US20040253607A1 - Screening of phage displayed peptides without clearing of the cell culture - Google Patents

Screening of phage displayed peptides without clearing of the cell culture Download PDF

Info

Publication number
US20040253607A1
US20040253607A1 US10/727,745 US72774503A US2004253607A1 US 20040253607 A1 US20040253607 A1 US 20040253607A1 US 72774503 A US72774503 A US 72774503A US 2004253607 A1 US2004253607 A1 US 2004253607A1
Authority
US
United States
Prior art keywords
phage
replicable genetic
scfv
cell culture
artificial sequence
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US10/727,745
Inventor
Steffen Nock
Paul Kassner
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Zyomyx Inc
Original Assignee
Zyomyx Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Zyomyx Inc filed Critical Zyomyx Inc
Priority to US10/727,745 priority Critical patent/US20040253607A1/en
Publication of US20040253607A1 publication Critical patent/US20040253607A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C40COMBINATORIAL TECHNOLOGY
    • C40BCOMBINATORIAL CHEMISTRY; LIBRARIES, e.g. CHEMICAL LIBRARIES
    • C40B40/00Libraries per se, e.g. arrays, mixtures
    • C40B40/02Libraries contained in or displayed by microorganisms, e.g. bacteria or animal cells; Libraries contained in or displayed by vectors, e.g. plasmids; Libraries containing only microorganisms or vectors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/10Processes for the isolation, preparation or purification of DNA or RNA
    • C12N15/1034Isolating an individual clone by screening libraries
    • C12N15/1037Screening libraries presented on the surface of microorganisms, e.g. phage display, E. coli display

