from IgM to IgG. Proliferation assays of T cells, taken from lymph nodes of BALB/c mice injected with bacteriophage particles in the presence or absence of Freund's complete adjuvant, indicated no difference in the immune response. Meola et al. (1995) tested if mimotopes could be useful in developing a vaccine against the human hepatitis B virus. They compared the humoral immune response of animals immunized either with a recombinant HbsAg vaccine or with mimotopes. Immunogens were prepared by fusing the mimotypes on different carrier molecules (phage coat protein pIII and pVIII, recombinant human H ferritin, HBV core peptide) and by synthesizing multiple antigenic peptides carrying the mimotypes' amino acid sequences. These immunogens were injected

I. Benhar / Biotechnology Advances 19 (2001) 133

19

into mice and rabbits and sera were collected and tested for the presence of HBsAg-specific antibodies. The results confirmed that mimotopes could induce a humoral immune response resembling that induced by the original antigen, and HBsAg mimotopes displayed on phage prove to be the best immunogens, inducing the most repro- ducible and potent immunization. Delmastro et al. (1997) showed that intranasal or intragastric administration of phage into mice induces an immunological response both to the wild type phage proteins and to mimotopes displayed on them. Using mimotopes of human hepatitis B virus surface antigen and of human hepatitis C virus peptides, they showed that the response induced by oral administration is specifically cross-reactive with the original antigen. Bastien et al. (1997) investigated whether a recombinant bacteriophage displaying a disease-specific protective epitope could be experimentally used as a vaccine to confer protection of immunized animals against infection. They genetically engineered a recombinant phage, fd, displaying at its surface a chimeric pIII coat protein (monovalent display) fused to the previously identified protective epitope 173187 from the glycoprotein G of the human respiratory syncytial virus. A selected recombinant fd phage elicited a strong immune response in mice, inducing a high level of circulating RSV-specific antibodies. Mice immunized with the recombinant phage acquired a complete resistance to RSV infection as evidenced by the lack of detectable virus particles in their lungs following intranasal challenge with live RSV. This was the first report of the ability of a phage presenting an immunogenic peptide to prevent infection of immunized animals by a pathogen. Recently, Zuercher et al. (2000) tested the effect of exposure of displaying phage to gastric fluid. This approach was examined, since an essential requirement for oral vaccines is the ability to survive the harsh environment of the stomach in an antigenically intact form. The feasibility of this approach was tested in a simulated gastric fluid using two different mimotopes as well as an anti-idiotypic Fab of the non-anaphylactogenic monoclonal anti-IgE antibody BSW17. All phage clones remained infective after this treatment. However, only epitopes displayed on the pVIII protein (polyvalent display) were still recognized by BSW17 whereas pIII-expressed (monovalent display) epitopes were rapidly inactivated. Surprisingly, when used for oral immunization of mice all phage clones induced anti-IgE antibodies. In contrast, oral immunization with the purified, pVIII protein that displayed the mimotope also induced anti-phage antibodies, but no anti-IgE antibodies were obtained. After feeding a single dose of mimotope-displaying bacteriophage, phage DNA could be detected in mouse feces for 10 days. These results have shown that epitope-displaying bacteriophage can be used to induce an epitope-specific antibody response via the oral route. In conclusion, phage that display epitopes or mimotopes may emerge as useful tools for the development of effective vaccines or may serve as vaccine-delivery vehicles themselves. For the application of phage as vaccines it seems that polyvalent rather than monovalent display are more likely to elicit an efficient immune response, that oral administration is the more effective route for immunization, and that adjuvant is usually not required. Such immunization protocols can be less expensive and more effective than the conventional method of synthesis and coupling peptide ligands to particulate synthetic carriers for immunization. The most common application of bacterial surface display has been in the development of live bacterial vaccine delivery systems. In contrast to phage display, which was a tool for epitope discovery and for vaccine delivery, displaying immunogenic peptides or proteins on

20

I. Benhar / Biotechnology Advances 19 (2001) 133

bacteria for the purpose of vaccine development followed a different scheme. Here, an already identified vaccine candidate (an immunogenic peptide or protein) was displayed on bacteria to be used as carriers for vaccine delivery. Gram-positive cells were earlier considered for vaccine delivery. Nguyen et al. (1993) developed a system for the surface expression of heterologous receptors on the cell surface of S. xylosus. That expression system was constructed by subcloning gene fragments encoding for peptides to be displayed on the cell surface into an E. colistaphylococci shuttle vector. The vector was designed for targeting of recombinant fusion proteins to the outer cell surface of the gram-positive host cell. A specific humoral immune response could be elicited in mice by oral immunization with the recombinant S. xylosus cells, suggesting that this type of gram-positive bacteria might offer potential vehicles for oral vaccination. Liljeqvist et al. (1999) assayed the surface expression in S. carnosus of three different fibronectin binding domains (FNBDs), derived from fibronectin binding proteins of Streptococcus dysgalactiae and S. aureus. All three surface-displayed FNBDs were demonstrated to bind fibronectin in whole-cell enzyme-linked binding assays. Furthermore, for one of the constructs, intranasal immunizations with the recombinant bacteria resulted in improved antibody responses to a model immunogen present within the chimeric surface proteins. To avoid the use of engineered pathogens for vaccine delivery, Fischetti et al. (1996) developed systems that allow the expression of heterologous antigens in commensal gram-positive bacteria. In some cases, both a serum IgG and secretory IgA response were induced to the recombinant protein after vaccination, verifying the validity of the approach. These live recombinant bacteria may be used in the future to introduce a protective immune response to pathogenic microorganisms after mucosal colonization. Many of the described gram-negative display systems have initially been evaluated in E. coli for surface display of various antigenic determinants, and subsequently applied to Salmonella spp. These species are of interest for the live, oral delivery of heterologous antigens for immunization. An early study demonstrated that bacteria that express recombinant proteins (although not at the surface in this particular example) could be used as live oral vaccines. In this study, Poirier et al. (1988) investigated the use of attenuated strains of Salmonella to deliver cloned antiphagocytic virulence determinants of unrelated bacteria. Thus, the aroA strain of S. typhimurium SL3261 was transformed with a low-copy plasmid that contained the cloned gene spm5, encoding streptococcal M protein (the major virulence factor of these organisms). The transformed Salmonella expressed type-5 M protein in the cytoplasmic fraction, and when fed orally to BALB/c mice, evoked both serum and salivary IgA, IgG, and IgM antibodies directed against type-5 M protein. The orally immunized mice were completely protected against both intranasal and intraperitoneal challenge infections with virulent S. typhimurium SL1344 or M5 streptococci. In a second study, mice were immunized with live Salmonella dublin, expressing a flagellum with an inserted cholera toxin, hepatitis B virus or influenza virus haemaglutinin epitopes. The immunized mice produced antigen-specific antibodies and in some cases partial protection from virus challenge (Wu et al., 1989; Newton et al., 1989). It was suggested that epitopes exposed at the bacterial cell surface induce antibody responses in a T-cell-independent manner and that the lipopolysaccharides may serve as adjuvants. These studies provided early evidence that an attenuated strain of Salmonella can be used effectively as a general vaccine vehicle to deliver antiphagocytic virulence determinants of unrelated bacteria.