Definitions

  • Phage display and related techniques have become powerful methods for the discovery of affinity binding reagents (Smith (1985) Science 228: 1315-1317).
  • Linear and constrained peptides, antibody fragments (e.g., scFvs, Fvs and Fabs), as well as a number of alternative binding domains have all been displayed on phage particles, for example, via fusion to one of the phage coat proteins.
  • phage proteins derived from gVIII, gVI, gVHI and gIX
  • gIII is the most widely used.
  • Phagemids containing a phage origin of replication, an antibiotic resistance marker, and a gene encoding a binding domain/gill fusion protein are readily constructed via conventional molecular biology techniques. Through large-scale ligation and transformation as well as recombination strategies, large libraries of 10 8 to 10 11 different recombinants are now being generated for use in affinity selection strategies (de Haard et al. (1999) J. Biol. Chem. 274: 18218-18230; Sblattero and Bradbury (2000) Nat. Biotechnol. 18: 75-80); Sheets et al. (1998) Proc. Natl. Acad. Sci., U.S.A. 95:6157-6162, published erratum appears in Proc. Natl. Acad. Sci., U.S.A. ( 1999) 96: 795).
  • HTS high-throughput screening
  • the present invention provides methods for screening a population of replicable genetic packages (e.g., phage, eukaryotic viruses, and the like) to obtain particles that display on their surface a fusion protein that specifically binds to a target molecule.
  • replicable genetic packages e.g., phage, eukaryotic viruses, and the like
  • the methods of the present invention involve contacting a target molecule with an uncleared cell culture that contains a population of replicable genetic packages.
  • Each replicable genetic package displays on its surface a fusion protein that has a surface-displayed replicable genetic package polypeptide and a potential binding polypeptide.
  • the replicable genetic package that specifically bind to the target molecule form complexes containing replicable genetic packages and target molecules.
  • the potential binding polypeptide can be encoded by a member of a library of nucleic acid molecules.
  • the nucleic acid molecules can be cDNA molecules or recombinant products.
  • the potential binding polypeptide can be, for example, an antibody, or derivative of an antibody.
  • the potential binding polypeptide can be a scFv or a Fab.
  • the methods of the invention are useful for obtaining polypeptides that bind to essentially any molecule.
  • the target molecule can be a polypeptide, an RNA, a DNA, a small organic molecule and a carbohydrate.
  • the target molecules can be immobilized directly or indirectly to a solid support.
  • Solid supports such as a bead, a chip, a microtiter plate, a eukaryotic cell, or a prokaryotic cell are present in some embodiments of the invention.
  • the solid supports of the present invention can contain a variety of materials, such as Sepharose, polystyrene, glass, silicon oxide, etc.
  • the methods also involve obtaining replicable genetic packages that specifically bind to the target molecule.
  • the replicable genetic packages that specifically bind to the target molecule can be separated from the bacterial cells after the binding of the phage to the target molecule.
  • the uncleared cell culture can be separated from a replicable genetic package-target complex(es) using aspiration.
  • some embodiments of the invention can further involve eluting the replicable genetic packages from the target molecule.
  • some embodiments involve identifying the replicable genetic packages that specifically bind to the target molecule with a detection reagent.
  • compositions containing an uncleared cell culture which contains: (a) a population of replicable genetic packages that display on their surfaces a fusion protein that includes a surface-displayed replicable genetic package polypeptide and a potential binding polypeptide; (b) a complex that is composed of one or members of the library of replicable genetic packages that specifically bind to the target molecule; and (c) cells in which the replicable genetic packages were amplified.
  • FIGS. 1A and 1B show that the binding of HP6054-scFv and HP6054-Fab phage to human lambda light chain is not affected by the presence of bacteria in the sample. Antigens were immobilized on 96-well plates (Nunc, Denmark) at 10 ⁇ g/ml. A phage ELISA was conducted using either an uncleared bacterial culture, or supernatants clarified by centrifugation or filtration. Each bar represents the mean ⁇ s.d. of duplicate samples.
  • FIG. 1A depicts representative results from the HP6054 scFv-phage
  • FIG. 1B is representative of HP6054 Fab-phage.
  • FIG. 2 shows that the sensitivity of phage ELISA is not impaired by the presence of bacteria.
  • Overnight cultures of HP6002 scFv-phage and HP6025 scFv-phage were mixed at various ratios and then supernatants or uncleared culture was tested in the phage ELISA against hIgG2 and hIgG4 (each at 10 ⁇ g/ml). Each bar represents the mean ⁇ s.d. of duplicate samples.
  • Replicable genetic packages include virions of bacteriophage, filamentous phage, or any other eukaryotic virus, bacterial virus or phage.
  • phage as used herein encompasses not only bacteriophage but also other types of replicable genetic packages, except where the term is used in a context that dictates a more specific meaning.
  • a molecule is “display(ed) on their surface” of a replicable genetic package if at least part of the molecule is accessible to the milieu surrounding a replicable genetic package.
  • the phrase “specifically (or selectively) binds to” in the context of a replicable genetic package refers to a binding reaction which is determinative of the presence of a replicable genetic package binding to a target molecule(s) in the presence of a population of other proteins, biologics, and replicable genetic packages.
  • a specifically binding replicable genetic package will bind to a particular molecule (e.g., target molecule) and under the same designated binding conditions, native replicable genetic packages do not bind to a particular molecule in a significant amount.
  • a replicable genetic package “specifically” binds to a target molecule when the number of replicable genetic packages displaying a potential binding polypeptide that are bound to the target molecules is at least twice the background binding observed using a native replicable genetic package as a control.
  • An “uncleared cell culture” is an aqueous medium containing bacterial or eukaryotic cells.
  • the “uncleared cell culture” is a growth of bacterial or eukaryotic cells infected with one or more replicable genetic package clones.
  • a “target molecule” is essentially any molecule that is being used as a probe to identify molecules that will bind to the target molecule.
  • target molecules include, without limitation, amino acids, peptides, proteins, polypeptides, carbohydrates, small organic molecules, inorganic molecules, etc.
  • a “surface-displayed replicable genetic package polypeptide” is a polypeptide that is, at least in part, exposed to the milieu surrounding the virion.
  • Examples of “surface-displayed replicable genetic package polypeptides” include, without limitation, pIII and pVIII.
  • a “potential binding polypeptide” is a polypeptide that may possibly bind to the target molecule.
  • a “potential binding polypeptide” can be screened for its ability to bind to a target molecule of choice.
  • a “replicable genetic package-target complex” is a complex in which a target molecule is bound to a replicable genetic package.
  • the target molecule is bound to the replicable genetic package through the binding domain portion of a polypeptide displayed on the surface of a replicable genetic package.
  • an “antibody” can be derived from sequence of a mammal, non-mammal (e.g., birds, chickens, fish, etc.), or fully synthetic antibody sequences.
  • a “mammal” is a member of the class Mammalia. Examples of mammals include, without limitation, humans, primates, chimpanzees, rodents, mice, rats, rabbits, sheep, and cows.
  • the term “antibody” also refers to fragments and substitutes for antibodies such as F(ab′) 2 , Fab′, and Fab fragments. Additionally the “antibodies” can be single chain antibodies known as ScFv fragments, which are obtained by recombinantly fusing the variable regions of the light and heavy chains of the antigen binding fragment of interest.
  • the present invention provides methods and compositions for screening replicable genetic package particles (e.g., phage, viruses, etc.) that display polypeptides for their ability to bind to a target molecule.
  • replicable genetic package particles e.g., phage, viruses, etc.
  • Such screening methods required clearing the host cells or bacteria from an uncleared cell culture and/or isolating a replicable genetic package stock before incubation with the target molecule.
  • the methods of the present invention do not require these procedures. Therefore, the invention provides significant advantages over previously available methods for screening phage and other particles, particularly when used in a high-throughput format.
  • methods of the invention involve infecting bacteria or other suitable host cells with phage particles (or incubating cells that are transfected with a phagemid expression vector with helper phage) to generate an uncleared cell culture that contains a library of phage particles. This uncleared culture is then incubated with a target molecule. Phage particles that display a polypeptide that binds to the target molecule form a complex with the target molecule. After an incubation period, the bacterial or other cells used to amplify the phage can be separated from the phage particles that bind to the target molecule. The phage particles that were able to bind to the target molecule can then be further purified, characterized, amplified, and/or detected, etc. These methods and compositions will be described in more detail below.
  • the methods of the invention are useful for screening a wide variety of phage display libraries.
  • Phage display and related techniques provides a powerful method for selecting proteins of interest from large libraries (Bass et al. (1990) Proteins: Struct. Funct. Genet. 8: 309; Lowman and Wells (1991) Methods: A Companion to Methods Enz. 3(3);205-216. Lowman and Wells (1993) J. Mol. Biol. 234;564-578).
  • Each phage or other particle displays a unique variant protein on its surface and packages the gene encoding that particular variant.
  • the libraries can be composed of homogenous or heterogenous populations of phage particles.
  • each phage in the library can display the same potential binding polypeptide, or each phage can display a different potential binding polypeptide.
  • Potential binding polypeptides can serve as epitopes, ligands, agonists, antagonists, enzymes, etc.
  • the potential binding polypeptides can encode scFvs and Fabs.
  • the methods of the invention are applicable to any of the genetic packages most frequently used for phage display libraries. These include, for example, bacteriophage, particularly filamentous phage, and especially phage M13, Fd and F1.
  • bacteriophage particularly filamentous phage, and especially phage M13, Fd and F1.
  • Webster (1996) Chapter 1 , Biology of the Filamentous Bacteriophage , in Kay et al., eds. (1996) Phage Display of Peptides and Proteins ).
  • Microbiological methods for growing, titering, and preparing filamentous phage particles, and phage DNA are known in the art (Rider et al. (1996) Chapter 4 , Microbiological Methods , in Kay et al., eds. (1996)) and their genomes are very well characterized.
  • filamentous phage have genes which encode the various capsid proteins and are known as genes III, VI, VII, VIII, and IX (Webster et al., (1996), supra).
  • the proteins the genes encode are known as pIII, pVI, pVII, pVIII, and pIX, respectively.
  • the most abundant capsid protein is pVIII, which has 2700 copies on the surface of the phage. Approximately 5 copies of pIII are displayed on the phage particle.
  • libraries of nucleic acid molecules are ligated into a phage-display vector and introduced into bacteria to create a library of particles displaying fusion proteins that consist of a surface-displayed phage polypeptide and a potential binding polypeptide.
  • nucleic acid libraries encoding polypeptides to be displayed into either a gIII or gVIII expression vector in order to produce phage-displayed fusion protein(s)
  • phage-displayed fusion protein(s) See, e.g., Dower, WO 91/19818; Devlin, WO 91/18989; MacCafferty, WO 92/01047 (gene III); Huse, WO 92/06204; Kang, WO 92/18619 (gene VIII)).
  • fusion proteins generally included a signal sequence, usually but not necessarily, from the phage coat protein, a polypeptide to be displayed and either the gene III or gene VIII protein or a fragment thereof.
  • Exogenous coding sequences are often inserted at or near the N-terminus of gene III or gene VIII, although other insertion sites are possible.
  • pVIII can only tolerate short inserts—about 5 to 6 amino acid residues. (Armstrong et al., (1996), supra). Larger peptides can be displayed as pVIII fusions if pVIII wild-type coat proteins are interspersed with the recombinant pVIII (Malik et al. (1996) Chapter 8 , Multiple Display of Foreign Peptide Epitopes on Filamentoisa Bacteriophage Virions , in Kay et al., eds. (1996)).
  • Eukaryotic replicable genetic packages such as eukaryotic viruses can also be used to display polypeptides in an analogous manner.
  • display of human heregulin fused to gp70 of Moloney murine leukemia virus has been reported by Han et al., (1995) Proc. Nat'l. Acad. Sci. USA 92: 9747-9751.
  • prokaryotic spores can be used as replicable genetic packages.
  • polypeptides are displayed from the outer surface of the spore.
  • spores from B. subtilis have been reported to be suitable. Sequences of coat proteins of these spores are described in Donovan et al., J. Mol. Biol. 196: 1-10 (1987).
  • spores can be used to display the potential binding polypeptides.
  • the nucleic acid libraries encoding the potential binding polypeptides can be constructed from nucleic acids from a variety of sources, including cDNA, genomic DNA, synthetic nucleotides and/or from oligomers encoding randomized peptides (see, e.g., Adey et al. (1996) Chapter 5 , Construction of Random Peptide Libraries in Bacteriophage M 13, in Kay et al., eds. (1996) for descriptions of randomized peptide libraries). Random peptide libraries have been constructed using synthetic degenerate oligonucleotides and expressed as fusions with pIII (Adey et al, (1996), supra).
  • libraries of antibody and antibody fragments can be expressed in phage display systems with pIII (McCafferty and Johnson (1996) Chapter 6 , Construction and Screening of Antibody Display Libraries , in Kay et al., eds. (1996)).
  • One method of constructing an antibody phage display library involves generating nucleic acids encoding antibody fragments from the amplification of variable domain gene sequences (McCafferty and Johnson (1996), supra). The fragments can be amplified from nucleic acids isolated from antigen immunized or non-immunized sources.
  • nucleic acids encoding variable heavy and light chain domains are then spliced together using overlap PCR and ligated into a phage-display vector to subsequently generate the antibody phage display library (McCafferty and Johnson (19.96), supra).
  • phage display libraries Numerous other methods for constructing phage display libraries are known in the art. For example, libraries expressing fragments of a protein can be used to map the epitopes of an antibody, which can serve as the target molecule (Plessis and Jordaan (1996) Chapter 9 , Phage Libraries Displaying Random Peptides Derived from a Target Sequence , in Kay et al., eds. (1996)). Also, once a recombinant phage has been isolated or constructed, it can be used to construct a second-generation phage-display library through DNA shuffling (Adey et al. (1996) Chapter 16 , Preparation of Second - Generation Phage Libraries , in Kay et al., eds. (1996)).
  • phage particles are obtained.
  • the vectors are introduced into appropriate host cells and amplified.
  • Uncleared cell cultures containing libraries of phage particles can be generated using methods well known in the art (see, e.g., Sparks et al. (1996) Chapter 13 , Screening Phage - Displayed Random Peptide Libraries , in Kay et al., eds. (1996)).
  • libraries of phage particles displaying the potential binding polypeptides can be used to infect bacteria (e.g, E. coli ) in order to generate an uncleared cell culture.
  • a library of nucleic acid molecules encoding the potential phage binding polypeptides can be introduced into bacteria, which are subsequently infected with a helper phage (see, e.g., Sparks et al., (1996) Ch. 13, supra). These procedures can generate a library of phage particles in an uncleared cell culture.
  • Phage display libraries are screened to obtain phage that display on their surfaces a polypeptide that has a desired activity (e.g., the ability to bind to a target molecule).
  • a desired activity e.g., the ability to bind to a target molecule.
  • Methods for screening phage-displayed libraries are known in the art (Sparks et al. (1996) Chapter 13, supra); McCafferty and Johnson (1996) Chapter 6, supra; McCafferty (1996) Chapter 15 , Phage Display: Factors Affecting Panning Efficiency , in Kay et al., eds. (1996)).
  • Screening involves selecting phage that display on their surface a polypeptide that has a desired biological activity. Often screening entails identifying phage whose potential binding polypeptides can bind to a target molecule. In general, enough clones or pfu should be screened to ensure an adequate representation of displayed peptides is being screened. Preferably about 10 5 -10 6 pfu would be screened, more preferably at least about 10 9 pfus would be screened, still more preferably at least about 10 11 -10 12 pfu, would be screened. Often more than one round of screening will be necessary to identify or sufficiently enrich the phage particles of interest.
  • Suitable target molecules include a wide variety of molecules and include a molecule for which a practitioner desires to identify or isolate a polypeptide that will bind to the target molecule.
  • the target molecule can be an antigen where a library of phage displaying antibodies (e.g., scFv or Fab) are being screened to identify antibody sequences that bind to that particular antigen.
  • targets e.g., peptides, proteins, carbohydrates, nucleic acids, peptide nucleic acids, RNA, DNA, small organic molecules (i.e., carbon containing molecules of 100 kDa or less, more preferably 50 kDa or less, still more preferably 10 kDa or less), inorganic molecules, etc.
  • the target can be any substance that can serve as a ligand for the potential binding polypeptide of the phage-displayed polypeptide. If possible, a positive control for the retention of binding activity of the target for a potential binding polypeptide of interest should be included in the screening process to ensure proper conditions for identifying the phage are maintained.
  • the immobilization of a target or target-binding molecule to a solid support can facilitate separation of replicable genetic packages that can bind to the target molecule from the cells and unbound replicable genetic packages that are present in the uncleared cell culture.
  • One or more species of target molecules can be immobilized directly or indirectly as an array (i.e., a two or three-dimensional arrangement of molecules) on a solid support.
  • the target molecule(s) can be directly or indirectly immobilized on a solid support (see below).
  • the target molecule can be immobilized directly to the solid support through covalent and non-covalent bonds.
  • the target molecule can be indirectly bound to the solid support by coating the solid support with a substance or molecule that can bind to the target molecule.
  • the solid support can be coated with strepavidin and the target molecule can be biotinylated (Sparks et al. (1996), supra).
  • the biotinylated target molecules can be immobilized to the strepavidin coated solid support through the biotin-strepavidin interaction.
  • immobilized metal affinity substrates can be used in the present invention to indirectly bind the target molecule to a solid support (see Ausubel et al., eds., (1994) for review of immobilized metal affinity technology).
  • Ni-NTA nickel-nitrilotriacetic acid
  • Ni-NTA Agarose Qiagen
  • Ni-NTA Magnetic Agarose Beads Qiagen
  • Ni-NTA Magnetic Agarose Beads are beads of agarose, containing magnetic particles and nitrilotriacetic acid (NTA) groups on their surfaces.
  • the replicable genetic package-target molecule complexes can be released from a Ni-NTA substrate by an increase in the concentration of an imidazole in the solution sufficient to disrupt the poly-histidine-Ni-NTA interaction.
  • Other suitable methods of indirectly immobilizing target molecules include the binding of a target having a ligand binding protein moiety to a support that contains a ligand for the binding protein, e.g., maltose binding protein and amylose (New England Biolabs); an antibody with an Fc domain and protein A (Sparks et al. (1996), supra); and glutathione-S-transferase and glutathione agarose (see e.g., Ausubel et al., eds., (1994), supra).
  • the target can be soluble, i.e., not immobilized on a solid support.
  • the uncleared cell culture is then incubated with the soluble target.
  • Any resulting replicable genetic package-target complexes can subsequently be captured on a solid support by a target-binding molecule (see, eg., Sparks et al. (1996) Ch. 13, supra; see also, methods for indirectly binding a target molecule above).
  • non-specific binding of phage to the solid support can be decreased with agents such as non-fat dry milk or BSA (bovine serum albumin).
  • BSA bovine serum albumin
  • a non-ionic detergent e.g., Tween-20 or Triton-X-100.
  • solid supports can be used in the present invention.
  • solid supports include, without limitation, bead, microtiter plates, chips, prokaryotic and eukaryotic cells.
  • Beads can be composed of materials such as Sepharose, agarose, polystyrene, etc. and can be paramagnetic.
  • Microtiter plates are commercially available in a variety of formats (e.g., 96, 384 and 1536 well plates) and materials (e.g., polystyrene).
  • Chips can be comprised of a variety of materials, layers and substrates (see, e.g, WO 00/04389).
  • substances for use solid supports can be selected from a group consisting of silicon, silica, quartz, glass, controlled pore glass, carbon, alumina, titania, tantalum oxide, germanium, silicon nitride, zeolites, and gallium arsenide.
  • Many metals such as gold, platinum, aluminum, copper, titanium, and their alloys are also options for solid supports of the present invention.
  • many ceramics and polymers may also be used as solid supports.
  • Polymers which may be used as solid supports include, but are not limited to, the following: polystyrene; poly(tetra)fluoroethylene (PTFE); polyvinylidenedifluoride; polycarbonate; polymethylmethacrylate; polyvinylethylene; polyethyleneimine; poly(etherether)ketone; polyoxymethylene (POM); polyvinylphenol; polylactides; polymethacrylimide (PMI); polyatkenesulfone (PAS); polypropylene; polyethylene; polyhydroxyethylmethacrylate (HEMA); polydimethylsiloxane; polyacrylamide; polyimide; and block-copolymers.
  • the solid support on which the target resides may also be a combination of any of the aforementioned solid support materials.
  • the solid support can also be comprised of a eukaryotic or prokaryotic cell.
  • a population of phage or other particles include members that can specifically bind to a target molecule, those particles will bind to, and form a complex with, the target molecule. It is often desirable to remove the unbound components of the uncleared cell culture from complex (for example, removing the phage particles that do not specifically bind to the target molecule, the cells, and other components of the uncleared cell culture can be removed).
  • the uncleared culture can be separated from the replicable genetic package-target complex using variety of separation methods known in the art.
  • the uncleared cell culture can be aspirated from the well, leaving behind those replicable genetic packages that are bound to the immobilized target.
  • the target can be immobilized on a bead and the uncleared cell culture can be passed through a filter with a pore size smaller than the bead, but larger than a bacterial cell (or a eukaryotic cell when using eukaryotic host cells).
  • Another separation method is the immobilization of a target on a paramagnetic bead, and the decantation of the uncleared cell culture leaving the replicable genetic package-target molecule complex behind bound to the paramagnetic bead held in place with a magnetic field.
  • any resulting replicable genetic package-target molecule complex(es) can be subsequently separated from the uncleared cell culture.
  • the replicable genetic package-target complex can be incubated in the presence of a third molecule, a target complex-binding molecule, that is immobilized on a solid support and does not disrupt the replicable genetic package-target molecule complex(es).
  • the target complex-binding molecule can bind to either the soluble target molecule or to the replicable genetic package. This permits the replicable genetic package-target complex to bind to the target-binding molecule, thereby indirectly immobilizing the replicable genetic package-target complex.
  • the uncleared cell culture can then be separated from the replicable genetic packages that bind specifically to the target molecule using the separation methods described above for the first category of replicable genetic package-target complexes.
  • At least 70% of the cells are separated from the replicable genetic packages that are specifically bound to the target molecule, more preferably, at least 80% of the cells are separated from the replicable genetic packages that are specifically bound to the target molecule, still more preferably, at least 90% of the cells are separated from the replicable genetic packages that are specifically bound to the target molecule, yet still more preferably, substantially all of the bacterial cells are separated from the replicable genetic packages that are specifically bound to the target molecule.
  • the wash can remove undesirable components of the cell cultures from the specifically bound replicable genetic packages.
  • the wash can remove cells, non-specifically bound replicable genetic packages, etc.
  • a wash buffer is used.
  • the wash buffer can contain a detergent, or other agents, and compositions that are compatible with replicable genetic package-target binding to increase the stringency of the screening process.
  • a wash buffer that can be used in the present invention is a solution of Tris buffered saline with 0.05% Tween-20, pH 7.4 (TBST).
  • replicable genetic packages that specifically bind to the target molecule.
  • the replicable genetic packages can then be used for, for example, further rounds of screening, amplification, detection, or characterization (e.g., nucleic acid sequencing). Elution can be accomplished using a variety of methods known in the art.
  • the replicable genetic packages can be eluted using pH changes, protein denaturants, or EGTA/EDTA if a metal ion is necessary for replicable genetic package-target interaction (See e.g., Sparks et al. (1996) Ch. 13, supra for elution techniques using phage-display).
  • the replicable genetic packages can be eluted using an acidic buffer (e.g., glycine-HCl, pH 2) (see, e.g., Sparks et al. (1996) Ch. 13, supra).
  • the eluate can then be removed and neutralized with the addition of a second buffer (e.g., NaPO 4 buffer pH 7.5) (see, e.g., Sparks et al. (1996) Ch. 13, supra).
  • a second buffer e.g., NaPO 4 buffer pH 7.5
  • natural or synthetic ligands that interrupt the replicable genetic package-target complex can be used to elute the replicable genetic package from the target (Sparks et al. (1996) Ch. 13, supra).
  • the replicable genetic package(s) can be amplified in order to increase the number of replicable genetic package, thus potentially increasing the chance that enough of the replicable genetic package(s) will be present in the next round for isolation, identification, or detection, etc.
  • Methods for amplifying replicable genetic packages in solid and liquid culture are known in the art (see, e.g., Sparks (1996), Ch. 13, supra, and Rider et al. (1996) Ch. 4, supra, for methods of amplifying filamentous bacteriophage).
  • screening methods of the present invention can be optimized. Furthermore, skilled artisans will recognize methods of optimizing to determine the effectiveness of steps and to increase the chances of identifying the replicable genetic package of interest. For example, the inclusion of positive and negative controls in the screening process can facilitate the trouble-shooting and/or optimization of a screening process.
  • Another aspect of the present invention is that the presence of cells and other components of an uncleared culture do not interfere with the detection of particles that specifically bind to a target molecule. Accordingly, some methods of the present invention involve contacting the particle-target molecule complex with a detection reagent prior to removing the cells and/or other components of the uncleared cell culture.
  • replicable genetic packages that bind to a target molecule can be detected using a variety of materials and methods known to those of skill in the art.
  • the replicable genetic package-target complexes can be incubated with a detection reagent.
  • a detection reagent is labeled with a substance that permits the qualitative or quantitative determination of the presence or absence of the replicable genetic package-target complex.
  • labeled refers to a composition is that is detectable, either directly or indirectly, by spectroscopic, photochemical, biochemical, immunochemical, or chemical means.
  • useful labels include radiolabels (e.g., H 3 , C 13 , C 14 , P 32 , S 35 , I 125 ), fluorescent dyes, fluorophores, electrondense reagents, enzymes and their substrates (e.g., as commonly used in enzyme-linked immunoassays, e.g., alkaline phosphatase and horse radish peroxidase), biotin-streptavidin, digoxigenin, or haptens and proteins for which antisera or monoclonal antibodies are available.
  • the label or detectable moiety is typically bound, either covalently, through a linker or chemical bound, or through ionic, van der Waals or hydrogen bonds to the molecule to be detected.
  • Detection reagents can include antibodies—such as antibodies that react with the native form of the phage being used, e.g., anti-M13KO7 antibody.
  • the antibody itself can be labeled.
  • horseradish peroxidase (HRP) can be conjugated to an anti-M13 antibody (Amersham-Pharmacia Biotech, Piscataway, N.J.).
  • HRP and o-Phenylenediamine Dihydrochloride OPD; Sigma, St. Louis Mo.
  • OPD o-Phenylenediamine Dihydrochloride
  • the detection of the replicable genetic package-target complex on the chip could be analyzed using a physical spectroscopy method, such as mass spectroscopy or surface plasmon resonance (U.S. Pat. No. 5,641,640).
  • Surface plasmon-resonance has been used to detect phage-displayed antibody-target interactions (de Haard et al., (1999)).
  • Chips and surface plasmon resonance instruments are commercially available (e.g., BIACORE, Uppsala, Sweden) for the detection of analytes.
  • Fluorescence polarization could also be employed by modifying the target molecule with an appropriate fluorescence label or fluorophore (Burke et al. (1996) Chapter 18 , Measurement of Peptide Binding Affinities Using Fluorescence Polarization , in Kay et al., eds. (1996)).
  • the replicable genetic package that are bound to a target can be further characterized as to the genetic or protein makeup of their potential binding polypeptide(s).
  • the nucleic acid sequence of the potential binding polypeptide can be determined by sequencing the phagemid vector contained in a particular phage (see e.g., Masecar et al. (1996) Chapter 17 , Nonradioactive Sequencing of Random Peptide Recombinant Phage, in Kay et al., eds. ( 1996)).
  • the protein makeup of a phage could be determined using methods known in the art, such as immunological assays (e.g., Western blots), two-dimensional gels, mass spectrometry, etc.
  • high-throughput analysis and screening of replicable genetic package-display libraries can be performed on a automated workstation (see e.g., U.S. Pat. No. 5,139,744, “Automated laboratory workstation having module identification means.”).
  • An “automated workstation” is typically a computer-controlled apparatus which can, through robotic functions, transfer, mix, and remove liquids from microtiter plates.
  • An automated workstation can also contain a built-in plate reader, which can read the absorbance of a liquid in a microtiter well.
  • the automated workstation can be programmed to carry out a series of mixing, transfer, and/or removal steps.
  • the automated workstation will typically have a multi-channel pipettor which can pipette small amounts of liquid (e.g., microliter amounts) from a vessel to the well.
  • the automated workstation can transfer uncleared cell culture(s) into a micro-titer plate.
  • the microtiter plate can have pre-immobilized target molecule(s) already in the wells.
  • the automated workstation can subsequently be used to remove uncleared cell cultures from the wells, wash the wells, or elute the replicable genetic packages from the immobilized target.
  • Detection of a replicable genetic package bound to an immobilized target molecule can also be carried out using an automated workstation.
  • the automated workstation can be used to add a detection reagent to the wells.
  • the automated workstation when equipped with a plate reader, can monitor the absorbance of the reaction of the detection reagent in the wells.
  • Assembled scFv or Fab DNA sequences were digested with SfiI and NotI, subcloned into the pCANTAB5E vector (Amersham-Pharmacia Biotech, Piscataway, N.J.), and transformed into TG1 or XL1-Blue competent E.coli .
  • Individual clones capable of specific binding to the target antigen were isolated by conventional methods and then used to explore alternatives to centrifugation. Single colonies were picked into 0.1 ml cultures (2 ⁇ YT supplemented with 2% glucose and 100 ⁇ g/ml Ampicillin) in a deep well 96-well plate and incubated at 37° C. with shaking for 5-6 hours when cultures reached mid-log phase.
  • Polystyrene plates were coated with protein antigens (hIgG1 ⁇ , hIgG1 ⁇ , or rabbit IgG) (1-10 ⁇ g/ml in 0.1 M sodium bicarbonate pH 9.6) overnight at 4° C., blocked with 3% non-fat dry milk (NFM) in Tris buffered saline with 0.05% Tween-20 (pH 7.4, TBST), and washed 3 ⁇ in TBST. Aliquots of bacterial culture were removed to a separate microtiter plate, or to wells in a 96-well filtration plate (MultiScreen plates from Millipore, Bedford Mass.), a vacuum was applied slowly, and filtrate collected in a microtiter plate. The remainder of the culture in the deep well plates was centrifuged at 1,725 ⁇ g (3,500 rpm in an Eppendorf 5804 equipped with an A2MTP rotor) for 30 minutes at room temperature (RT).
  • RT room temperature
  • HRP horseradish peroxidase
  • Assays were performed in duplicate and repeated 2 or 3 times with similar results.
  • the scFv display phage derived from HP6054 bind to the human lambda light chain antigen (in association with IgG1), but not to the kappa light chain (hIgG1 Kappa) or to Rabbit IgG (FIG. 1A). Filtrate generated from the same culture yields an equivalent level of binding as observed for the supernatant.
  • the uncleared bacterial culture demonstrated similar levels of binding to the immobilized antigen, indicating that removal of bacteria by time consuming centrifugation or costly filtration is not necessary. Furthermore, no increase in binding was observed to either of the two non-specific antigens tested, hIgG1 ⁇ and rabbit IgG.
  • assay of a polyclonal population of phage producing bacteria would be necessary, e.g, library screening.
  • ELISA electrospray-activated protein ase Ig., Ag. Ag. Ag. Ag. Ag. Ag. Ag. Ag. Ag. Ag. Ag. Ag. Ag. Ag. Ag. Ag. Ag. Ag. Ag. Ag. Ag. culture involves competition between individuals, which affects the yield of specific phage. However, the bias that is introduced during polyclonal growth would exist regardless of the means of analysis of that culture.
  • E.coli carrying phagemid expressing scFv derived from HP6002 (recognizing hIgG2) or HP6025 (recognizing hIgG4) were grown overnight in the absence of IPTG. Cultures that attained different densities (OD 600 for HP6002 was 1.2 and 1.0, and HP6025 was 2.1 and 3.3 in two separate trials) were mixed on the basis of volume. Final volume ratios ranged from a 0.001 to 1000 of HP6025/HP6002. Aliquots of the mixed culture were compared to supernatants clarified by centrifugation in the phage ELISA. FIG. 2 demonstrates that clarified phage supernatant and bacterial culture do not exhibit significant differences in binding at any of the ratios tested. Therefore, sensitivity of the ELISA does not appear to be compromised by the presence of bacteria during the binding of the phage an immobilized antigen.