I. Benhar / Biotechnology Advances 19 (2001) 133

21

Schorr et al. (1991) used E. coli OmpA as a carrier for the expression of foreign antigens on the surface of gram-negative bacteria. Immunogenic moieties of P. falciparum blood stage antigens were expressed in an attenuated S. typhimurium strain. Live bacteria expressing the fusion proteins reacted with malarial antigen sera. Mice that were immunized orally with S. typhimurium cells expressing malarial antigens on their surface showed a humoral immune response. Leclerc et al. (1991) targeted two foreign B cell antigenic viral epitopes to different locations in the E. coli cell to examine what effect this had on antibody responses elicited by the recombinant bacteria. The two epitopes were peptides derived from the PreS2 region of hepatitis B virus and the C3 neutralization epitope of poliovirus type 1. They were each expressed in two forms either on the surface, as part of the outer-membrane protein LamB, or soluble in the periplasm, as part of the periplasmic protein MalE. When live bacteria expressing the foreign epitopes at the cell surface were used for immunization of mice, they induced T-cell-independent antibody responses characterized by a rapid induction of IgM and IgG antibodies. In contrast, when the same foreign epitopes were inserted into the MalE protein, the antibody response was only detectable after 3 weeks, belonged only to the IgG class and was strictly T-cell dependent. This study has therefore identified two major pathways by which epitopes expressed by bacterial cells can stimulate specific antibody responses. The first pathway is mediated by direct activation of B cells by bacterial cell-surface antigen and does not require T-cell help. The second pathway is T-cell dependent and concerns antigen that can be released from the bacteria in a soluble form. Stentebjerg-Olesen et al. (1997) assessed the potential of the major structural protein of type 1 fimbriae as a display system for heterologous sequences. As a reporter-epitope, a heterologous sequence mimicking a neutralizing epitope of the cholera toxin B chain was inserted into the fimA gene. The chimeric proteins were exposed on the bacterial surface and the cholera toxin epitope was authentically displayed, i.e. it was recognized on bacteria by specific antiserum. Immunization of rabbits with purified chimeric fimbriae resulted in serum, which specifically recognized cholera toxin B chain, confirming the utility of the employed strategy. It can be concluded that several of the developed bacterial surface-display systems, of both gram-positive and gram-negative bacteria merit additional future consideration for application as live or attenuated vaccines. 9. Phage and cell display come together As described in the previous sections, phage display has been used mostly for affinity screening of combinatorial peptide and protein libraries, to define epitopes for monoclonal antibodies, to select enzyme substrates and to screen cloned antibody repertoires. Phage libraries are enriched for specific binding clones by subjecting the phage to repetitive rounds of affinity selection (biopanning) that include binding, washing and elution steps, reinfection of bacteria and growth to re-express the binding molecule on the phage surface (Fig. 1). Several selection schemes may be applied to isolate antigen-specific phage from a heterogeneous population composed mostly of nonbinders. They include capture with immobilized antigen, capture with soluble ligand followed by isolation of phageligand complexes, selection on antigen columns, selection on living or fixed cells and selection in whole animals (Bradbury et al., 1993; Hoogenboom, 1997; Hoogenboom et al., 1998; Persic

22

I. Benhar / Biotechnology Advances 19 (2001) 133

et al., 1999; Pasqualini and Ruoslahti, 1996; Poul and Marks, 1999; Becerril et al., 1999). With most selection approaches, successive panning results in the diminution of diversity with a few dominant phage clones (usually those having the highest affinity or display efficiency) being recovered. It is therefore desirable to isolate binders following fewer, preferably a single panning cycle. This, in principle, could be achieved by linking antigen recognition with phage infectivity, as has been demonstrated in SIP (selectively infective phage) and similar selection strategies (Malmborg et al., 1997; Krebber et al., 1997). However, even with those powerful selection strategies, enrichment factors rarely exceed 104 per cycle. Recently we developed a novel selection scheme that combines bacterial surface display and phage display of polypeptides for the highly efficient isolation of binders, where antigendisplaying E. coli cells are used to capture antibody-displaying phage. Bradbury et al. (1993) used a similar approach to select phage on living or fixed antigen-displaying F bacteria. In that case, an epitope derived from the oncoprotein p21ras was expressed within the E. coli outer membrane protein, LamB. A library of single-chain Fvs was selected on the displaying bacteria. Bound phage were eluted at a high pH and used to infect F + bacteria for phage expansion. In the reported example, an enrichment factor of 13 000 was obtained in a single cycle. Our recently described system (Benhar et al., 2000) was named DIP (delayed infectivity panning). In this approach, the enrichment factor exceeded 106 per cycle. DIP takes advantage of the versatile Lpp-OmpAH bacterial surface-display system (Georgiou et al., 1997) where protein-coding sequences are fused to an Lpp-OmpAH hybrid sequence. In addition to the successful display of enzymes, antibodies, and cellulose-binding proteins that were described in the earlier sections, we could display receptor domains and a bacterial toxin fragment by this system. DIP combines bacterial display with conventional phage-display libraries (Fig. 3) where the selecting bacterial cells are also being infected by the captured phage. This is made possible by the fact that F + E. coli cells grown at low temperatures do not express the F pilus, which is required for phage infection (Novotny and Fives-Taylor, 1974; Novotny and Lavin, 1971). After phage capture and washing procedures, the bacteria are transferred to 37C so pilus expression is induced and infection ensues. Some antigens (in particular cell-surface receptors) are notoriously difficult to produce as recombinant proteins. In DIP, however, a purified antigen is not required for phage selection and identification. Using two model antibodyantigen pairs, we demonstrated enrichment of over 106-fold in a single DIP cycle for phage, which display a specific antibody, over those displaying an irrelevant antibody. We further showed the successful isolation of anti-toxin and anti-receptor antibodies from an immune phage library, a shuffled library and a large synthetic human library. DIP selection is far more efficient than panning with immobilized antigen or compared to other systems where antigen-displaying cells are used to capture antibody-displaying phage. Several factors may contribute to this observation. First, the selection `matrix' is bacteria that are essentially identical to those used to propagate the phage (except that the latter do not display the antigen). This may be regarded as a `counter-selection' step, where most of the antibody-displaying phage that recognize E. coli cell-surface epitopes adhere to the cells that were used to produce them and are thus eliminated. Second, we may have been fortunate in selecting the Lpp-OmpAH bacterial display format as the platform for DIP,

I. Benhar / Biotechnology Advances 19 (2001) 133

23

Fig. 3. Scheme of the DIP selection method. (A) Map of plasmid pIB-Tx used for bacterial display of antigens. (B) Scheme of DIP selection: (1) Antigen displaying E. coli grown at 16C, when the F pilus is not expressed, are mixed with antibody displaying phage. Antigen-specific phages bind to the cells. (2) Unbound phages are washed out. (3) Cells are shifted to 37C, so F pilus expression is induced, bound phage dissociate from antigen and infect the cell through the F pilus. (4) Infected cells where bacterial display plasmid and phage genome co-reside.

in that the antigen density and accessibility to antibodies may be better than antigens adsorbed to solid surfaces. Third, the antigen concentration used in DIP is relatively low: assuming 104 105 displayed molecules per bacterium (Francisco and Georgiou, 1994; Francisco et al., 1993b; Georgiou et al., 1997; Daugherty et al., 1998), with 1010 bacteria per milliliter we are in the nanomolar concentration range. Thus, our selection is more stringent than using micromolar concentrations of antigen, which is the general practice with immobilized antigens. The effectiveness of DIP makes it suitable for the efficient isolation of rare clones present in large libraries. Since a multitude of phage- and cell-based systems for surface display of polypeptides exists, DIP should be suitable for the isolation and characterisation of various proteinprotein interacting pairs. References
Abedi MR, Caponigro G, Kamb A. Green fluorescent protein as a scaffold for intracellular presentation of peptides. Nucleic Acids Res 1998;26:623 30. Agterberg M, Adriaanse H, van Bruggen A, Karperien M, Tommassen J. Outer-membrane PhoE protein of Escherichia coli K-12 as an exposure vector: possibilities and limitations. Gene 1990;88:37 45. Agterberg M, Adriaanse H, Lankhof H, Meloenn R, Tommassen J. Outer membrane PhoE protein of Escherichia coli as a carrier for foreign antigenic determinants: immunogenicity of epitopes of foot-andmouth disease virus. Vaccine 1990;8:85 91. Arbabi Ghahroudi M, Desmyter A, Wyns L, Hamers R, Muyldermans S. Selection and identification of single domain antibody fragments from camel heavy-chain antibodies. FEBS Lett 1997;414:521 6.

24

I. Benhar / Biotechnology Advances 19 (2001) 133

Arkin MR, Wells JA. Probing the importance of second sphere residues in an esterolytic antibody by phage display. J Mol Biol 1998;284:1083 94. Atwell S, Wells JA. Selection for improved subtiligases by phage display. Proc Natl Acad Sci USA 1999; 96:9497 502. Baca M, Scanlan TS, Stephenson RC, Wells JA. Phage display of a catalytic antibody to optimize affinity for transition-state analog binding. Proc Natl Acad Sci USA 1997;94:10063 8. Barbas CF, Kang AS, Lerner RA, Benkovic SJ. Assembly of combinatorial antibody libraries on phage surfaces: the gene III site. Proc Natl Acad Sci USA 1991;88:7978 82. Barbas CF 3rd, Collet TA, Amberg W, Roben P, Binley JM, Hoekstra D, Cababa D, Jones TM, Williamson RA, Pilkington GR, Haigwood NL, Cabezas E, Satterthwait AC, Sanz I, Burton DR. Molecular profile of an antibody response to HIV-1 as probed by combinatorial libraries. J Mol Biol 1993;230:812 23. Bass S, Greene R, Wells JA. Hormone phage: an enrichment method for variant proteins with altered binding properties. Proteins 1990;8:309 14. Bastien N, Trudel M, Simard C. Protective immune responses induced by the immunization of mice with a recombinant bacteriophage displaying an epitope of the human respiratory syncytial virus. Virology 1997; 234:118 22. Becerril B, Poul MA, Marks JD. Toward selection of internalizing antibodies from phage libraries. Biochem Biophys Res Commun 1999;255:386 93. Benhar I, Nahary L, Shaky S, Azriel R, Tamarkin A, Wels W. DIP selection of phage-antibodies mediated by antigen expression on live bacteria. J Mol Biol 2000;301:893 904. Berdichevsky Y, Ben-Zeev E, Lamed R, Benhar I. Phage display of a cellulose binding domain from Clostridium thermocellum and its application as a tool for antibody engineering. J Immunol Methods 1999; 228:151 62. Better M, Chang CP, Robinson RR, Horwitz AH. Escherichia coli secretion of an active chimeric antibody fragment. Science 1988;240:1041 3. Bingle WH, Nomellini JF, Smit J. Cell-surface display of a Pseudomonas aeruginosa strain K pilin peptide within the paracrystalline S-layer of Caulobacter crescentus. Mol Microbiol 1997;26:277 88. Bradbury A, Persic L, Werge T, Cattaneo A. Use of living columns to select specific phage antibodies. Biotechnology 1993;11:1565 9. Buchli PJ, Wu Z, Ciardelli TL. The functional display of interleukin-2 on filamentous phage. Arch Biochem Biophys 1997;339:79 84. Burton DR, Barbas CF. Human antibodies from combinatorial libraries. Adv Immunol 1994;57:191 280. Burton D, Barbas CF, Persson M, Koenig S, Chanock R, Lerner RA. A large array of human monoclonal antibodies to type 1 human immunodeficiency virus from combinatorial libraries of asymptomatic seropositive individuals. Proc Natl Acad Sci USA 1991;88:10134 7. Cabilly S. The basic structure of filamentous phage and its use in the display of combinatorial peptide libraries. Mol Biotechnol 1999;12:143 8. Cai X, Garen A. Anti-melanoma antibodies from melanoma patients immunized with genetically modified autologous tumor cells: selection of specific antibodies from single-chain Fv fusion phage libraries. Proc Natl Acad Sci USA 1995;92:6537 41. Cai X, Garen A. A melanoma-specific VH antibody cloned from a fusion phage library of a vaccinated melanoma patient. Proc Natl Acad Sci USA 1996;93:6280 5. Chang H, Lo SJ. Modification with a phosphorylation tag of PKA in the TraT-based display vector of Escherichia coli. J Biotechnol 2000;78:115 22. Chang HJ, Sheu SY, Lo SJ. Expression of foreign antigens on the surface of Escherichia coli by fusion to the outer membrane protein traT. J Biomed Sci 1999;6:64 70. Charbit A, Molla A, Saurin W, Hofnung M. Versatility of a vector for expressing foreign polypeptides at the surface of gram-negative bacteria. Gene 1988;70:181 9. Chen G, Cloud J, Georgiou G, Iverson BL. A quantitative immunoassay utilizing Escherichia coli cells possessing surface-expressed single chain Fv molecules. Biotechnol Prog 1996;12:572 4. Chen G, Dubrawsky I, Mendez P, Georgiou G, Iverson BL. In vitro scanning saturation mutagenesis of all the specificity determining residues in an antibody binding site. Protein Eng 1999;12:349 56.

I. Benhar / Biotechnology Advances 19 (2001) 133

25

Chester KA, Begent RH, Robson L, Keep P, Pedley RB, Boden JA, Boxer G, Green A, Winter G, Cochet O, Hawkins RE. Phage libraries for generation of clinically useful antibodies. Lancet 1994;343:455 6. Choo Y, Klug A. Toward a code for the interactions of zinc fingers with DNA: selection of randomized fingers displayed on phage. Proc Natl Acad Sci USA 1994;91:11163 7. Choo Y, Castellanos A, Garcia-Hernandez B, Sanchez-Garcia I, Klug A. Promoter-specific activation of gene expression directed by bacteriophage-selected zinc fingers. J Mol Biol 1997;273:525 32. Chowdhury PS, Pastan I. Improving antibody affinity by mimicking somatic hypermutation in vitro. Nat Biotechnol 1999;17:568 72. Clackson T, Hoogenboom HR, Griffiths AD, Winter G. Making antibody fragments using phage display libraries. Nature 1991;352:624 8. Corey DR, Shiau AK, Yang Q, Janowski BA, Craik CS. Trypsin display on the surface of bacteriophage. Gene 1993;128:129 34. Cortese R, Felici F, Galfre G, Luzzago A, Monaci P, Nicosia A. Epitope discovery using peptide libraries displayed on phage. Trends Biotechnol 1994;12:262 7. Cortese R, Monaci P, Luzzago A, Santini C, Bartoli F, Cortese I, Fortugno P, Galfre G, Nicosia A, Felici F. Selection of biologically active peptides by phage display of random peptide libraries. Curr Opin Biotechnol 1996;7:616 21. Crameri A, Cwirla S, Stemmer WP. Construction and evolution of antibody-phage libraries by DNA shuffling. Nat Med 1996;2:100 2. Cwirla SE, Peters EA, Barrett RW, Dower WJ. Peptides on phage: a vast library of peptides for identifying ligands. Proc Natl Acad Sci USA 1990;87:6378 82. Daniels DA, Lane DP. Phage peptide libraries. Methods 1996;9:494 507. Daugherty PS, Chen G, Olson MJ, Iverson BL, Georgiou G. Antibody affinity maturation using bacterial surface display. Protein Eng 1998;11:825 32. Daugherty PS, Chen G, Iverson BL, Georgiou G. Quantitative analysis of the effect of the mutation frequency on the affinity maturation of single chain Fv antibodies. Proc Natl Acad Sci USA 2000;97:2029 34. Davies EL, Smith JS, Birkett CR, Manser JM, Anderson-Dear DV, Young JR. Selection of specific phage-display antibodies using libraries derived from chicken immunoglobulin genes. J Immunol Methods 1995; 186:125 35. de Haard HJ, van Neer N, Reurs A, Huftonn SE, Roovers RC, Henderikx P, de Bruine AP, Arends JW, Hoogenboom HR. A large non-immunized human Fab fragment phage library that permits rapid isolation and kinetic analysis of high affinity antibodies. J Biol Chem 1999;274:18218 30. de Kruif J, Terstappen L, Boel E, Logtenberg T. Rapid selection of cell subpopulation-specific human monoclonal antibodies from a synthetic phage antibody library. Proc Natl Acad Sci USA 1995;92:3938 42. de la Cruz VF, Lal AA, McCutchan TF. Immunogenicity and epitope mapping of foreign sequences via genetically engineered filamentous phage. J Biol Chem 1988;263:4318 22. Delmastro P, Meola A, Monaci P, Cortese R, Galfre G. Immunogenicity of filamentous phage displaying peptide mimotopes after oral administration. Vaccine 1997;15:1276 85. Demangel C, Lafaye P, Mazie JC. Reproducing the immune response against the Plasmodium vivax merozoite surface protein 1 with mimotopes selected from a phage-displayed peptide library. Mol Immunol 1996; 33:909 16. Demartis S, Huber A, Viti F, Lozzi L, Giovannoni L, Neri P, Winter G, Neri D. A strategy for the isolation of catalytic activities from repertoires of enzymes displayed on phage. J Mol Biol 1999;286:617 33. Devlin JJ, Panganiban LC, Devlin PE. Random peptide libraries: a source of specific protein binding molecules. Science 1990;249:404 6. Dhillon JK, Drew PD, Porter AJ. Bacterial surface display of an anti-pollutant antibody fragment. Lett Appl Microbiol 1999;28:350 4. di Marzo Veronese F, Willis AE, Boyer-Thompson C, Appella E, Perham RN. Structural mimicry and enhanced immunogenicity of peptide epitopes displayed on filamentous bacteriophage. The V3 loop of HIV-1 gp120. J Mol Biol 1994;243:167 72. Dimasi N, Martin F, Volpari C, Brunetti M, Biasiol G, Altamura S, Cortese R, De Francesco R, Steinkuhler C, Sollazzo M. Characterization of engineered hepatitis C virus NS3 protease inhibitors affinity selected from human pancreatic secretory trypsin inhibitor and minibody repertoires. J Virol 1997;71:7461 9.