Landscapes

  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Genetics & Genomics (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Molecular Biology (AREA)
  • Microbiology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • Biotechnology (AREA)
  • Biomedical Technology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Biophysics (AREA)
  • Physics & Mathematics (AREA)
  • Crystallography & Structural Chemistry (AREA)
  • Plant Pathology (AREA)
  • Virology (AREA)
  • Medicinal Chemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Health & Medical Sciences (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Investigating Or Analysing Biological Materials (AREA)

Abstract

This invention provides methods for screening populations of phage-displayed polypeptides that are particularly well-suited for high-throughput screening. The methods do not require the clearing of cells from a culture used to obtain the population of phage or other replicable genetic packages. Accordingly, the invention provides methods for forming complexes between a replicable genetic package displaying a polypeptide fusion and a target molecule in an uncleared cell culture containing replicable genetic package. Compositions made up of an uncleared cell culture containing replicable genetic packages displaying a polypeptide fusion and a target molecule are provided in the invention as well.

Description

    CROSS-REFERENCES TO RELATED APPLICATIONS
  • This application claims the benefit of U.S. Provisional Patent application Ser. No. 60/209503, filed on Jun. 5, 2000, the teachings of which are herein incorporated by reference.[0001]
  • STATEMENT AS TO RIGHTS TO INVENTIONS MADE UNDER FEDERALLY SPONSORED RESEARCH AND DEVELOPMENT
  • Not applicable. [0002]
  • BACKGROUND OF THE INVENTION
  • Phage display and related techniques have become powerful methods for the discovery of affinity binding reagents (Smith (1985) [0003] Science 228: 1315-1317). Linear and constrained peptides, antibody fragments (e.g., scFvs, Fvs and Fabs), as well as a number of alternative binding domains have all been displayed on phage particles, for example, via fusion to one of the phage coat proteins. Although several phage proteins (derived from gVIII, gVI, gVHI and gIX) have all been used as fusion partners for display of recombinant proteins, gIII is the most widely used. Phagemids containing a phage origin of replication, an antibiotic resistance marker, and a gene encoding a binding domain/gill fusion protein are readily constructed via conventional molecular biology techniques. Through large-scale ligation and transformation as well as recombination strategies, large libraries of 108 to 1011 different recombinants are now being generated for use in affinity selection strategies (de Haard et al. (1999) J. Biol. Chem. 274: 18218-18230; Sblattero and Bradbury (2000) Nat. Biotechnol. 18: 75-80); Sheets et al. (1998) Proc. Natl. Acad. Sci., U.S.A. 95:6157-6162, published erratum appears in Proc. Natl. Acad. Sci., U.S.A. (1999) 96: 795).
  • Once a library of phage displaying potential binding agents is generated, individual phage with the capacity to bind to a chosen target must be isolated from an enormous excess of non-binding phage. To screen large numbers of phage to identify those that display polypeptides having a desired activity, it is desirable to develop high-throughput screening (HTS) methods. Preferably, such HTS methods would automate the phage screening process so that large numbers of phage could be screened with little human intervention. Although HTS methods are available for many types of screening, previously known phage display protocols include steps that are not readily automatable. In particular, phage display protocols require, prior to screening, separation of the phage from the host cells in which the phage are amplified. [0004]
  • Traditionally, overnight cultures of bacteria producing phage are centrifuged or filtered to pellet bacteria and phage supernatants are used in the screening (See generally, Kay et al., eds. (1996) [0005] Phage display of peptides and proteins: a laboratory manual. Academic Press Inc., San Diego Calif.). Alternatively, phage can be purified and concentrated from cleared supernatants by precipitation (e.g., with polyethylene glycol). However, these clearing methods are not readily performed by robotic systems (e.g., automated workstations). Therefore, time-consuming and expensive human intervention is required. These drawbacks are exacerbated as the numbers of samples are increased and during high-throughput screening. Therefore, a need exists for more fully automated methods for screening of phage display libraries. The present invention fulfills this and other needs.
  • SUMMARY OF THE INVENTION
  • The present invention provides methods for screening a population of replicable genetic packages (e.g., phage, eukaryotic viruses, and the like) to obtain particles that display on their surface a fusion protein that specifically binds to a target molecule. Unlike previous methods, which involve clearing a culture of cells prior to screening the methods of the present invention involve contacting a target molecule with an uncleared cell culture that contains a population of replicable genetic packages. Each replicable genetic package displays on its surface a fusion protein that has a surface-displayed replicable genetic package polypeptide and a potential binding polypeptide. The replicable genetic package that specifically bind to the target molecule form complexes containing replicable genetic packages and target molecules. In some cases, the potential binding polypeptide can be encoded by a member of a library of nucleic acid molecules. For example, the nucleic acid molecules can be cDNA molecules or recombinant products. In other cases, the potential binding polypeptide can be, for example, an antibody, or derivative of an antibody. For example, the potential binding polypeptide can be a scFv or a Fab. [0006]
  • The methods of the invention are useful for obtaining polypeptides that bind to essentially any molecule. For example, the target molecule can be a polypeptide, an RNA, a DNA, a small organic molecule and a carbohydrate. The target molecules can be immobilized directly or indirectly to a solid support. Solid supports such as a bead, a chip, a microtiter plate, a eukaryotic cell, or a prokaryotic cell are present in some embodiments of the invention. The solid supports of the present invention can contain a variety of materials, such as Sepharose, polystyrene, glass, silicon oxide, etc. [0007]
  • In some embodiments, the methods also involve obtaining replicable genetic packages that specifically bind to the target molecule. For example, the replicable genetic packages that specifically bind to the target molecule can be separated from the bacterial cells after the binding of the phage to the target molecule. For example, the uncleared cell culture can be separated from a replicable genetic package-target complex(es) using aspiration. Once the replicable genetic packages are bound to the target molecule, some embodiments of the invention can further involve eluting the replicable genetic packages from the target molecule. Also, some embodiments involve identifying the replicable genetic packages that specifically bind to the target molecule with a detection reagent. [0008]
  • The present invention also provides compositions containing an uncleared cell culture, which contains: (a) a population of replicable genetic packages that display on their surfaces a fusion protein that includes a surface-displayed replicable genetic package polypeptide and a potential binding polypeptide; (b) a complex that is composed of one or members of the library of replicable genetic packages that specifically bind to the target molecule; and (c) cells in which the replicable genetic packages were amplified.[0009]
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIGS. 1A and 1B show that the binding of HP6054-scFv and HP6054-Fab phage to human lambda light chain is not affected by the presence of bacteria in the sample. Antigens were immobilized on 96-well plates (Nunc, Denmark) at 10 μg/ml. A phage ELISA was conducted using either an uncleared bacterial culture, or supernatants clarified by centrifugation or filtration. Each bar represents the mean±s.d. of duplicate samples. FIG. 1A depicts representative results from the HP6054 scFv-phage, and FIG. 1B is representative of HP6054 Fab-phage. [0010]
  • FIG. 2 shows that the sensitivity of phage ELISA is not impaired by the presence of bacteria. Overnight cultures of HP6002 scFv-phage and HP6025 scFv-phage were mixed at various ratios and then supernatants or uncleared culture was tested in the phage ELISA against hIgG2 and hIgG4 (each at 10 μg/ml). Each bar represents the mean±s.d. of duplicate samples.[0011]
  • DESCRIPTION OF THE SPECIFIC EMBODIMENTS
  • Definitions [0012]
  • “Replicable genetic packages” include virions of bacteriophage, filamentous phage, or any other eukaryotic virus, bacterial virus or phage. The term “phage” as used herein encompasses not only bacteriophage but also other types of replicable genetic packages, except where the term is used in a context that dictates a more specific meaning. [0013]
  • A molecule is “display(ed) on their surface” of a replicable genetic package if at least part of the molecule is accessible to the milieu surrounding a replicable genetic package. [0014]
  • The phrase “specifically (or selectively) binds to” in the context of a replicable genetic package refers to a binding reaction which is determinative of the presence of a replicable genetic package binding to a target molecule(s) in the presence of a population of other proteins, biologics, and replicable genetic packages. Thus, under designated binding conditions, a specifically binding replicable genetic package will bind to a particular molecule (e.g., target molecule) and under the same designated binding conditions, native replicable genetic packages do not bind to a particular molecule in a significant amount. Typically, a replicable genetic package “specifically” binds to a target molecule when the number of replicable genetic packages displaying a potential binding polypeptide that are bound to the target molecules is at least twice the background binding observed using a native replicable genetic package as a control. [0015]
  • An “uncleared cell culture” is an aqueous medium containing bacterial or eukaryotic cells. Typically, the “uncleared cell culture” is a growth of bacterial or eukaryotic cells infected with one or more replicable genetic package clones. [0016]
  • A “target molecule” is essentially any molecule that is being used as a probe to identify molecules that will bind to the target molecule. Examples of target molecules include, without limitation, amino acids, peptides, proteins, polypeptides, carbohydrates, small organic molecules, inorganic molecules, etc. [0017]
  • A “surface-displayed replicable genetic package polypeptide” is a polypeptide that is, at least in part, exposed to the milieu surrounding the virion. Examples of “surface-displayed replicable genetic package polypeptides” include, without limitation, pIII and pVIII. [0018]
  • A “potential binding polypeptide” is a polypeptide that may possibly bind to the target molecule. A “potential binding polypeptide” can be screened for its ability to bind to a target molecule of choice. [0019]
  • A “replicable genetic package-target complex” is a complex in which a target molecule is bound to a replicable genetic package. The target molecule is bound to the replicable genetic package through the binding domain portion of a polypeptide displayed on the surface of a replicable genetic package. [0020]
  • An “antibody” can be derived from sequence of a mammal, non-mammal (e.g., birds, chickens, fish, etc.), or fully synthetic antibody sequences. A “mammal” is a member of the class Mammalia. Examples of mammals include, without limitation, humans, primates, chimpanzees, rodents, mice, rats, rabbits, sheep, and cows. The term “antibody” also refers to fragments and substitutes for antibodies such as F(ab′)[0021] 2, Fab′, and Fab fragments. Additionally the “antibodies” can be single chain antibodies known as ScFv fragments, which are obtained by recombinantly fusing the variable regions of the light and heavy chains of the antigen binding fragment of interest.
  • I. Introduction [0022]
  • The present invention provides methods and compositions for screening replicable genetic package particles (e.g., phage, viruses, etc.) that display polypeptides for their ability to bind to a target molecule. Traditionally, such screening methods required clearing the host cells or bacteria from an uncleared cell culture and/or isolating a replicable genetic package stock before incubation with the target molecule. The methods of the present invention, however, do not require these procedures. Therefore, the invention provides significant advantages over previously available methods for screening phage and other particles, particularly when used in a high-throughput format. [0023]
  • Briefly, methods of the invention involve infecting bacteria or other suitable host cells with phage particles (or incubating cells that are transfected with a phagemid expression vector with helper phage) to generate an uncleared cell culture that contains a library of phage particles. This uncleared culture is then incubated with a target molecule. Phage particles that display a polypeptide that binds to the target molecule form a complex with the target molecule. After an incubation period, the bacterial or other cells used to amplify the phage can be separated from the phage particles that bind to the target molecule. The phage particles that were able to bind to the target molecule can then be further purified, characterized, amplified, and/or detected, etc. These methods and compositions will be described in more detail below. [0024]
  • II. Replicable Genetic Package Display Libraries [0025]
  • The methods of the invention are useful for screening a wide variety of phage display libraries. Phage display and related techniques provides a powerful method for selecting proteins of interest from large libraries (Bass et al. (1990) [0026] Proteins: Struct. Funct. Genet. 8: 309; Lowman and Wells (1991) Methods: A Companion to Methods Enz. 3(3);205-216. Lowman and Wells (1993) J. Mol. Biol. 234;564-578). Each phage or other particle displays a unique variant protein on its surface and packages the gene encoding that particular variant. For example, the libraries can be composed of homogenous or heterogenous populations of phage particles. That is, each phage in the library can display the same potential binding polypeptide, or each phage can display a different potential binding polypeptide. Potential binding polypeptides can serve as epitopes, ligands, agonists, antagonists, enzymes, etc. For example, the potential binding polypeptides can encode scFvs and Fabs.
  • Some recent reviews on the phage display technique include, for example, McGregor (1996) [0027] Mol Biotechnol. 6(2): 155-62; Dunn (1996) Curr. Opin. Biotechnol. 7(5):547-53; Hill et al. (1996) Mol Microbiol 20(4):685-92; Phage Display of peptides and Proteins: A Laboratory Manual. B K. Kay, J. Winter, J, McCafferty eds., Academic Press 1996; O'Neil et al. (1995) Curr. Opin. Struct. Biol. 5(4):443-9; Phizicky et al. (1995) Microbiol. Rev. 59(1):94123; Clackson et al. (1994) Trends Biotechnol. 12(5):173-84; Felici et al. (1995) Biotechnol. Annu. Rev. 1:149-83; Burton (1995) Immunotechnology 1(2):87-94.) See, also, Cwirla et al., Proc. Natl. Acad. Sci. USA 87:6378-6382 (1990); Devlin et al., Science 249: 404-406 (1990), Scott & Smith, Science 249: 386-388 (1990); Ladner et al., U.S. Pat. No. 5,571,698.
  • The methods of the invention are applicable to any of the genetic packages most frequently used for phage display libraries. These include, for example, bacteriophage, particularly filamentous phage, and especially phage M13, Fd and F1. (Webster (1996) Chapter 1[0028] , Biology of the Filamentous Bacteriophage, in Kay et al., eds. (1996) Phage Display of Peptides and Proteins). Microbiological methods for growing, titering, and preparing filamentous phage particles, and phage DNA are known in the art (Rider et al. (1996) Chapter 4, Microbiological Methods, in Kay et al., eds. (1996)) and their genomes are very well characterized. These filamentous phage have genes which encode the various capsid proteins and are known as genes III, VI, VII, VIII, and IX (Webster et al., (1996), supra). The proteins the genes encode are known as pIII, pVI, pVII, pVIII, and pIX, respectively. The most abundant capsid protein is pVIII, which has 2700 copies on the surface of the phage. Approximately 5 copies of pIII are displayed on the phage particle.