26

I. Benhar / Biotechnology Advances 19 (2001) 133

Fashena SJ, Serebriiskii I, Golemis EA. The continued evolution of two-hybrid screening approaches in yeast: how to outwit different preys with different baits. Gene 2000;250:1 14. Felici F, Castagnoli L, Musacchio A, Jappelli R, Cesareni G. Selection of antibody ligands from a large library of oligopeptides expressed on a multivalent exposition vector. J Mol Biol 1991;222:301 10. Felici F, Luzzago A, Monaci P, Nicosia A, Sollazzo M, Traboni C. Peptide and protein display on the surface of filamentous bacteriophage. Biotechnol Annu Rev 1995;1:149 83. Fischetti VA, Medaglini D, Pozzi G. Gram-positive commensal bacteria for mucosal vaccine delivery. Curr Opin Biotechnol 1996;7:659 66. Folgori A, Tafi R, Meola A, Felici F, Galfre G, Cortese R, Monaci P, Nicosia A. A general strategy to identify mimotopes of pathological antigens using only random peptide libraries and human sera. EMBO J 1994; 13:2236 43. Forrer P, Jung S, Pluckthun A. Beyond binding: using phage display to select for structure, folding and enzymatic activity in proteins. Curr Opin Struct Biol 1999;9:514 20. Francisco JA, Georgiou G. The expression of recombinant proteins on the external surface of Escherichia coli. Biotechnological applications. Ann NY Acad Sci 1994;745:372 82. Francisco JA, Earhart CF, Georgiou G. Transport and anchoring of betalactamase to the external surface of Escherichia coli. Proc Natl Acad Sci USA 1992;89:2713 7. Francisco JA, Stathopoulos C, Warren RA, Kilburn DG, Georgiou G. Specific adhesion and hydrolysis of cellulose by intact Escherichia coli expressing surface anchored cellulase or cellulose binding domains. Biotechnology 1993a;11:491 5. Francisco JA, Campbell R, Iverson BL, Georgiou G. Production and fluorescence-activated cell sorting of Escherichia coli expressing a functional antibody fragment on the external surface. Proc Natl Acad Sci USA 1993b;90:10444 8. Freeman A, Abramov S, Georgiou G. Site-protected fixation and immobilization of Escherichia coli cells displaying surface-anchored betalactamase. Biotechnol Bioeng 1999;62:155 9. Fuchs P, Breitling F, Dubel S, Seehaus T, Little M. Targeting recombinant antibodies to the surface of Escherichia coli: fusion to a peptidoglycan associated lipoprotein. Biotechnology 1991;9:1369 72. Fuchs P, Weichel W, Dubel S, Breitling F, Little M. Separation of E. coli expressing functional cell-wall bound antibody fragments by FACS. Immunotechnology 1996;2:97 102. Fujii I, Fukuyama S, Iwabuchi Y, Tanimura R. Evolving catalytic antibodies in a phage-displayed combinatorial library. Nat Biotechnol 1998;16:463 7. 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 30. Gargus JJ. Mutant isolation and gene transfer as tools in study of transport proteins. Am J Physiol 1987; 252:457 67. Garrard LJ, Henner DJ. Selection of an anti-IGF-1 Fab from a Fab phage library created by mutagenesis of multiple CDR loops. Gene 1993;128:103 9. Garrard LJ, Yang M, O'Connell MP, Kelley RF, Henner DJ. Fab assembly and enrichment in a monovalent phage display system. Biotechnology 1991;9:1373 7. Georgiou G, Stathopoulos C, Daugherty PS, Nayak AR, Iverson BL, Curtiss R. 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. Gram H, Marconi LA, Barbas CF, Collet TA, Lerner RA, Kang AS. In vitro selection and affinity maturation of antibodies from a naive combinatorial immunoglobulin library. Proc Natl Acad Sci USA 1992;89:3576 80. Gram H, Strittmatter U, Lorenz M, Gluck D, Zenke G. Phage display as a rapid gene expression system: production of bioactive cytokine-phage and generation of neutralizing monoclonal antibodies. J Immunol Methods 1993;161:169 76. Griffiths AD, Duncan AR. Strategies for selection of antibodies by phage display. Curr Opin Biotechnol 1998; 9:102 8. Griffiths AD, Malmqvist M, Marks JD, Bye JM, Embleton MJ, McCafferty J, Baier M, Holliger KP, Gorick BD, Hughes-Jones NC. Human anti-self antibodies with high specificity from phage display libraries. EMBO J 1993;12:725 34.