  • Typically, libraries of nucleic acid molecules are ligated into a phage-display vector and introduced into bacteria to create a library of particles displaying fusion proteins that consist of a surface-displayed phage polypeptide and a potential binding polypeptide. Most work has involved inserting nucleic acid libraries encoding polypeptides to be displayed into either a gIII or gVIII expression vector in order to produce phage-displayed fusion protein(s) (See, e.g., Dower, WO 91/19818; Devlin, WO 91/18989; MacCafferty, WO 92/01047 (gene III); Huse, WO 92/06204; Kang, WO 92/18619 (gene VIII)). These fusion proteins generally included a signal sequence, usually but not necessarily, from the phage coat protein, a polypeptide to be displayed and either the gene III or gene VIII protein or a fragment thereof. Exogenous coding sequences are often inserted at or near the N-terminus of gene III or gene VIII, although other insertion sites are possible. pVIII, however, can only tolerate short inserts—about 5 to 6 amino acid residues. (Armstrong et al., (1996), supra). Larger peptides can be displayed as pVIII fusions if pVIII wild-type coat proteins are interspersed with the recombinant pVIII (Malik et al. (1996) Chapter 8[0029] , Multiple Display of Foreign Peptide Epitopes on Filamentoisa Bacteriophage Virions, in Kay et al., eds. (1996)).
  • A variety of vectors for displaying pIII and pVIII fusion proteins in a phage display library have been described (Armstrong et al. (1996) Chapter 3[0030] , Vectors for Phage Display, in Kay et al., eds. (1996); Dottavio (1996) Chapter 7, Phagemid-Displayed Peptide Libraries, in Kay et al., eds. (1996); (Malik et al. (1996) Chapter 8, Multiple Display of Foreign Peptide Epitopes on Filamentous Bacteriophage Virions, in Kay et al., eds. (1996)) and are commercially available (e.g., pCANTAB5E, Pharmacia; λSurfZap, Stratagene).
  • Eukaryotic replicable genetic packages such as eukaryotic viruses can also be used to display polypeptides in an analogous manner. For example, display of human heregulin fused to gp70 of Moloney murine leukemia virus has been reported by Han et al., (1995) [0031] Proc. Nat'l. Acad. Sci. USA 92: 9747-9751.
  • Alternatively, prokaryotic spores can be used as replicable genetic packages. In this case, polypeptides are displayed from the outer surface of the spore. For example, spores from [0032] B. subtilis have been reported to be suitable. Sequences of coat proteins of these spores are described in Donovan et al., J. Mol. Biol. 196: 1-10 (1987). Thus, spores can be used to display the potential binding polypeptides.
  • The nucleic acid libraries encoding the potential binding polypeptides can be constructed from nucleic acids from a variety of sources, including cDNA, genomic DNA, synthetic nucleotides and/or from oligomers encoding randomized peptides (see, e.g., Adey et al. (1996) Chapter 5[0033] , Construction of Random Peptide Libraries in Bacteriophage M13, in Kay et al., eds. (1996) for descriptions of randomized peptide libraries). Random peptide libraries have been constructed using synthetic degenerate oligonucleotides and expressed as fusions with pIII (Adey et al, (1996), supra). Also, libraries of antibody and antibody fragments (Fv, scFv and Fab) can be expressed in phage display systems with pIII (McCafferty and Johnson (1996) Chapter 6, Construction and Screening of Antibody Display Libraries, in Kay et al., eds. (1996)). One method of constructing an antibody phage display library involves generating nucleic acids encoding antibody fragments from the amplification of variable domain gene sequences (McCafferty and Johnson (1996), supra). The fragments can be amplified from nucleic acids isolated from antigen immunized or non-immunized sources. The nucleic acids encoding variable heavy and light chain domains are then spliced together using overlap PCR and ligated into a phage-display vector to subsequently generate the antibody phage display library (McCafferty and Johnson (19.96), supra).
  • Molecular biological methods that can be used to isolate, manipulate, and generate the nucleic acid libraries of the present invention are well known in the art and are detailed in Sambrook et al., [0034] Molecular Cloning, A Laboratory Manual (2nd ed. 1989); Kriegler, Gene Transfer and Expression: A Laboratory Manual (1990); and Current Protocols in Molecular Biology (Ausubel et al., eds., (1994)).
  • Numerous other methods for constructing phage display libraries are known in the art. For example, libraries expressing fragments of a protein can be used to map the epitopes of an antibody, which can serve as the target molecule (Plessis and Jordaan (1996) Chapter 9[0035] , Phage Libraries Displaying Random Peptides Derived from a Target Sequence, in Kay et al., eds. (1996)). Also, once a recombinant phage has been isolated or constructed, it can be used to construct a second-generation phage-display library through DNA shuffling (Adey et al. (1996) Chapter 16, Preparation of Second-Generation Phage Libraries, in Kay et al., eds. (1996)).
  • Once the nucleic acids have been introduced into an appropriate expression vector, phage particles are obtained. The vectors are introduced into appropriate host cells and amplified. Uncleared cell cultures containing libraries of phage particles can be generated using methods well known in the art (see, e.g., Sparks et al. (1996) Chapter 13[0036] , Screening Phage-Displayed Random Peptide Libraries, in Kay et al., eds. (1996)). For example, libraries of phage particles displaying the potential binding polypeptides can be used to infect bacteria (e.g, E. coli) in order to generate an uncleared cell culture. Alternatively, a library of nucleic acid molecules encoding the potential phage binding polypeptides (e.g., phagemid vectors) can be introduced into bacteria, which are subsequently infected with a helper phage (see, e.g., Sparks et al., (1996) Ch. 13, supra). These procedures can generate a library of phage particles in an uncleared cell culture.
  • III. Screening Replicable Genetic Package-Display Libraries [0037]
  • Phage display libraries are screened to obtain phage that display on their surfaces a polypeptide that has a desired activity (e.g., the ability to bind to a target molecule). Methods for screening phage-displayed libraries are known in the art (Sparks et al. (1996) Chapter 13, supra); McCafferty and Johnson (1996) Chapter 6, supra; McCafferty (1996) Chapter 15[0038] , Phage Display: Factors Affecting Panning Efficiency, in Kay et al., eds. (1996)). To date, however, these methods involve either clearing a cell culture (e.g., by centrifugation, filtration) or isolating the entire phage library in the culture (e.g., by precipitation, centrifugation, etc.) for subsequent screening. This represents an extra step that necessitates the expenditure of extra time and effort to transfer the container or plate containing the uncleared cell culture to another format suitable for centrifugation, filtration, etc. For example, centrifugation of uncleared cell cultures that have been transferred to or grown in a microtiter plate requires transferring the plate to a centrifuge. This requires an operator to move the plate from the bench to the centrifuge, wait for the centrifugation to take place, and then remove the cleared culture from the plate to continue with the screening. These time consuming and unnecessary steps for clarifyng a bacterial culture in the screening of a phage display library can be eliminated using the methods of the present invention.
  • Screening involves selecting phage that display on their surface a polypeptide that has a desired biological activity. Often screening entails identifying phage whose potential binding polypeptides can bind to a target molecule. In general, enough clones or pfu should be screened to ensure an adequate representation of displayed peptides is being screened. Preferably about 10[0039] 5-106 pfu would be screened, more preferably at least about 109 pfus would be screened, still more preferably at least about 1011-1012 pfu, would be screened. Often more than one round of screening will be necessary to identify or sufficiently enrich the phage particles of interest.
  • Suitable target molecules include a wide variety of molecules and include a molecule for which a practitioner desires to identify or isolate a polypeptide that will bind to the target molecule. For example, the target molecule can be an antigen where a library of phage displaying antibodies (e.g., scFv or Fab) are being screened to identify antibody sequences that bind to that particular antigen. Thus, a variety of targets (e.g., peptides, proteins, carbohydrates, nucleic acids, peptide nucleic acids, RNA, DNA, small organic molecules (i.e., carbon containing molecules of 100 kDa or less, more preferably 50 kDa or less, still more preferably 10 kDa or less), inorganic molecules, etc.) can be used to probe a phage-display library. Essentially, the target can be any substance that can serve as a ligand for the potential binding polypeptide of the phage-displayed polypeptide. If possible, a positive control for the retention of binding activity of the target for a potential binding polypeptide of interest should be included in the screening process to ensure proper conditions for identifying the phage are maintained. [0040]
  • The immobilization of a target or target-binding molecule to a solid support can facilitate separation of replicable genetic packages that can bind to the target molecule from the cells and unbound replicable genetic packages that are present in the uncleared cell culture. One or more species of target molecules can be immobilized directly or indirectly as an array (i.e., a two or three-dimensional arrangement of molecules) on a solid support. [0041]
  • Those of skill in the art will recognize a variety of methods to immobilize a target molecule to a solid support. For example, the target molecule(s) can be directly or indirectly immobilized on a solid support (see below). The target molecule can be immobilized directly to the solid support through covalent and non-covalent bonds. [0042]
  • Alternatively, the target molecule can be indirectly bound to the solid support by coating the solid support with a substance or molecule that can bind to the target molecule. For example, the solid support can be coated with strepavidin and the target molecule can be biotinylated (Sparks et al. (1996), supra). Thus, the biotinylated target molecules can be immobilized to the strepavidin coated solid support through the biotin-strepavidin interaction. Those of skill in the art will also recognize that immobilized metal affinity substrates can be used in the present invention to indirectly bind the target molecule to a solid support (see Ausubel et al., eds., (1994) for review of immobilized metal affinity technology). For example, solid supports containing Ni-NTA (nickel-nitrilotriacetic acid) such as Ni-NTA Agarose (Qiagen) or Ni-NTA Magnetic Agarose Beads (Qiagen) can be used to bind target molecules having an N-terminal or C-terminal stretch of poly-histidine (e.g., 6 or more histidines). Ni-NTA Magnetic Agarose Beads are beads of agarose, containing magnetic particles and nitrilotriacetic acid (NTA) groups on their surfaces. The replicable genetic package-target molecule complexes can be released from a Ni-NTA substrate by an increase in the concentration of an imidazole in the solution sufficient to disrupt the poly-histidine-Ni-NTA interaction. Other suitable methods of indirectly immobilizing target molecules include the binding of a target having a ligand binding protein moiety to a support that contains a ligand for the binding protein, e.g., maltose binding protein and amylose (New England Biolabs); an antibody with an Fc domain and protein A (Sparks et al. (1996), supra); and glutathione-S-transferase and glutathione agarose (see e.g., Ausubel et al., eds., (1994), supra). [0043]
  • Alternatively the target can be soluble, i.e., not immobilized on a solid support. The uncleared cell culture is then incubated with the soluble target. Any resulting replicable genetic package-target complexes can subsequently be captured on a solid support by a target-binding molecule (see, eg., Sparks et al. (1996) Ch. 13, supra; see also, methods for indirectly binding a target molecule above). [0044]
  • After immobilizing the target on the solid support, non-specific binding of phage to the solid support can be decreased with agents such as non-fat dry milk or BSA (bovine serum albumin). Those of skill in the art will recognize other agents that can be used alone or in combination to decrease non-specific binding such as a non-ionic detergent (e.g., Tween-20 or Triton-X-100). [0045]
  • A variety of solid supports can be used in the present invention. Examples of solid supports include, without limitation, bead, microtiter plates, chips, prokaryotic and eukaryotic cells. Beads can be composed of materials such as Sepharose, agarose, polystyrene, etc. and can be paramagnetic. Microtiter plates are commercially available in a variety of formats (e.g., 96, 384 and 1536 well plates) and materials (e.g., polystyrene). Chips can be comprised of a variety of materials, layers and substrates (see, e.g, WO 00/04389). For example, substances for use solid supports can be selected from a group consisting of silicon, silica, quartz, glass, controlled pore glass, carbon, alumina, titania, tantalum oxide, germanium, silicon nitride, zeolites, and gallium arsenide. Many metals such as gold, platinum, aluminum, copper, titanium, and their alloys are also options for solid supports of the present invention. In addition, many ceramics and polymers may also be used as solid supports. Polymers which may be used as solid supports include, but are not limited to, the following: polystyrene; poly(tetra)fluoroethylene (PTFE); polyvinylidenedifluoride; polycarbonate; polymethylmethacrylate; polyvinylethylene; polyethyleneimine; poly(etherether)ketone; polyoxymethylene (POM); polyvinylphenol; polylactides; polymethacrylimide (PMI); polyatkenesulfone (PAS); polypropylene; polyethylene; polyhydroxyethylmethacrylate (HEMA); polydimethylsiloxane; polyacrylamide; polyimide; and block-copolymers. The solid support on which the target resides may also be a combination of any of the aforementioned solid support materials. The solid support can also be comprised of a eukaryotic or prokaryotic cell. [0046]
  • IV. Separating the Replicable Genetic Package-Target Complex(Es) from the Uncleared Cell Culture [0047]
  • If a population of phage or other particles include members that can specifically bind to a target molecule, those particles will bind to, and form a complex with, the target molecule. It is often desirable to remove the unbound components of the uncleared cell culture from complex (for example, removing the phage particles that do not specifically bind to the target molecule, the cells, and other components of the uncleared cell culture can be removed). In cases where the target is immobilized, directly or indirectly, on a solid support, the uncleared culture can be separated from the replicable genetic package-target complex using variety of separation methods known in the art. There are many separation methods known in the art (e.g., filtering, sedimenting, centrifuging, decanting, precipitation, etc.) that can be used or adapted for use in the present invention. For example, the where the target is immobilized on a microtiter plate, the uncleared cell culture can be aspirated from the well, leaving behind those replicable genetic packages that are bound to the immobilized target. Alternatively, the target can be immobilized on a bead and the uncleared cell culture can be passed through a filter with a pore size smaller than the bead, but larger than a bacterial cell (or a eukaryotic cell when using eukaryotic host cells). Another separation method is the immobilization of a target on a paramagnetic bead, and the decantation of the uncleared cell culture leaving the replicable genetic package-target molecule complex behind bound to the paramagnetic bead held in place with a magnetic field. [0048]
  • If a target is used that is free in solution, any resulting replicable genetic package-target molecule complex(es) can be subsequently separated from the uncleared cell culture. For example, the replicable genetic package-target complex can be incubated in the presence of a third molecule, a target complex-binding molecule, that is immobilized on a solid support and does not disrupt the replicable genetic package-target molecule complex(es). The target complex-binding molecule can bind to either the soluble target molecule or to the replicable genetic package. This permits the replicable genetic package-target complex to bind to the target-binding molecule, thereby indirectly immobilizing the replicable genetic package-target complex. The uncleared cell culture can then be separated from the replicable genetic packages that bind specifically to the target molecule using the separation methods described above for the first category of replicable genetic package-target complexes. [0049]
  • In preferred embodiments, at least 70% of the cells are separated from the replicable genetic packages that are specifically bound to the target molecule, more preferably, at least 80% of the cells are separated from the replicable genetic packages that are specifically bound to the target molecule, still more preferably, at least 90% of the cells are separated from the replicable genetic packages that are specifically bound to the target molecule, yet still more preferably, substantially all of the bacterial cells are separated from the replicable genetic packages that are specifically bound to the target molecule. [0050]
  • It is sometimes desirable to wash the replicable genetic package-target complex. The wash can remove undesirable components of the cell cultures from the specifically bound replicable genetic packages. The wash can remove cells, non-specifically bound replicable genetic packages, etc. Often, a wash buffer is used. The wash buffer can contain a detergent, or other agents, and compositions that are compatible with replicable genetic package-target binding to increase the stringency of the screening process. For example, a wash buffer that can be used in the present invention is a solution of Tris buffered saline with 0.05% Tween-20, pH 7.4 (TBST). [0051]
  • For some applications, it is desirable to elute the replicable genetic packages that specifically bind to the target molecule. The replicable genetic packages can then be used for, for example, further rounds of screening, amplification, detection, or characterization (e.g., nucleic acid sequencing). Elution can be accomplished using a variety of methods known in the art. The replicable genetic packages can be eluted using pH changes, protein denaturants, or EGTA/EDTA if a metal ion is necessary for replicable genetic package-target interaction (See e.g., Sparks et al. (1996) Ch. 13, supra for elution techniques using phage-display). For example, the replicable genetic packages can be eluted using an acidic buffer (e.g., glycine-HCl, pH 2) (see, e.g., Sparks et al. (1996) Ch. 13, supra). The eluate can then be removed and neutralized with the addition of a second buffer (e.g., NaPO[0052] 4 buffer pH 7.5) (see, e.g., Sparks et al. (1996) Ch. 13, supra). Alternatively, natural or synthetic ligands that interrupt the replicable genetic package-target complex can be used to elute the replicable genetic package from the target (Sparks et al. (1996) Ch. 13, supra).
  • If desired, the replicable genetic package(s) can be amplified in order to increase the number of replicable genetic package, thus potentially increasing the chance that enough of the replicable genetic package(s) will be present in the next round for isolation, identification, or detection, etc. Methods for amplifying replicable genetic packages in solid and liquid culture are known in the art (see, e.g., Sparks (1996), Ch. 13, supra, and Rider et al. (1996) Ch. 4, supra, for methods of amplifying filamentous bacteriophage). [0053]
  • Those of skill in the art will recognize that screening methods of the present invention can be optimized. Furthermore, skilled artisans will recognize methods of optimizing to determine the effectiveness of steps and to increase the chances of identifying the replicable genetic package of interest. For example, the inclusion of positive and negative controls in the screening process can facilitate the trouble-shooting and/or optimization of a screening process. [0054]
  • V. Detection and Characterization of Replicable Genetic Packages [0055]
  • Another aspect of the present invention is that the presence of cells and other components of an uncleared culture do not interfere with the detection of particles that specifically bind to a target molecule. Accordingly, some methods of the present invention involve contacting the particle-target molecule complex with a detection reagent prior to removing the cells and/or other components of the uncleared cell culture. [0056]
  • The presence of replicable genetic packages that bind to a target molecule can be detected using a variety of materials and methods known to those of skill in the art. For example, the replicable genetic package-target complexes can be incubated with a detection reagent. Typically, a detection reagent is labeled with a substance that permits the qualitative or quantitative determination of the presence or absence of the replicable genetic package-target complex. The term “labeled” refers to a composition is that is detectable, either directly or indirectly, by spectroscopic, photochemical, biochemical, immunochemical, or chemical means. For example, useful labels include radiolabels (e.g., H[0057] 3, C13, C14, P32, S35, I125), fluorescent dyes, fluorophores, electrondense reagents, enzymes and their substrates (e.g., as commonly used in enzyme-linked immunoassays, e.g., alkaline phosphatase and horse radish peroxidase), biotin-streptavidin, digoxigenin, or haptens and proteins for which antisera or monoclonal antibodies are available. The label or detectable moiety is typically bound, either covalently, through a linker or chemical bound, or through ionic, van der Waals or hydrogen bonds to the molecule to be detected.
  • Detection reagents can include antibodies—such as antibodies that react with the native form of the phage being used, e.g., anti-M13KO7 antibody. The antibody itself can be labeled. For example, horseradish peroxidase (HRP) can be conjugated to an anti-M13 antibody (Amersham-Pharmacia Biotech, Piscataway, N.J.). The absorbance of the reaction produce of HRP and o-Phenylenediamine Dihydrochloride (OPD; Sigma, St. Louis Mo.) can be monitored with a 490 nm filter (Biorad, Hercules Calif.) after stopping the reaction with acid. [0058]
  • The detection of the replicable genetic package-target complex on the chip could be analyzed using a physical spectroscopy method, such as mass spectroscopy or surface plasmon resonance (U.S. Pat. No. 5,641,640). Surface plasmon-resonance has been used to detect phage-displayed antibody-target interactions (de Haard et al., (1999)). Chips and surface plasmon resonance instruments are commercially available (e.g., BIACORE, Uppsala, Sweden) for the detection of analytes. [0059]
  • Fluorescence polarization could also be employed by modifying the target molecule with an appropriate fluorescence label or fluorophore (Burke et al. (1996) Chapter 18[0060] , Measurement of Peptide Binding Affinities Using Fluorescence Polarization, in Kay et al., eds. (1996)).
  • The replicable genetic package that are bound to a target can be further characterized as to the genetic or protein makeup of their potential binding polypeptide(s). In some embodiments, the nucleic acid sequence of the potential binding polypeptide can be determined by sequencing the phagemid vector contained in a particular phage (see e.g., Masecar et al. (1996) Chapter 17[0061] , Nonradioactive Sequencing of Random Peptide Recombinant Phage, in Kay et al., eds. (1996)). The protein makeup of a phage could be determined using methods known in the art, such as immunological assays (e.g., Western blots), two-dimensional gels, mass spectrometry, etc.
  • VI. High-Throughput Screening on an Automated Workstation. [0062]
  • In the present invention, high-throughput analysis and screening of replicable genetic package-display libraries can be performed on a automated workstation (see e.g., U.S. Pat. No. 5,139,744, “Automated laboratory workstation having module identification means.”). An “automated workstation” is typically a computer-controlled apparatus which can, through robotic functions, transfer, mix, and remove liquids from microtiter plates. An automated workstation can also contain a built-in plate reader, which can read the absorbance of a liquid in a microtiter well. The automated workstation can be programmed to carry out a series of mixing, transfer, and/or removal steps. The automated workstation will typically have a multi-channel pipettor which can pipette small amounts of liquid (e.g., microliter amounts) from a vessel to the well. [0063]
  • For example, in some embodiments of the present invention, the automated workstation can transfer uncleared cell culture(s) into a micro-titer plate. The microtiter plate can have pre-immobilized target molecule(s) already in the wells. The automated workstation can subsequently be used to remove uncleared cell cultures from the wells, wash the wells, or elute the replicable genetic packages from the immobilized target. Detection of a replicable genetic package bound to an immobilized target molecule can also be carried out using an automated workstation. The automated workstation can be used to add a detection reagent to the wells. The automated workstation, when equipped with a plate reader, can monitor the absorbance of the reaction of the detection reagent in the wells. [0064]
  • EXAMPLES
  • The following examples are offered to illustrate, but not to limit the present invention. [0065]
  • Example 1 [0066]
  • Comparison of Detection of Phage from Uncleared Bacterial Cultures and Cleared Bacterial Cultures
  • This Example describes experiments in which alternatives to centrifugation or filtration prior to the screening of a phage-display library were explored. Phage displaying scFvs or Fabs were generated by PCR amplification of cDNA corresponding to the heavy and light chain variable regions from the HP6002, HP6025, and HP6054 hybridomas (Reimer et al. (1984) [0067] Hybridoma 3: 263-275) (cells obtained from ATCC Manassas, Va.; CRL-1788, CRL-1775 and CRL-1763 respectively). The regions were amplified using the primers (SEQ ID NOS: 1-84) set out in Table 1:
    TABLE 1
    Primer sequences for ScFv
    and Fab library generation
    Name Mer Sequence
    MCH1-G1R 48 ATTGGCGCGCCTTATTAACAATCCCTGG
    GCACAATTTTCTTGTCCACC
    MCH1-G2A 44 ATTGGCGCGCCTTATTAACAGGGCTTGA
    TTGTGGGCCCTCTGGG
    MCH1-G2B 45 ATTGGCGCGCCTTATTAACAGGGGTTGA
    TTGTTGAAATGGGCCCG
    MHV-Back1 50 TTATTACTCGCGGCCCAGCCGGCCATGG
    CCGATGTGAAGCTTCAGGAGTC
    MHV-Back2 50 TTATTACTCGCGGCCCAGCCGGCCATGG
    CCCAGGTGCAGCTGAAGGAGTC
    MHV-Back3 50 TTATTACTCGCGGCCCAGCCGGCCATGG
    CCCAGGTGCAGCTGAAGCAGTC
    MHV-Back4 50 TTATTACTCGCGGCCCAGCCGGCCATGG
    CCCAGGTTACTCTGAAAGAGTC
    MHV-Back5 51 TTATTACTCGCGGCCCAGCCGGCCATGG
    CCGAGGTCCAGCTGCAACAATCT
    MHV-Back6 50 TTATTACTCGCGGCCCAGCCGGCCATGG
    CCGAGGTCCAGCTGCAGCAGTC
    MHV-Back7 51 TTATTACTCGCGGCCCAGCCGGCCATGG
    CCCAGGTCCAACTGCAGCAGCCT
    MHV-Back8 50 TTATTACTCGCGGCCCAGCCGGCCATGG
    CCGAGGTGAAGCTGGTGGAGTC
    MHV-Back9 50 TTATTACTCGCGGCCCAGCCGGCCATGG
    CCGAGGTGAAGCTGGTGGAATC
    MHV-Back10 50 TTATTACTCGCGGCCCAGCCGGCCATGG
    CCGATGTGAACTTGGAAGTGTC
    MHV-For1 33 ACCTGGCGCGCCTGCAGAGACAGTGACC
    AGAGT
    MHV-for1b 42 ACCGCCTCCACCTGGCGCGCCTGCAGAG
    ACAGTGACCAGAGT
    MHV-For2 33 ACCTGGCGCGCCTGAGGAGACTGTGAGA
    GTGGT
    MHV-for2b 42 ACCGCCTCCACCTGGCGCGCCTGAGGAG
    ACTGTGAGAGTGGT
    MHV-For3 33 ACCTGGCGCGCCTGAGGAGACGGTGACT
    GAGGT
    MHV-for3b 42 ACCGCCTCCACCTGGCGCGCCTGAGGAG
    ACGGTGACTGAGGT
    MHV-For4 33 ACCTGGCGCGCCTGAGGAGACGGTGACC
    GTGGT
    MHV-for4b 42 ACCGCCTCCACCTGGCGCGCCTGAGGAG
    ACGGTGACCGTGGT
    MKV-back1 39 TCTGGCGGTGGCGGATCGGATGTTTTGA
    TGACCCAAACT
    MKV-Back2 39 TCTGGCGGTGGCGGATCGGATATTGTGA
    TGACGCAGGCT
    MKV-Back3 36 TCTGGCGGTGGCGGATCGGATATTGTGA
    TAACCCAG
    MKV-Back4 39 TCTGGCGGTGGCGGATCGGACATTGTGC
    TGACCCAATCT
    MKV-Back5 39 TCTGGCGGTGGCGGATCGGACATTGTGA
    TGACCCAGTCT
    MKV-Back6 39 TCTGGCGGTGGCGGATCGGATATTGTGC
    TAACTCAGTCT
    MKV-Back7 39 TCTGGCGGTGGCGGATCGGATATCCAGA
    TGACACAGACT
    MKV-Back8 39 TCTGGCGGTGGCGGATCGGACATCCAGC
    TGACTCAGTCT
    MKV-Back9 39 TCTGGCGGTGGCGGATCGCAAATTGTTC
    TCACCCAGTCT
    MKV-For1 38 GATGGTGATGTGCGGCCGCCCGTTTCAG
    CTCCAGCTTG
    MKV-For2 40 GATGGTGATGTGCGGCCGCCCGTTTTAT
    TTCCAGCTTGGT
    MKV-For3 39 GATGGTGATGTGCGGCCGCCCGTTTTAT
    TTCCAACTTTG
    MKV-For4 40 GATGGTGATGTGCGGCCGCGGATACAGT
    TGGTGCAGCATC
    MVH1 55 CCTTTCTATGCGGCCCAGCCGGCCATGG
    CCGAGGTRMAGCTTCAGGAGTCAGGAC
    MVH2 55 CCTTTCTATGCGGCCCAGCCGGCCATGG
    CCGAGGTSCAGCTKCAGCAGTCAGGAC
    MVH3 53 CCTTTCTATGCGGCCCAGCCGGCCATGG
    CCCAGGTGCAGCTGAAGSASTCAGG
    MVH4 55 CCTTTCTATGCGGCCCAGCCGGCCATGG
    CCGAGCTGCAGCTTCAGGAGTCSGGAC
    MVH5 55 CCTTTCTATGCGGCCCAGCCGGCCATGG
    CCGARGTCCAGCTGCAACAGTCYGGAC
    MVH6 53 CCTTTCTATGCGGCCCAGCCGGCCATGG
    CCCAGGTCCAGCTKCAGCAATCTGG
    MVH7 53 CCTTTCTATGCGGCCCAGCCGGCCATGG
    CCCAGSTBCAGCTGCAGCAGTCTGG
    MVH8 55 CCTTTCTATGCGGCCCAGCCGGCCATGG
    CCCAGGTYCAGCTGCAGCAGTCTGGRC
    MVH9 53 CCTTTCTATGCGGCCCAGCCGGCCATGG
    CCGAGGTYCAGCTYCAGCAGTCTGG
    MVH10 56 CCTTTCTATGCGGCCCAGCCGGCCATGG
    CCGAGGTCCARCTGCAACAATCTGGACC
    MVH11 54 CCTTTCTATGCGGCCCAGCCGGCCATGG
    CCCAGGTCCACGTGAAGCAGTCTGGG
    MVH12 52 CCTTTCTATGCGGCCCAGCCGGCCATGG
    CCGAGGTGAASSTGGTGGAATCTG
    MVH13 52 CCTTTCTATGCGGCCCAGCCGGCCATGG
    CCGAVGTGAAGYTGGTGGAGTCTG
    MVH14 55 CCTTTCTATGCGGCCCAGCCGGCCATGG
    CCGAGGTGCAGSKGGTGGAGTCTGGGG
    MVH15 54 CCTTTCTATGCGGCCCAGCCGGCCATGG
    CCGAKGTGCAMCTGGTGCAGTCTGGG
    MVH16 53 CCTTTCTATGCGGCCCAGCCGGCCATGG
    CCGAGGTGAAGCTGATGGARTCTGG
    MVH17 55 CCTTTCTATGCGGCCCAGCCGGCCATGG
    CCGAGGTGCARCTTGTTGAGTCTGGTG
    MVH18 54 CCTTTCTATGCGGCCCAGCCGGCCATGG
    CCGARGTRAAGCTTCTCGAGTCTGGA
    MVH19 53 CCTTTCTATGCGGCCCAGCCGGCCATGG
    CCGAAGTGAARSTTGAGGAGTCTGG
    MVH20 54 CCTTTCTATGCGGCCCAGCCGGCCATGG
    CCGAAGTGATGCTGGTGGAGTCTGGG
    MVH21 55 CCTTTCTATGCGGCCCAGCCGGCCATGG
    CCCAGGTTACTCTRAAAGWGTSTGGCC
    MVH22 53 CCTTTCTATGCGGCCCAGCCGGCCATGG
    CCCAGGTCCAACTVCAGCARCCTGG
    MVH23 52 CCTTTCTATGCGGCCCAGCCGGCCATGG
    CCCAGGTYCARCTGCAGCAGTCTG
    MVH24 53 CCTTTCTATGCGGCCCAGCCGGCCATGG
    CCGATGTGAACTTGGAAGTGTCTGG
    MVH25 53 CCTTTCTATGCGGCCCAGCCGGCCATGG
    CCGAGGTGAAGGTCATCGAGTCTGG
    MVK1 38 TTACTCCGGTCCGCGGACATTGTTCTCA
    CCCAGTCTCC
    MVK2 38 TTACTCCGGTCCGCGGACATTGTGCTSA
    CCCAGTCTCC
    MVK3 38 TTACTCCGGTCCGCGGACATTGTGATGA
    CTCAGTCTCC
    MVK4 38 TTACTCCGGTCCGCGGACATTGTGCTMA
    CTCAGTCTCC
    MVK5 38 TTACTCCGGTCCGCGGACATTGTGYTRA
    CACAGTCTCC
    MVK6 38 TTACTCCGGTCCGCGGACATTGTRATGA
    CACAGTCTCC
    MVK7 38 TTACTCCGGTCCGCGGACATTMAGATRA
    CCCAGTCTCC
    MVK8 38 TTACTCCGGTCCGCGGACATTCAGATGA
    MCCAGTCTCC
    MVK9 38 TTACTCCGGTCCGCGGACATTCAGATGA
    CDCAGTCTCC
    MVK10 38 TTACTCCGGTCCGCGGACATTCAGATGA
    CACAGACTAC
    MVK11 38 TTACTCCGGTCCGCGGACATTCAGATCA
    TTCAGTCTCC
    MVK12 38 TTACTCCGGTCCGCGGACATTGTTCTCA
    WCCAGTCTCC
    MVK13 38 TTACTCCGGTCCGCGGACATTGTTCTCT
    CCCAGTCTCC
    MVK14 38 TTACTCCGGTCCGCGGACATTGWGCTSA
    CCCAATCTCC
    MVK15 37 TTACTCCGGTCCGCGGACATTSTGATGA
    CCCARTCTC
    MVK16 38 TTACTCCGGTCCGCGGACATTKTGATGA
    CCCARACTCC
    MVK17 38 TTACTCCGGTCCGCGGACATTGTGATGA
    CTCAGGCTAC
    MVK18 38 TTACTCCGGTCCGCGGACATTGTGATGA
    CBCAGGCTGC
    MVK19 37 TTACTCCGGTCCGCGGACATTGTGATAA
    CYCAGGATG
    MVK20 38 TTACTCCGGTCCGCGGACATTGTGATGA
    CCCAGTTTCG
    MVK21 38 TTACTCCGGTCCGCGGACATTGTGATGA
    CACAACCTGC
    MVK22 38 TTACTCCGGTCCGCGGACATTTTGCTGA
    CTCAGTCTCC
    MVK23 38 TTACTCCGGTCCGCGGACATTTTGCTGA
    CTCAGTCTCC
    MVK24 38 TTACTCCGGTCCGCGGACATTGTAATGA
    CCCAATCTCC
    MVK25 38 TTACTCCGGTCCGCGGACATTGTGATGA
    CCCACACTCC