I. Benhar / Biotechnology Advances 19 (2001) 133

27

Griffiths AD, Williams SC, Hartley O, Tomlinson IM, Waterhouse P, Crosby WL, Kontermann RE, Jones PT, Low NM, Allison TJ, Prospero TD, Hoogenboom H, Nissim A, Cox JPL, Harrison JL, Zaccolo M, Gherardi E, Winter G. Isolation of high affinity human antibodies directly from large synthetic repertoires. EMBO J 1994;13:3245 60. Grothaus MC, Srivastava N, Smithson SL, Kieber-Emmons T, Williams DB, Carlone GM, Westerink MA. Selection of an immunogenic peptide mimic of the capsular polysaccharide of Neisseria meningitidis serogroup A using a peptide display library. Vaccine 2000;18:1253 63. Gunneriusson E, Samuelson P, Uhlen M, Nygren PA, Stahl S. Surface display of a functional single-chain Fv antibody on staphylococci. J Bacteriol 1996;178:1341 6. Gunneriusson E, Samuelson P, Ringdahl J, Gronlund H, Nygren PA, Stahl S. Staphylococcal surface display of immunoglobulin A (IgA)- and IgE-specific in vitro-selected binding proteins (affibodies) based on Staphylococcus aureus protein A. Appl Environ Microbiol 1999;65:4134 40. Hanski E, Horwitz PA, Caparon MG. Expression of protein F, the fibronectin-binding protein of Streptococcus pyogenes JRS4, in heterologous streptococcal and enterococcal strains promotes their adherence to respiratory epithelial cells. Infect Immun 1992;60:5119 25. Hansson M, Stahl S, Nguyen TN, Bachi T, Robert A, Binz H, Sjolander A, Uhlen M. Expression of recombinant proteins on the surface of the coagulase-negative bacterium Staphylococcus xylosus. J Bacteriol 1992; 174:4239 45. Hansson LO, Widersten M, Mannervik B. Mechanism-based phage display selection of active site mutants of human glutathione transferase A1-1 catalyzing SNAr reactions. Biochemistry 1997;36:11252 60. Hawkins R, Russell S, Winter G. Selection of phage antibodies by binding affinity. Mimicking affinity maturation. J Mol Biol 1992;226:889 96. Heal KG, Hill HR, Stockley PG, Hollingdale MR, Taylor-Robinson AW. Expression and immunogenicity of a liver stage malaria epitope presented as a foreign peptide on the surface of RNA free MS2 bacteriophage capsids. Vaccine 1999;18:251 8. Hedegaard L, Klemm P. Type 1 fimbriae of Escherichia coli as carriers of heterologous antigenic sequences. Gene 1989;85:115 24. Hofnung M. Expression of foreign polypeptides at the Escherichia coli cell surface. Methods Cell Biol 1991; 34:77 105. Hoogenboom HR. Designing and optimizing library selection strategies for generating high-affinity antibodies. Trends Biotechnol 1997;15:62 70. Hoogenboom HR, Winter G. By-passing immunisation. Human antibodies from synthetic repertoires of germline VH gene segments rearranged in vitro. J Mol Biol 1992;227:381 8. Hoogenboom HR, Griffiths AD, Johnson KS, Chiswell DJ, Hudson P, Winter G. Multi-subunit proteins on the surface of filamentous phage: methodologies for displaying antibody (Fab) heavy and light chains. Nucleic Acids Res 1991;19:4133 7. Hoogenboom HR, de Bruine AP, Hufton SE, Hoet RM, Arends JW, Roovers RC. Antibody phage display technology and its applications. Immunotechnology 1998;4:1 20. Houshmand H, Froman G, Magnusson G. Use of bacteriophage T7 displayed peptides for determination of monoclonal antibody specificity and biosensor analysis of the binding reaction. Anal Biochem 1999; 268:363 70. Huang W, Petrosino J, Palzkill T. Display of functional beta-lactamase inhibitory protein on the surface of M13 bacteriophage. Antimicrob Agents Chemother 1998;42:2893 7. Hufton SE, Moerkerk PT, Meulemans EV, de Bruine A, Arends J, Hoogenboom HR. Phage display of cDNA repertoires: the pVI display system and its applications for the selection of immunogenic ligands. J Immunol Methods 1999;231:39 51. Irving RA, Kortt AA, Hudson PJ. Affinity maturation of recombinant antibodies using E. coli mutator cells. Immunotechnology 1996;2:127 43. Jamieson AC, Kim SH, Wells JA. In vitro selection of zinc fingers with altered DNA-binding specificity. Biochemistry 1994;33:5689 95. Janda KD, Lo CH, Li T, Barbas CF, Wirsching P, Lerner RA. Direct selection for a catalytic mechanism from combinatorial antibody libraries. Proc Natl Acad Sci USA 1994;91:2532 6.

28

I. Benhar / Biotechnology Advances 19 (2001) 133

Janda KD, Lo LC, Lo CHL, Sim MM, Wang R, Wong CH, Lerner RA. Chemical selection for catalysis in combinatorial antibody libraries. Science 1997;275:945 8. Janssen R, Tommassen J. PhoE protein as a carrier for foreign epitopes. Int Rev Immunol 1994;11:113 21. 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. Biotechnology 1995;13:378 82. Jestin JL, Kristensen P, Winter G. A method for the selection of catalytic activity using phage display and proximity coupling. Angew Chem Int Ed Engl 1999;38:1124 7. Jiang J, Abu-Shilbayeh L, Rao VB. Display of a PorA peptide from Neisseria meningitidis on the bacteriophage T4 capsid surface. Infect Immun 1997;65:4770 7. Jung HC, Park JH, Park SH, Lebeault JM, Pan JG. Expression of carboxymethylcellulase on the surface of Escherichia coli using Pseudomonas syringae ice nucleation protein. Enzyme Microb Technol 1998a;22:348 54. Jung HC, Lebeault JM, Pan JG. Surface display of Zymomonas mobilis levansucrase by using the ice-nucleation protein of Pseudomonas syringae. Nat Biotechnol 1998b;16:576 80. Jung S, Honegger A, Pluckthun A. Selection for improved protein stability by phage display. J Mol Biol 1999; 294:163 80. Kang AS, Barbas CF, Janda KD, Benkovic SJ, Lerner RA. Linkage of recognition and replication functions by assembling combinatorial antibody Fab libraries along phage surfaces. Proc Natl Acad Sci USA 1991; 88:4363 6. Kay BK, Winter J, McCaferty J. Phage display of peptides and proteins. A laboratory manual. San Diego, CA: Academic Press, 1996. Ke SH, Coombs GS, Tachias K, Navre M, Corey DR, Madison EL. Distinguishing the specificities of closely related proteases. Role of P3 in substrate and inhibitor discrimination between tissue-type plasminogen activator and urokinase. J Biol Chem 1997;272:16603 9. Ke SH, Coombs GS, Tachias K, Corey DR, Madison EL. Optimal subsite occupancy and design of a selective inhibitor of urokinase. J Biol Chem 1997;272:20456 62. Kettleborough CA, Ansell KH, Allen RW, Rosell-Vives E, Gussow DH, Bendig MM. Isolation of tumor cellspecific single-chain Fv from immunized mice using phage-antibody libraries and the re-construction of whole antibodies from these antibody fragments. Eur J Immunol 1994;24:952 8. Kim YS, Jung HC, Pan JG. Bacterial cell surface display of an enzyme library for selective screening of improved cellulase variants. Appl Environ Microbiol 2000;66:788 93. Klauser T, Pohlner J, Meyer TF. Extracellular transport of cholera toxin B subunit using Neisseria IgA protease beta-domain: conformation-dependent outer membrane translocation. EMBO J 1990;9:1991 9. Kornacker MG, Pugsley AP. The normally periplasmic enzyme beta-lactamase is specifically and efficiently translocated through the Escherichia coli outer membrane when it is fused to the cell-surface enzyme pullulanase. Mol Microbiol 1990;4:1101 9. Kotrba P, Pospisil P, de Lorenzo V, Ruml T. Enhanced metallosorption of Escherichia coli cells due to surface display of beta- and alpha-domains of mammalian metallothionein as a fusion to LamB protein. J Recept Signal Transduction Res 1999;19:703 15. Krebber A, Burmester J, Pluckthun A. Inclusion of an upstream transcriptional terminator in phage display vectors abolishes background expression of toxic fusions with coat protein g3p. Gene 1996;178:71 4. Krebber C, Spada S, Desplancq D, Krebber A, Ge L, Pluckthun A. Selectively-infective phage (SIP): a mechan istic dissection of a novel in vivo selection for protein ligand interactions. J Mol Biol 1997;268:607 18. Ku J, Schultz PG. Alternate protein frameworks for molecular recognition. Proc Natl Acad Sci USA 1995; 92:6552 6. Kuwabara I, Maruyama H, Mikawa YG, Zuberi RI, Liu FT, Maruyama IN. Efficient epitope mapping by bacteriophage lambda surface display. Nat Biotechnol 1997;15:74 8. Kwak YD, Yoo SK, Kim EJ. Cell surface display of human immunodeficiency virus type 1 gp120 on Escherichia coli by using ice nucleation protein. Clin Diagn Lab Immunol 1999;6:499 503. Ladner RC. Constrained peptides as binding entities. Trends Biotechnol 1995;13:426 30. Lane DP, Stephen CW. Epitope mapping using bacteriophage peptide libraries. Curr Opin Immunol 1993;5:268 71.

I. Benhar / Biotechnology Advances 19 (2001) 133

29

Lang IM, Barbas CF, Schleef RR. Recombinant rabbit Fab with binding activity to type-1 plasminogen activator inhibitor derived from a phage-display library against human alpha-granules. Gene 1996;172:295 8. Leclerc C, Charbit A, Martineau P, Deriaud E, Hofnung M. The cellular location of a foreign B cell epitope expressed by recombinant bacteria determines its T cell-independent or T cell-dependent characteristics. J Immunol 1991;147:3545 52. Legendre D, Laraki N, Graslund T, Bjornvad ME, Bouchet M, Nygren PA, Borchert TV, Fastrez J. Display of active subtilisin 309 on phage: analysis of parameters influencing the selection of subtilisin variants with changed substrate specificity from libraries using phosphonylating inhibitors. J Mol Biol 2000; 296:87 102. Li Y, Cockburn W, Kilpatrick JB, Whitelam GC. High affinity ScFvs from a single rabbit immunized with multiple haptens. Biochem Biophys Res Commun 2000a;268:398 404. Li Y, Kilpatrick J, Whitelam GC. Sheep monoclonal antibody fragments generated using a phage display system. J Immunol Methods 2000b;236:133 46. Light J, Lerner RA. Random mutagenesis of staphylococcal nuclease and phage display selection. Bioorg Med Chem 1995;3:955 67. Liljeqvist S, Cano F, Nguyen TN, Uhlen M, Robert A, Stahl S. Surface display of functional fibronectin-binding domains on Staphylococcus carnosus. FEBS Lett 1999;446:299 304. Lindqvist BH, Naderi S. Peptide presentation by bacteriophage P4. FEMS Microbiol Rev 1995;17:33 9. Little M, Fuchs P, Breitling F, Dubel S. Bacterial surface presentation of proteins and peptides: an alternative to phage technology? Trends Biotechnol 1993;11:3 5. Low NM, Holliger PH, Winter G. Mimicking somatic hypermutation: affinity maturation of antibodies displayed on bacteriophage using a bacterial mutator strain. J Mol Biol 1996;260:359 68. Lowman HB. Bacteriophage display and discovery of peptide leads for drug development. Annu Rev Biophys Biomol Struct 1997;26:401 24. Lowman HB, Bass SH, Simpson N, Wells JA. Selecting high-affinity binding proteins by monovalent phage display. Biochemistry 1991;30:10832 8. Lu Z, Murray KS, Van Cleave V, LaVallie ER, Stahl ML, McCoy JM. Expression of thioredoxin random peptide libraries on the Escherichia coli cell surface as functional fusions to flagellin: a system designed for exploring protein protein interactions. Biotechnology 1995;13:366 72. Lutz R, Bujard H. Independent and tight regulation of transcriptional units in Escherichia coli via the LacR/O, the TetR/O and AraC/I1 I2 regulatory elements. Nucleic Acids Res 1997;25:1203 10. Malmborg AC, Duenas M, Ohlin M, Soderlind E, Borrebaeck CA. Selection of binders from phage displayed antibody libraries using the BIAcore biosensor. J Immunol Methods 1996;198:51 7. Malmborg AC, Soderlind E, Frost L, Borrebaeck CA. Selective phage infection mediated by epitope expression on F pilus. J Mol Biol 1997;273:544 51. Markland W, Roberts BL, Saxena MJ, Guterman SK, Ladner RC. Design, construction and function of a multicopy display vector using fusions to the major coat protein of bacteriophage M13. Gene 1991;109:13 9. Markland W, Ley AC, Ladner RC. Iterative optimization of high-affinity protease inhibitors using phage display: 2. Plasma kallikrein and thrombin. Biochemistry 1996;35:8058 67. Marks JD, Hoogenboom HR, Bonnert TP, McCafferty J, Griffiths AD, Winter G. By-passing immunization. Human antibodies from V-gene libraries displayed on phage. J Mol Biol 1991;222:581 97. Marks JD, Griffiths AD, Malmqvist M, Clackson TP, Bye JM, Winter G. By-passing immunization: building high affinity human antibodies by chain shuffling. Biotechnology 1992;10:779 83. Martin F, Volpari C, Steinkuhler C, Dimasi N, Brunetti M, Biasiol G, Altamura S, Cortese R, De Francesco R, Sollazzo M. Affinity selection of a camelized V(H) domain antibody inhibitor of hepatitis C virus NS3 protease. Protein Eng 1997;10:607 14. Maruyama IN, Maruyama HI, Brenner S. Lambda foo: a lambda phage vector for the expression of foreign proteins. Proc Natl Acad Sci USA 1994;91:8273 7. Maser RL, Calvet JP. Analysis of differential gene expression in the kidney by differential cDNA screening, subtractive cloning, and mRNA differential display. Semin Nephrol 1995;15:29 42. Mastico RA, Talbot SJ, Stockley PG. Multiple presentation of foreign peptides on the surface of an RNA-free spherical bacteriophage capsid. J Gen Virol 1993;74:541 8.

30

I. Benhar / Biotechnology Advances 19 (2001) 133

Matthews DJ, Wells JA. Substrate phage: selection of protease substrates by monovalent phage display. Science 1993;260:1113 7. Maurer J, Jose J, Meyer TF. Autodisplay: one-component system for efficient surface display and release of soluble recombinant proteins from Escherichia coli. J Bacteriol 1997;179:794 804. McCafferty J, Griffiths AD, Winter G, Chiswell DJ. Phage antibodies: filamentous phage displaying antibody variable domains. Nature 1990;348:552 4. McCafferty J, Jackson RH, Chiswell DJ. Phage-enzymes: expression and affinity chromatography of functional alkaline phosphatase on the surface of bacteriophage. Protein Eng 1991;4:955 61. McCarrey JR, Williams SA. Construction of cDNA libraries from limiting amounts of material. Curr Opin Biotechnol 1994;5:34 9. Mennuni C, Santini C, Dotta F, Farilla L, Di Mario U, Fierabracci A, Bottazzo G, Cortese R, Luzzago A. Selection of phage-displayed peptides mimicking type 1 diabetes-specific epitopes. J Autoimmun 1996; 9:431 6. Meola A, Delmastro P, Monaci P, Luzzago A, Nicosia A, Felici F, Cortese R, Galfre G. Derivation of vaccines from mimotopes. Immunologic properties of human hepatitis B virus surface antigen mimotopes displayed on filamentous phage. J Immunol 1995;154:3162 72. Mesnage S, Weber-Levy M, Haustant M, Mock M, Fouet A. Cell surface-exposed tetanus toxin fragment C produced by recombinant Bacillus anthracis protects against tetanus toxin. Infect Immun 1999;67:4847 50. Mikawa YG, Maruyama IN, Brenner S. Surface display of proteins on bacteriophage lambda heads. J Mol Biol 1996;262:21 30. Moe GR, Tan S, Granoff DM. Molecular mimetics of polysaccharide epitopes as vaccine candidates for prevention of Neisseria meningitidis serogroup B disease. FEMS Immunol Med Microbiol 1999;26:209 26. Mullaney JM, Black LW. Capsid targeting sequence targets foreign proteins into bacteriophage T4 and permits proteolytic processing. J Mol Biol 1996;261:372 85. Mullaney JM, Black LW. Activity of foreign proteins targeted within the bacteriophage T4 head and prohead: implications for packaged DNA structure. J Mol Biol 1998;283:913 29. Muyldermans S, Lauwereys M. Unique single-domain antigen binding fragments derived from naturally occurring camel heavy-chain antibodies. J Mol Recognit 1999;12:131 40. Newton SM, Jacob CO, Stocker BA. Immune response to cholera toxin epitope inserted in Salmonella flagellin. Science 1989;244:70 2. Nguyen TN, Hansson M, Stahl S, Bachi T, Robert A, Domzig W, Binz H, Uhlen M. Cell-surface display of heterologous epitopes on Staphylococcus xylosus as a potential delivery system for oral vaccination. Gene 1993;128:89 94. Nguyen TN, Gourdon MH, Hansson M, Robert A, Samuelson P, Libon C, Andreoni C, Nygren PA, Binz H, Uhlen M, Stahl S. Hydrophobicity engineering to facilitate surface display of heterologous gene products on Staphylococcus xylosus. J Biotechnol 1995;42:207 19. Nissim A, Hoogenboom HR, Tomlinson IM, Flynn G, Midgley C, Lane D, Winter G. Antibody fragments from a ``single pot'' phage display library as immunochemical reagents. EMBO J 1994;13:692 8. Novotny CP, Fives-Taylor P. Retraction of F pili. J Bacteriol 1974;117:1306 11. Novotny CP, Lavin K. Some effects of temperature on the growth of F pili. J Bacteriol 1971;107:671 82. Nygren PA, Uhlen M. Scaffolds for engineering novel binding sites in proteins. Curr Opin Struct Biol 1997; 7:463 9. O'Boyle DR, Pokornowski KA, McCann P, Weinheimer SP. Identification of a novel peptide substrate of HSV-1 protease using substrate phage display. Virology 1997;236:338 47. Oggioni MR, Medaglini D, Maggi T, Pozzi G. Engineering the gram-positive cell surface for construction of bacterial vaccine vectors. Methods 1999;19:163 73. Ohkubo S, Miyadera K, Sugimoto Y, Matsuo K, Wierzba K, Yamada Y. Identification of substrate sequences for membrane type-1 matrix metalloproteinase using bacteriophage peptide display library. Biochem Biophys Res Commun 1999;266:308 13. Pallesen L, Poulsen LK, Christiansen G, Klemm P. Chimeric FimH adhesin of type 1 fimbriae: a bacterial surface display system for heterologous sequences. Microbiology 1995;141:2839 48. Pannekoek H, van Meijer M, Schleef RR, Loskutoff DJ, Barbas CF. Functional display of human plasminogen-

I. Benhar / Biotechnology Advances 19 (2001) 133

31

activator inhibitor 1 (PAI-1) on phages: novel perspectives for structure function analysis by error-prone DNA synthesis. Gene 1993;128:135 40. Parmley SF, Smith GP. Filamentous fusion phage cloning vectors for the study of epitopes and design of vaccines. Adv Exp Med Biol 1989;251:215 8. Pasqualini R, Ruoslahti E. Organ targeting in vivo using phage display peptide libraries. Nature 1996;380:364 6. Pedersen H, Holder S, Sutherlin DP, Schwitter U, King DS, Schultz PG. A method for directed evolution and functional cloning of enzymes. Proc Natl Acad Sci USA 1998;95:10523 8. Persic L, Horn IR, Rybak S, Cattaneo A, Hoogenboom HR, Bradbury A. Single-chain variable fragments selected on the 57 76 p21Ras neutralising epitope from phage antibody libraries recognise the parental protein. FEBS Lett 1999;443:112 6. Phalipon A, Folgori A, Arondel J, Sgaramella G, Fortugno P, Cortese R, Sansonetti PJ, Felici F. Induction of anticarbohydrate antibodies by phage library-selected peptide mimics. Eur J Immunol 1997;27:2620 5. Pistor S, Hobom G. Expression of viral hemagglutinin on the surface of E. coli. Klin Wochenschr 1988;66:110 6. Ploug M, Ostergaard S, Hansen LB, Holm A, Dano K. Photoaffinity labeling of the human receptor for urokinasetype plasminogen activator using a decapeptide antagonist. Evidence for a composite ligand-binding site and a short interdomain separation. Biochemistry 1998;37:3612 22. Poirier TP, Kehoe MA, Beachey EH. Protective immunity evoked by oral administration of attenuated aroA Salmonella typhimurium expressing cloned streptococcal M protein. J Exp Med 1988;168:25 32. Poul MA, Marks JD. Targeted gene delivery to mammalian cells by filamentous bacteriophage. J Mol Biol 1999;288:203 11. Pozzi G, Contorni M, Oggioni MR, Manganelli R, Tommasino M, Cavalieri F, Fischetti VA. Delivery and expression of a heterologous antigen on the surface of streptococci. Infect Immun 1992;60:1902 7. Proba K, Honegger A, Pluckthun A. A natural antibody missing a cysteine in VH: consequences for thermo dynamic stability and folding. J Mol Biol 1997;265:161 72. Rader C, Barbas CF. Phage display of combinatorial antibody libraries. Curr Opin Biotechnol 1997;8:503 8. Rakonjac J, Jovanovic G, Model P. Filamentous phage infection-mediated gene expression: construction and propagation of the gIII deletion mutant helper phage R408d3. Gene 1997;198:99 103. Rebar EJ, Pabo CO. Zinc finger phage: affinity selection of fingers with new DNA-binding specificities. Science 1994;263:671 3. Ren Z, Black LW. Phage T4 SOC and HOC display of biologically active, full-length proteins on the viral capsid. Gene 1998;215:439 44. Roberts BL, Markland W, Ley AC, Kent RB, White DW, Guterman SK, Ladner RC. Directed evolution of a protein: selection of potent neutrophil elastase inhibitors displayed on M13 fusion phage. Proc Natl Acad Sci USA 1992;89:2429 33. Rodi DJ, Makowski L. Phage-display technology finding a needle in a vast molecular haystack. Curr Opin Biotechnol 1999;10:87 93. Rottgen P, Collins J. A human pancreatic secretory trypsin inhibitor presenting a hypervariable highly constrained epitope via monovalent phagemid display. Gene 1995;164:243 50. Saggio I, Gloaguen I, Laufer R. Functional phage display of ciliary neurotrophic factor. Gene 1995;152:35 9. Samuelson P, Hansson M, Ahlborg N, Andreoni C, Gotz F, Bachi T, Nguyen TN, Binz H, Uhlen M, Stahl S. Cell surface display of recombinant proteins on Staphylococcus carnosus. J Bacteriol 1995;177:1470 6. 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 35. Sastry L, Mubaraki M, Janda KD, Benkovic SJ, Lerner RA. Screening combinatorial antibody libraries for catalytic acyl transfer reactions. Ciba Found Symp 1995;159:145 51. Scarselli E, Esposito G, Traboni C. Receptor phage display of functional domains of the human high affinity IgE receptor on the M13 phage surface. FEBS Lett 1993;329:223 6. Schier R, Bye J, Apell G, McCal LA, Adams G, Malmqvist M, Weiner L, Marks J. Isolation of highaffinity monomeric human anti-c-erbB-2 single chain Fv using affinity-driven selection. J Mol Biol 1996; 255:28 43. Schneewind O, Model P, Fischetti VA. Sorting of protein A to the staphylococcal cell wall. Cell 1992;70:267 81.

32

I. Benhar / Biotechnology Advances 19 (2001) 133

Schneewind O, Mihaylova-Petkov D, Model P. Cell wall sorting signals in surface proteins of gram-positive bacteria. EMBO J 1993;12:4803 11. Schneewind O, Fowler A, Faull KF. Structure of the cell wall anchor of surface proteins in Staphylococcus aureus. Science 1995;268:103 6. Schorr J, Knapp B, Hundt E, Kupper HA, Amann E. Surface expression of malarial antigens in Salmonella typhimurium: induction of serum antibody response upon oral vaccination of mice. Vaccine 1991;9:675 81. Schreuder MP, Deen C, Boersma WJ, Pouwels PH, Klis FM. Yeast expressing hepatitis B virus surface antigen determinants on its surface: implications for a possible oral vaccine. Vaccine 1991;14:383 8. Scott JK, Smith GP. Searching for peptide ligands with an epitope library. Science 1990;249:386 90. Sewani CR, Bagdasarian MM, Ireland JJ, Bagdasarian M. Display of an inhibin epitope in a surface-exposed loop of the E. coli heat-labile enterotoxin B subunit. Vaccine 1998;16:1611 9. Silver SC, Hunt SW. Techniques for cloning cDNAs encoding interactive transcriptional regulatory proteins. Mol Biol Rep 1993;17:155 65. Skerra A, Pluckthun A. Assembly of a functional immunoglobulin Fv fragment in Escherichia coli. Science 1988;240:1038 41. Smith GP. Filamentous fusion phage: novel expression vectors that display cloned antigens on the virion surface. Science 1985;228:1315 7. Smith GP, Patel SU, Windass JD, Thornton JM, Winter G, Griffiths AD. Small binding proteins selected from a combinatorial repertoire of knottins displayed on phage. J Mol Biol 1998;277:317 32. Soumillion PLJ, Bouchet M, Marchard-Brynaert J, Sartiaux P, Fastrez J. Phage display of enzymes and in vitro selection for catalytic activity. Appl Biochem Biotechnol 1994;47:175 89. Sousa C, Cebolla A, de Lorenzo V. Enhanced metalloadsorption of bacterial cells displaying poly-His peptides. Nat Biotechnol 1996;14:1017 20. Spada S, Honegger A, Pluckthun A. Reproducing the natural evolution of protein structural features with the selectively infective phage (SIP) technology. The kink in the first strand of antibody kappa domains. J Mol Biol 1998;283:395 407. Stathopoulos C, Georgiou G, Earhart CF. Characterization of Escherichia coli expressing an LppHOmpA(46 159)PhoA fusion protein localized in the outer membrane. Appl Microbiol Biotechnol 1996;45:112 9. Steidler L, Remaut E, Fiers W. Pap pili as a vector system for surface exposition of an immunoglobulin G-binding domain of protein A of Staphylococcus aureus in Escherichia coli. J Bacteriol 1993;175:7639 43. Stentebjerg-Olesen B, Pallesen L, Jensen LB, Christiansen G, Klemm P. Authentic display of a cholera toxin epitope by chimeric type 1 fimbriae: effects of insert position and host background. Microbiology 1997; 143:2027 38. Sternberg N, Hoess RH. Display of peptides and proteins on the surface of bacteriophage lambda. Proc Natl Acad Sci USA 1995;92:1609 13. Stover CK, Bansal GP, Hanson MS, Burlein JE, Palaszynski SR, Young JF, Koenig S, Young DB, Sadziene A, Barbour AG. Protective immunity elicited by recombinant bacille Calmette-Guerin (BCG) expressing outer surface protein A (OspA) lipoprotein: a candidate Lyme disease vaccine. J Exp Med 1993;178:197 209. Strauss A, Gotz F. In vivo immobilization of enzymatically active polypeptides on the cell surface of Staphylococcus carnosus. Mol Microbiol 1996;21:491 500. Thompson J, Pope T, Tung J-S, Chan C, Hollis G, Mark G, Johnson K. Affinity maturation of a high-affinity human monoclonal antibody against the third hypervariable loop of human immunodeficiency virus: use of phage display to improve affinity and broaden strain reactivity. J Mol Biol 1996;256:77 88. Tommassen J, Leunissen J, van Damme-Jongsten M, Overduin P. Failure of E. coli K-12 to transport PhoE LacZ hybrid proteins out of the cytoplasm. EMBO J 1985;4:1041 7. Tommassen J, Agterberg M, Janssen R, Spierings G. Use of the enterobacterial outer membrane protein PhoE in the development of new vaccines and DNA probes. Zentralbl Bakteriol 1994;278:396 406. Tramontano A, Bianchi E, Venturini S, Martin F, Pessi A, Sollazzo M. The making of the minibody: an engineered beta-protein for the display of conformationally constrained peptides. J Mol Recognit 1994;7:9 24. Van Ewijk W, de Kruif J, Germeraad WT, Berendes P, Ropke C, Platenburg PP, Logtenberg T. Subtractive isolation of phage-displayed single-chain antibodies to thymic stromal cells by using intact thymic fragments. Proc Natl Acad Sci USA 1997;94:3903 8.

I. Benhar / Biotechnology Advances 19 (2001) 133

33

van Meijer M, Roelofs Y, Neels J, Horrevoets AJ, van Zonneveld AJ, Pannekoek H. Selective screening of a large phage display library of plasminogen activator inhibitor 1 mutants to localize interaction sites with either thrombin or the variable region 1 of tissue-type plasminogen activator. J Biol Chem 1996;271:7423 8. Vaughan TJ, Williams AJ, Pritchard K, Osbourn JK, Pope AR, Earnshaw JC, McCafferty J, Hooits RA, Wilton J, Johnson KS. Human antibodies with sub-nanomolar affinities isolated from a large non-immunized phage display library. Nat Biotechnol 1996;14:309 14. Wang L, Radic MZ, Siegel D, Chang T, Bracy J, Galili U. Cloning of anti-Gal Fabs from combinatorial phage display libraries: structural analysis and comparison of Fab expression in pComb3H and pComb8 phage. Mol Immunol 1997;34:609 18. Widersten M, Mannervik B. Glutathione transferases with novel active sites isolated by phage display from a library of random mutants. J Mol Biol 1995;250:115 22. Willis AE, Perham RN, Wraith D. Immunological properties of foreign peptides in multiple display on a filamentous bacteriophage. Gene 1993;128:79 83. Wilson DR, Finlay BB. Phage display: applications, innovations, and issues in phage and host biology. Can J Microbiol 1998;44:313 29. Winter G, Griffiths AD, Hawkins RE, Hoogenboom HR. Making antibodies by phage display technology. Annu Rev Immunol 1994;12:433 55. Wu JY, Newton S, Judd A, Stocker B, Robinson WS. Expression of immunogenic epitopes of hepatitis B surface antigen with hybrid flagellin proteins by a vaccine strain of Salmonella. Proc Natl Acad Sci USA 1989; 86:4726 30. Xu Z, Lee SY. Display of polyhistidine peptides on the Escherichia coli cell surface by using outer membrane protein C as an anchoring motif. Appl Environ Microbiol 1999;65:5142 7. Yamamoto M, Kominato Y, Yamamoto F. Phage display cDNA cloning of protein with carbohydrate affinity. Biochem Biophys Res Commun 1999;255:194 9. Yamanaka HI, Inoue T, Ikeda-Tanaka O. Chicken monoclonal antibody isolated by a phage display system. J Immunol 1996;157:1156 62. Yang WP, Green K, Pinz-Sweeney S, Briones AT, Burton DR, Barbas CF. CDR walking mutagenesis for the affinity maturation of a potent human anti-HIV-1 antibody into the picomolar range. J Mol Biol 1995; 254:392 403. Zacher AN, Stock CA, Golden JWD, Smith GP. A new filamentous phage cloning vector: fd-tet. Gene 1980; 9:127 40. Zahn G, Skerra A, Hohne W. Investigation of a tetracycline-regulated phage display system. Protein Eng 1999; 12:1031 4. Zuercher AW, Miescher SM, Vogel M, Rudolf MP, Stadler MB, Stadler BM. Oral anti-IgE immunization with epitope-displaying phage. Eur J Immunol 2000;30:128 35.

Вам также может понравиться