  • Assembled scFv or Fab DNA sequences were digested with SfiI and NotI, subcloned into the pCANTAB5E vector (Amersham-Pharmacia Biotech, Piscataway, N.J.), and transformed into TG1 or XL1-Blue competent [0068] E.coli. Individual clones capable of specific binding to the target antigen were isolated by conventional methods and then used to explore alternatives to centrifugation. Single colonies were picked into 0.1 ml cultures (2×YT supplemented with 2% glucose and 100 μg/ml Ampicillin) in a deep well 96-well plate and incubated at 37° C. with shaking for 5-6 hours when cultures reached mid-log phase. Cultures received M13KO7 helper phage (˜1×109 pfu in 5 μl) and were incubated for 1 hour at 37° C. with shaking. A 50 μl aliquot was removed to a duplicate deep well plate and 1 ml of media (2×YT supplemented with 100 μg/ml Ampicillin, 50 μg/ml Kanamycin with or without 1 mM IPTG) was added to wells for overnight growth at 30° C. Polystyrene plates were coated with protein antigens (hIgG1κ, hIgG1λ, or rabbit IgG) (1-10 μg/ml in 0.1 M sodium bicarbonate pH 9.6) overnight at 4° C., blocked with 3% non-fat dry milk (NFM) in Tris buffered saline with 0.05% Tween-20 (pH 7.4, TBST), and washed 3× in TBST. Aliquots of bacterial culture were removed to a separate microtiter plate, or to wells in a 96-well filtration plate (MultiScreen plates from Millipore, Bedford Mass.), a vacuum was applied slowly, and filtrate collected in a microtiter plate. The remainder of the culture in the deep well plates was centrifuged at 1,725×g (3,500 rpm in an Eppendorf 5804 equipped with an A2MTP rotor) for 30 minutes at room temperature (RT).
  • Aliquots (80 μl), of clarified phage supernatant, filtrate, or uncleared bacterial culture were added to the ELISA plate and mixed with 20 μl of 10% NFM/5×PBS directly in the wells. Plates were incubated without shaking for 1.5 hours at 37° C., then washed 4× with TBST (using a Wellwash 4 MK2 platewasher, Labsystems). [0069]
  • Horseradish peroxidase (HRP)-conjugated anti-M13 antibody(Amersham-Pharmacia Biotech, Piscataway, N.J.) was diluted 1:5000 into 3% NFM/TBST and incubated in wells for 1 hour at 37° C. Following 4 washes with TBST, 100 μl of o-Phenylenediamine Dihydrochloride (OPD; Sigma, St. Louis Mo.) substrate was added to wells for approximately 5 minutes prior to stopping the reaction with 25 μl 3N HCl. Plates were read on a microplate reader with a 490 nm filter (Biorad, Hercules Calif.). Assays were performed in duplicate and repeated 2 or 3 times with similar results. [0070]
  • The scFv display phage derived from HP6054 bind to the human lambda light chain antigen (in association with IgG1), but not to the kappa light chain (hIgG1 Kappa) or to Rabbit IgG (FIG. 1A). Filtrate generated from the same culture yields an equivalent level of binding as observed for the supernatant. The uncleared bacterial culture demonstrated similar levels of binding to the immobilized antigen, indicating that removal of bacteria by time consuming centrifugation or costly filtration is not necessary. Furthermore, no increase in binding was observed to either of the two non-specific antigens tested, hIgG1κ and rabbit IgG. [0071]
  • Similar results were observed for phage displaying a Fab also derived from HP6054 (FIG. 1B). Addition of IPTG adversely affects the binding of HP6054 scFv-phage (due to reduced bacterial growth and phage production) and increases the binding of HP6054 Fab-phage (due to increased Fab:gIII fusion production). Although the level of IPTG did affect the overall binding of the phage populations, there were no significant differences in levels of binding observed when ELISA was performed directly on bacteria containing cultures, or cultures that were clarified by centrifugation or filtration. [0072]
  • Example 2 Sensitivity of Phase ELISA is not Impaired by the Presence of Bacteria
  • In some applications, assay of a polyclonal population of phage producing bacteria would be necessary, e.g, library screening. For example, following several rounds of selection, one might wish to test the population by ELISA to verify that binding members have been selected and are present in the population. To demonstrate that bacteria remaining during the ELISA would not present a problem when low levels of binding were expected, we grew independent cultures prior to mixing them at various ratios and then performed the ELISA on the mixed cultures or centrifuge-clarified culture supernatants derived from the same mixtures. A mixture of cultures from two clones was used as a model system to simulate a polyclonal culture. Growth of a polyclonal culture involves competition between individuals, which affects the yield of specific phage. However, the bias that is introduced during polyclonal growth would exist regardless of the means of analysis of that culture. [0073]
  • [0074] E.coli carrying phagemid expressing scFv derived from HP6002 (recognizing hIgG2) or HP6025 (recognizing hIgG4) were grown overnight in the absence of IPTG. Cultures that attained different densities (OD600 for HP6002 was 1.2 and 1.0, and HP6025 was 2.1 and 3.3 in two separate trials) were mixed on the basis of volume. Final volume ratios ranged from a 0.001 to 1000 of HP6025/HP6002. Aliquots of the mixed culture were compared to supernatants clarified by centrifugation in the phage ELISA. FIG. 2 demonstrates that clarified phage supernatant and bacterial culture do not exhibit significant differences in binding at any of the ratios tested. Therefore, sensitivity of the ELISA does not appear to be compromised by the presence of bacteria during the binding of the phage an immobilized antigen.
  • Our results demonstrate the phage ELISAs can be performed directly on bacterial culture and that there is no need to clarify by centrifugation or filtration. We have successfully used culture from scFv (5 different antibodies) and Fab (2 different antibodies) display-phage in our ELISA. [0075]
  • Additionally, we have used this method for analysis of both peptides and proteins displayed on the major coat protein (gene VIII protein) of filamentous phage. To date, no significant differences between culture and clarified supernatant have been observed for any display agents or antigen tested by this method. [0076]
  • Furthermore, this procedure has worked well with two common [0077] E.coli strains (TG1 cells and XL1-Blue) and overnight cultures of various densities (OD600 from 0.1 to 3.3).
  • It is understood that the examples and embodiments described herein are for illustrative purposes only and that various modifications or changes in light thereof will be suggested to persons skilled in the art and are to be included within the spirit and purview of this application and scope of the appended claims. All publications, patents, and patent applications cited herein are hereby incorporated by reference for all purposes. [0078]
  • 1 84 1 48 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MCH1G1R 1 attggcgcgc cttattaaca atccctgggc acaattttct tgtccacc 48 2 44 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MCH1-G2A 2 attggcgcgc cttattaaca gggcttgatt gtgggccctc tggg 44 3 45 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)-MCH1-G2B 3 attggcgcgc cttattaaca ggggttgatt gttgaaatgg gcccg 45 4 50 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MHV-Back2 4 ttattactcg cggcccagcc ggccatggcc gatgtgaagc ttcaggagtc 50 5 50 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MHC-Back2 5 ttattactcg cggcccagcc ggccatggcc caggtgcagc tgaaggagtc 50 6 50 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MHC-Back3 6 ttattactcg cggcccagcc ggccatggcc caggtgcagc tgaagcagtc 50 7 50 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MHV-Back4 7 ttattactcg cggcccagcc ggccatggcc caggttactc tgaaagagtc 50 8 51 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MHV-Back5 8 ttattactcg cggcccagcc ggccatggcc gaggtccagc tgcaacaatc t 51 9 50 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MHV-Back6 9 ttattactcg cggcccagcc ggccatggcc gaggtccagc tgcagcagtc 50 10 51 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MHV-Back7 10 ttattactcg cggcccagcc ggccatggcc caggtccaac tgcagcagcc t 51 11 50 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MHV-Back8 11 ttattactcg cggcccagcc ggccatggcc gaggtgaagc tggtggagtc 50 12 50 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MHV-Back9 12 ttattactcg cggcccagcc ggccatggcc gaggtgaagc tggtggaatc 50 13 50 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MHV-Back10 13 ttattactcg cggcccagcc ggccatggcc gatgtgaact tggaagtgtc 50 14 33 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MHV-For1 14 acctggcgcg cctgcagaga cagtgaccag agt 33 15 42 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MHV-For1b 15 accgcctcca cctggcgcgc ctgcagagac agtgaccaga gt 42 16 33 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MHV-For2 16 acctggcgcg cctgaggaga ctgtgagagt ggt 33 17 42 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MHV-for2b 17 accgcctcca cctggcgcgc ctgaggagac tgtgagagtg gt 42 18 33 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MHV-For3 18 acctggcgcg cctgaggaga cggtgactga ggt 33 19 42 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MHV-for3b 19 accgcctcca cctggcgcgc ctgaggagac ggtgactgag gt 42 20 33 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MHV-For4 20 acctggcgcg cctgaggaga cggtgaccgt ggt 33 21 42 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MHV-for4b 21 accgcctcca cctggcgcgc ctgaggagac ggtgaccgtg gt 42 22 39 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MKV-back1 22 tctggcggtg gcggatcgga tgttttgatg acccaaact 39 23 39 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MKV-Back2 23 tctggcggtg gcggatcgga tattgtgatg acgcaggct 39 24 36 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MKV-Back3 24 tctggcggtg gcggatcgga tattgtgata acccag 36 25 39 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MKV-Back4 25 tctggcggtg gcggatcgga cattgtgctg acccaatct 39 26 39 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MKV-Back5 26 tctggcggtg gcggatcgga cattgtgatg acccagtct 39 27 39 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MKV-Back6 27 tctggcggtg gcggatcgga tattgtgcta actcagtct 39 28 39 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MKV-Back7 28 tctggcggtg gcggatcgga tatccagatg acacagact 39 29 39 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MKV-Back8 29 tctggcggtg gcggatcgga catccagctg actcagtct 39 30 39 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MKV-Back9 30 tctggcggtg gcggatcgca aattgttctc acccagtct 39 31 38 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MKV-For1 31 gatggtgatg tgcggccgcc cgtttcagct ccagcttg 38 32 40 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MKV-For2 32 gatggtgatg tgcggccgcc cgttttattt ccagcttggt 40 33 39 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MKV-For3 33 gatggtgatg tgcggccgcc cgttttattt ccaactttg 39 34 40 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)-MKV-For4 34 gatggtgatg tgcggccgcg gatacagttg gtgcagcatc 40 35 55 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MVH1 35 cctttctatg cggcccagcc ggccatggcc gaggtrmagc ttcaggagtc aggac 55 36 55 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MVH2 36 cctttctatg cggcccagcc ggccatggcc gaggtscagc tkcagcagtc aggac 55 37 53 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MVH3 37 cctttctatg cggcccagcc ggccatggcc caggtgcagc tgaagsastc agg 53 38 55 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MVH4 38 cctttctatg cggcccagcc ggccatggcc gaggtgcagc ttcaggagtc sggac 55 39 55 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MVH5 39 cctttctatg cggcccagcc ggccatggcc gargtccagc tgcaacagtc yggac 55 40 53 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MVH6 40 cctttctatg cggcccagcc ggccatggcc caggtccagc tkcagcaatc tgg 53 41 53 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MVH7 41 cctttctatg cggcccagcc ggccatggcc cagstbcagc tgcagcagtc tgg 53 42 55 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MVH8 42 cctttctatg cggcccagcc ggccatggcc caggtycagc tgcagcagtc tggrc 55 43 53 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MHV9 43 cctttctatg cggcccagcc ggccatggcc gaggtycagc tycagcagtc tgg 53 44 56 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MHV10 44 cctttctatg cggcccagcc ggccatggcc gaggtccarc tgcaacaatc tggacc 56 45 54 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MHV11 45 cctttctatg cggcccagcc ggccatggcc caggtccacg tgaagcagtc tggg 54 46 52 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MHV12 46 cctttctatg cggcccagcc ggccatggcc gaggtgaass tggtggaatc tg 52 47 52 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MHV13 47 cctttctatg cggcccagcc ggccatggcc gavgtgaagy tggtggagtc tg 52 48 55 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MHV14 48 cctttctatg cggcccagcc ggccatggcc gaggtgcags kggtggagtc tgggg 55 49 54 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MVH15 49 cctttctatg cggcccagcc ggccatggcc gakgtgcamc tggtgcagtc tggg 54 50 53 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MVH16 50 cctttctatg cggcccagcc ggccatggcc gaggtgaagc tgatggartc tgg 53 51 55 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MVH17 51 cctttctatg cggcccagcc ggccatggcc gaggtgcarc ttgttgagtc tggtg 55 52 54 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MVH18 52 cctttctatg cggcccagcc ggccatggcc gargtraagc ttctcgagtc tgga 54 53 53 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)0 MVH19 53 cctttctatg cggcccagcc ggccatggcc gaagtgaars ttgaggagtc tgg 53 54 54 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MVH20 54 cctttctatg cggcccagcc ggccatggcc gaagtgatgc tggtggagtc tggg 54 55 55 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MVH21 55 cctttctatg cggcccagcc ggccatggcc caggttactc traaagwgts tggcc 55 56 53 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MVH22 56 cctttctatg cggcccagcc ggccatggcc caggtccaac tvcagcarcc tgg 53 57 52 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MVH23 57 cctttctatg cggcccagcc ggccatggcc caggtycarc tgcagcagtc tg 52 58 53 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)-MVH24 58 cctttctatg cggcccagcc ggccatggcc gatgtgaact tggaagtgtc tgg 53 59 53 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MVH25 59 cctttctatg cggcccagcc ggccatggcc gaggtgaagg tcatcgagtc tgg 53 60 38 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MVK1 60 ttactccggt ccgcggacat tgttctcacc cagtctcc 38 61 38 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MVK2 61 ttactccggt ccgcggacat tgtgctsacc cagtctcc 38 62 38 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MVK3 62 ttactccggt ccgcggacat tgtgatgact cagtctcc 38 63 38 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)MVK4 63 ttactccggt ccgcggacat tgtgctmact cagtctcc 38 64 38 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MVK5 64 ttactccggt ccgcggacat tgtgytraca cagtctcc 38 65 38 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)-MVK6 65 ttactccggt ccgcggacat tgtratgaca cagtctcc 38 66 38 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)-MVK7 66 ttactccggt ccgcggacat tmagatracc cagtctcc 38 67 38 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)-MVK8 67 ttactccggt ccgcggacat tcagatgamc cagtctcc 38 68 38 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MVK9 68 ttactccggt ccgcggacat tcagatgacd cagtctcc 38 69 38 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MVK10 69 ttactccggt ccgcggacat tcagatgaca cagactac 38 70 38 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MVK11 70 ttactccggt ccgcggacat tcagatcatt cagtctcc 38 71 38 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MVK12 71 ttactccggt ccgcggacat tgttctcawc cagtctcc 38 72 38 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MVK13 72 ttactccggt ccgcggacat tgttctctcc cagtctcc 38 73 38 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MVK14 73 ttactccggt ccgcggacat tgwgctsacc caatctcc 38 74 36 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MVK15 74 ttactccggt ccgcggacat ttgatgaccc artctc 36 75 38 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MVK16 75 ttactccggt ccgcggacat tktgatgacc caractcc 38 76 38 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MVK17 76 ttactccggt ccgcggacat tgtgatgact caggctac 38 77 38 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MVK18 77 ttactccggt ccgcggacat tgtgatgacb caggctgc 38 78 37 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MVK19 78 ttactccggt ccgcggacat tgtgataacy caggatg 37 79 38 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MVK20 79 ttactccggt ccgcggacat tgtgatgacc cagtttcg 38 80 38 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MVK21 80 ttactccggt ccgcggacat tgtgatgaca caacctgc 38 81 38 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MVK22 81 ttactccggt ccgcggacat tttgctgact cagtctcc 38 82 38 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MVK23 82 ttactccggt ccgcggacat tttgctgact cagtctcc 38 83 38 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MVK24 83 ttactccggt ccgcggacat tgtaatgacc caatctcc 38 84 38 DNA Artificial Sequence Primers for ScFv and Fab library generation (Table I)- MVK25 84 ttactccggt ccgcggacat tgtgatgacc cacactcc 38

Claims (9)

1-19. (canceled)
20. An uncleared cell culture comprising:
(a) a population of replicable genetic packages, each of which displays on its surface a fusion protein that comprises a surface-displayed replicable genetic package polypeptide and an exogenous polypeptide;
(b) a complex that comprises a target molecule and one or more members of the population of replicable genetic packages that specifically bind to said target molecule via said exogenous polypeptide; and
(c) cells in which the replicable genetic packages were amplified prior to contact with the target molecule.
21. The uncleared cell culture of claim 20, wherein said replicable genetic packages are selected from the group consisting of bacteriophage and eukaryotic viruses.
22. The uncleared cell culture of claim 20, wherein said target molecule is immobilized on a solid support.
23. The uncleared cell culture of claim 22, wherein said solid support is selected from the group consisting of: a bead, a chip, a microtiter plate, a prokaryotic cell and a eukaryotic cell.
24. The uncleared cell culture of claim 20, wherein said target molecule is selected from the group consisting of: a polypeptide, a nucleic acid, an RNA, a DNA, a small organic molecule, and a carbohydrate.
25. The uncleared cell culture of claim 20, wherein said exogenous polypeptide is an antibody.
26. The uncleared cell culture of claim 25, wherein said antibody is a scFv or a Fab.
27. The uncleared cell culture of claim 20, wherein the uncleared cell culture further comprises a detection reagent that specifically binds to the replicable genetic packages.
US10/727,745 2000-06-05 2003-12-03 Screening of phage displayed peptides without clearing of the cell culture Abandoned US20040253607A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US10/727,745 US20040253607A1 (en) 2000-06-05 2003-12-03 Screening of phage displayed peptides without clearing of the cell culture

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US20950300P 2000-06-05 2000-06-05
US09/874,547 US6686154B2 (en) 2000-06-05 2001-06-04 Screening of phage displayed peptides without clearing of the cell culture
WOPCT/US01/18421 2001-06-05
PCT/US2001/018421 WO2001094950A2 (en) 2000-06-05 2001-06-05 Screening of phage displayed peptides without clearing of the cell culture
US10/727,745 US20040253607A1 (en) 2000-06-05 2003-12-03 Screening of phage displayed peptides without clearing of the cell culture

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/874,547 Division US6686154B2 (en) 2000-06-05 2001-06-04 Screening of phage displayed peptides without clearing of the cell culture

Publications (1)

Publication Number Publication Date
US20040253607A1 true US20040253607A1 (en) 2004-12-16

Family

ID=26904223

Family Applications (2)

Application Number Title Priority Date Filing Date
US09/874,547 Expired - Fee Related US6686154B2 (en) 2000-06-05 2001-06-04 Screening of phage displayed peptides without clearing of the cell culture
US10/727,745 Abandoned US20040253607A1 (en) 2000-06-05 2003-12-03 Screening of phage displayed peptides without clearing of the cell culture

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US09/874,547 Expired - Fee Related US6686154B2 (en) 2000-06-05 2001-06-04 Screening of phage displayed peptides without clearing of the cell culture

Country Status (3)

Country Link
US (2) US6686154B2 (en)
AU (1) AU2001268225A1 (en)
WO (1) WO2001094950A2 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090203536A1 (en) * 2006-06-06 2009-08-13 Vermette Patrick Assay supports comprising a peg support, said support attached from a peg solution in cloud point (theta solvent) conditions

Families Citing this family (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7179638B2 (en) * 1999-07-30 2007-02-20 Large Scale Biology Corporation Microarrays and their manufacture by slicing
EP1301632A2 (en) * 2000-07-19 2003-04-16 Pointilliste, Inc. Nested sorting and high throughput screening
EP1343914A2 (en) * 2000-12-11 2003-09-17 HK Pharmaceuticals, Inc. Multiplexed protein expression and activity assay
US20030143612A1 (en) * 2001-07-18 2003-07-31 Pointilliste, Inc. Collections of binding proteins and tags and uses thereof for nested sorting and high throughput screening
WO2003049530A2 (en) * 2001-12-07 2003-06-19 Dyax Corporation Method and apparatus for washing magnetically responsive particles
CA2472030A1 (en) * 2002-01-24 2003-07-31 Pointilliste, Inc. Use of collections of binding sites for sample profiling and other applications
US8557743B2 (en) 2002-09-05 2013-10-15 Dyax Corp. Display library process
CN1852765A (en) * 2002-10-18 2006-10-25 普罗梅加公司 Compositions for separating molecules
US6862879B2 (en) * 2002-11-21 2005-03-08 Ford Global Technologies, Llc Diesel aftertreatment system
DE10350474C5 (en) * 2003-10-23 2009-09-17 C-Lecta Gmbh Method for the selection of biomolecules from variant libraries of biomolecules
BRPI0520438A2 (en) * 2005-06-28 2009-05-12 Scripps Research Inst distribution of active proteins to the central nervous system using phage vectors
WO2007081791A2 (en) * 2006-01-04 2007-07-19 The Johns Hopkins University Compare-ms:method rapid, sensitive and accurate detection of dna methylation
WO2011153464A2 (en) * 2010-06-03 2011-12-08 New York University In situ oriented immobilization of proteins on a support
CN111549061A (en) * 2020-05-07 2020-08-18 西南大学 Method for high-throughput screening of target genes of eukaryotic cells responding to extreme pH of environment

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6451527B1 (en) * 1997-08-29 2002-09-17 Selective Genetics, Inc. Methods using genetic package display for selecting internalizing ligands for gene delivery

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SE9304060D0 (en) 1993-12-06 1993-12-06 Bioinvent Int Ab Methods to select specific bacteriophages
US5516637A (en) 1994-06-10 1996-05-14 Dade International Inc. Method involving display of protein binding pairs on the surface of bacterial pili and bacteriophage
IT1303776B1 (en) 1998-11-19 2001-02-23 S I S S A Scuola Internaz Supe PROCESS FOR THE PREPARATION OF GENOTECHE, OF POLYPEPTIDES USING THESE GENOTECHE AND THE OCTOPUS OBTAINED.

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6451527B1 (en) * 1997-08-29 2002-09-17 Selective Genetics, Inc. Methods using genetic package display for selecting internalizing ligands for gene delivery

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090203536A1 (en) * 2006-06-06 2009-08-13 Vermette Patrick Assay supports comprising a peg support, said support attached from a peg solution in cloud point (theta solvent) conditions

Also Published As

Publication number Publication date
US20020058269A1 (en) 2002-05-16
WO2001094950A3 (en) 2002-05-10
US6686154B2 (en) 2004-02-03
AU2001268225A1 (en) 2001-12-17
WO2001094950A2 (en) 2001-12-13

Similar Documents

Publication Publication Date Title
US7674752B2 (en) Functional protein arrays
US6686154B2 (en) Screening of phage displayed peptides without clearing of the cell culture
Wells et al. Rapid evolution of peptide and protein binding properties in vitro
US7534574B2 (en) Epitope-captured antibody display
US8735330B2 (en) pVII phage display
AU2001278613A1 (en) Functional protein arrays
US8969253B2 (en) Method for screening phage display libraries against each other
Scholle et al. In vivo biotinylated proteins as targets for phage-display selection experiments
Crameri pJuFo: a phage surface display system for cloning genes based on protein-ligand interaction
RU2540010C2 (en) PHAGE DISPLAY ON pIX PROTEIN INDEPENDENT FROM SIGNAL SEQUENCE
Lu et al. An alternating selection strategy for cloning phage display antibodies
Ehrlich et al. Phage display technology: affinity selection by biopanning
WO2015166036A1 (en) Peptide libraries
US6586190B2 (en) Parallel high throughput method and kit
WO2004023140A1 (en) Method for screening anti-ligand libraries for identifying anti-ligands specific for differentially and infrequently expressed ligands
AU2001289811A1 (en) High throughput method and kit
Phipps et al. Detection of antibody display phage without clearing of bacterial culture
Liotta Hot Methods Clinic: PHAGE EPITOPE DISPLAY LIBRARIES
Palombo 10 Phage selection for biopanning
Stolle Development of bispecific filamentous bacteriophages for the generation of a novel automated screening system based on phage display technology
Heinis et al. Calmodulin-tagged phage and two-filter sandwich assays for the identification of enzymatic activities

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION