WO1995034648A1 - A method for displaying proteins - Google Patents

A method for displaying proteins Download PDF

Info

Publication number
WO1995034648A1
WO1995034648A1 PCT/US1995/007541 US9507541W WO9534648A1 WO 1995034648 A1 WO1995034648 A1 WO 1995034648A1 US 9507541 W US9507541 W US 9507541W WO 9534648 A1 WO9534648 A1 WO 9534648A1
Authority
WO
WIPO (PCT)
Prior art keywords
protein
dna
phage
pilin
encoding
Prior art date
Application number
PCT/US1995/007541
Other languages
French (fr)
Inventor
Grace P. Huang
Peter R. Rhode
Jeffrey R. Stinson
Hing C. Wong
Original Assignee
Dade International 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 Dade International Inc. filed Critical Dade International Inc.
Priority to AU28282/95A priority Critical patent/AU697865B2/en
Priority to JP8502436A priority patent/JPH09504181A/en
Priority to EP95923861A priority patent/EP0722495A1/en
Publication of WO1995034648A1 publication Critical patent/WO1995034648A1/en

Links

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
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/195Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria
    • C07K14/24Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria from Enterobacteriaceae (F), e.g. Citrobacter, Serratia, Proteus, Providencia, Morganella, Yersinia
    • C07K14/245Escherichia (G)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • 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
    • 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
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/70Vectors or expression systems specially adapted for E. coli
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/02Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving viable microorganisms
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/70Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving virus or bacteriophage
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/02Fusion polypeptide containing a localisation/targetting motif containing a signal sequence

Definitions

  • the present invention relates generally to the exportation and display of polypeptides and proteins on the surface of bacteria. Methods are disclosed providing for display, modification, selection and purification of proteins, including antigenically active proteins, specific binding proteins and enzymatically active proteins.
  • recombinant proteins including antigens and antibodies
  • the recombinant protein is fused to the phage coat proteins expressed by either gene III (minor coat protein) or gene VIII (major coat protein).
  • the display phage can be selectively enriched based on the binding properties of the recombinant protein.
  • the phage carries a vector for expression of the recombinant protein-gene III fusion allowing propagation of the display phage.
  • One of the advantages of this system is that a large library of different proteins such as Fab or single-chain Fv antibody fragments can be displayed on the phage and selected for on the basis of their binding characteristics.
  • One disadvantage is that the number of heterologous protein molecules displayed by the phage is low, thus complicating the selection process.
  • Another disadvantage with phage systems, as well as current bacterial systems is that the enrichment or panning process requires a significant amount of purified binding protein, e.g., antigen, and involves repeated rounds of selection and re- amplification that may result in the isolation of recombinant proteins, e.g., single-chain antibodies, with low binding affinities.
  • a display system combining the benefits of bacterial display and phage display has yet to be developed. Such a system would be very desirable.
  • the present invention relates to a fusion protein, comprising a pilin protein or a portion thereof and a heterologous polypeptide (target protein) .
  • target protein a heterologous polypeptide
  • it relates to a method for displaying the target protein on the outer surface of a bacterial host cell capable of forming pilus.
  • the pilus is a receptor for bacteriophage attachment and infection.
  • the F pilus is preferred.
  • the fusion protein is expressed from a chimeric DNA having a DNA segment encoding a leader amino acid sequence capable of mediating secretion of the fusion protein, a DNA segment encoding pilin subunits, e.g., the traA gene product, and a DNA segment encoding the target protein.
  • the DNA segments are positioned such that expression of the fusion protein results in display of the target protein on the surface of the pilus.
  • the pilus is preferably anchored to the cell surface of a bacteria forming what is referred to as a "display bacteria.”
  • the chimeric DNA may be integrated into the bacterial cell chromosome or be carried by a vector.
  • expression of the fusion protein may be regulated by an inducible promoter, e.g., lac.
  • Bacteria displaying a particular protein may be selected, for example, using antibody affinity.
  • the fusion protein can be detached from selected cells. If desired, the target protein may be separated from the pilin protein and further purified.
  • the present invention further relates to a method for selecting and isolating specific binding pairs, e.g., antigen-antibody, receptor-ligand.
  • a display bacteria is formed in which one protein of the specific binding pair is displayed and replaces the natural receptor for bacteriophage infection.
  • the phage is also altered such that the normal pilin interaction domain is substituted with the other member of the specific binding pair or a library of proteins containing potential binding members.
  • the bacteria display a library of protein containing potential binding proteins.
  • the display phage is then contacted with the display bacteria. Phage displaying one member of the specific binding pair recognize and infect the display bacteria displaying the other member based on the protein-protein interactions between the displayed proteins.
  • the phage genome is then internalized by the display bacteria. These bacteria can then be selected by, for example, identifying of a marker gene, i.e., antibiotic resistance, transferred from the phage to the display bacteria.
  • phage displaying high affinity binding proteins infect and replicate at a higher rate than the phage displaying lower affinity binding proteins. This allows phage displaying a library of potential binding proteins to be screened for high affinity binding since these phage will be selectively enriched with continued growth in cultures of the display bacteria. DNA encoding members of the specific binding pair can then isolated from the display bacterial host.
  • bacteriophage also include phage rescued from an E. coli host carrying a phagemid vector encoding the fusion protein. While such phage are capable of infecting the display bacteria, since they lack the necessary phage genes they cannot produce particles for reinfection and thus cannot be used in method where reinfection is desired.
  • the DNA encoding a member of a specific binding pair is mutaginized, e.g., by use of a mutator strain, and the method of the present invention used to select a member of a specific binding pair having an altered binding affinity, e.g., increased affinity.
  • compounds can be tested for their ability to affect, e.g., inhibit or potentiate, the specific binding pair interaction.
  • Target proteins useful in the present invention include peptides, proteins, e.g., hormones, enzymes, inhibitors, and receptors, antigens, antibodies including antibody fragments, single-chain antibodies and a member of a specific binding pair.
  • the target protein may be a derivative or analog of any such proteins.
  • Specific binding pairs include any pair of molecules, either naturally derived or synthetically produced in which one of the pair has an area which binds to the other molecule. Examples of such specific binding pairs include, for example, antigen- antibody, hormone-hormone receptor, receptor-ligand, enzyme-substrate and IgG-protein A.
  • protein display methods of the present invention include, for example, epitope mapping, screening of antibody libraries and live bacterial vaccines.
  • Figure 1 is a diagram showing the features of the traA expression vector pPR35.
  • Figure 2 sets forth the construction of pPR35.
  • Figure 3 sets forth oligonucleotides (SEQ ID NOS: 1 -1 6) used in PCR amplification and vector construction. Relevant restriction sites are underlined.
  • Figure 4 sets forth the nucleotide (SEQ ID NO: 1 7) and amino acid sequence (SEQ ID NO: 18) of the pPR35 traA fusion region.
  • Figure 5 sets forth the construction of pGH21 .
  • Figure 6 demonstrates the detection of antigen on the surface of a bacterial host cell by colony immunoblotting.
  • Figure 7 is a graph demonstrating antigen tag expression on the eel surface.
  • Figure 8 is a graph demonstrating recombinant antibody expression on the cell surface.
  • Figure 9 is a colony blot showing detection of anti-CK-MB activity of the cell surface.
  • Figure 10 shows the fd gene III protein structure function analysis.
  • Figure 1 1 illustrates the bacteriophage/pilin interaction system.
  • Figure 1 2 shows the recombinant protein/gene III p ⁇ BS fusion region.
  • Figure 1 3 shows the scheme for constructing the r-protein/gene III p ⁇ BS display phagemid.
  • the Oligonucleotides used in cloning are set forth below:
  • OPR1 5'-GGG GGG AGC TCT CTG CAA AGG AGA CAG TCA TAA TGA AAT ACC TAT TGC CTA CGG CAG CCG CTG GAT TG-3' (SEQ ID NO:19)
  • OPR2 5'-GGG GGG CCG CGG CCG CGG CCA TGG CCG GCT GGG CCG CGA GTA ATA ACA ATC CAG CGG CTG CCG TAG-3' (SEQ ID N0:20)
  • Figure 14 shows the construction of the r-protein/gene III p ⁇ BS display phage vector.
  • Figure 1 5 shows a Western blot analysis of partially purified TA-1 scFv/EE tag/traA fusion protein.
  • XL1 -B/pGH21 cells 1 L were grown and TA-1 /EE tag/traA fusion protein expression was induced with IPTG as described in example 2.
  • the bacteria pilin protein was partially purified by shearing the pili from the cells and PEG precipitation as described by Moore et al. (J. Bacteriology, 146 ( 1 ):251 -259, 1 981 ) .
  • the fusion proteins present in the induced cells (cell lysate lane) and in the partially purified protein (PEG ppt.
  • the immunoreactive material at the top of the stacking gel is aggregated fusion protein that does not enter the resolving gel.
  • F pili are filaments found on the surface of cells carrying the F plasmid. They are essential for establishing competent mating pairs during bacterial conjugation [see, Ippen-lhler and Minkley, Ann. Rev. Genet. , 20:593-624 ( 1 986)] and are the site of attachment for three classes of bacteriophage, R1 7, QB, and fd [Paranchych, Cold Spring Harbor Laboratory, pp. 85-1 1 1 ( 1 975)]. The top of the pilus is thought to be involved in the recognition of a recipient cell (mating pair formation) or another donor cell (surface exclusion) and is the site of attachment of the filamentous phage, fd.
  • the sides of the pilus are the site of attachment of two types of spherical phages, exemplified by R1 7 and QB.
  • Synthesis of the F pilus requires 1 3 or more gene products encoded by the transfer region on the F plasmid [Ippen-lhler and Minkley, supra ( 1 986)].
  • the F pilus is composed of a single subunit of 7,200 daltons encoded by the traA gene.
  • the initial traA gene product is propilin (1 3,200 daltons) which contains 51 -amino acid leader sequence.
  • the pilin subunit is acetylated at the amino-terminus and traG is thought to be involved in this process [Ippen-lhler and Minkley supra ( 1 986); Willetts and Skurray, American Society for Microbiology, 2: 1 1 10-1 1 33 (1 987)].
  • F-like plasmids encode four known types of pili which can be distinguished serologically [Lawn and Meynell J. Hyg. , 68:683-694 ( 1 978); Meynell International Conference on Pili, pp. 207-234 (1 978)], by phage sensitivity patterns or surface exclusion [Willetts and Maule, Genet. Res. 47: 1 -1 1 (1 986)]. These pili are also thought to recognize different receptors on the surface of the recipient cell [Havekes, et al., Mol. Gen.
  • pilin genes from four types have been sequenced (Frost, et al., J. Bacteriol. , 1 64: 1238-1 247 (1 985)] and the changes in protein sequence are found in the amino-terminus [Finlay, et al., J. Bacteriol. , 1 63:331 -335 ( 1 985)], with the carboxy- terminus influencing the antigenicity of the protein [Frost, et al., supra (1 985)].
  • the four pilus types vary in their ability to attach to F-specific phages [Meynell, supra (1 978)], which reflects changes in pilin sequence.
  • the method of the present invention relates to displaying a heterologous polypeptide (target protein) on the outer surface of a bacterial host cell.
  • This method comprises expression of a fusion protein, comprising a pilin protein or a portion thereof and the target protein in a bacterial host cell capable of forming a pilus.
  • the fusion protein being expressed from a chimeric DNA having a DNA segment encoding a leader amino acid sequence capable of mediating secretion of the fusion protein, a DNA segment encoding the pilin protein and a DNA segment encoding the target protein, said DNA segments being operably linked such that the host cell displays the target protein on its surface.
  • Any bacterial strain capable of forming a pili can be used as a bacterial host cell for the expression of the chimeric DNA.
  • Strain capable of forming an F or F-like pili are preferred.
  • Such strains include E. coli
  • E. coli is the preferred host cell.
  • Particularly preferred E. coli strains include XL1 B [Bullock, W.O. et al. ( 1 987) Bio/Techniques 5, 376-378] and DH5 ⁇ F ' [Woodcock, D.M. et al.
  • E. coli strains that overexpress pili are preferred.
  • Such strains include, for example, those that carry the depressed F-like plasmid pED208 [Frost, L.S., et al., (1 985) J. Bacteriol. 1 64, 1238-1 247].
  • the first component of the chimeric DNA is a DNA segment encoding a leader amino acid sequence capable of mediating secretion of the fusion protein, i.e., directing the fusion protein to the external membrane surface.
  • sequences include, for example, the traA leader sequence, the phoA leader or the pelB leader. The traA leader sequence is preferred.
  • the traA leader sequence may be obtained by PCR amplification from an F plasmid template.
  • F plasmids are available, for example, from bacterial cells such as E. coli XL1 B.
  • a representative traA leader sequence is set forth in Figure 4.
  • the second component of the chimeric DNA is a DNA segment encoding the pilin protein subunit or a portion thereof capable of displaying the target protein on the cell surface. Mutation analysis suggest that the region of the pilin subunit between amino acids 1 8 to 68 contain elements required for pilus assembly (Frost et al., Mol. Gen. Genet. 21 3: 134-1 39 ( 1 988)). The traA gene product is preferred.
  • TraA genes have been cloned and sequenced from F and a number of related F-like plasmids, including ColB2 (Group II), R1 -1 9 (Group III), R100-1 (Group IV), and pED208 (Group VHFinlay, B.B. et al, (1 984) J. Bactriol. 1 60:402-407; Frost, L. S. et al (1 984) J. Bacteriol. 1 60:395-401 ; Frost, L. S. et al (1 985); Finlay, B. B. et al ( 1 986) J. Bacteriol. 1 68:990-998].
  • Target proteins encoded by the third component of the chimeric DNA, can include peptides, proteins, e.g., hormones, enzymes, inhibitors, and receptors, antigens, antibodies including antibody fragments (e.g., Fab, Fab' and F(ab') 2 ) single-chain antibodies and a member of a specific binding pair.
  • the target protein may be a derivative or analog of any such proteins.
  • Specific binding pairs include any pair of molecules, either naturally derived or synthetically produced, in which one of the pair has an area which binds to the other molecule. Examples of such specific binding pairs include, for example, antigen-antibody, hormone-hormone receptor, receptor-ligand, enzyme-substrate and IgG-protein A.
  • nucleotide sequence of many target proteins are readily available through a number of computer data bases, for example, GenBank, EMBL and Swiss-Prot. Using this information, a DNA segment encoding the desired target protein may be chemically synthesized or, alternatively, the such a DNA segment may be obtained using routine procedures in the art, e.g, PCR amplification. The DNA segments are positioned such that expression of the fusion protein results in the display of the target protein on the cell surface, forming what is referred to as a "display bacteria. "
  • the target protein may be fused to any portion of the pilin protein that is capable of displaying the target protein on the cell surface. Fusion to the amino terminal region of the pilin protein is preferred.
  • Successful display of the target protein on the cell surface can be detected using a number of methods, for example, if the target peptide can be specifically labelled by a procedure that does not operate through the membrane, its cell surface display can be readily demonstrated. This can be done by iodination ( 125 l) of tyrosyl residues in the presence of lactoperoxidase [Marchalonis, et al., J. Biochem. , 124:921 -927 ( 1 971 ); King and Swanson, Infect. Immunol. ,21 :575-584 (1 978)].
  • the action of the protease can be monitored by looking at the cleavage of the polypeptide by SDS-PAGE, or by examining if other properties of the polypeptide are affected (enzyme activity, antigenicity, etc.).
  • nitrocefin is such a substrate for Mactamase [O'Callaghan, et al., Res. Microbiol. , 141 :963-969 ( 1 972); Komacker and Pugsley, Mol. Microbiol. , 4(7): 1 101 - 1 109 (1990)]. It is important to ensure that the outer membrane is indeed impermeable to the substrate when the hybrid protein is expressed. Antibodies against the target protein may also be used. However, these methods have limitations.
  • the fusion protein may be constrained in conformation where the target polypeptide is not detected by the antibody used [Charbit, et al., Embo, J., 5:3029-3037 ( 1 986); Maclntyre, et al., J. Biol. Chem., 263: 1 9053-1 9059 ( 1 988)].
  • the antibody is targeted to a short peptide within the target (for example, an epitope included within 10 residues)
  • the results will only give information on this epitope; thus a positive result may indicate that only this short peptide is exposed, whereas a negative result may indicate that part of the epitope is not accessible, which does not mean that some other part of the target protein is not exposed.
  • Binding of the antibodies to the bacteria can be examined with a number of different techniques. Such methods include, bacterial agglutination, immunofluorescence, ELISA with intact cells, RIA with intact cells, immunoelectron microscopy, and targeted action of complement [see, M. Hofmung, Methods in Cell Biology, 34:77 (1 991 )].
  • the chimeric DNA may be integrated into the host cell chromosome or be carried within a vector.
  • Methods of integrating DNA into a host cell chromosome are well known in the art and include, for example, homologous recombination. See,Winona, et al. J. Bacteriol 1 61 :21 9-21 ( 1 985).
  • the chimeric DNA may also be carried within a recombinant vector, e.g., a plasmid. Recombinant vectors are preferred.
  • the recombinant vectors of the present invention comprise a vector backbone and the chimeric DNA.
  • the recombinant vectors may include an inducible promoter sequence operably linked to the chimeric DNA. Promoters are well known in the art and can readily be selected depending on what cell type is to be used for expression of the fusion protein.
  • the DNA segment encoding the leader is preferably positioned downstream of the promoter sequence.
  • the traA leader sequence is preferred.
  • the DNA segment encoding the target peptide is positioned downstream of the leader sequence.
  • the DNA segment encoding the traA gene product is preferably positioned downstream of the DNA encoding the target peptide.
  • Plasmids useful as the vector backbone include plasmids containing replicon and control sequences which are derived from species compatible with the host cell. For example, if E. coli is used as a host cell, plasmids such as pUC1 9, pUC18 or pBR322 may be used.
  • Vectors can also be constructed comprising the traA leader DNA segments and the traA DNA segment with a cloning site incorporated between the DNA segments to allow insertion of DNA encoding a target protein or insertion of a DNA library.
  • the vector may also contain an inducible promoter and marker gene, e.g., antibiotic resistance.
  • a preferred recombinant vector of the present invention is plasmid pPR35.
  • This plasmid contains a traA leader DNA segment and a traA DNA segment downstream of the inducible lacZ promoter of pUC1 9.
  • Cloning sites for Ncol, Sfi ⁇ and Not ⁇ are incorporated between the traA leader and traA protein sequences to allow insertion of the DNA segments encoding the target peptide.
  • a DNA sequence encoding the EE tag antigen is positioned between the traA leader and traA protein sequences to allow for detection of the fusion protein and characterization of the expression-display system.
  • Introduction of the chimeric DNA to the host cell may be effected by any method known to those skilled in the art. For example, if the DNA is carried by a recombinant vector, the vector can be introduced, for example, by transformation, electroporation, or phage transfection.
  • the detection techniques noted above can be used initially to verify that the method of the present invention is working, i.e., that the fusion pilin protein has been expressed and transported to the bacterial cell surface and is orientated so that the target protein is accessible i.e., displayed.
  • Cells that display the target may be separated from those which do not, using, for example, affinity separation techniques.
  • affinity separation techniques include affinity column chromatography, batch elution from affinity matrix material and fluorescent-activated cell sorting.
  • a bacterial display library produced in accordance with the present invention can be separated by affinity chromatography just as with the phage. Because bacterial cells are larger, care must be taken during loading to prevent plugging and the non-specific retention of bacteria in the column. Subsequently, the cells can be eluted either by passing free antigen through the column or by low pH. Even though gram-negative bacteria are not as resistant to low pH as are phage, there is no meaningful decrease in cell vitality for at least 10 minutes at pH 3.3 [Martineul, et al., Bio/Technology, 9: 1 70 ( 1 991 )]. Thus, elution by low pH and rapid neutralization can be employed for the isolation of strong binding clones.
  • the host cells displaying the desired target protein may then be further cultured and used to obtain the fusion protein. If desired, the target protein may be separated from the pilin protein and further purified using pilin purification techniques familiar to the artisan [J. Bacteriology, 146( 1 ):251 -259 ( 1 981 )].
  • mutate the DNA encoding the heterologous polypeptide e.g., by use of a mutator strain, and use affinity separation technology to identify and select peptides that bind to one or more targets.
  • the display method of the present invention can be used for the detection and characterization of recombinant proteins.
  • the method can be used to map an uncharacterized epitope as follows: Sequences encoding either a library of (1 ) random peptides or (2) peptides derived from the immunoreactive protein of interest can be cloned into a traA expression vector of the present invention, e.g., pPR35. E. coli host cells capable of forming F pili (e.g. XL1 B) are then transformed with the vector bank and the peptide library-traA fusion proteins are displayed on the bacterial cell surface.
  • a solid substrate e.g., a nylon membrane
  • the resulting bacteria are screened for expression of the fusion protein that react with labeled antibody. Reactive colonies can then be picked and the vectors isolated. Sequence analysis of the DNA insert would reveal which of the cloned peptides sequences corresponded to the epitopes recognized by the antibody.
  • the display method of the present invention can also be for detecting recombinant protein activity e.g., antibodies.
  • the method can readily be applied to screening libraries of recombinant antibody-traA fusion proteins. These libraries may include combinatorial single-chain gene banks of heavy and light variable region genes or mutational libraries of specific recombinant antibody genes [Reviewed in Whitlow, M & Filpula, D. (1991 ).
  • the present invention can also be used as a primary cloning system.
  • a cDNA library can be constructed and inserted in a vector of the present invention and the library screened for the ability to bind a ligand.
  • the ligand/binding molecule combination could include any pair of molecules with an ability to specifically bind to one another, e.g., receptor/ligand, enzyme/substrate (or analog), nucleic acid binding protein/nucleic acid, etc. If one member of the complementary pair is available, this may be a preferred way of isolating a clone for the other of the pair.
  • mutator strains can be used.
  • a mutator strain is a strain which contains a genetic defect which causes DNA replicated with in it to be mutated with respect to its parent DNA.
  • Such strains include those carrying the mut D5 mutation such as ES 1 578. Therefore, if a population of genes is introduced into these strains, it will be further diversified and can be transferred to a non-mutator strain if desired, for display and selection.
  • the display method of the present invention can make use of a binding protein on the phage to target the phage genome to a particular bacterial cell displaying a protein recognized by the phage.
  • a binding protein on the phage instead of having the pilus/bacteriophage interaction that allows the phage to enter the cell, an antigen/antibody interaction can be used to allow the bacteriophage to interact with the pili and then enter the cell.
  • the product of gene III acts as the attachment protein, it is believed, through interactions with residues near the N- terminus of the pilin protein.
  • the gene III protein is made up of specific domains involved in incorporation into the page coat, phage morphology, interactions with the bacteria pilus, and entry into the bacteria cells, as depicted in Figure 10.
  • RNA bacteriophage In addition to filamentous bacteriophage, RNA bacteriophage, such as Q ⁇ , MS2, f2 and R17, specifically interact with the F pilus and infect the cells. The ability to absorb to the pilus is conferred by maturation A protein (or A 2 for Q ⁇ ) which is present in one copy per virion [Paranchych, W. ( 1 975) in RNA Phages, ed. N.D.Zinder. (Cold Spring Harbor Laboratory:New York), pp.85-1 12]. Like the gene III protein of filamentous bacteriophage, the RNA phage maturation A protein can be used to form fusions without affecting infectivity.
  • maturation A protein or A 2 for Q ⁇
  • the RNA phage maturation A protein can be used to form fusions without affecting infectivity.
  • a display bacteria is formed in which one protein of the specific binding pair is displayed and replaces the natural receptor for bacteriophage infection.
  • a bacteriophage is also altered such that the normal pilin interaction domain is substituted with the other member of the specific binding pair. This is accomplished by removing the region of the phage attachment protein (e.g. gene III protein of filamentous phage or the A protein of RNA phage) that encodes the pilin binding domain and inserting in its place DNA that encodes the second member of the specific binding pair.
  • the chimeric gene may be incorporated into the phage genome or a recombinant phagemid expression vector. The gene is then expressed in the appropriate strain, e.g. E.
  • coli and the fusion protein and the corresponding phage (or phagemid) genome are packaged into the bacteriophage particles.
  • the phage is then contacted with the display bacteria under standard conditions. Phage displaying one member of the specific binding pair recognize and infect the bacteria displaying the other member based on the protein-protein interactions between the displayed proteins.
  • the phage genome is then internalized by the display bacteria. Display bacteria infected with the phage genome can then be selected by, for example, identifying of a marker gene i.e., antibiotic resistance, transferred from the phage to the display bacteria. DNA encoding members of the specific binding pair can then isolated from the display bacterial host.
  • the phage is altered such that the normal pilin interaction domain (e.g., amino acid 107 to 1 97 of the gene III protein) is removed and replaced by a polypeptide which will specifically bind the target protein displayed on the display bacteria.
  • the display phage recognizes and infects the display bacteria solely based on the protein-protein interactions between the displayed recombinant proteins.
  • Figure 1 1 shows the general characteristics of this system.
  • the phage or phagemid genome is then internalized and expressed. Control signals for transcription, translation and replication can be present. It is particularly useful if the phage or phagemid genome contain sequences useful in selecting for the desired target cell. Useful sequences include, for example, those conferring antibiotic resistance to the target cell.
  • Bacteriophage useful in the method of the present invention include filamentous phage and RNA phage that utilize as a receptor the pilin protein.
  • Such phage include MS2, Q ⁇ , M13, f1 , fd and fd-tet.
  • phagemid expression vectors derived from such filamentous phage can also be used. These vectors can carry plasmid and phage origins of replication and genes that confer antibiotic resistance.
  • the preferred phage is fd-tet [Zacher, A.N., Stock, C.A., Golden, J.W. and Smith, G.P. ( 1 980) Gene
  • phagemid is a derivative of fl phage such as pBC (Stratagene).
  • the EE tag antigen is displayed on the bacteria pili as a traA fusion using a phagemid expression vector that has been developed to allow for recombinant proteins to be displayed on the surface of bacteriophage.
  • the anti-EE tag scFv is displayed on the surface of the bacteriophage particles as a fusion with the gene III protein that has the pilin binding region (amino acid 107-1 97) deleted (this protein will be referred to as genelllp ⁇ BS as shown in Figure 10).
  • Interactions between the anti-EE tag scFv antibody and the EE tag antigen are measured by the ability of the display phage to infect the display bacteria. Specific strategies for generating the display bacteriophage and for measuring infection are set out in Example 8 below.
  • recombinant proteins can be displayed on the bacteriophage and bacterial cell surface. These can include libraries of scFv genes displayed on the phage and a specific antigen peptide on the display bacteria. Screening for specific scFv-antigen interactions involves 1 ) rescue of the scFv display phagemid particles and 2) mixing the phage with the antigen displaying bacteria and testing for the presence of a marker e.g., infectivity by growth on agar plates containing antibiotics (chloramphenicol).
  • the method of the present invention does not require antigen purification or the multiple rounds of enrichment and phage amplification steps that are currently required in phage display systems.
  • Phage or phagemid DNA would be isolated from the resulting antibiotic resistant colonies and the candidate scFv genes could be sequenced. Once the initial characterization is completed, the candidate scFv genes could be subcloned into bacteria expression vectors for the production and further characterization of the single-chain antibodies.
  • a bacteriophage vector based system can also be constructed for display of the recombinant proteins.
  • Such a method has the advantages that it can be used to genetically select for high affinity protein-protein interactions and for binding affinity improvement when coupled with random or site-directed mutagenesis of the recombinant protein.
  • an expression vector is constructed from the fd-tet phage by replacing the normal genelll with the anti-EE tag scFv-genelllp ⁇ BS fusion gene as outlined in Figure 13.
  • DH5- ⁇ F' cells are transformed to tetracycline resistance with the phage expression vector.
  • the transformed cells are be grown overnight, for example, in 100 ml of 2xYT media containing 1 5 ⁇ g/ml tetracycline.
  • the cells are removed by centrifugation and the phage particles in the culture media can be concentrated by precipitation with, for example, 5% PEG and 0.5 M NaCI.
  • the resulting phage particles carry the genelllp ⁇ BS phage vector and display the anti-EE tag scFv-genelllp ⁇ BS fusion protein of the bacteriophage surface. These phage particles are used to infect DH5- ⁇ F' cells carrying the EE tag-traA - 23 -
  • Infectivity can be tested by selection of tetracycline resistant colonies on agar plates as previously described.
  • infectivity can be characterized by the formation of plaques on a lawn of the display bacteria or the propagation of the phage is liquid cultures of the display bacteria. Plaque size or phage titer liquid media provides an indication of the strength of the recombinant protein- protein interactions responsible for the phage infectivity and propagation. In other words, the highest affinity recombinant protein-protein interactions between the display phage and the display bacteria results the highest infectivity rates.
  • the specificity of the infection can be tested with cells that do not display the EE tag antigen.
  • This system is useful in screening libraries of recombinant protein such as scFv.
  • Phage displaying the high affinity scFv can infect and replicate in the antigen displaying bacteria at a higher rates than the phage displaying low affinity scFv.
  • the phage displaying the high affinity scFv will be selectively enriched with continued growth of the culture. This is true for other specific binding pairs as well.
  • the resulting phage DNA can be isolated and the candidate scFv genes and proteins further characterized by sequence and affinity analyses.
  • This system can further be used to screen compounds, i.e., inhibitors or co-factors, that affect specific binding pair interaction.
  • the display bacteria and the display phage are mixed and infectivity of the display phage or phagemid particles is measured as previously described.
  • One such detection method would be antibiotic- resistant growth of the display bacteria following infection with the display phage carrying the antibiotic resistance gene.
  • Candidate compounds are added to the binding reaction and the effect on the level of phage infectivity is measured. For example, the suppression of growth of the display bacteria in appropriate selective media is one means of screening a large number of candidate inhibitor molecules.
  • Compounds potentiating binding can be selected by screening for increased growth.
  • a system was designed to allow inducible expression and display of polypeptides fused to the amino terminus of the pilin protein on the surface of bacteria.
  • the gene encoding the polypeptide of interest was cloned into the traA vector, pPR35 and expressed in an F + bacteria strain.
  • the traA expression vector is based on the multicopy pUC1 9 vector with features shown in Figure 1 .
  • the traA leader and traA protein (pilin) DNA fragments were cloned downstream of the inducible lac ⁇ promoter of pUC1 9.
  • the traA leader allows for proper processing and display of the pilin fusion protein.
  • Cloning sites for Nco ⁇ , Sfi ⁇ and Not ⁇ were incorporated between the traA leader and pilin polypeptide sequences to allow insertion of foreign DNA sequences.
  • a DNA sequence encoding the EE tag antigen was cloned between the traA leader and traA protein sequences to allow for detection of the fusion protein and characterization of the expression-display system.
  • Typical PCR amplification reactions 100 /I contained 10 5 boiled XL1 B bacteria cells carrying the F plasmid as source of template DNA, 10 pmoles of the appropriate primers, 2.5 units of Taq polymerase, 100/yM dNTP, 50mM KCI, 10mM Tris-HCI, pH 8.3, 1 .5mM MgCI 2 , 0.01 % gelatin.
  • the template was denatured by an initial incubation at 96°C for 5 min. during which the Taq polymerase was added to hot-start the reaction.
  • the desired products were amplified by 10 thermal cycles of 55°C for 1 min., 70°C for 1 min. and 96°C for 1 min. followed by 20-step cycles of 70°C for 1 min. and 96°C for 1 min.
  • Amplification with the primers results in the addition of an £coRI site on the 5' end of ⁇ col and Bam ⁇ sites on the 3' end of the traA leader fragment and Bam ⁇ and Kas ⁇ sites on the 5' end and an Xba ⁇ site on the 3' end of the traA protein fragment.
  • the PCR products from 5 reactions were pooled, precipitated with 2 volumes of ethanol/0.3M sodium acetate, and the resulting products (about 0.2 ⁇ g of DNA) were resuspended in water.
  • the traA leader PCR product was digested with EcoRI and Bam ⁇ and the traA protein PCR fragment was digested with Bam ⁇ and Xba ⁇ .
  • the digested fragments were resolved by agarose gel electrophoresis and purified by elution from the agarose gel.
  • pPR5 was generated by digesting pUS1 8 DNA with Kas ⁇ , filling-in the site with Klenow DNA polymerase and religating the blunt ends.
  • the traA fusion vector has been designed to express both peptide antigens such as the EE antigen as well as other recombinant proteins such as single chain antibodies.
  • single-chain antibody genes were created in which the heavy and light variable regions of a particular monoclonal antibody were joined together by a flexible polypeptides linker.
  • Single-chain antibody (scFv) genes were generated from a monoclonal antibody (TA1 ) directed against the prothrombin polypeptide F1 .2 and from a monoclonal antibody directed against creatine kinase-MB ( ⁇ -CKMB) as described below and outlined in Figure 5.
  • the first step involved poly-A RNA isolation from TA1 hybridoma cells by using the Fast-track RNA isolation kit (Invitrogen) according to manufacturer's procedures.
  • This RNA 1 /10 of the mRNA isolated was used
  • was converted to cDNA using Superscript-MLV Reverse Transcriptase (GIBCO-BRL) and oligo-dT specific priming according to manufacturer's procedures.
  • GMBCO-BRL Superscript-MLV Reverse Transcriptase
  • oligo-dT specific priming Of the 20 ⁇ of cDNA generated, 2 ⁇ l was used as template DNA for PCR.
  • the PCR primers for amplifying the TA1 mAb heavy and light chain variable region genes are JS1 35/JS1 34 and JS1 33/JS1 53, respectively, as shown in Figure 2.
  • the PCR buffer conditions are the same as described in Example 1 .
  • the template was denatured by an initial incubation at 96°C for 5 min. during which the Taq polymerase was added to hot-start the reaction.
  • the immunoglobulin variable region gene fragments were amplified by 10 thermal cycles of 48°C for 1 min., 70°C for 1 min., and 96°C for 1 min. followed by 25- step cycles of 70°C for 1 min. and 96°C for 1 min.
  • the desired products (about 260 bp) were resolved by agarose gel electrophoresis and purified by elution from the agarose gel.
  • PCR primers used in the linker attachment are JS135/JS1 39 and JS137/JS1 53 for the heavy and light chain variable gene fragments, respectively.
  • the PCR conditions were 10 thermal cycles of 48°C for 1 min., 70°C for 1 min., and 96°C for 1 min., followed by 25-step cycles of 70°C for 1 min., and 96°C for 1 min.
  • Sequence-overlap extension PCR was used to link the heavy and light chain variable gene fragments by first annealing and extending the heavy chain + light chain variable + linker gene fragments for 10 thermal cycles of 52°C for 1 min., 70°C for 1 min., and 96°C for 1 min. The linked fragments were then amplified by the addition of JS135/JS1 53 primers and 1 5 additional step cycles of 70°C for 1 min. and 96°C for 1 min.
  • the desired products were then amplified by the addition of JS135/JS1 53 primers and 1 5 additional step cycles of 70°C for 1 min. and 96°C for 1 min.
  • the TA1 scFv gene fragment was digested with Ncol and Spel and ligated into the pJS102 cloning vector digested with Nco ⁇ ISpe ⁇ . The resulting construct was sequenced to verify that it contains the TA1 scFv gene.
  • the pJS102/TA1 scFv plasmid was then used as template DNA to PCR the TA1 scFv gene fragment in order to add a Not ⁇ site to the 3' end of the light chain variable gene.
  • the primers used were JS1 35/JS1 53 and the PCR conditions were 10 thermal cycles of 48°C for 1 min., 70°C for 1 min.
  • the desired products (about 720 bp) were resolved by agarose gel electrophoresis and purified by elution from agarose gel.
  • the TA1 scFv gene fragments were digested with Nco ⁇ and Not ⁇ and ligated into the pPR35 traA expression vector digested with Nco ⁇ INot ⁇ , resulting the creation of the TA1 scFv/EE tag/traA fusion vector, pGH21 .
  • CKMB scFv gene fragment was digested Ncol and Spe ⁇ and ligated into the pGH21 traA expression vector digested with Nco ⁇ ISpe ⁇ , essentially swapping the TA1 scFv gene for the ⁇ -CKMB scFv gene.
  • the resulting construct is referred to as p ⁇ -CKMB scFv-traA.
  • the traA expression system was characterized in several ways.
  • bacterial expression of the TA1 scFv-EE tag-traA or ⁇ CKMB scFv-EE tag-traA fusion protein was examined by immunoblot analysis.
  • the pGH21 and p ⁇ -CKMB scFv-traA vectors were transformed into XL1 B cells carrying the F plasmid. Correct candidates were screened by restriction analysis of alkaline-SDS miniprep DNA and verified by DNA sequencing.
  • 60 ⁇ l of an overnight culture was used to inoculate 3 ml of 2xLB media, 50 ⁇ g/ml ampicillin, 1 5 ⁇ g/ml tetracycline.
  • IPTG isopropyl-1 -thio ⁇ -D- galactoside
  • the supernatant (10 ⁇ l) was mixed with SDS/ ?-mercaptoethanol loading buffer and boiled for 5 min. to denature the proteins.
  • the samples were resolved by SDS- polyacrylamide gel electrophoresis on 1 2.5% polyacrylamide gels.
  • the material in the gels was transferred to PVDF nylon membranes using a semi-dry transblot apparatus. The membrane was blocked overnight at 4°C with 20 ml of blocking buffer (0.5% NP-40, 0.5% non-fat dried milk in PBS) and probed with 20 ml of 43ng/ml anti-EE tag mAb conjugated to horseradish preoxidase (anti-EE tag mAb-HRP).
  • the anti-EE tag mAb-HRP was detected by the ECL reagent (Amersham) .
  • the signal for the ⁇ -CKMB scFv-traA fusion protein was detected at the expected molecular weight of 40 kD, while lysates from XL1 B/vector alone showed no signal.
  • the TA1 scFv-traA fusion protein migrates at 46 kD, however, the TA1 scFv protein migrates through SDS-PA gels at a higher molecular weight than expected.
  • the TA1 scFv-traA fusion protein was also detected in the growth media, consistent with the fact that F pili can detach from the cell surface and be found in the media.
  • XL1 -B/pGH21 cells ( 1 L) were grown and TA-1 /EE tag/traA fusion protein expression was induced with IPTG as described in Example 2.
  • the bacteria pilin protein was partially purified by shearing the pili from the cells and PEG precipitation as described by Moore, et al. [J. Bacteriology, 146( 1 ):251 -259 ( 1 981 )].
  • the fusion proteins present in the induced cells (cell lysate lane) and in the partially purified protein (PEG ppt lane) were examined by Western analysis using the anti-EE tag mAb-HRP as a probe. See, Figure 1 5.
  • the band corresponding to the TA-1 /EE tag/traA fusion protein is indicated.
  • the immunoreactive material at the top of the stacking gel is aggregated fusion protein that does not enter the resolving gel.
  • the traA expression system was used to develop improved methods for the detection of recombinant proteins. Two simple detection methods were performed to test whether the antigen-traA fusion protein was displayed on the surface of the bacteria cells. The first was an immunodetection method for screening for bacterial colonies grown on nylon membranes.
  • the XL1 B strain expressing the TA1 -EE tag-traA fusion protein was spread on a nylon membrane and the membrane was placed on 2xLB agar plate containing 50 ⁇ g/ml ampicillin and 1 5 ⁇ g/ml tetracycline for selection of the vector and XL1 B strain, respectively.
  • the membrane was prewet with 10mM IPTG.
  • anti-EE tag mAb-HRP recognized the IPTG-induced XL1 B/TA1 -EE tag-traA colony but not the non-induced XL1 B/TA1 -EE tag-traA colony.
  • XL1 B cells carrying a control vector (no EE tag-traA) failed to give any signal.
  • the specificity of binding of anti-EE tag mAb on cell surface was also determined by incubating the colony membrane with an antibody to a different peptide tag (KT3). No signal was detected on these membranes.
  • the bacteria colony immunodetection method was also applied to epitope mapping analysis.
  • mixtures of XL1 B cells carrying either the TA1 -EE tag-traA or the control vector (no EE tag-traA insert) were grown overnight on 2xLB agar plates containing 50 ⁇ g/ml ampicillin and 1 5 ⁇ g/ml tetracycline overnight.
  • the colonies were replica- plated onto nylon membranes and placed on 2xLB agar plates containing 10mM IPTG, 50 ⁇ g/ml ampicillin and 1 5 ⁇ g/ml tetracycline.
  • the colonies on the membranes were probed with anti-EE tag mAb-HRP as described above.
  • positive signals were detected for single colonies as shown in Figure 6.
  • the corresponding colonies were picked from the master plate for characterization and were found to carrying the TA1 -EE tag-traA vector.
  • the second method to test the accessibilty of the antigen-traA fusion protein on the surface of the bacteria was an ELISA method with intact cells.
  • Cell grown to early log-phase were induced by IPTG for 4 hours at 37°C.
  • the cells were harvested and resuspended in cold PBS to 1 .0 0D 595 /ml. This step will remove any traA fusion protein present in the media that is not associated with the cells.
  • Microtiter plates were coated with 100 ⁇ l of bacterial dilution per well.
  • the induced XL1 B/TA1 -EE tag-traA samples showed greater than six-fold higher readings than the non-induced sample or the XL1 B/control vector (no EE tag-traA insert) sample as shown in Figure 7, indicating that this method is applicable to specifically detecting antigens presented on the cell surface.
  • this method could be used to quantitative antibody/antigen binding.
  • the epitope could be characterized in a comparative ELISA assay format where the effect of different peptides on antibody/antigen-traA fusion protein interaction is determined.
  • Cells carrying either the ⁇ -CKMB scFv-EE tag-traA fusion vector or the control vector (TA1 -EE tag-traA fusion vector or a vector without an insert) were grown to early log-phase at 37°C. At that point, expression of the fusion protein was induced by the addition of 0.2mM IPTG for 4 hours at 37°C.
  • the cells were harvested and resuspended in cold PBS to 10.0 0D 595 /ml.
  • Microtiter plates were coated with 100 ⁇ g/ml anti-CK-BB mAb in coating buffer (0.1 M Tris-HCI, pH8.5) and were incubated overnight at 4°C.
  • Unattached anti-CK-BB mAb was discarded and the wells were washed once with washing buffer (0.1 M Tris-HCI, pH 7.4, 1 .0M NaCI, 0.1 % NaN 3 ).
  • the wells were incubated with 100 ⁇ l of 0.3 ⁇ g/ml CK-MB in dilution buffer (2% gelatin, 0.1 % Tween 20 in 0.01 m Tris-HCI, pH7.3, 0.1 5 M NaCI) at room temperature for 1 hour with agitation.
  • the wells were washed once with rinse buffer (0.01 M Tris-HCI, pH7.3, 0.1 5M NaCI, 0.2% BSA, 0.05% Tween-20, 0.2% NaN 3 ).
  • This example demonstrates the successful detection of a recombinant protein activity expressed on the surface of the bacterial cell colony.
  • a mixture of the XL1 B strain carrying the ⁇ -CKMB scFv-EE tag- traA fusion vector and the strain carrying the TA1 -EE tag-traA fusion vector was distributed evenly on a nylon membrane.
  • the membrane was placed on a 2xLB agar plate containing 50 ⁇ g/ml ampicillin and 1 5 ⁇ g/ml tetracycline and incubated at 37°C until small bacterial colonies appeared.
  • a replica membrane was made by overlaying the master membrane with a new membrane. The replica membrane was then removed and cut in half.
  • This method provides an easy rapid procedure for detecting recombinant single-chain antibody activity. It could be readily applied to screening libraries of recombinant antibody-traA fusion proteins. These libraries may include combinatorial single-chain gene banks of heavy and light variable region genes or mutational libraries of specific recombinant antibody genes. On the basis of the results indicating that the ⁇ -CKMB scFv-traA fusion protein is folded into a biologically active conformation, this method could have general application to detection of recombinant protein activities expressed on the surface of the bacterial cell colony. The activities to be detected could include binding activities, catalytic activities, inhibitory activities and altered structural conformations.
  • a phagemid vector was designed for the expression and display of genelllp ⁇ BS fusion proteins on the surface of the bacteriophage particle.
  • the phagemid vector is based on the pBC phagemid vector (Stratagene) with features shown in Figure 1 2.
  • This vector carries the ColEI replication origin for plasmid propagation, the f1 filamentous phage replication origin for recovery of phagemid DNA following co-infection with helper phage and the chloramphenicol resistance gene for antibiotic selection.
  • the pe/B leader and genelllp ⁇ BS DNA fragments were cloned downstream of the inducible lacZ promoter of pBC.
  • the pe/B leader was designed to allow for proper processing and display of the fusion protein on the bacteriophage particle. Cloning sites for Nc ⁇ l, Sfi ⁇ , Spe ⁇ , and Nofl were incorporated between the pe/B leader and genelllp ⁇ BS sequences to allow insertion of foreign D ⁇ A sequences. The steps involved in constructing this vector (referred to as LE2) are shown in Figure 12.
  • the anti-EE tag scFv gene are isolated from monoclonal hybridoma mR ⁇ A as outlined in Example 2 and are inserted at the Nofl and Spe ⁇ sites of LE2.
  • An F' host strain, DH5- ⁇ F' [Woodcock, D:M. et al ( 1 989) ⁇ ucl. Acids. Res. 1 7,3469-3478] is used to propagate these vectors by growth in media containing 30 ⁇ g/ml chloramphenicol.
  • DH5- ⁇ F' cells carrying phagemid expression vector are transformed with the fKN1 6 phage DNA to tetracycline resistance.
  • the fKN1 6 phage derivative was constructed from the tetracycline-resistance phage, fd-tet, by deleting a 507 bp segment of gene III (Nelson, et a/., Virology, 108:338-350 (1 981 )) .
  • This phage is non- infective due to the gene III deletion but provides the helper phage proteins necessary for replication and packaging of the phagemid expression vector.
  • DH5- ⁇ F' cells carrying both the phagemid expression vector and fKN1 6 are gown overnight in 100 ml of 2xYT media containing 30 ⁇ g/ml chloramphenicol and 1 5 ⁇ g/ml tetracycline.
  • the cells are removed by centrifugation and the phage particles in the culture media are concentrated by precipitation with 5% PEG and 0.5 M NaCI.
  • the resulting phage particles carry either the fKN1 6 phage (tet r ) or the genelllp ⁇ BS phagemid vector (chl r ). Both the defective fKN1 6 gene protein and the anti-EE tag scFv-genelllp ⁇ BS fusion protein are displayed on the bacteriophage surface.
  • the rescued phage are used to infect XL-1 B cells carrying the EE tag-traA fusion vector.
  • the bacteriophage displaying the anti-EE tag scFv- genelllp ⁇ BS fusion protein bind the EE tag-traA fusion protein and infect these cells resulting in chloramphenicol resistant clones carrying the phagemid expression vector.
  • a bacteriophage vector based system can also be constructed for the display of recombinant proteins.
  • the expression vector can be constructed from the fd-tet phage by replacing the normal genelll with the anti-EE tag scFv-genelllp ⁇ BS fusion gene as outlined in figure 14.
  • DH5- ⁇ F' cells are transformed to tetracycline resistance with the phage expression vector.
  • the transformed cells are grown overnight in 100 ml of 2xYT media containing 1 5 ⁇ g/ml tetracycline. The cells will be removed by centrifugation and the phage particles in the culture media will be concentrated by precipitation with 5% PEG and 0.5 M NaCI.
  • the resulting phage particles will carry the genelllp ⁇ BS phage vector and display the anti-EE tag scFv-genelllp ⁇ BS fusion protein of the bacteriophage surface. These phage particles will be used to infect DH5- ⁇ F' cells carrying the EE tag-traA fusion vector.
  • Infectivity can then be tested by selection of tetracycline resistant colonies on agar plates as described for the phagemid system.
  • infectivity can be characterized by the formation of plaques on a lawn of the display bacteria or the propagation of the phage in liquid cultures of the display bacteria. Plaque size or phage titer in the liquid media provide an indication of the strength of the recombinant protein-protein interactions responsible for the phage infectivity and propagation. In other words, the highest affinity recombinant protein-protein interactions between the display phage and the display bacteria provide the highest infectivity rates. The specificity of the infection can also be tested with cells that do not display the EE tag antigen.
  • This system is useful in screening libraries of recombinant protein such as scFvs.
  • the phage displaying the high affinity scFv will infect and replicate in the antigen displaying bacteria at a higher rate that the phage displaying low affinity scFv.
  • the phage displaying the high affinity scFv will be selectively enriched with continued growth of the culture.
  • the resulting phage DNA can be isolated and the candidate scFv genes and proteins could be further characterized by sequence and affinity analyses.
  • ADDRESSEE David G. Conlin; DIKE, BRONSTEIN,
  • TGCAGGCATG CAAGCTTGGC ACTGGCCGTC GTTTTACAAC GTCGTGACTG GGAAAACCCT 540

Abstract

The present invention relates to a fusion protein, comprising a pilin protein or a portion thereof and a heterologous polypeptide (target protein). In a preferred embodiment it relates to a method for displaying the target protein on the outer surface of a bacterial host cell capable of forming pilus. In certain embodiments, it is desirable that the pilus is a receptor for bacteriophage attachment and infection. The F pilus is preferred.

Description

A METHOD FOR DISPLAYING PROTEINS
The present invention relates generally to the exportation and display of polypeptides and proteins on the surface of bacteria. Methods are disclosed providing for display, modification, selection and purification of proteins, including antigenically active proteins, specific binding proteins and enzymatically active proteins.
BACKGROUND OF THE INVENTION
The expression of polypeptides on the surface of bacteria and bacteriophage has been pursued for several years, in part because of interest in recombinant antibody production. Many other potential applications exist, including the production of genetically-engineered whole cell adsorbents, construction of "peptide libraries", cell bound enzymes, and use as live vaccines or immunogens to generate antibodies. [See, W092/01047 and W093/10214.]
In bacteria, one approach to obtaining surface expressed foreign proteins has been the use of native membrane proteins as a carrier for a foreign protein. In general, most attempts to develop methods of anchoring proteins on a bacterial surface have focused on fusion of the desired recombinant polypeptide to a native protein that is normally exposed on the cell's exterior with the hope that the resulting hybrid will also be localized on the surface. However, in most cases, the foreign protein interferes with localization, and thus, the fusion protein is unable to reach the cell surface. These fusions either end up at incorrect cellular locations or become anchored in the membrane with a secreted protein domain facing the periplasm [Murphy, et al. , J. Bacteriol., 1 72:2736 (1990)].
Francisco, et al., [Proc. Nat/. Acad. Sci. , 89:271 3 ( 1 992)] reported constructing a surface-expression vehicle consisting of the Ipp N-terminal targeting sequence fused to a sequence derived from ompA leaving the C- terminus exposed on the external side of the outer membrane. These fusions have been reported to export a number of heterologous proteins to the E. coli surface, including Mactomase, single-chain Fv antibody and a cellulose binding protein [W093/10214]. In addition, Fuschs, et al.,
[Bio/Technology, 9: 1369 (1991 )] reported that a fusion between the E. coli peptidoglycan-associated lipoprotein (pal) and a lysozyme-binding single- chain Fv antibody fragment could be detected on the surface of bacteria. However, in these systems, the displayed proteins were affixed to the cell surface, and thus in order to isolate purified protein, the DNA encoding the protein must be subcloned to another system.
Systems have been developed for displaying recombinant proteins, including antigens and antibodies, on the surface of filamentous bacteriophage [see, for example, W092/01047]. In these systems, the recombinant protein is fused to the phage coat proteins expressed by either gene III (minor coat protein) or gene VIII (major coat protein). The display phage can be selectively enriched based on the binding properties of the recombinant protein. In addition, the phage carries a vector for expression of the recombinant protein-gene III fusion allowing propagation of the display phage. One of the advantages of this system is that a large library of different proteins such as Fab or single-chain Fv antibody fragments can be displayed on the phage and selected for on the basis of their binding characteristics. One disadvantage is that the number of heterologous protein molecules displayed by the phage is low, thus complicating the selection process. Another disadvantage with phage systems, as well as current bacterial systems is that the enrichment or panning process requires a significant amount of purified binding protein, e.g., antigen, and involves repeated rounds of selection and re- amplification that may result in the isolation of recombinant proteins, e.g., single-chain antibodies, with low binding affinities.
A display system combining the benefits of bacterial display and phage display has yet to be developed. Such a system would be very desirable.
It would also be desirable to have a method that can be used for cloning and protein purification with out the need for subcloning.
It would be desirable to have a display and selection method that eliminates the need for panning and purification of binding protein.
SUMMARY OF THE INVENTION
The present invention relates to a fusion protein, comprising a pilin protein or a portion thereof and a heterologous polypeptide (target protein) . In a preferred embodiment it relates to a method for displaying the target protein on the outer surface of a bacterial host cell capable of forming pilus. In certain embodiments, it is desirable that the pilus is a receptor for bacteriophage attachment and infection. The F pilus is preferred.
The fusion protein is expressed from a chimeric DNA having a DNA segment encoding a leader amino acid sequence capable of mediating secretion of the fusion protein, a DNA segment encoding pilin subunits, e.g., the traA gene product, and a DNA segment encoding the target protein. The DNA segments are positioned such that expression of the fusion protein results in display of the target protein on the surface of the pilus. The pilus is preferably anchored to the cell surface of a bacteria forming what is referred to as a "display bacteria."
The chimeric DNA may be integrated into the bacterial cell chromosome or be carried by a vector. In certain preferred embodiments, expression of the fusion protein may be regulated by an inducible promoter, e.g., lac. Bacteria displaying a particular protein may be selected, for example, using antibody affinity. The fusion protein can be detached from selected cells. If desired, the target protein may be separated from the pilin protein and further purified.
The present invention further relates to a method for selecting and isolating specific binding pairs, e.g., antigen-antibody, receptor-ligand. In accordance with this method, a display bacteria is formed in which one protein of the specific binding pair is displayed and replaces the natural receptor for bacteriophage infection. The phage is also altered such that the normal pilin interaction domain is substituted with the other member of the specific binding pair or a library of proteins containing potential binding members. Alternatively, the bacteria display a library of protein containing potential binding proteins. The display phage is then contacted with the display bacteria. Phage displaying one member of the specific binding pair recognize and infect the display bacteria displaying the other member based on the protein-protein interactions between the displayed proteins. The phage genome is then internalized by the display bacteria. These bacteria can then be selected by, for example, identifying of a marker gene, i.e., antibiotic resistance, transferred from the phage to the display bacteria. In addition, phage displaying high affinity binding proteins infect and replicate at a higher rate than the phage displaying lower affinity binding proteins. This allows phage displaying a library of potential binding proteins to be screened for high affinity binding since these phage will be selectively enriched with continued growth in cultures of the display bacteria. DNA encoding members of the specific binding pair can then isolated from the display bacterial host.
As used herein, bacteriophage also include phage rescued from an E. coli host carrying a phagemid vector encoding the fusion protein. While such phage are capable of infecting the display bacteria, since they lack the necessary phage genes they cannot produce particles for reinfection and thus cannot be used in method where reinfection is desired.
In one embodiment, the DNA encoding a member of a specific binding pair is mutaginized, e.g., by use of a mutator strain, and the method of the present invention used to select a member of a specific binding pair having an altered binding affinity, e.g., increased affinity. In another embodiment, compounds can be tested for their ability to affect, e.g., inhibit or potentiate, the specific binding pair interaction.
Target proteins useful in the present invention include peptides, proteins, e.g., hormones, enzymes, inhibitors, and receptors, antigens, antibodies including antibody fragments, single-chain antibodies and a member of a specific binding pair. Alternatively, the target protein may be a derivative or analog of any such proteins. Specific binding pairs include any pair of molecules, either naturally derived or synthetically produced in which one of the pair has an area which binds to the other molecule. Examples of such specific binding pairs include, for example, antigen- antibody, hormone-hormone receptor, receptor-ligand, enzyme-substrate and IgG-protein A.
Other uses for the protein display methods of the present invention include, for example, epitope mapping, screening of antibody libraries and live bacterial vaccines.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 is a diagram showing the features of the traA expression vector pPR35.
Figure 2 sets forth the construction of pPR35.
Figure 3 sets forth oligonucleotides (SEQ ID NOS: 1 -1 6) used in PCR amplification and vector construction. Relevant restriction sites are underlined.
Figure 4 sets forth the nucleotide (SEQ ID NO: 1 7) and amino acid sequence (SEQ ID NO: 18) of the pPR35 traA fusion region.
Figure 5 sets forth the construction of pGH21 .
Figure 6 demonstrates the detection of antigen on the surface of a bacterial host cell by colony immunoblotting.
Figure 7 is a graph demonstrating antigen tag expression on the eel surface. Figure 8 is a graph demonstrating recombinant antibody expression on the cell surface.
Figure 9 is a colony blot showing detection of anti-CK-MB activity of the cell surface.
Figure 10 shows the fd gene III protein structure function analysis.
Figure 1 1 illustrates the bacteriophage/pilin interaction system.
Figure 1 2 shows the recombinant protein/gene III pΔBS fusion region.
Figure 1 3 shows the scheme for constructing the r-protein/gene III pΔBS display phagemid. The Oligonucleotides used in cloning are set forth below:
OPR1 - 5'-GGG GGG AGC TCT CTG CAA AGG AGA CAG TCA TAA TGA AAT ACC TAT TGC CTA CGG CAG CCG CTG GAT TG-3' (SEQ ID NO:19) OPR2 - 5'-GGG GGG CCG CGG CCG CGG CCA TGG CCG GCT GGG CCG CGA GTA ATA ACA ATC CAG CGG CTG CCG TAG-3' (SEQ ID N0:20)
0PR3 - 5'-GGG GGG CCG CGG CCG CGG AGG AAG AAG AGT ACA ACC CGA ACG AAG GCG CCG CCT AGA CTG TTG AAA GTT GTT TAG CAA AAC CTC-3'(SEQ ID N0:21)
0PR4 - 5'-GGG CCG AAT TCC TAT TAA GAC TCC TTA TTA CGC AGT ATG TTA GC-3' (SEQ ID NO:22) 0GH1 - 5'-GGG GGG ACT AGT GCG GCC GCG GGC GCC GCT GAA ACT GTT GAA AGTTGTTTA GC-3' (SEQ ID N0:23)
OGH107 - 5'-GGG GGG GGA TCC AGA GGG TTG ATA TAA GTA TAG CC-3' (SEQ ID N0:24) Figure 14 shows the construction of the r-protein/gene III pΔBS display phage vector.
Figure 1 5 shows a Western blot analysis of partially purified TA-1 scFv/EE tag/traA fusion protein. XL1 -B/pGH21 cells ( 1 L) were grown and TA-1 /EE tag/traA fusion protein expression was induced with IPTG as described in example 2. The bacteria pilin protein was partially purified by shearing the pili from the cells and PEG precipitation as described by Moore et al. (J. Bacteriology, 146 ( 1 ):251 -259, 1 981 ) . The fusion proteins present in the induced cells (cell lysate lane) and in the partially purified protein (PEG ppt. lane) were examined by western analysis using the anti- EE tag mAb-HRP as a probe, the band corresponding to the TA-1 /EE tag/traA fusion protein is indicated. The immunoreactive material at the top of the stacking gel is aggregated fusion protein that does not enter the resolving gel.
DETAILED DESCRIPTION OF THE INVENTION
F pili are filaments found on the surface of cells carrying the F plasmid. They are essential for establishing competent mating pairs during bacterial conjugation [see, Ippen-lhler and Minkley, Ann. Rev. Genet. , 20:593-624 ( 1 986)] and are the site of attachment for three classes of bacteriophage, R1 7, QB, and fd [Paranchych, Cold Spring Harbor Laboratory, pp. 85-1 1 1 ( 1 975)]. The top of the pilus is thought to be involved in the recognition of a recipient cell (mating pair formation) or another donor cell (surface exclusion) and is the site of attachment of the filamentous phage, fd. The sides of the pilus are the site of attachment of two types of spherical phages, exemplified by R1 7 and QB. Synthesis of the F pilus requires 1 3 or more gene products encoded by the transfer region on the F plasmid [Ippen-lhler and Minkley, supra ( 1 986)]. The F pilus is composed of a single subunit of 7,200 daltons encoded by the traA gene. The initial traA gene product is propilin (1 3,200 daltons) which contains 51 -amino acid leader sequence. The pilin subunit is acetylated at the amino-terminus and traG is thought to be involved in this process [Ippen-lhler and Minkley supra ( 1 986); Willetts and Skurray, American Society for Microbiology, 2: 1 1 10-1 1 33 (1 987)].
F-like plasmids encode four known types of pili which can be distinguished serologically [Lawn and Meynell J. Hyg. , 68:683-694 ( 1 978); Meynell International Conference on Pili, pp. 207-234 (1 978)], by phage sensitivity patterns or surface exclusion [Willetts and Maule, Genet. Res. 47: 1 -1 1 (1 986)]. These pili are also thought to recognize different receptors on the surface of the recipient cell [Havekes, et al., Mol. Gen.
Genet. , 1 55: 1 85-1 89 (1 977)]. Representative pilin genes from four types have been sequenced (Frost, et al., J. Bacteriol. , 1 64: 1238-1 247 (1 985)] and the changes in protein sequence are found in the amino-terminus [Finlay, et al., J. Bacteriol. , 1 63:331 -335 ( 1 985)], with the carboxy- terminus influencing the antigenicity of the protein [Frost, et al., supra (1 985)]. The four pilus types vary in their ability to attach to F-specific phages [Meynell, supra (1 978)], which reflects changes in pilin sequence. However, the amino-terminus does not seem to be involved in phage attachment since pili with different amino-terminal attach fd phage equally [Frost, et al., supra ( 1 985); Finlay, et al. J. Bacteriol. , 1 68:990-998
(1 986)]. The changes in sequence which probably affect phage binding occur at residues 1 1 and 14 in type IV pilin (represented by the R100-1 plasmid) and at the carboxy-terminus in Type III pilin (represented by the R1 -1 9 plasmid). Studies with polyclonal antisera [Worobec, et al., J. Bacteriol. , 1 67:660-665 ( 1 986)] and monoclonal antisera [Frost, et al., J. Bacteriol. , 1 68: 1 92-198 (1 986)] have shown that the major epitope at the amino-terminus is exposed in a tip-specific manner at the end of the pilus. The minor epitope(s) which involve the carboxy-terminus of the pilin protein are exposed on the sides of the pilus.
The method of the present invention relates to displaying a heterologous polypeptide (target protein) on the outer surface of a bacterial host cell. This method comprises expression of a fusion protein, comprising a pilin protein or a portion thereof and the target protein in a bacterial host cell capable of forming a pilus. The fusion protein being expressed from a chimeric DNA having a DNA segment encoding a leader amino acid sequence capable of mediating secretion of the fusion protein, a DNA segment encoding the pilin protein and a DNA segment encoding the target protein, said DNA segments being operably linked such that the host cell displays the target protein on its surface.
Any bacterial strain capable of forming a pili can be used as a bacterial host cell for the expression of the chimeric DNA. Strain capable of forming an F or F-like pili are preferred. Such strains include E. coli
(Ippen-lhler, et al.), Salmonella typhimurium [Artz, S. Holzschu, D., Blum, P., and Shand, R. ( 1 983) Gene 26, 147-1 58], as well as other gram- negative bacterial carrying F-like plasmids. E. coli is the preferred host cell. Particularly preferred E. coli strains include XL1 B [Bullock, W.O. et al. ( 1 987) Bio/Techniques 5, 376-378] and DH5σF' [Woodcock, D.M. et al.
( 1 989) Nucleic Acids Res. 1 7,3469-3478]. In certain embodiments, E. coli strains that overexpress pili are preferred. Such strains include, for example, those that carry the depressed F-like plasmid pED208 [Frost, L.S., et al., (1 985) J. Bacteriol. 1 64, 1238-1 247]. The first component of the chimeric DNA is a DNA segment encoding a leader amino acid sequence capable of mediating secretion of the fusion protein, i.e., directing the fusion protein to the external membrane surface. Such sequences include, for example, the traA leader sequence, the phoA leader or the pelB leader. The traA leader sequence is preferred. The traA leader sequence may be obtained by PCR amplification from an F plasmid template. F plasmids are available, for example, from bacterial cells such as E. coli XL1 B. A representative traA leader sequence is set forth in Figure 4.
The second component of the chimeric DNA is a DNA segment encoding the pilin protein subunit or a portion thereof capable of displaying the target protein on the cell surface. Mutation analysis suggest that the region of the pilin subunit between amino acids 1 8 to 68 contain elements required for pilus assembly (Frost et al., Mol. Gen. Genet. 21 3: 134-1 39 ( 1 988)). The traA gene product is preferred.
Hydropathy profiles the F-pilin suggests that the molecule is organized into four domains [Paiva, W.D., et al., (1 992) J. Biol. Chem. 267, 261 91 -261 97]. Variability in the number and type of amino acids present in the N-terminal domain is observed for different F-like pilin proteins [Frost, L.S. et al (1 985)], suggesting that this region may be dispensable for pili assembly and display of on the cell surface. As described above, the traA gene encodes the 51 amino acid pilin leader and the 70 amino acid mature pilin protein. TraA genes have been cloned and sequenced from F and a number of related F-like plasmids, including ColB2 (Group II), R1 -1 9 (Group III), R100-1 (Group IV), and pED208 (Group VHFinlay, B.B. et al, (1 984) J. Bactriol. 1 60:402-407; Frost, L. S. et al (1 984) J. Bacteriol. 1 60:395-401 ; Frost, L. S. et al (1 985); Finlay, B. B. et al ( 1 986) J. Bacteriol. 1 68:990-998]. These genes show a high homology with each other and encode pilin proteins that comprise morphologically and functionally similar structures, as emphasized by the formation of mixed pili by cells carrying different F-like plasmids [Lawn, A.M., et al (1971 ) Ann. Institute Pasteur 120:3-8]. Since the sequences of various traA genes are available, the DNA encoding the traA gene product can be readily isolated from a number of sources, including for example, PCR amplification from an F plasmid template. See, Example 1 for the details of the PCR amplification. A representative F plasmid traA gene sequence is set forth in Figure 4.
Target proteins, encoded by the third component of the chimeric DNA, can include peptides, proteins, e.g., hormones, enzymes, inhibitors, and receptors, antigens, antibodies including antibody fragments (e.g., Fab, Fab' and F(ab')2) single-chain antibodies and a member of a specific binding pair. Alternatively, the target protein may be a derivative or analog of any such proteins. Specific binding pairs include any pair of molecules, either naturally derived or synthetically produced, in which one of the pair has an area which binds to the other molecule. Examples of such specific binding pairs include, for example, antigen-antibody, hormone-hormone receptor, receptor-ligand, enzyme-substrate and IgG-protein A.
The nucleotide sequence of many target proteins are readily available through a number of computer data bases, for example, GenBank, EMBL and Swiss-Prot. Using this information, a DNA segment encoding the desired target protein may be chemically synthesized or, alternatively, the such a DNA segment may be obtained using routine procedures in the art, e.g, PCR amplification. The DNA segments are positioned such that expression of the fusion protein results in the display of the target protein on the cell surface, forming what is referred to as a "display bacteria. "
The target protein may be fused to any portion of the pilin protein that is capable of displaying the target protein on the cell surface. Fusion to the amino terminal region of the pilin protein is preferred.
Successful display of the target protein on the cell surface can be detected using a number of methods, for example, if the target peptide can be specifically labelled by a procedure that does not operate through the membrane, its cell surface display can be readily demonstrated. This can be done by iodination (125l) of tyrosyl residues in the presence of lactoperoxidase [Marchalonis, et al., J. Biochem. , 124:921 -927 ( 1 971 ); King and Swanson, Infect. Immunol. ,21 :575-584 (1 978)].
In addition, one can examine if the target polypeptide is accessible to proteases added from the outside to intact cells. The action of the protease can be monitored by looking at the cleavage of the polypeptide by SDS-PAGE, or by examining if other properties of the polypeptide are affected (enzyme activity, antigenicity, etc.).
If the target polypeptide displays enzymatic activity, one may use such activity to demonstrate cell surface display. This can be done if a substrate unable to cross the outer membrane is available: nitrocefin is such a substrate for Mactamase [O'Callaghan, et al., Res. Microbiol. , 141 :963-969 ( 1 972); Komacker and Pugsley, Mol. Microbiol. , 4(7): 1 101 - 1 109 (1990)]. It is important to ensure that the outer membrane is indeed impermeable to the substrate when the hybrid protein is expressed. Antibodies against the target protein may also be used. However, these methods have limitations. First, the fusion protein may be constrained in conformation where the target polypeptide is not detected by the antibody used [Charbit, et al., Embo, J., 5:3029-3037 ( 1 986); Maclntyre, et al., J. Biol. Chem., 263: 1 9053-1 9059 ( 1 988)]. Second, if the antibody is targeted to a short peptide within the target (for example, an epitope included within 10 residues), the results will only give information on this epitope; thus a positive result may indicate that only this short peptide is exposed, whereas a negative result may indicate that part of the epitope is not accessible, which does not mean that some other part of the target protein is not exposed.
Binding of the antibodies to the bacteria can be examined with a number of different techniques. Such methods include, bacterial agglutination, immunofluorescence, ELISA with intact cells, RIA with intact cells, immunoelectron microscopy, and targeted action of complement [see, M. Hofmung, Methods in Cell Biology, 34:77 (1 991 )].
The chimeric DNA may be integrated into the host cell chromosome or be carried within a vector. Methods of integrating DNA into a host cell chromosome are well known in the art and include, for example, homologous recombination. See,Winona, et al. J. Bacteriol 1 61 :21 9-21 ( 1 985). The chimeric DNA may also be carried within a recombinant vector, e.g., a plasmid. Recombinant vectors are preferred.
The recombinant vectors of the present invention comprise a vector backbone and the chimeric DNA. The recombinant vectors may include an inducible promoter sequence operably linked to the chimeric DNA. Promoters are well known in the art and can readily be selected depending on what cell type is to be used for expression of the fusion protein. The DNA segment encoding the leader is preferably positioned downstream of the promoter sequence. The traA leader sequence is preferred. The DNA segment encoding the target peptide is positioned downstream of the leader sequence. The DNA segment encoding the traA gene product is preferably positioned downstream of the DNA encoding the target peptide.
Plasmids useful as the vector backbone include plasmids containing replicon and control sequences which are derived from species compatible with the host cell. For example, if E. coli is used as a host cell, plasmids such as pUC1 9, pUC18 or pBR322 may be used.
Vectors can also be constructed comprising the traA leader DNA segments and the traA DNA segment with a cloning site incorporated between the DNA segments to allow insertion of DNA encoding a target protein or insertion of a DNA library. The vector may also contain an inducible promoter and marker gene, e.g., antibiotic resistance.
A preferred recombinant vector of the present invention is plasmid pPR35. This plasmid contains a traA leader DNA segment and a traA DNA segment downstream of the inducible lacZ promoter of pUC1 9. Cloning sites for Ncol, Sfi\ and Not\ are incorporated between the traA leader and traA protein sequences to allow insertion of the DNA segments encoding the target peptide. In addition, a DNA sequence encoding the EE tag antigen is positioned between the traA leader and traA protein sequences to allow for detection of the fusion protein and characterization of the expression-display system. Introduction of the chimeric DNA to the host cell may be effected by any method known to those skilled in the art. For example, if the DNA is carried by a recombinant vector, the vector can be introduced, for example, by transformation, electroporation, or phage transfection.
The detection techniques noted above can be used initially to verify that the method of the present invention is working, i.e., that the fusion pilin protein has been expressed and transported to the bacterial cell surface and is orientated so that the target protein is accessible i.e., displayed.
Cells that display the target may be separated from those which do not, using, for example, affinity separation techniques. Such techniques include affinity column chromatography, batch elution from affinity matrix material and fluorescent-activated cell sorting.
A bacterial display library produced in accordance with the present invention can be separated by affinity chromatography just as with the phage. Because bacterial cells are larger, care must be taken during loading to prevent plugging and the non-specific retention of bacteria in the column. Subsequently, the cells can be eluted either by passing free antigen through the column or by low pH. Even though gram-negative bacteria are not as resistant to low pH as are phage, there is no meaningful decrease in cell vitality for at least 10 minutes at pH 3.3 [Martineul, et al., Bio/Technology, 9: 1 70 ( 1 991 )]. Thus, elution by low pH and rapid neutralization can be employed for the isolation of strong binding clones.
The host cells displaying the desired target protein (display bacteria) may then be further cultured and used to obtain the fusion protein. If desired, the target protein may be separated from the pilin protein and further purified using pilin purification techniques familiar to the artisan [J. Bacteriology, 146( 1 ):251 -259 ( 1 981 )].
Once a desired target protein has been displayed, one can mutate the DNA encoding the heterologous polypeptide, e.g., by use of a mutator strain, and use affinity separation technology to identify and select peptides that bind to one or more targets.
The display method of the present invention can be used for the detection and characterization of recombinant proteins. For example, the method can be used to map an uncharacterized epitope as follows: Sequences encoding either a library of (1 ) random peptides or (2) peptides derived from the immunoreactive protein of interest can be cloned into a traA expression vector of the present invention, e.g., pPR35. E. coli host cells capable of forming F pili (e.g. XL1 B) are then transformed with the vector bank and the peptide library-traA fusion proteins are displayed on the bacterial cell surface. Following growth on a a solid substrate, e.g., a nylon membrane, the resulting bacteria are screened for expression of the fusion protein that react with labeled antibody. Reactive colonies can then be picked and the vectors isolated. Sequence analysis of the DNA insert would reveal which of the cloned peptides sequences corresponded to the epitopes recognized by the antibody.
The display method of the present invention can also be for detecting recombinant protein activity e.g., antibodies. For example, the method can readily be applied to screening libraries of recombinant antibody-traA fusion proteins. These libraries may include combinatorial single-chain gene banks of heavy and light variable region genes or mutational libraries of specific recombinant antibody genes [Reviewed in Whitlow, M & Filpula, D. (1991 ). Methods: A Companion to Methods in Enzymology 2:97-105.] On the basis of the results set forth in Examples 6 and 7 indicating that the σ-CKMB scFv-traA fusion protein is folded into a biologically active conformation, this method has general application to detection of recombinant protein activities displayed on the surface of the bacterial cell colony. The activities to be detected could include binding activities, catalytic activities, inhibitory activities and altered structural conformations.
The present invention can also be used as a primary cloning system. For example, a cDNA library can be constructed and inserted in a vector of the present invention and the library screened for the ability to bind a ligand. The ligand/binding molecule combination could include any pair of molecules with an ability to specifically bind to one another, e.g., receptor/ligand, enzyme/substrate (or analog), nucleic acid binding protein/nucleic acid, etc. If one member of the complementary pair is available, this may be a preferred way of isolating a clone for the other of the pair.
As discussed above, it will often be necessary to increase the diversity of a population of genes cloned for the display of their proteins on a bacterial surface or to mutate individual nucleotide sequence. In vitro or in vivo mutagenesis techniques can be used for either purpose and are well known to the skilled artisan. Alternatively, mutator strains can be used. A mutator strain is a strain which contains a genetic defect which causes DNA replicated with in it to be mutated with respect to its parent DNA. Such strains include those carrying the mut D5 mutation such as ES 1 578. Therefore, if a population of genes is introduced into these strains, it will be further diversified and can be transferred to a non-mutator strain if desired, for display and selection.
Since the F pili acts a receptor for the RNA bacteriophage and filamentous DNA phage, the display method of the present invention can make use of a binding protein on the phage to target the phage genome to a particular bacterial cell displaying a protein recognized by the phage. For example, instead of having the pilus/bacteriophage interaction that allows the phage to enter the cell, an antigen/antibody interaction can be used to allow the bacteriophage to interact with the pili and then enter the cell. For filamentous phage, the product of gene III acts as the attachment protein, it is believed, through interactions with residues near the N- terminus of the pilin protein. The gene III protein is made up of specific domains involved in incorporation into the page coat, phage morphology, interactions with the bacteria pilus, and entry into the bacteria cells, as depicted in Figure 10.
In addition to filamentous bacteriophage, RNA bacteriophage, such as Qβ, MS2, f2 and R17, specifically interact with the F pilus and infect the cells. The ability to absorb to the pilus is conferred by maturation A protein (or A2 for Qβ) which is present in one copy per virion [Paranchych, W. ( 1 975) in RNA Phages, ed. N.D.Zinder. (Cold Spring Harbor Laboratory:New York), pp.85-1 12]. Like the gene III protein of filamentous bacteriophage, the RNA phage maturation A protein can be used to form fusions without affecting infectivity.
In accordance with this method, a display bacteria is formed in which one protein of the specific binding pair is displayed and replaces the natural receptor for bacteriophage infection. A bacteriophage is also altered such that the normal pilin interaction domain is substituted with the other member of the specific binding pair. This is accomplished by removing the region of the phage attachment protein (e.g. gene III protein of filamentous phage or the A protein of RNA phage) that encodes the pilin binding domain and inserting in its place DNA that encodes the second member of the specific binding pair. The chimeric gene may be incorporated into the phage genome or a recombinant phagemid expression vector. The gene is then expressed in the appropriate strain, e.g. E. coli, and the fusion protein and the corresponding phage (or phagemid) genome are packaged into the bacteriophage particles. The phage is then contacted with the display bacteria under standard conditions. Phage displaying one member of the specific binding pair recognize and infect the bacteria displaying the other member based on the protein-protein interactions between the displayed proteins. The phage genome is then internalized by the display bacteria. Display bacteria infected with the phage genome can then be selected by, for example, identifying of a marker gene i.e., antibiotic resistance, transferred from the phage to the display bacteria. DNA encoding members of the specific binding pair can then isolated from the display bacterial host. Using fd phage, for example, the phage is altered such that the normal pilin interaction domain (e.g., amino acid 107 to 1 97 of the gene III protein) is removed and replaced by a polypeptide which will specifically bind the target protein displayed on the display bacteria. Thus, the display phage recognizes and infects the display bacteria solely based on the protein-protein interactions between the displayed recombinant proteins. Figure 1 1 shows the general characteristics of this system. The phage or phagemid genome is then internalized and expressed. Control signals for transcription, translation and replication can be present. It is particularly useful if the phage or phagemid genome contain sequences useful in selecting for the desired target cell. Useful sequences include, for example, those conferring antibiotic resistance to the target cell.
Bacteriophage useful in the method of the present invention include filamentous phage and RNA phage that utilize as a receptor the pilin protein. Such phage include MS2, Qβ, M13, f1 , fd and fd-tet. In addition, phagemid expression vectors derived from such filamentous phage can also be used. These vectors can carry plasmid and phage origins of replication and genes that confer antibiotic resistance. The preferred phage is fd-tet [Zacher, A.N., Stock, C.A., Golden, J.W. and Smith, G.P. ( 1 980) Gene
9, 127-140] and the preferred phagemid is a derivative of fl phage such as pBC (Stratagene).
As an example of this method, the EE tag antigen is displayed on the bacteria pili as a traA fusion using a phagemid expression vector that has been developed to allow for recombinant proteins to be displayed on the surface of bacteriophage. Using this system, the anti-EE tag scFv is displayed on the surface of the bacteriophage particles as a fusion with the gene III protein that has the pilin binding region (amino acid 107-1 97) deleted (this protein will be referred to as genelllp ΔBS as shown in Figure 10). Interactions between the anti-EE tag scFv antibody and the EE tag antigen are measured by the ability of the display phage to infect the display bacteria. Specific strategies for generating the display bacteriophage and for measuring infection are set out in Example 8 below.
Other recombinant proteins can be displayed on the bacteriophage and bacterial cell surface. These can include libraries of scFv genes displayed on the phage and a specific antigen peptide on the display bacteria. Screening for specific scFv-antigen interactions involves 1 ) rescue of the scFv display phagemid particles and 2) mixing the phage with the antigen displaying bacteria and testing for the presence of a marker e.g., infectivity by growth on agar plates containing antibiotics (chloramphenicol). The method of the present invention does not require antigen purification or the multiple rounds of enrichment and phage amplification steps that are currently required in phage display systems.
Phage or phagemid DNA would be isolated from the resulting antibiotic resistant colonies and the candidate scFv genes could be sequenced. Once the initial characterization is completed, the candidate scFv genes could be subcloned into bacteria expression vectors for the production and further characterization of the single-chain antibodies.
A bacteriophage vector based system can also be constructed for display of the recombinant proteins. Such a method has the advantages that it can be used to genetically select for high affinity protein-protein interactions and for binding affinity improvement when coupled with random or site-directed mutagenesis of the recombinant protein. As an example, an expression vector is constructed from the fd-tet phage by replacing the normal genelll with the anti-EE tag scFv-genelllp ΔBS fusion gene as outlined in Figure 13. DH5-σF' cells are transformed to tetracycline resistance with the phage expression vector. The transformed cells are be grown overnight, for example, in 100 ml of 2xYT media containing 1 5 μg/ml tetracycline. The cells are removed by centrifugation and the phage particles in the culture media can be concentrated by precipitation with, for example, 5% PEG and 0.5 M NaCI. The resulting phage particles carry the genelllp ΔBS phage vector and display the anti-EE tag scFv-genelllp ΔBS fusion protein of the bacteriophage surface. These phage particles are used to infect DH5-σF' cells carrying the EE tag-traA - 23 -
fusion vector. Infectivity can be tested by selection of tetracycline resistant colonies on agar plates as previously described. Alternatively, since the expression phage is able to replicate and re-infect bacteria displaying the EE tag, infectivity can be characterized by the formation of plaques on a lawn of the display bacteria or the propagation of the phage is liquid cultures of the display bacteria. Plaque size or phage titer liquid media provides an indication of the strength of the recombinant protein- protein interactions responsible for the phage infectivity and propagation. In other words, the highest affinity recombinant protein-protein interactions between the display phage and the display bacteria results the highest infectivity rates. The specificity of the infection can be tested with cells that do not display the EE tag antigen.
This system is useful in screening libraries of recombinant protein such as scFv. Phage displaying the high affinity scFv can infect and replicate in the antigen displaying bacteria at a higher rates than the phage displaying low affinity scFv. Thus, the phage displaying the high affinity scFv will be selectively enriched with continued growth of the culture. This is true for other specific binding pairs as well. The resulting phage DNA can be isolated and the candidate scFv genes and proteins further characterized by sequence and affinity analyses.
This system can further be used to screen compounds, i.e., inhibitors or co-factors, that affect specific binding pair interaction. In this screening method, the display bacteria and the display phage are mixed and infectivity of the display phage or phagemid particles is measured as previously described. One such detection method would be antibiotic- resistant growth of the display bacteria following infection with the display phage carrying the antibiotic resistance gene. Candidate compounds are added to the binding reaction and the effect on the level of phage infectivity is measured. For example, the suppression of growth of the display bacteria in appropriate selective media is one means of screening a large number of candidate inhibitor molecules. Compounds potentiating binding can be selected by screening for increased growth.
The present invention is further illustrated by the following Examples. These Examples are provided to aid in the understanding of the invention and are not construed as a limitation thereof.
The references cited above and below are herein incorporated by reference.
EXAMPLE 1
CONSTRUCTION OF A traA FUSION VECTOR FOR
EXPRESSING PROTEINS ON THE BACTERIAL SURFACE
A system was designed to allow inducible expression and display of polypeptides fused to the amino terminus of the pilin protein on the surface of bacteria. In this system, the gene encoding the polypeptide of interest was cloned into the traA vector, pPR35 and expressed in an F+ bacteria strain. The traA expression vector is based on the multicopy pUC1 9 vector with features shown in Figure 1 . The traA leader and traA protein (pilin) DNA fragments were cloned downstream of the inducible lac∑ promoter of pUC1 9. The traA leader allows for proper processing and display of the pilin fusion protein. Cloning sites for Nco\, Sfi\ and Not\ were incorporated between the traA leader and pilin polypeptide sequences to allow insertion of foreign DNA sequences. In addition, a DNA sequence encoding the EE tag antigen was cloned between the traA leader and traA protein sequences to allow for detection of the fusion protein and characterization of the expression-display system.
The steps required to construct the pPR35 vector are outlined in Figure 2 and detailed as follows. The traA leader and traA protein gene fragments were amplified separately by PCR from an F plasmid template. The primers used in the amplification are described in Figure 2 and Figure 3. Typical PCR amplification reactions ( 100 /I) contained 105 boiled XL1 B bacteria cells carrying the F plasmid as source of template DNA, 10 pmoles of the appropriate primers, 2.5 units of Taq polymerase, 100/yM dNTP, 50mM KCI, 10mM Tris-HCI, pH 8.3, 1 .5mM MgCI2, 0.01 % gelatin. The template was denatured by an initial incubation at 96°C for 5 min. during which the Taq polymerase was added to hot-start the reaction. The desired products were amplified by 10 thermal cycles of 55°C for 1 min., 70°C for 1 min. and 96°C for 1 min. followed by 20-step cycles of 70°C for 1 min. and 96°C for 1 min. Amplification with the primers results in the addition of an £coRI site on the 5' end of Λ col and Bam \ sites on the 3' end of the traA leader fragment and Bam \ and Kas\ sites on the 5' end and an Xba\ site on the 3' end of the traA protein fragment. The PCR products from 5 reactions were pooled, precipitated with 2 volumes of ethanol/0.3M sodium acetate, and the resulting products (about 0.2 μg of DNA) were resuspended in water. The traA leader PCR product was digested with EcoRI and Bam \ and the traA protein PCR fragment was digested with Bam \ and Xba\. The digested fragments were resolved by agarose gel electrophoresis and purified by elution from the agarose gel. In order to clone these fragments, a vector referred to as pPR5 was generated by digesting pUS1 8 DNA with Kas\, filling-in the site with Klenow DNA polymerase and religating the blunt ends. The purified digested PCR products were then ligated into Eco~\IXba\ digested pPR5. Bacteria transformed with this ligation mix were screened for the product on the three fragment ligation. Shown in Figure 2, this vector is referred to as pPR2. Finally, the EE tag linker sequence was generated by two complementary oligonucleotide which was annealed have a Ncσl sticky end at the 5' end and a Kas\ sticky end at the 3' end. The annealed oligonucleotide were ligated into NcollKas\ digested pPR2 to give the traA fusion vector, pPR35. The sequence of pPR35 is shown in Figure 4.
EXAMPLE 2
ISOLATION OF SINGLE-CHANGE ANTIBODY GENE AND CLONING INTO THE traA FUSION VECTOR
The traA fusion vector has been designed to express both peptide antigens such as the EE antigen as well as other recombinant proteins such as single chain antibodies. For the purpose of this example, single-chain antibody genes were created in which the heavy and light variable regions of a particular monoclonal antibody were joined together by a flexible polypeptides linker. Single-chain antibody (scFv) genes were generated from a monoclonal antibody (TA1 ) directed against the prothrombin polypeptide F1 .2 and from a monoclonal antibody directed against creatine kinase-MB (σ-CKMB) as described below and outlined in Figure 5. For the TA1 -ScFv, the first step involved poly-A RNA isolation from TA1 hybridoma cells by using the Fast-track RNA isolation kit (Invitrogen) according to manufacturer's procedures. This RNA ( 1 /10 of the mRNA isolated was used) was converted to cDNA using Superscript-MLV Reverse Transcriptase (GIBCO-BRL) and oligo-dT specific priming according to manufacturer's procedures. Of the 20 μ\ of cDNA generated, 2 μl was used as template DNA for PCR. The PCR primers for amplifying the TA1 mAb heavy and light chain variable region genes are JS1 35/JS1 34 and JS1 33/JS1 53, respectively, as shown in Figure 2. The PCR buffer conditions are the same as described in Example 1 . The template was denatured by an initial incubation at 96°C for 5 min. during which the Taq polymerase was added to hot-start the reaction. The immunoglobulin variable region gene fragments were amplified by 10 thermal cycles of 48°C for 1 min., 70°C for 1 min., and 96°C for 1 min. followed by 25- step cycles of 70°C for 1 min. and 96°C for 1 min. The desired products (about 260 bp) were resolved by agarose gel electrophoresis and purified by elution from the agarose gel. These fragments were then used as DNA templates in PCRs to attach a 45 nucleotide linker sequence to the 3' end of the heavy chain and the 5' end of the light chain variable gene fragment, resulting in the addition of a flexible 1 5 amino acid peptide linker to the variable region polypeptides. The PCR primers used in the linker attachment are JS135/JS1 39 and JS137/JS1 53 for the heavy and light chain variable gene fragments, respectively. The PCR conditions were 10 thermal cycles of 48°C for 1 min., 70°C for 1 min., and 96°C for 1 min., followed by 25-step cycles of 70°C for 1 min., and 96°C for 1 min. Following resolution by agarose gel electrophoresis, the desired products (about 400 bp) purified by elution from the agarose gel. Sequence-overlap extension PCR was used to link the heavy and light chain variable gene fragments by first annealing and extending the heavy chain + light chain variable + linker gene fragments for 10 thermal cycles of 52°C for 1 min., 70°C for 1 min., and 96°C for 1 min. The linked fragments were then amplified by the addition of JS135/JS1 53 primers and 1 5 additional step cycles of 70°C for 1 min. and 96°C for 1 min. The desired products
(about 720 bp) were purified as described above. Initially, the TA1 scFv gene fragment was digested with Ncol and Spel and ligated into the pJS102 cloning vector digested with Nco\ISpe\. The resulting construct was sequenced to verify that it contains the TA1 scFv gene. The pJS102/TA1 scFv plasmid was then used as template DNA to PCR the TA1 scFv gene fragment in order to add a Not\ site to the 3' end of the light chain variable gene. The primers used were JS1 35/JS1 53 and the PCR conditions were 10 thermal cycles of 48°C for 1 min., 70°C for 1 min. and 96°C for 1 min. followed by 25-step cycles of 70°C for 1 min. and 96°C for 1 min. The desired products (about 720 bp) were resolved by agarose gel electrophoresis and purified by elution from agarose gel. The TA1 scFv gene fragments were digested with Nco\ and Not\ and ligated into the pPR35 traA expression vector digested with Nco\INot\, resulting the creation of the TA1 scFv/EE tag/traA fusion vector, pGH21 .
The same strategy was used to isolate the variable region genes from Conan σ-CKMB hybridoma cell line and to construct the σ-CKMB scFv gene. The corresponding heavy and light chain PCR primers are shown in Figure 3. Following the sequence-overlap expression PCR step, the σ-
CKMB scFv gene fragment was digested Ncol and Spe\ and ligated into the pGH21 traA expression vector digested with Nco\ISpe\, essentially swapping the TA1 scFv gene for the σ-CKMB scFv gene. The resulting construct is referred to as pσ-CKMB scFv-traA.
EXAMPLE 3
PRODUCTION OF traA FUSION PROTEINS
The traA expression system was characterized in several ways.
First, bacterial expression of the TA1 scFv-EE tag-traA or σCKMB scFv-EE tag-traA fusion protein was examined by immunoblot analysis. The pGH21 and pσ-CKMB scFv-traA vectors were transformed into XL1 B cells carrying the F plasmid. Correct candidates were screened by restriction analysis of alkaline-SDS miniprep DNA and verified by DNA sequencing. To induce the expression of the traA fusion protein, 60 μl of an overnight culture was used to inoculate 3 ml of 2xLB media, 50 μg/ml ampicillin, 1 5 μg/ml tetracycline. Following a 2 hour incubation at 37 °C, isopropyl-1 -thio β-D- galactoside (IPTG) was added to 2mM final concentration. After 4 hours at 37°C, the OD600 of the culture was determined and 2 ml of the culture was harvested by microcentrifugation for 5 min. The cell pellet was frozen at -70°C and then was resuspended at 10 ODs/ml in cold TxTBS (0.1 % Triton C-100, 10mM Tris-HCI, pH 7.4, 0.1 5M NaCI). The cells were sonicated for 3 to 5 min. and the cell debris removed by microcentrifugation at 10,000 x g for 10 min. at 4°C. The supernatant (10 μl) was mixed with SDS/ ?-mercaptoethanol loading buffer and boiled for 5 min. to denature the proteins. The samples were resolved by SDS- polyacrylamide gel electrophoresis on 1 2.5% polyacrylamide gels. The material in the gels was transferred to PVDF nylon membranes using a semi-dry transblot apparatus. The membrane was blocked overnight at 4°C with 20 ml of blocking buffer (0.5% NP-40, 0.5% non-fat dried milk in PBS) and probed with 20 ml of 43ng/ml anti-EE tag mAb conjugated to horseradish preoxidase (anti-EE tag mAb-HRP). The anti-EE tag mAb-HRP was detected by the ECL reagent (Amersham) . The signal for the σ-CKMB scFv-traA fusion protein was detected at the expected molecular weight of 40 kD, while lysates from XL1 B/vector alone showed no signal. The TA1 scFv-traA fusion protein migrates at 46 kD, however, the TA1 scFv protein migrates through SDS-PA gels at a higher molecular weight than expected. The TA1 scFv-traA fusion protein was also detected in the growth media, consistent with the fact that F pili can detach from the cell surface and be found in the media. XL1 -B/pGH21 cells ( 1 L) were grown and TA-1 /EE tag/traA fusion protein expression was induced with IPTG as described in Example 2. The bacteria pilin protein was partially purified by shearing the pili from the cells and PEG precipitation as described by Moore, et al. [J. Bacteriology, 146( 1 ):251 -259 ( 1 981 )]. The fusion proteins present in the induced cells (cell lysate lane) and in the partially purified protein (PEG ppt lane) were examined by Western analysis using the anti-EE tag mAb-HRP as a probe. See, Figure 1 5. The band corresponding to the TA-1 /EE tag/traA fusion protein is indicated. The immunoreactive material at the top of the stacking gel is aggregated fusion protein that does not enter the resolving gel.
EXAMPLE 4
DETECTION OF THE ANTIGEN-traA FUSION PROTEIN
ON THE BACTERIAL SURFACE BY CLONING SCREENING
The traA expression system was used to develop improved methods for the detection of recombinant proteins. Two simple detection methods were performed to test whether the antigen-traA fusion protein was displayed on the surface of the bacteria cells. The first was an immunodetection method for screening for bacterial colonies grown on nylon membranes. The XL1 B strain expressing the TA1 -EE tag-traA fusion protein was spread on a nylon membrane and the membrane was placed on 2xLB agar plate containing 50 μg/ml ampicillin and 1 5 μg/ml tetracycline for selection of the vector and XL1 B strain, respectively. For induction of the traA fusion gene expression, the membrane was prewet with 10mM IPTG. Following overnight incubation at 37°C, the membrane was removed from plate and washed 3 times by cold Imidazole buffer saline (IBS - 40 mM Imidazole, pH 7.0, 0.1 5M NaCI). Membrane was blocked with 0.5% milk-PBS with agitation at 4°C for 1 hour, and then incubated for 1 to 2 hours with 43 ng/ml anti-EE tag mAb-HRP in IBS at 4°C. Following 5 washes with IBS at 4°C, the membranes were reacted with ECL reagents and the immunoreactive material was detected. By this colony immunoblot methodology, anti-EE tag mAb-HRP recognized the IPTG-induced XL1 B/TA1 -EE tag-traA colony but not the non-induced XL1 B/TA1 -EE tag-traA colony. XL1 B cells carrying a control vector (no EE tag-traA) failed to give any signal. The specificity of binding of anti-EE tag mAb on cell surface was also determined by incubating the colony membrane with an antibody to a different peptide tag (KT3). No signal was detected on these membranes.
The bacteria colony immunodetection method was also applied to epitope mapping analysis. To test this method, mixtures of XL1 B cells carrying either the TA1 -EE tag-traA or the control vector (no EE tag-traA insert) were grown overnight on 2xLB agar plates containing 50 μg/ml ampicillin and 1 5 μg/ml tetracycline overnight. The colonies were replica- plated onto nylon membranes and placed on 2xLB agar plates containing 10mM IPTG, 50 μg/ml ampicillin and 1 5 μg/ml tetracycline. Following growth at 37°C, the colonies on the membranes were probed with anti-EE tag mAb-HRP as described above. In the IPTG induced samples, positive signals were detected for single colonies as shown in Figure 6. The corresponding colonies were picked from the master plate for characterization and were found to carrying the TA1 -EE tag-traA vector.
EXAMPLE 5
WHOLE CELL ELISA TO DETECT ANTIGEN EXPRESSED ON THE BACTERIAL SURFACE The second method to test the accessibilty of the antigen-traA fusion protein on the surface of the bacteria was an ELISA method with intact cells. Cell grown to early log-phase were induced by IPTG for 4 hours at 37°C. The cells were harvested and resuspended in cold PBS to 1 .0 0D595/ml. This step will remove any traA fusion protein present in the media that is not associated with the cells. Microtiter plates were coated with 100μl of bacterial dilution per well. After overnight incubation at 4°C, unattached cells were discarded and wells were blocked with PBS containing 1 % bovine serum albumin for 1 hour at 4°C. Following the blocking step, the wells were incubated with 100 ul of 0.34 μg/ml anti-EE tag antibody HRP. After 5 times washes with PBS, antigen-antibody complexes were developed by HRP-ELISA substrate (H2O2, ABTS peroxidaes substrate). Reaction values were recorded by ELISA reader. The OD450 reading indicates the amount of anti-EE tag mAB-HRP activity captured in each well and correlates with the amount of EE tag fusion protein expressed on the cell surface. The induced XL1 B/TA1 -EE tag-traA samples showed greater than six-fold higher readings than the non-induced sample or the XL1 B/control vector (no EE tag-traA insert) sample as shown in Figure 7, indicating that this method is applicable to specifically detecting antigens presented on the cell surface. By adding known amounts of peptide antigen and antibody to the binding reaction, this method could be used to quantitative antibody/antigen binding. In addition, the epitope could be characterized in a comparative ELISA assay format where the effect of different peptides on antibody/antigen-traA fusion protein interaction is determined.
EXAMPLE 6
WHOLE CELL ELISA TO DETECT THE ACTIVITY OF A RECOMBINANT ANTIBODY DISPLAYED ON THE BACTERIAL SURFACE The results from Examples 4 and 5 indicate that antigens fused to the traA protein could be displayed on the surface of bacteria and could be specifically detected by the corresponding antibody. Similar experiments were carried out to determine if a functional recombinant protein could be displayed of the bacterial surface as described below. To detect the activity of recombinant single chain anti-CKMB Ab displayed on the bacteria cell surface, an ELISA method with intact cells was performed. Cells carrying either the σ-CKMB scFv-EE tag-traA fusion vector or the control vector (TA1 -EE tag-traA fusion vector or a vector without an insert) were grown to early log-phase at 37°C. At that point, expression of the fusion protein was induced by the addition of 0.2mM IPTG for 4 hours at 37°C. The cells were harvested and resuspended in cold PBS to 10.0 0D595/ml. Microtiter plates were coated with 100 μg/ml anti-CK-BB mAb in coating buffer (0.1 M Tris-HCI, pH8.5) and were incubated overnight at 4°C. Unattached anti-CK-BB mAb was discarded and the wells were washed once with washing buffer (0.1 M Tris-HCI, pH 7.4, 1 .0M NaCI, 0.1 % NaN3). The wells were incubated with 100μl of 0.3 μg/ml CK-MB in dilution buffer (2% gelatin, 0.1 % Tween 20 in 0.01 m Tris-HCI, pH7.3, 0.1 5 M NaCI) at room temperature for 1 hour with agitation. The wells were washed once with rinse buffer (0.01 M Tris-HCI, pH7.3, 0.1 5M NaCI, 0.2% BSA, 0.05% Tween-20, 0.2% NaN3). 100 μl of cell suspension was added to each well and the plate was incubated at room temperature for 1 hour with agitation. The unattached cells were discarded and the wells were washed twice with rinse buffer. The wells were incubated with 100μl of 0.34μg/ml anti-EE tag mAb-HRP conjugate in dilution buffer at room temperature for 1 hour with agitation. After washing the wells twice with rinse buffer, the HRP-ELIA substrate (H2O2, ABTS peroxidase substrate) was added and developed for 20 min. The amount of color development as determined by an ELISA reader at 450 nm corresponds to the amount of scFv-EE tag-traA fusion protein detected in the well. These values correlate with the amount of anti-CKMB scFv activity displayed of the cell surface that is captured by the immobilized CK-MB. Typical results are shown in Figure 8. The IPTG-induced XL1 B/σ-CKMB scFv-EE tag-traA plasmid sample showed 29-fold higher levels of captured σ-CKMB scFv-EE tag-traA fusion protein than the non-induced XL1 B/TA1 -EE tag-traA cells that express a different recombinant antibody which does not recognize CK-MB. The XL1 B cells carrying the control vector did not express EE tag- traA protein and showed no activity in this assay.
The results of these experiments indicate that the σ-CKMB scFv domain of the fusion protein is folded into a biologically active conformation and is displayed on the surface of the cells. The combination of the single-chain antibody and the EE antigen tag on the same display protein allows for versatility in the development of ELISA formats. The sandwich capture ELISA format used in this Example is just one of the many possibilities.
EXAMPLE 7
DETECTION OF FUNCTIONAL SINGLE-CHAIN ACTIVITY
OF THE BACTERIAL SURFACE BY COLONY SCREENING
This example demonstrates the successful detection of a recombinant protein activity expressed on the surface of the bacterial cell colony. A mixture of the XL1 B strain carrying the σ-CKMB scFv-EE tag- traA fusion vector and the strain carrying the TA1 -EE tag-traA fusion vector was distributed evenly on a nylon membrane. The membrane was placed on a 2xLB agar plate containing 50μg/ml ampicillin and 1 5μg/ml tetracycline and incubated at 37°C until small bacterial colonies appeared. At this time, a replica membrane was made by overlaying the master membrane with a new membrane. The replica membrane was then removed and cut in half. One half was incubated on a 2xLB agar plate containing 10mM IPTG, 50μg/ml ampicillin and 1 5μg/ml tetracycline and the other half was on a 2xLB agar plate containing just 50μg/ml ampicillin and 1 5μg/ml tetracycline. Following the overnight incubation at 37°C, the IPTG-induced and non-induced membranes were removed and washed 3 times by IBS. Membranes were blocked with 20ml of 0.5% milk-IBS with agitation at 4°C for 1 hour, and then incubated for 1 hour at 4°C with 20 ml of 0.3 μg/ml CKMB in the dilution buffer. Following 3 washes with cold IBS, the membranes were blocked at 4°C for 1 hour, and with anti-CK-BB mAb conjugated to alkaline phosphatase. Following 5 washes with cold IBS, the membranes were reacted with Lumiphos 53 (Boehringer Mannheim). The immunoreactive material was detected by fluorography and the typical results are shown in Figure 9. Strong positive signals corresponding to single colonies were detected in the IPTG-induced samples. These colonies were picked from the master plate and found to carry the σ-CKMB scFv-EE tag-traA vector. The colonies corresponding to negative signal on the film contained the control vector. The results indicate that the σ-CKMB scFv-traA fusion protein is displayed of the bacterial surface and the single-chain antibody is folded into a biologically active conformation.
This method provides an easy rapid procedure for detecting recombinant single-chain antibody activity. It could be readily applied to screening libraries of recombinant antibody-traA fusion proteins. These libraries may include combinatorial single-chain gene banks of heavy and light variable region genes or mutational libraries of specific recombinant antibody genes. On the basis of the results indicating that the σ-CKMB scFv-traA fusion protein is folded into a biologically active conformation, this method could have general application to detection of recombinant protein activities expressed on the surface of the bacterial cell colony. The activities to be detected could include binding activities, catalytic activities, inhibitory activities and altered structural conformations.
EXAMPLE 8 BACTERIOPHAGE/PILIN INTERACTION SYSTEM
A phagemid vector was designed for the expression and display of genelllp ΔBS fusion proteins on the surface of the bacteriophage particle. The phagemid vector is based on the pBC phagemid vector (Stratagene) with features shown in Figure 1 2. This vector carries the ColEI replication origin for plasmid propagation, the f1 filamentous phage replication origin for recovery of phagemid DNA following co-infection with helper phage and the chloramphenicol resistance gene for antibiotic selection. The pe/B leader and genelllp ΔBS DNA fragments were cloned downstream of the inducible lacZ promoter of pBC. The pe/B leader was designed to allow for proper processing and display of the fusion protein on the bacteriophage particle. Cloning sites for Ncσl, Sfi\, Spe\, and Nofl were incorporated between the pe/B leader and genelllp ΔBS sequences to allow insertion of foreign DΝA sequences. The steps involved in constructing this vector (referred to as LE2) are shown in Figure 12.
The anti-EE tag scFv gene are isolated from monoclonal hybridoma mRΝA as outlined in Example 2 and are inserted at the Nofl and Spe\ sites of LE2. An F' host strain, DH5-σF' [Woodcock, D:M. et al ( 1 989) Νucl. Acids. Res. 1 7,3469-3478] is used to propagate these vectors by growth in media containing 30 μg/ml chloramphenicol. In order to rescue phagemid particles, DH5-σF' cells carrying phagemid expression vector are transformed with the fKN1 6 phage DNA to tetracycline resistance. The fKN1 6 phage derivative was constructed from the tetracycline-resistance phage, fd-tet, by deleting a 507 bp segment of gene III (Nelson, et a/., Virology, 108:338-350 (1 981 )) . This phage is non- infective due to the gene III deletion but provides the helper phage proteins necessary for replication and packaging of the phagemid expression vector. DH5-σF' cells carrying both the phagemid expression vector and fKN1 6 are gown overnight in 100 ml of 2xYT media containing 30 μg/ml chloramphenicol and 1 5 μg/ml tetracycline. The cells are removed by centrifugation and the phage particles in the culture media are concentrated by precipitation with 5% PEG and 0.5 M NaCI. The resulting phage particles carry either the fKN1 6 phage (tetr) or the genelllp ΔBS phagemid vector (chlr). Both the defective fKN1 6 gene protein and the anti-EE tag scFv-genelllp ΔBS fusion protein are displayed on the bacteriophage surface. The rescued phage are used to infect XL-1 B cells carrying the EE tag-traA fusion vector. Since these cell express the EE tag antigen on their pili, the bacteriophage displaying the anti-EE tag scFv- genelllp ΔBS fusion protein bind the EE tag-traA fusion protein and infect these cells resulting in chloramphenicol resistant clones carrying the phagemid expression vector.
A bacteriophage vector based system can also be constructed for the display of recombinant proteins. The expression vector can be constructed from the fd-tet phage by replacing the normal genelll with the anti-EE tag scFv-genelllp ΔBS fusion gene as outlined in figure 14. DH5- σF' cells are transformed to tetracycline resistance with the phage expression vector. The transformed cells are grown overnight in 100 ml of 2xYT media containing 1 5 μg/ml tetracycline. The cells will be removed by centrifugation and the phage particles in the culture media will be concentrated by precipitation with 5% PEG and 0.5 M NaCI. The resulting phage particles will carry the genelllp ΔBS phage vector and display the anti-EE tag scFv-genelllp ΔBS fusion protein of the bacteriophage surface. These phage particles will be used to infect DH5-σ F' cells carrying the EE tag-traA fusion vector.
Infectivity can then be tested by selection of tetracycline resistant colonies on agar plates as described for the phagemid system. Alternatively, since the expression phage is able to replicate and re-infect bacteria displaying the EE tag, infectivity can be characterized by the formation of plaques on a lawn of the display bacteria or the propagation of the phage in liquid cultures of the display bacteria. Plaque size or phage titer in the liquid media provide an indication of the strength of the recombinant protein-protein interactions responsible for the phage infectivity and propagation. In other words, the highest affinity recombinant protein-protein interactions between the display phage and the display bacteria provide the highest infectivity rates. The specificity of the infection can also be tested with cells that do not display the EE tag antigen.
This system is useful in screening libraries of recombinant protein such as scFvs. The phage displaying the high affinity scFv will infect and replicate in the antigen displaying bacteria at a higher rate that the phage displaying low affinity scFv. Thus, the phage displaying the high affinity scFv will be selectively enriched with continued growth of the culture. The resulting phage DNA can be isolated and the candidate scFv genes and proteins could be further characterized by sequence and affinity analyses. It should be 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 and purview of this Applications and the scope of the appended claims.
SEQUENCE LISTING
(1) GENERAL INFORMATION:
(i) APPLICANT: Huang, Grace P. Rhode, Peter R. Stinson, Jeffrey R. Wong, Hing C.
(ii) TITLE OF INVENTION: A METHOD FOR DISPLAYING
PROTEINS
(iii) NUMBER OF SEQUENCES: 24
(iv) CORRESPONDENCE ADDRESS:
(A) ADDRESSEE: David G. Conlin; DIKE, BRONSTEIN,
ROBERTS & CUSHMAN
(B) STREET: 130 WATER STREET
(C) CITY: BOSTON
(D) STATE: MASSACHUSETTS
(E) COUNTRY: US
(F) ZIP: 02109
(v) COMPUTER READABLE FORM:
(A) MEDIUM TYPE: Floppy disk
(B) COMPUTER: IBM PC compatible
(C) OPERATING SYSTEM: PC-DOS/MS-DOS
(D) SOFTWARE: Patentin Release #1.0, Version #1.25
(vi) CURRENT APPLICATION DATA:
(A) APPLICATION NUMBER: US 08/258026
(B) FILING DATE: 10-JUN-1994
(C) CLASSIFICATION:
(viii) ATTORNEY/AGENT INFORMATION:
(A) NAME: Resnick, David R.
(B) REGISTRATION NUMBER: 34235
(C) REFERENCE/DOCKET NUMBER: 42838
(ix) TELECOMMUNICATION INFORMATION:
(A) TELEPHONE: (617) 523-3400
(B) TELEFAX: (617) 523-6400
(C) TELEX: 200291 STRE UR
(2) INFORMATION FOR SEQ ID Nθ:l:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 35 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: unknown
(D) TOPOLOGY: unknown
(xi) SEQUENCE DESCRIPTION: SEQ ID Nθ:l: GGGGGGAATT CTATCCGAAA TTGAGGTAAC TTATG 35
(2) INFORMATION FOR SEQ ID NO:2:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 39 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: unknown (D) TOPOLOGY: unknown
(xi) SEQUENCE DESCRIPTION: SEQ ID Nθ:2: GGGGGGTCTA GATTATCAGA GGCCAACGAC GGCCATAAC 39
(2) INFORMATION FOR SEQ ID NO:3:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 41 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: unknown
(D) TOPOLOGY: unknown
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:3: GGGGGGATCC CCATGGCCAG CTGCGGGAAG AACATCATCA G 41
(2) INFORMATION FOR SEQ ID NO:4:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 40 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: unknown
(D) TOPOLOGY: unknown
(xi) SEQUENCE DESCRIPTION: SEQ ID Nθ:4: GGGGGGATCC GGCGCCGGCA GCAGTGGTCA GGACCTGATG 40
(2) INFORMATION FOR SEQ ID NO:5:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 33 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: unknown
(D) TOPOLOGY: unknown
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:5: CACTTGGCCA TGGCCGAGGT TCAGCTGCAG CAG 33
(2) INFORMATION FOR SEQ ID NO:6:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 34 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: unknown
(D) TOPOLOGY: unknown
(xi) SEQUENCE DESCRIPTION: SEQ ID Nθ:6: GCTGCCACCG CCACCTGAGG AGACGGTGAC TGAG 34
(2) INFORMATION FOR SEQ ID NO:7:
(i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 35 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: unknown
(D) TOPOLOGY: unknown
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:7: GGAGGCGGCG GTTCTGATAT TGTGATGACT CAGGC 35
(2) INFORMATION FOR SEQ ID NO:8:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 40 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: unknown
(D) TOPOLOGY: unknown
(xi) SEQUENCE DESCRIPTION: SEQ ID Nθ:8: TTCATAGGCG GCCGCACTAG TAGCMCGTTT CAGYTCCARC 40
(2) INFORMATION FOR SEQ ID NO:9:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 33 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: unknown
(D) TOPOLOGY: unknown
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:9: GCCGGCCATG GCCCAGGTBC ARCTKMARSA RTC 33
(2) INFORMATION FOR SEQ ID NO:10:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 35 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: unknown
(D) TOPOLOGY: unknown
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:10: GCTGCCACCG CCACCTGMRG AGACDGTGAS TGARG 35
(2) INFORMATION FOR SEQ ID NO:11:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 35 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: unknown
(D) TOPOLOGY: unknown
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:11: GGAGGCGGCG GTTCTGACAT TGTGMTGWCA CAGTC 35 (2) INFORMATION FOR SEQ ID NO: 12:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 40 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: unknown
(D) TOPOLOGY: unknown
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:12: TTCATAGGCG GCCGCACTAG TAGCMCGTTT KATYTCCARC 40
(2) INFORMATION FOR SEQ ID NO:13:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 45 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: unknown
(D) TOPOLOGY: unknown
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:13: GGTGGCGGTG GCAGCGGCGG TGGTGGTTCC GGAGGCGGCG GTTCT 45
(2) INFORMATION FOR SEQ ID NO: 14:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 45 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: unknown
(D) TOPOLOGY: unknown
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:14: AGAACCGCCG CCTCCGGAAG GAGGACCGCC GCTGCCACCG CCACC 45
(2) INFORMATION FOR SEQ ID NO:15:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 53 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: unknown
(D) TOPOLOGY: unknown
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:15: CATGGCGGCC GGCAGCGCGG CCGCTGAGGA AGAAGAGTAC ATGCCGATGG AAC 53
(2) INFORMATION FOR SEQ ID NO:16:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 53 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: unknown
(D) TOPOLOGY: unknown
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 16: GCGCCTTCCA TCGGCATGTA CTCTTCTTCC TCAGCGGCCG CGCTGCCGGC CGC 53
(2) INFORMATION FOR SEQ ID NO: 17:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 540 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: unknown
(D) TOPOLOGY: unknown
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 17:
GAAACAGCTA TGACCATGAT TACGAATTCT ATCCGAAATT GAGGTAACTT ATGAATGCTG 60
TTTTAAGTGT TCAGGGTGCT TCTGCGCCCG TCAAAAAGAA GTCGTTTTTT TCCAAATTCA 120
CTCGTCTGAA TATGCTTCGC CTGGCTCGCG CAGTGATCCC GGCTGCTGTT CTGATGATGT 180
TCTTCCCGCA GCTGGCCATG GCGGCCGGCA GCGCGGCCGC TGAGGAAGAA GAGTACATGC 240
CGATGGAAGG CGCCGGCAGC AGTGGTCAGG ACCTGATGGC AAGCGGTAAC ACCACGGTTA 300
AGGCGACCTT CGGTAAGGAC TCCAGTGTTG TTAAATGGGT TGTTCTGGCT GAAGTTCTGG 360
TCGGTGCTGT CATGTACATG ATGACCAAAA ACGTCAAGTT CCTGGCCGGT TTTGCCATCA 420
TCTCTGTATT TATTGCTGTG GTTATGGCCG TCGTTGGCCT CTGATAATCT AGAGTCGACC 480
TGCAGGCATG CAAGCTTGGC ACTGGCCGTC GTTTTACAAC GTCGTGACTG GGAAAACCCT 540
(2) INFORMATION FOR SEQ ID NO:18:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 137 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS: unknown
(D) TOPOLOGY: unknown
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:18:
Met Asn Ala Val Leu Ser Val Gin Gly Ala Ser Ala Pro Val Lys Lys 1 5 10 15
Lys Ser Phe Phe Ser Lys Phe Thr Arg Leu Asn Met Leu Arg Leu Ala 20 25 30
Arg Ala Val lie Pro Ala Ala Val Leu Met Met Phe Phe Pro Gin Leu 35 40 45
Ala Met Ala Ala Gly Ser Ala Ala Ala Glu Glu Glu Glu Tyr Met Pro 50 55 60
Met Glu Gly Ala Gly Ser Ser Gly Gin Asp Leu Met Ala Ser Gly Asn 65 70 75 80
Thr Thr Val Lys Ala Thr Phe Gly Lys Asp Ser Ser Val Val Lys Trp 85 90 95
Val Val Leu Ala Glu Val Leu Val Gly Ala Val Met Tyr Met Met Thr 100 105 110
Lys Asn Val Lys Phe Leu Ala Gly Phe Ala lie lie Ser Val Phe lie 115 120 125
Ala Val Val Met Ala Val Val Gly Leu 130 135
(2) INFORMATION FOR SEQ ID NO:19:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 67 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: unknown
(D) TOPOLOGY: unknown
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:19: GGGGGGAGCT CTCTGCAAAG AGACAGTCAT AATGAAATAC CTATTGCCTA CGGCAGCCGC 60 TGGATTG 67
(2) INFORMATION FOR SEQ ID NO:20:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 66 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: unknown
(D) TOPOLOGY: unknown
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:20: GGGGGGCCGC GGCCGCGGCC ATGGCCGGCT GGGCCGCGAG TAATAACAAT CCAGCGGCTG 60 CCGTAG 66
(2) INFORMATION FOR SEQ ID NO:21:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 84 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: unknown
(D) TOPOLOGY: unknown
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:21: GGGGGGCCGC GGCCGCGGAG GAAGAAGAGT ACAACCCGAA CGAAGGCGCC GCCTAGACTG 60 TTGAAAGTTG TTTAGCAAAA CCTC 84
(2) INFORMATION FOR SEQ ID NO:22:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 44 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: unknown
(D) TOPOLOGY: unknown
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:22: GGGCCGAATT CCTATTAAGA CTCCTTATTA CGCAGTATGT TAGC 44 (2) INFORMATION FOR SEQ ID NO:23:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 53 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: unknown
(D) TOPOLOGY: unknown
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:23: GGGGGGACTA GTGCGGCCGC GGGCGCCGCT GAAACTGTTG AAAGTTGTTT AGC 53
(2) INFORMATION FOR SEQ ID NO:24:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 35 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: unknown
(D) TOPOLOGY: unknown
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:24: GGGGGGGGAT CCAGAGGGTT GATATAAGTA TAGCC 35

Claims

WHAT IS CLAIMED IS:
1 . A method for displaying a target protein on a bacterial host cell comprising: expressing a fusion protein in a bacterial host capable of forming a pilus, said fusion protein encoded by a chimeric DNA comprising a DNA segment encoding a leader amino acid sequence capable of mediating secretion of the fusion protein, a DNA segment encoding pilin subunits capable of forming said pilus, and a DNA segment encoding the target protein, said DNA segments being operably linked such that the host cell displays said target protein at its surface.
2. The method of claim 1 , wherein said pilus is a receptor for bacteriophage attachment and infection.
3. The method of claim 2, wherein said pilus is an F pilus.
4. The method of claim 3, wherein the DNA segment encoding a leader segment encodes the traA leader sequence.
5. The method of claim 3, wherein the DNA segment encoding the pilin subunit encodes the tra A gene product.
6. The method of claim 1 , wherein the DNA segment encoding the target protein is linked between the DNA segment encoding the leader and the 5' end of the DNA segment encoding the pilin subunit.
7. The method of claim 1 , wherein the bacterial host cell is E. coli.
8. The method of claim 1 , wherein the chimeric DNA further comprises an inducible promoter operably linked thereto.
9. The method of claim 1 , wherein said chimeric DNA is carried within a recombinant vector.
10. A method for selecting a recombinant target protein from a DNA library including DNA encoding said recombinant target protein comprising:
(a) displaying the recombinant proteins encoded by said DNA library by the method of claim 1 ; and
(b) selecting bacterial host cells displaying the desired target recombinant protein.
1 1 . The method of claim 1 or 10, wherein the bacterial host is a mutator strain which introduces genetic diversity into the target protein.
12. The method of claim 10, wherein the target protein is an antigenic determinant polypeptide.
1 3. The method of claim 10, wherein the bacterial host cells displaying the desired target recombinant protein is selected by immunoreactivity against an antibody.
14. A method for isolating a member of a specific binding pair comprising:
(a) expressing a fusion protein in a bacterial host capable of forming a pilus that is a receptor for bacteriophage attachment and infection, said fusion protein encoded by a chimeric DNA comprising a DNA segment encoding a leader amino acid sequence capable of mediating secretion of the fusion protein, DNA segment encoding a pilin subunits capable of forming a pilus, and a DNA encoding said member;
(b) contacting the bacterial host of step (a) with a bacteriophage displaying an attachment protein having a pilin interaction domain wherein the DNA encoding the pilin interaction domain has been substituted with DNA encoding the other member of the specific binding pair; and
(c) selecting bacterial host cells recognized by the phage.
1 5. The method of claim 14, wherein said pilus is an F pilus.
1 6. The method of claim 1 5, wherein the phage is a filamentous phage or RNA bacteriophage.
1 7. The method of claim 1 6, wherein the phage is an fd phage.
1 8. The method of claim 14, wherein the DNA encoding the member is from a DNA library.
1 9. The method of claim 14, wherein the DNA encoding the other member is from a DNA library.
20. The method of claim 14, wherein the DNA encoding the member encodes an antigenic determinant and the DNA encoding the other member encodes an antibody.
21 . The method of claim 14, wherein the DNA encoding the member encodes an antibody and the DNA encoding the other member encodes an antigenic determinant.
22. The method of claim 17, wherein the bacteriophage attachment protein is the gene III protein.
23. The method of claim 14, wherein the bacterial host recognized by the phage is selected by identification of a marker gene transferred from the phage to the host.
24. The method of claim 14, wherein the DNA encoding a member of the specific binding pair is mutagenized and specific binding pairs having an increased affinity are selected.
25. A method of screening for compounds affecting specific binding pair interaction comprising:
(a) contacting a bacterial cell having a specific binding pair-pilin fusion protein displayed on its surface with a bacteriophage altered such that the pilin interaction domain is substituted by the other member of the specific binding pair;
(b) adding a test compound; and
(c) measuring the effect of the test compound on the phage/ bacterial cell interaction.
26. The method of claim 25, wherein the effect of the test compound on the phage bacterial cell interaction is measured by measuring the effect of the test compound on the infectivity of the display phage.
27. A method of screening for a member of a specific binding pair having increased binding affinity comprising:
(a) contacting a display bacteria displaying a pilin-specific binding member fusion with a bacteriophage altered such that the normal pilin binding domain is substituted with a library of proteins containing the other member of the specific binding pair under conditions that allow phage infection; and
(b) identifying specific binding pairs having increased binding affinity by identifying bacteriophage having an increase frequency of infectivity.
28. A bacterial host cell displaying a target protein-pilin fusion.
29. The bacterial host cell of claim 28, wherein the host is E. coli.
PCT/US1995/007541 1994-06-10 1995-06-12 A method for displaying proteins WO1995034648A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
AU28282/95A AU697865B2 (en) 1994-06-10 1995-06-12 A method for displaying proteins
JP8502436A JPH09504181A (en) 1994-06-10 1995-06-12 How to display protein
EP95923861A EP0722495A1 (en) 1994-06-10 1995-06-12 A method for displaying proteins

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US08/258,026 1994-06-10
US08/258,026 US5516637A (en) 1994-06-10 1994-06-10 Method involving display of protein binding pairs on the surface of bacterial pili and bacteriophage

Publications (1)

Publication Number Publication Date
WO1995034648A1 true WO1995034648A1 (en) 1995-12-21

Family

ID=22978788

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1995/007541 WO1995034648A1 (en) 1994-06-10 1995-06-12 A method for displaying proteins

Country Status (6)

Country Link
US (1) US5516637A (en)
EP (1) EP0722495A1 (en)
JP (1) JPH09504181A (en)
AU (1) AU697865B2 (en)
CA (1) CA2169358A1 (en)
WO (1) WO1995034648A1 (en)

Cited By (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997010330A1 (en) * 1995-09-13 1997-03-20 Bioinvent International Ab Combined ligand and receptor display
WO1999010485A1 (en) * 1997-08-29 1999-03-04 Selective Genetics, Inc. Methods using phage display for selecting internalizing ligands for gene delivery
WO1999011777A1 (en) * 1997-09-03 1999-03-11 Biovation Limited Methods for protein screening
EP0932670A4 (en) * 1996-03-25 1999-08-04
EP0944737A1 (en) * 1996-11-07 1999-09-29 RAMOT UNIVERSITY AUTHORITY FOR APPLIED RESEARCH & INDUSTRIAL DEVELOPMENT LTD. Representations of bimolecular interactions
US6040136A (en) * 1990-12-03 2000-03-21 Genentech, Inc. Enrichment method for variant proteins with altered binding properties
WO2000023465A2 (en) * 1998-10-19 2000-04-27 Mitotix, Inc. Methods and reagents for isolating biologically active peptides
WO2000029555A1 (en) * 1998-11-17 2000-05-25 Selective Genetics, Inc. Methods using genetic package display for detecting and identifying protein-protein interactions
WO2001094950A2 (en) * 2000-06-05 2001-12-13 Zyomyx, Inc. Screening of phage displayed peptides without clearing of the cell culture
WO2002046213A2 (en) * 2000-11-07 2002-06-13 Gpc Biotech Inc. Methods and reagents for isolating angiogenic modulators
US6448083B1 (en) 1997-08-29 2002-09-10 Selective Genetics, Inc. Receptor-mediated gene delivery using bacteriophage vectors
US6589730B1 (en) 1997-08-29 2003-07-08 Selective Genetics, Inc. Methods for identifying protein-protein interactions by selective transduction
US6723512B2 (en) 1997-08-29 2004-04-20 Selective Genetics Inc. Methods using genetic package display for detecting and identifying protein-protein interactions that facilitate internalization and transgene expression and cells or tissues competent for the same and methods for evolving gene delivery vectors
US6927025B1 (en) 1997-09-03 2005-08-09 Biovation Limited Methods for protein screening
EP2014303A2 (en) 2000-07-27 2009-01-14 Genentech, Inc. APO-2L receptor agonist and CPT-11 synergism
US8685893B2 (en) 1998-07-27 2014-04-01 Genentech, Inc. Phage display
EP2857516A1 (en) 2000-04-11 2015-04-08 Genentech, Inc. Multivalent antibodies and uses therefor

Families Citing this family (557)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7597886B2 (en) * 1994-11-07 2009-10-06 Human Genome Sciences, Inc. Tumor necrosis factor-gamma
US7820798B2 (en) * 1994-11-07 2010-10-26 Human Genome Sciences, Inc. Tumor necrosis factor-gamma
US7429646B1 (en) 1995-06-05 2008-09-30 Human Genome Sciences, Inc. Antibodies to human tumor necrosis factor receptor-like 2
US7888466B2 (en) 1996-01-11 2011-02-15 Human Genome Sciences, Inc. Human G-protein chemokine receptor HSATU68
US5922537A (en) * 1996-11-08 1999-07-13 N.o slashed.AB Immunoassay, Inc. Nanoparticles biosensor
US7041441B1 (en) * 1997-04-11 2006-05-09 The United States Of America As Represented By The Department Of Health And Human Services Phage display of intact domains at high copy number
ZA988460B (en) * 1997-09-17 1999-03-19 Genentech Inc Polypeptides and nucleic acids encoding the same
US6475785B1 (en) 1997-12-18 2002-11-05 Spectral Diagnostics, Inc. Single-chain polypeptides comprising troponin I N-terminal fragments and troponin C
US5981249A (en) * 1998-02-05 1999-11-09 Spectral Diagnostics, Inc. Single-chain polypeptides comprising creatine kinase M and creatine kinase B
AU3072799A (en) 1998-03-19 1999-10-11 Human Genome Sciences, Inc. Cytokine receptor common gamma chain like
EP1161451A4 (en) 1999-02-26 2006-05-17 Human Genome Sciences Inc Human endokine alpha and methods of use
WO2000053232A1 (en) * 1999-03-12 2000-09-14 Georgetown University Matriptase, a serine protease and its applications
CA2721199A1 (en) 1999-05-05 2000-11-16 Phylogica Limited Isolating biological modulators from biodiverse gene fragment libraries
US7803765B2 (en) * 1999-05-05 2010-09-28 Phylogica Limited Methods of constructing biodiverse gene fragment libraries and biological modulators isolated therefrom
US7270969B2 (en) 1999-05-05 2007-09-18 Phylogica Limited Methods of constructing and screening diverse expression libraries
US20040001826A1 (en) * 1999-06-30 2004-01-01 Millennium Pharmaceuticals, Inc. Glycoprotein VI and uses thereof
US7291714B1 (en) * 1999-06-30 2007-11-06 Millennium Pharmaceuticals, Inc. Glycoprotein VI and uses thereof
US6479280B1 (en) * 1999-09-24 2002-11-12 Vlaams Interuniversitair Institutuut Voor Biotechnologie Vzw Recombinant phages capable of entering host cells via specific interaction with an artificial receptor
US20060073509A1 (en) * 1999-11-18 2006-04-06 Michael Kilpatrick Method for detecting and quantitating multiple subcellular components
GB9928787D0 (en) * 1999-12-03 2000-02-02 Medical Res Council Direct screening method
IL149809A0 (en) * 1999-12-15 2002-11-10 Genentech Inc Shotgun scanning, a combinatorial method for mapping functional protein epitopes
CA2747325A1 (en) 2000-04-12 2001-10-25 Human Genome Sciences, Inc. Albumin fusion proteins
US7297762B2 (en) * 2000-04-24 2007-11-20 Yale University Modified avian pancreatic polypeptide miniature binding proteins
US7495070B2 (en) * 2000-04-24 2009-02-24 Yale University Protein binding miniature proteins
US20030031675A1 (en) 2000-06-06 2003-02-13 Mikesell Glen E. B7-related nucleic acids and polypeptides useful for immunomodulation
CA2413160A1 (en) 2000-06-15 2001-12-20 Human Genome Sciences, Inc. Human tumor necrosis factor delta and epsilon
CA2407910C (en) 2000-06-16 2013-03-12 Steven M. Ruben Antibodies that immunospecifically bind to blys
GB0022978D0 (en) 2000-09-19 2000-11-01 Oxford Glycosciences Uk Ltd Detection of peptides
US7060462B2 (en) * 2000-11-02 2006-06-13 National University Of Singapore AopB gene, protein,homologs, fragments and variants thereof, and their use for cell surface display
EP2412384A1 (en) 2000-11-28 2012-02-01 MedImmune, LLC Methods of administering/dosing anti-RSV antibodies for prophylaxis and treatment
ES2727425T3 (en) 2000-12-12 2019-10-16 Medimmune Llc Molecules with prolonged half-lives, compositions and uses thereof
WO2002051438A2 (en) 2000-12-22 2002-07-04 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. Use of repulsive guidance molecule (rgm) and its modulators
CN1564826A (en) 2001-02-09 2005-01-12 人类基因组科学公司 Human G-protein chemokine receptor (CCR5) HDGNR10
US20030044406A1 (en) * 2001-03-02 2003-03-06 Christine Dingivan Methods of preventing or treating inflammatory or autoimmune disorders by administering CD2 antagonists in combination with other prophylactic or therapeutic agents
JP4295513B2 (en) * 2001-03-07 2009-07-15 チルドレンズ メディカル センター コーポレーション Methods for screening peptide display libraries using minicell display
US8981061B2 (en) 2001-03-20 2015-03-17 Novo Nordisk A/S Receptor TREM (triggering receptor expressed on myeloid cells) and uses thereof
US20090081199A1 (en) * 2001-03-20 2009-03-26 Bioxell S.P.A. Novel receptor trem (triggering receptor expressed on myeloid cells) and uses thereof
US8231878B2 (en) * 2001-03-20 2012-07-31 Cosmo Research & Development S.P.A. Receptor trem (triggering receptor expressed on myeloid cells) and uses thereof
CA2342376C (en) * 2001-03-20 2013-11-12 Marco Colonna A receptor trem (triggering receptor expressed on myeloid cells) and uses thereof
JP2004536579A (en) 2001-04-13 2004-12-09 ヒューマン ジノーム サイエンシーズ, インコーポレイテッド Vascular endothelial growth factor 2
WO2002098280A2 (en) * 2001-06-04 2002-12-12 Ikonisys Inc. Method for detecting infectious agents using computer controlled automated image analysis
EP1417328A2 (en) * 2001-07-20 2004-05-12 Children's Medical Center Corporation Invasion complex and methods of targeting
US6867189B2 (en) * 2001-07-26 2005-03-15 Genset S.A. Use of adipsin/complement factor D in the treatment of metabolic related disorders
AU2002347404A1 (en) * 2001-09-14 2003-04-01 Cytos Biotechnology Ag In vivo activation of antigen presenting cells for enhancement of immune responses induced by virus like particles
US7175983B2 (en) * 2001-11-02 2007-02-13 Abmaxis, Inc. Adapter-directed display systems
EP1463751B1 (en) 2001-12-21 2013-05-22 Human Genome Sciences, Inc. Albumin fusion proteins
WO2003062395A2 (en) * 2002-01-18 2003-07-31 Bristol-Myers Squibb Company Predictor sets for tyrosine kinase pathways
DE60329020D1 (en) * 2002-03-01 2009-10-08 Siemens Healthcare Diagnostics ASSAYS FOR THE MONITORING OF CANCER PATIENTS BASED ON THE MIRROR OF THE EXTRACELLULAR DOMAIN (ECD) ANALYSIS OF THE EPIDERMAL GROWTH FACTOR RECEPTOR (EGFR), ALONE OR IN COMBINATION WITH OTHER ANALYTES, IN SAMPLES FROM BODY FLUIDS
US20030180819A1 (en) * 2002-03-01 2003-09-25 Carney Walter P. Assays for cancer patient monitoring based on levels of analyte components of the plasminogen activator system in body fluid samples
GB0207533D0 (en) 2002-04-02 2002-05-08 Oxford Glycosciences Uk Ltd Protein
EP1499352A4 (en) 2002-04-12 2006-10-11 Medimmune Inc Recombinant anti-interleukin-9 antibodies
US20040010116A1 (en) * 2002-05-10 2004-01-15 Children's Medical Center Corp. Minicell display and products therefrom
WO2003104415A2 (en) * 2002-06-05 2003-12-18 Sopherion Therapeutics, Inc. Method to screen ligands using eukaryotic cell display
EP1912069A3 (en) 2002-06-05 2008-07-09 Sopherion Therapeutics, Inc. Method to screen ligands using eukaryotic cell display
US7563882B2 (en) * 2002-06-10 2009-07-21 University Of Rochester Polynucleotides encoding antibodies that bind to the C35 polypeptide
US7425618B2 (en) 2002-06-14 2008-09-16 Medimmune, Inc. Stabilized anti-respiratory syncytial virus (RSV) antibody formulations
US7425620B2 (en) 2002-08-14 2008-09-16 Scott Koenig FcγRIIB-specific antibodies and methods of use thereof
WO2004022097A1 (en) * 2002-09-05 2004-03-18 Medimmune, Inc. Methods of preventing or treating cell malignancies by administering cd2 antagonists
WO2004035537A2 (en) 2002-10-16 2004-04-29 Euro-Celtique S.A. Antibodies that bind cell-associated ca 125/o772p and methods of use thereof
KR20040036176A (en) * 2002-10-23 2004-04-30 (주)엘피스바이오텍 Protein interaction mediated selective infection of bacteriophage and there use of
AU2003301804A1 (en) * 2002-11-05 2004-06-07 Institut National De La Sante Et De La Recherche Medicale Dc-sign blockers and their use for preventing or treating viral infections.
US7427469B2 (en) * 2002-11-05 2008-09-23 Institut Pasteur Method of treating cytomegalovirus with DC-SIGN blockers
JP2006518997A (en) * 2003-01-21 2006-08-24 ブリストル−マイヤーズ スクイブ カンパニー Novel acyl coenzyme A: polynucleotide encoding monoacylglycerol acyltransferase-3 (MGAT3) and uses thereof
DE10303974A1 (en) 2003-01-31 2004-08-05 Abbott Gmbh & Co. Kg Amyloid β (1-42) oligomers, process for their preparation and their use
WO2004073656A2 (en) 2003-02-20 2004-09-02 Seattle Genetics, Inc. Anti-cd70 antibody-drug conjugates and their use for the treatment of cancer and immune disorders
ES2537015T3 (en) * 2003-03-19 2015-06-01 Biogen Ma Inc. NOGO receptor binding protein
US7294701B2 (en) * 2003-04-02 2007-11-13 Technion Research & Development Foundation Ltd. Antibody fragment capable of modulating multidrug resistance and compositions and kits and methods using same
KR20110094361A (en) 2003-04-11 2011-08-23 메디뮨 엘엘씨 Recombinant il-9 antibodies and uses thereof
CA2532250A1 (en) * 2003-07-15 2005-02-03 Barros Research Institute Compositions and methods for immunotherapy of cancer and infectious diseases
WO2005010163A2 (en) * 2003-07-15 2005-02-03 Barros Research Institute Compositions and methods for immunotherapy of human immunotherapy of human immunodeficiency virus (hiv)
CA2445420A1 (en) * 2003-07-29 2005-01-29 Invitrogen Corporation Kinase and phosphatase assays
US7619059B2 (en) * 2003-07-29 2009-11-17 Life Technologies Corporation Bimolecular optical probes
US7727752B2 (en) * 2003-07-29 2010-06-01 Life Technologies Corporation Kinase and phosphatase assays
CA2534661A1 (en) 2003-08-08 2005-02-17 Genenews Inc. Osteoarthritis biomarkers and uses thereof
US20060228350A1 (en) * 2003-08-18 2006-10-12 Medimmune, Inc. Framework-shuffling of antibodies
EP2272566A3 (en) 2003-08-18 2013-01-02 MedImmune, LLC Humanisation of antibodies
IL158287A0 (en) 2003-10-07 2004-05-12 Yeda Res & Dev Antibodies to nik, their preparation and use
WO2005035569A2 (en) * 2003-10-10 2005-04-21 Five Prime Therapeutics, Inc. Kiaa0779, splice variants thereof, and methods of their use
GB0325836D0 (en) * 2003-11-05 2003-12-10 Celltech R&D Ltd Biological products
PL1983000T3 (en) * 2003-11-21 2016-02-29 Ucb Biopharma Sprl Method for the treatment of multiple sclerosis by inhibiting IL-17 activity
EP1708751B1 (en) * 2003-12-04 2011-09-28 Vaccinex, Inc. Methods of killing tumor cells by targeting internal antigens exposed on apoptotic tumor cells
GB0329825D0 (en) * 2003-12-23 2004-01-28 Celltech R&D Ltd Biological products
US20050266425A1 (en) * 2003-12-31 2005-12-01 Vaccinex, Inc. Methods for producing and identifying multispecific antibodies
PL1729795T3 (en) 2004-02-09 2016-08-31 Human Genome Sciences Inc Albumin fusion proteins
EP2207033B1 (en) 2004-04-15 2014-06-18 University of Florida Research Foundation, Inc. Neural proteins as biomarkers for nervous system injury and other neural disorders
JP2008501638A (en) 2004-04-23 2008-01-24 ドイツ連邦共和国 Method of treating T cell mediated pathology by in vivo depletion of ICOS positive cells
US20060292554A1 (en) * 2004-05-18 2006-12-28 Genentech, Inc. Major coat protein variants for C-terminal and bi-terminal display
DK1754052T3 (en) * 2004-06-03 2015-09-28 Phylogica Ltd Modulators with biochemical properties
US8486893B2 (en) 2004-06-24 2013-07-16 Biogen Idec Ma Inc. Treatment of conditions involving demyelination
US6986264B1 (en) * 2004-07-15 2006-01-17 Carrier Corporation Economized dehumidification system
WO2006017673A2 (en) 2004-08-03 2006-02-16 Biogen Idec Ma Inc. Taj in neuronal function
WO2006034292A2 (en) 2004-09-21 2006-03-30 Medimmune, Inc. Antibodies against and methods for producing vaccines for respiratory syncytial virus
WO2006047417A2 (en) 2004-10-21 2006-05-04 University Of Florida Research Foundation, Inc. Detection of cannabinoid receptor biomarkers and uses thereof
US20060121042A1 (en) 2004-10-27 2006-06-08 Medimmune, Inc. Modulation of antibody specificity by tailoring the affinity to cognate antigens
GB0426146D0 (en) 2004-11-29 2004-12-29 Bioxell Spa Therapeutic peptides and method
WO2006086242A2 (en) 2005-02-07 2006-08-17 Genenews, Inc. Mild osteoarthritis biomarkers and uses thereof
AU2006214121B9 (en) 2005-02-15 2013-02-14 Duke University Anti-CD19 antibodies and uses in oncology
US20090142338A1 (en) * 2005-03-04 2009-06-04 Curedm, Inc. Methods and Compositions for Treating Type 1 and Type 2 Diabetes Mellitus and Related Conditions
JP2008531730A (en) * 2005-03-04 2008-08-14 キュアーディーエム、インク. Methods and pharmaceutical compositions for treating type I diabetes mellitus and other conditions
JP5153613B2 (en) 2005-03-18 2013-02-27 メディミューン,エルエルシー Antibody framework shuffle
GB0506912D0 (en) 2005-04-05 2005-05-11 Celltech R&D Ltd Biological products
ES2707152T3 (en) 2005-04-15 2019-04-02 Macrogenics Inc Covalent diabodies and uses thereof
US20060269556A1 (en) * 2005-04-18 2006-11-30 Karl Nocka Mast cell activation using siglec 6 antibodies
ES2477765T3 (en) 2005-04-19 2014-07-17 Seattle Genetics, Inc. Binding agents to humanized anti-cd70 and their uses
CA2607281C (en) 2005-05-05 2023-10-03 Duke University Anti-cd19 antibody therapy for autoimmune disease
WO2006128083A2 (en) * 2005-05-25 2006-11-30 Curedm, Inc. Human proislet peptide, derivatives and analogs thereof, and methods of using same
WO2007002543A2 (en) 2005-06-23 2007-01-04 Medimmune, Inc. Antibody formulations having optimized aggregation and fragmentation profiles
NZ596992A (en) * 2005-06-30 2013-07-26 Abbott Lab Il-12/p40 binding proteins
EP1904652A2 (en) * 2005-07-08 2008-04-02 Brystol-Myers Squibb Company Single nucleotide polymorphisms associated with dose-dependent edema and methods of use thereof
KR101245462B1 (en) 2005-07-08 2013-03-20 바이오겐 아이덱 엠에이 인코포레이티드 Sp35 antibodies and uses thereof
JP2009501522A (en) * 2005-07-12 2009-01-22 コドン デバイシズ インコーポレイテッド Compositions and methods for biocatalytic engineering
NZ565631A (en) 2005-08-03 2011-01-28 Adelaide Res & Innovation Pty Polysaccharide synthases
US20070202512A1 (en) * 2005-08-19 2007-08-30 Bristol-Myers Squibb Company Human single nucleotide polymorphisms associated with dose-dependent weight gain and methods of use thereof
US20090215992A1 (en) * 2005-08-19 2009-08-27 Chengbin Wu Dual variable domain immunoglobulin and uses thereof
EP2500358A3 (en) 2005-08-19 2012-10-17 Abbott Laboratories Dual variable domain immunoglobulin and uses thereof
WO2007024715A2 (en) 2005-08-19 2007-03-01 Abbott Laboratories Dual variable domain immunoglobin and uses thereof
US7612181B2 (en) 2005-08-19 2009-11-03 Abbott Laboratories Dual variable domain immunoglobulin and uses thereof
ES2382879T3 (en) * 2005-09-14 2012-06-14 Ucb Pharma, S.A. Antibody-comb polymer conjugate.
CN101277974A (en) * 2005-09-30 2008-10-01 阿伯特有限及两合公司 Binding domains of proteins of the repulsive guidance molecule (RGM) protein family and functional fragments thereof, and their use
US8249814B2 (en) 2005-10-21 2012-08-21 Genenews Inc. Method, computer readable medium, and system for determining a probability of colorectal cancer in a test subject
JP2009517340A (en) 2005-11-04 2009-04-30 バイオジェン・アイデック・エムエイ・インコーポレイテッド Methods for promoting neurite outgrowth and survival of dopaminergic neurons
CA2628238A1 (en) 2005-11-07 2007-05-18 The Scripps Research Institute Compositions and methods for controlling tissue factor signaling specificity
EP2567973B1 (en) 2005-11-28 2014-05-14 Zymogenetics, Inc. IL-21 antagonists
PT1976877E (en) 2005-11-30 2014-04-29 Abbvie Inc Monoclonal antibodies against amyloid beta protein and uses thereof
EP1954718B1 (en) 2005-11-30 2014-09-03 AbbVie Inc. Anti-a globulomer antibodies, antigen-binding moieties thereof, corresponding hybridomas, nucleic acids, vectors, host cells, methods of producing said antibodies, compositions comprising said antibodies, uses of said antibodies and methods of using said antibodies
US20090175872A1 (en) 2005-12-02 2009-07-09 Biogen Idec Ma Inc. Treatment of Conditions Involving Demyelination
KR101422523B1 (en) * 2005-12-09 2014-07-24 유씨비 파마, 에스.에이. Antibody molecules having specificity for human il-6
PT1981902E (en) 2006-01-27 2015-11-02 Biogen Ma Inc Nogo receptor antagonists
WO2007090126A2 (en) * 2006-01-30 2007-08-09 Invitrogen Corporation Compositions and methods for detecting and quantifying toxic substances in disease states
US8575070B2 (en) * 2006-02-20 2013-11-05 Phylogica Limited Methods of constructing and screening libraries of peptide structures
US8389688B2 (en) 2006-03-06 2013-03-05 Aeres Biomedical, Ltd. Humanized anti-CD22 antibodies and their use in treatment of oncology, transplantation and autoimmune disease
GB0611116D0 (en) 2006-06-06 2006-07-19 Oxford Genome Sciences Uk Ltd Proteins
WO2010123874A1 (en) 2009-04-20 2010-10-28 Oxford Biotherapeutics Ltd. Antibodies specific to cadherin-17
BRPI0713426A2 (en) 2006-06-14 2012-10-09 Macrogenics Inc methods of treating, slowing the progression, or ameliorating one or more symptoms of a disorder, and preventing or delaying the onset of a disorder
SI2029173T1 (en) 2006-06-26 2016-12-30 Macrogenics, Inc. Fc riib-specific antibodies and methods of use thereof
US7572618B2 (en) 2006-06-30 2009-08-11 Bristol-Myers Squibb Company Polynucleotides encoding novel PCSK9 variants
JP2009543862A (en) 2006-07-14 2009-12-10 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニア Cancer biomarkers and methods of use thereof
CN101622276B (en) * 2006-07-18 2015-04-22 赛诺菲-安万特 Antagonist antibody against EphA2 for the treatment of cancer
MY162024A (en) 2006-08-28 2017-05-31 La Jolla Inst Allergy & Immunology Antagonistic human light-specific human monoclonal antibodies
CA2914170C (en) 2006-09-08 2018-10-30 Abbvie Bahamas Ltd. Interleukin-13 binding proteins
WO2008034161A1 (en) * 2006-09-19 2008-03-27 Phylogica Limited Neuroprotective peptide inhibitors of ap-1 signaling and uses therefor
EP2407548A1 (en) 2006-10-16 2012-01-18 MedImmune, LLC Molecules with reduced half-lives, compositions and uses thereof
GB0620729D0 (en) 2006-10-18 2006-11-29 Ucb Sa Biological products
EP1914242A1 (en) * 2006-10-19 2008-04-23 Sanofi-Aventis Novel anti-CD38 antibodies for the treatment of cancer
US8785400B2 (en) * 2006-11-22 2014-07-22 Curedm Group Holdings, Llc Methods and compositions relating to islet cell neogenesis
US8455626B2 (en) 2006-11-30 2013-06-04 Abbott Laboratories Aβ conformer selective anti-aβ globulomer monoclonal antibodies
PL2099823T5 (en) 2006-12-01 2023-02-20 Seagen Inc. Variant target binding agents and uses thereof
CN101678100A (en) 2006-12-06 2010-03-24 米迪缪尼有限公司 methods of treating systemic lupus erythematosus
KR20090098880A (en) 2006-12-12 2009-09-17 바이오렉시스 파마슈티칼 코포레이션 Transferrin fusion protein libraries
LT2740744T (en) 2007-01-09 2018-05-10 Biogen Ma Inc. Sp35 antibodies and uses thereof
US8128926B2 (en) 2007-01-09 2012-03-06 Biogen Idec Ma Inc. Sp35 antibodies and uses thereof
US8685666B2 (en) * 2007-02-16 2014-04-01 The Board Of Trustees Of Southern Illinois University ARL-1 specific antibodies and uses thereof
US8114606B2 (en) * 2007-02-16 2012-02-14 The Board Of Trustees Of Southern Illinois University ARL-1 specific antibodies
WO2008104803A2 (en) 2007-02-26 2008-09-04 Oxford Genome Sciences (Uk) Limited Proteins
EP2121745A2 (en) 2007-02-26 2009-11-25 Oxford Genome Sciences (UK) Limited Proteins
US20100311767A1 (en) 2007-02-27 2010-12-09 Abbott Gmbh & Co. Kg Method for the treatment of amyloidoses
KR101605908B1 (en) 2007-03-22 2016-03-23 바이오젠 엠에이 인코포레이티드 Binding proteins, including antibodies, antibody derivatives and antibody fragments, that specifically bind cd154 and uses thereof
JP5456658B2 (en) 2007-03-30 2014-04-02 メディミューン,エルエルシー Antibody preparation
US7807168B2 (en) * 2007-04-10 2010-10-05 Vaccinex, Inc. Selection of human TNFα specific antibodies
EP2164868B1 (en) 2007-05-04 2015-03-25 Technophage, Investigação E Desenvolvimento Em Biotecnologia, SA Engineered rabbit antibody variable domains and uses thereof
EP2068925A4 (en) 2007-05-07 2011-08-31 Medimmune Llc Anti-icos antibodies and their use in treatment of oncology, transplantation and autoimmune disease
ES2558689T3 (en) 2007-05-14 2016-02-08 Medimmune, Llc Methods to reduce eosinophil levels
US20090232801A1 (en) * 2007-05-30 2009-09-17 Abbot Laboratories Humanized Antibodies Which Bind To AB (1-42) Globulomer And Uses Thereof
PE20090329A1 (en) * 2007-05-30 2009-03-27 Abbott Lab HUMANIZED ANTIBODIES AGAINST GLOBULOMER AB (20-42) AND ITS USES
WO2008154700A1 (en) * 2007-06-20 2008-12-24 Phylogica Limited Compositions and uses thereof for the treatment of acute respiratory distress syndrome (ards) and clinical disorders associated with therewith
CN107226864A (en) 2007-06-21 2017-10-03 宏观基因有限公司 Covalent diabodies and application thereof
WO2009020477A1 (en) * 2007-08-06 2009-02-12 Yale University Modified miniature proteins
CN101896499B (en) * 2007-08-30 2014-02-12 库尔Dm股份有限公司 Compositions and methods of using proislet peptides and analogs thereof
EP2033971A1 (en) * 2007-09-06 2009-03-11 Abbott GmbH & Co. KG Bone Morphogenetic Protein (BMP) binding domains of proteins of the Repulsive Guidance Molecule (RGM) protein family and functional fragments thereof and their application
GB0717337D0 (en) 2007-09-06 2007-10-17 Ucb Pharma Sa Method of treatment
CN101842387B (en) 2007-09-26 2014-05-07 Ucb医药有限公司 Dual specificity antibody fusions
EP2205071B1 (en) * 2007-10-11 2015-07-22 Biogen MA Inc. Lingo-1 antagonists and trkb agonists for use in the treatment of glaucoma
WO2009061818A1 (en) 2007-11-05 2009-05-14 Medimmune, Llc Methods of treating scleroderma
JP5754046B2 (en) * 2007-11-08 2015-07-22 バイオジェン・エムエイ・インコーポレイテッドBiogen MA Inc. Use of a LINGO-4 antagonist in the treatment of conditions involving demyelination
EP2728017B1 (en) 2007-11-19 2016-08-24 Celera Corporation Lung cancer markers and uses thereof
JP5490714B2 (en) * 2007-11-28 2014-05-14 メディミューン,エルエルシー Protein preparation
AU2008341050B2 (en) 2007-12-26 2013-10-24 Vaccinex, Inc. Anti-C35 antibody combination therapies and methods
GB0800277D0 (en) 2008-01-08 2008-02-13 Imagination Tech Ltd Video motion compensation
EP2388323B1 (en) 2008-01-11 2016-04-13 Gene Techno Science Co., Ltd. Humanized anti-9 integrin antibodies and the uses thereof
KR20100107501A (en) 2008-01-18 2010-10-05 메디뮨 엘엘씨 Cysteine engineered antibodies for site-specific conjugation
BR122020023189B1 (en) 2008-02-08 2022-02-01 Astrazeneca Ab Use of a pharmaceutical composition comprising an antibody specific for ifnar1
US8962803B2 (en) * 2008-02-29 2015-02-24 AbbVie Deutschland GmbH & Co. KG Antibodies against the RGM A protein and uses thereof
US20110020368A1 (en) 2008-03-25 2011-01-27 Nancy Hynes Treating cancer by down-regulating frizzled-4 and/or frizzled-1
CA2720365C (en) 2008-04-02 2019-01-15 Macrogenics, Inc. Bcr-complex-specific antibodies and methods of using same
CN102046195A (en) 2008-04-02 2011-05-04 宏观基因有限公司 HER2/neu-specific antibodies and methods of using same
DK2276509T3 (en) 2008-04-11 2016-09-19 Seattle Genetics Inc DETECTION AND TREATMENT OF CANCER IN PANCREAS, ovarian and other cancers
GB0807413D0 (en) 2008-04-23 2008-05-28 Ucb Pharma Sa Biological products
CA2721716C (en) 2008-04-24 2019-09-24 Gene Techno Science Co., Ltd. Humanized antibodies specific for amino acid sequence rgd of an extracellular matrix protein and the uses thereof
US20090269786A1 (en) * 2008-04-25 2009-10-29 The Board Of Trustees Of The University Of Illinois RHO1-Gamma Amino Butyric Acid C Receptor-Specific Antibodies
US20100260668A1 (en) * 2008-04-29 2010-10-14 Abbott Laboratories Dual Variable Domain Immunoglobulins and Uses Thereof
NZ588554A (en) 2008-04-29 2013-03-28 Abbott Lab Dual variable domain immunoglobulins and uses thereof
CA2723197C (en) 2008-05-02 2017-09-19 Seattle Genetics, Inc. Methods and compositions for making antibodies and antibody derivatives with reduced core fucosylation
SG188142A1 (en) 2008-05-09 2013-03-28 Abbott Gmbh & Co Kg Antibodies to receptor of advanced glycation end products (rage) and uses thereof
EP2304439A4 (en) 2008-05-29 2012-07-04 Nuclea Biotechnologies Llc Anti-phospho-akt antibodies
AR072001A1 (en) 2008-06-03 2010-07-28 Abbott Lab IMMUNOGLOBULIN WITH DUAL VARIABLE DOMAIN AND USES OF THE SAME
CN102112494A (en) 2008-06-03 2011-06-29 雅培制药有限公司 Dual variable domain immunoglobulins and uses thereof
WO2010033279A2 (en) 2008-06-04 2010-03-25 Macrogenics, Inc. Antibodies with altered binding to fcrn and methods of using same
TW201014602A (en) 2008-07-08 2010-04-16 Abbott Lab Prostaglandin E2 binding proteins and uses thereof
WO2010006060A2 (en) * 2008-07-08 2010-01-14 Abbott Laboratories Prostaglandin e2 dual variable domain immunoglobulins and uses thereof
CA2729961C (en) 2008-07-09 2018-05-01 Biogen Idec Ma Inc. Li113, li62 variant co2, anti-lingo antibodies
EP2341924A4 (en) 2008-10-02 2013-01-23 David Gladstone Inst Methods of treating hepatitis c virus infection
US20120251502A1 (en) 2008-10-24 2012-10-04 The Government of the US as Represented by the Secretary of the Dept. of health Human Ebola Virus Species and Compositions and Methods Thereof
JP5933975B2 (en) 2008-11-12 2016-06-15 メディミューン,エルエルシー Antibody preparation
RU2011127198A (en) * 2008-12-04 2013-01-10 Эбботт Лэборетриз IMMUNOGLOBULINS WITH DOUBLE VARIABLE DOMAINS AND THEIR APPLICATION
AU2009335798B2 (en) 2008-12-19 2014-11-27 Macrogenics, Inc. Covalent diabodies and uses thereof
CN102341411A (en) 2008-12-31 2012-02-01 比奥根艾迪克Ma公司 Anti-lymphotoxin antibodies
GB0900425D0 (en) 2009-01-12 2009-02-11 Ucb Pharma Sa Biological products
CA2749572A1 (en) 2009-01-14 2010-07-22 Iq Therapeutics Bv Combination antibodies for the treatment and prevention of disease caused by bacillus anthracis and related bacteria and their toxins
WO2010085510A1 (en) 2009-01-20 2010-07-29 Zadeh Homayoun H Antibody mediated osseous regeneration
US20120058131A1 (en) 2009-01-21 2012-03-08 Oxford Biotherapeutics Ltd Pta089 protein
WO2010088393A2 (en) * 2009-01-28 2010-08-05 Antigen Express, Inc. Li-kay hybrid peptides that modulate the immune response to influenza
CA2749966A1 (en) * 2009-01-29 2010-08-05 Abbott Laboratories Il-1 binding proteins
US20110165063A1 (en) * 2009-01-29 2011-07-07 Abbott Laboratories Il-1 binding proteins
WO2010087927A2 (en) 2009-02-02 2010-08-05 Medimmune, Llc Antibodies against and methods for producing vaccines for respiratory syncytial virus
US20110014190A1 (en) 2009-02-12 2011-01-20 Human Genome Sciences, Inc. Use of b lymphocyte stimulator protein antagonists to promote transplantation tolerance
MX2011008697A (en) * 2009-02-17 2011-11-18 Ucb Pharma Sa Antibody molecules having specificity for human ox40.
JP5836807B2 (en) 2009-03-05 2015-12-24 アッヴィ・インコーポレイテッド IL-17 binding protein
EP2405920A1 (en) 2009-03-06 2012-01-18 Novartis Forschungsstiftung, Zweigniederlassung Friedrich Miescher Institute For Biomedical Research Novel therapy for anxiety
WO2010104208A1 (en) 2009-03-10 2010-09-16 Gene Techno Science Co., Ltd. Generation, expression and characterization of the humanized k33n monoclonal antibody
GB0904214D0 (en) 2009-03-11 2009-04-22 Ucb Pharma Sa Biological products
EP2241323A1 (en) 2009-04-14 2010-10-20 Novartis Forschungsstiftung, Zweigniederlassung Friedrich Miescher Institute For Biomedical Research Tenascin-W and brain cancers
CN106390107B (en) 2009-06-10 2019-12-31 纽约大学 Immune targeting of pathological TAU proteins
CN102802661B (en) 2009-06-22 2016-01-13 米迪缪尼有限公司 For the engineered Fc district of site-specific conjugation
UY32808A (en) * 2009-07-29 2011-02-28 Abbott Lab IMMUNOGLOBULINS AS A DUAL VARIABLE DOMAIN AND USES OF THE SAME
PL2464664T3 (en) 2009-08-13 2016-02-29 Crucell Holland Bv Antibodies against human respiratory syncytial virus (rsv) and methods of use
EP2292266A1 (en) 2009-08-27 2011-03-09 Novartis Forschungsstiftung, Zweigniederlassung Treating cancer by modulating copine III
CN105131112A (en) 2009-08-29 2015-12-09 Abbvie公司 Therapeutic dll4 binding proteins
US20110059111A1 (en) 2009-09-01 2011-03-10 Los Angeles Biomedical Research Institute At Harbor-Ucla Medical Center Mammalian receptors as targets for antibody and active vaccination therapy against mold infections
EP2473524A4 (en) * 2009-09-01 2013-05-22 Abbott Lab Dual variable domain immunoglobulins and uses thereof
US20120283415A1 (en) 2009-09-10 2012-11-08 Ucb Pharma S.A. Multivalent Antibodies
WO2011036118A1 (en) 2009-09-22 2011-03-31 Novartis Forschungsstiftung, Zweigniederlassung Friedrich Miescher Institute For Biomedical Research Treating cancer by modulating mex-3
GB201005063D0 (en) 2010-03-25 2010-05-12 Ucb Pharma Sa Biological products
AR078470A1 (en) 2009-10-02 2011-11-09 Sanofi Aventis ANTIBODIES THAT SPECIFICALLY JOIN THE EPHA2 RECEIVER
US20120231004A1 (en) 2009-10-13 2012-09-13 Oxford Biotherapeutic Ltd. Antibodies
WO2011045352A2 (en) 2009-10-15 2011-04-21 Novartis Forschungsstiftung Spleen tyrosine kinase and brain cancers
EP2488658A4 (en) 2009-10-15 2013-06-19 Abbvie Inc Dual variable domain immunoglobulins and uses thereof
US9234037B2 (en) 2009-10-27 2016-01-12 Ucb Biopharma Sprl Method to generate antibodies to ion channels
WO2011051350A1 (en) 2009-10-27 2011-05-05 Ucb Pharma S.A. Function modifying nav 1.7 antibodies
GB0922435D0 (en) 2009-12-22 2010-02-03 Ucb Pharma Sa Method
GB0922434D0 (en) 2009-12-22 2010-02-03 Ucb Pharma Sa antibodies and fragments thereof
UY32979A (en) * 2009-10-28 2011-02-28 Abbott Lab IMMUNOGLOBULINS WITH DUAL VARIABLE DOMAIN AND USES OF THE SAME
US20120213801A1 (en) 2009-10-30 2012-08-23 Ekaterina Gresko Phosphorylated Twist1 and cancer
WO2011053707A1 (en) 2009-10-31 2011-05-05 Abbott Laboratories Antibodies to receptor for advanced glycation end products (rage) and uses thereof
US20120282177A1 (en) 2009-11-02 2012-11-08 Christian Rohlff ROR1 as Therapeutic and Diagnostic Target
WO2011058087A1 (en) 2009-11-11 2011-05-19 Gentian As Immunoassay for assessing related analytes of different origin
GB0920127D0 (en) 2009-11-17 2009-12-30 Ucb Pharma Sa Antibodies
GB0920324D0 (en) 2009-11-19 2010-01-06 Ucb Pharma Sa Antibodies
MX2012006560A (en) 2009-12-08 2012-10-05 Abbott Gmbh & Co Kg Monoclonal antibodies against the rgm a protein for use in the treatment of retinal nerve fiber layer degeneration.
GB201000467D0 (en) 2010-01-12 2010-02-24 Ucb Pharma Sa Antibodies
MY160628A (en) 2010-03-02 2017-03-15 Abbvie Inc Therapeutic DLL4 Binding Proteins
EP2542578A1 (en) 2010-03-05 2013-01-09 Novartis Forschungsstiftung, Zweigniederlassung Friedrich Miescher Institute For Biomedical Research Smoc1, tenascin-c and brain cancers
WO2011117653A1 (en) 2010-03-25 2011-09-29 Ucb Pharma S.A. Disulfide stabilized dvd-lg molecules
GB201005064D0 (en) 2010-03-25 2010-05-12 Ucb Pharma Sa Biological products
JP2013523182A (en) 2010-04-15 2013-06-17 アボット・ラボラトリーズ Amyloid beta-binding protein
US20130034543A1 (en) 2010-04-19 2013-02-07 Novartis Forschungsstiftung, Zweigniederlassung Friedrich Miescher Institute For Biomedical Resear Modulating xrn1
EP2380909A1 (en) 2010-04-26 2011-10-26 Max-Planck-Gesellschaft zur Förderung der Wissenschaften e.V. PTK-7 protein involved in breast cancer
EP3508854A1 (en) 2010-04-27 2019-07-10 The Regents of The University of California Cancer biomarkers and methods of use thereof
ES2623799T3 (en) 2010-04-30 2017-07-12 Alexion Pharmaceuticals, Inc. Anti-C5a antibodies and methods for the use of antibodies
JP6038777B2 (en) 2010-05-03 2016-12-07 ユニバーシティー オブ ロチェスター Anti-glucosaminidase passive immunization for Staphylococcus aureus infection
US20110293629A1 (en) 2010-05-14 2011-12-01 Bastid Jeremy Methods of Treating and/or Preventing Cell Proliferation Disorders with IL-17 Antagonists
PE20130205A1 (en) 2010-05-14 2013-03-24 Abbvie Inc IL-1 BINDING PROTEINS
EP2580239A1 (en) 2010-06-10 2013-04-17 Novartis Forschungsstiftung, Zweigniederlassung Friedrich Miescher Institute For Biomedical Research Treating cancer by modulating mammalian sterile 20-like kinase 3
WO2012006500A2 (en) 2010-07-08 2012-01-12 Abbott Laboratories Monoclonal antibodies against hepatitis c virus core protein
UY33492A (en) 2010-07-09 2012-01-31 Abbott Lab IMMUNOGLOBULINS WITH DUAL VARIABLE DOMAIN AND USES OF THE SAME
NZ718973A (en) 2010-07-09 2019-01-25 Janssen Vaccines & Prevention Bv Anti-human respiratory syncytial virus (rsv) antibodies and methods of use
US9127057B2 (en) 2010-07-20 2015-09-08 Teva Pharmaceuticals Ausralia Pty Ltd Anti-IL-23 heterodimer specific antibodies
ES2667100T3 (en) 2010-08-02 2018-05-09 Macrogenics, Inc. Covalent Diabodies and Their Uses
EP3252072A3 (en) 2010-08-03 2018-03-14 AbbVie Inc. Dual variable domain immunoglobulins and uses thereof
EP2603524A1 (en) 2010-08-14 2013-06-19 AbbVie Inc. Amyloid-beta binding proteins
EP2606067B1 (en) 2010-08-19 2018-02-21 Zoetis Belgium S.A. Anti-ngf antibodies and their use
GB201014033D0 (en) 2010-08-20 2010-10-06 Ucb Pharma Sa Biological products
KR20130139884A (en) 2010-08-26 2013-12-23 애브비 인코포레이티드 Dual variable domain immunoglobulins and uses thereof
EP2614080A1 (en) 2010-09-10 2013-07-17 Novartis Forschungsstiftung, Zweigniederlassung Friedrich Miescher Institute For Biomedical Research Phosphorylated twist1 and metastasis
EP2640738A1 (en) 2010-11-15 2013-09-25 Novartis Forschungsstiftung, Zweigniederlassung Friedrich Miescher Institute For Biomedical Research Anti-fungal agents
SG191312A1 (en) 2010-12-21 2013-07-31 Abbvie Inc Il-1 -alpha and -beta bispecific dual variable domain immunoglobulins and their use
US20120275996A1 (en) 2010-12-21 2012-11-01 Abbott Laboratories IL-1 Binding Proteins
EA201390923A1 (en) 2010-12-22 2013-12-30 Сефалон Острэйлиа Пти Лтд. MODIFIED ANTIBODY WITH AN IMPROVED ELEMENTATION SEMIUM PERIOD
GB201100282D0 (en) 2011-01-07 2011-02-23 Ucb Pharma Sa Biological methods
US10208349B2 (en) 2011-01-07 2019-02-19 Ucb Biopharma Sprl Lipocalin 2 as a biomarker for IL-17 inhibitor therapy efficacy
AU2012206431B2 (en) 2011-01-14 2015-04-30 UCB Biopharma SRL Antibody molecules which bind IL-17A and IL-17F
DK2668210T3 (en) 2011-01-26 2020-08-24 Celldex Therapeutics Inc ANTI-KIT ANTIBODIES AND USES THEREOF
KR101970025B1 (en) 2011-04-20 2019-04-17 메디뮨 엘엘씨 Antibodies and other molecules that bind b7-h1 and pd-1
EP3103810A3 (en) 2011-04-21 2017-03-08 Garvan Institute of Medical Research Modified variable domain molecules and methods for producing and using them
EP2714079B2 (en) 2011-05-21 2019-08-28 MacroGenics, Inc. Deimmunized serum-binding domains and their use for extending serum half-life
WO2012168259A1 (en) 2011-06-06 2012-12-13 Novartis Forschungsstiftung, Zweigniederlassung Protein tyrosine phosphatase, non-receptor type 11 (ptpn11) and triple-negative breast cancer
US9244074B2 (en) 2011-06-07 2016-01-26 University Of Hawaii Biomarker of asbestos exposure and mesothelioma
US9561274B2 (en) 2011-06-07 2017-02-07 University Of Hawaii Treatment and prevention of cancer with HMGB1 antagonists
ES2664972T3 (en) 2011-06-10 2018-04-24 Medimmune Limited Pseudomonas anti-Psl binding molecules and uses thereof
RS55716B1 (en) 2011-06-28 2017-07-31 Oxford Biotherapeutics Ltd Therapeutic and diagnostic target
ES2640960T3 (en) 2011-06-28 2017-11-07 Oxford Biotherapeutics Ltd. Antibodies for ADP-ribosyl cyclase 2
WO2013009521A2 (en) 2011-07-13 2013-01-17 Abbvie Inc. Methods and compositions for treating asthma using anti-il-13 antibodies
GB201112056D0 (en) 2011-07-14 2011-08-31 Univ Leuven Kath Antibodies
CA2845536A1 (en) 2011-08-15 2013-02-21 Amplimmune, Inc. Anti-b7-h4 antibodies and their uses
EP2753697A1 (en) 2011-09-05 2014-07-16 ETH Zürich Biosynthetic gene cluster for the production of peptide/protein analogues
JP6216317B2 (en) 2011-09-09 2017-10-18 メディミューン リミテッド Anti-Siglec-15 antibody and use thereof
EA035018B1 (en) 2011-09-30 2020-04-17 Тева Фармасьютикал Австралия Пти Лтд. ANTIBODIES AGAINST TL1a AND USES THEREOF
UY34411A (en) 2011-10-24 2013-05-31 Abbvie Inc IMMUNO LINKERS AGAINST SCLEROSTINE
CA2852709A1 (en) 2011-10-28 2013-05-02 Patrys Limited Pat-lm1 epitopes and methods for using same
CA2853951A1 (en) 2011-11-01 2013-05-10 Bionomics, Inc. Antibodies and methods of treating cancer
US10598653B2 (en) 2011-11-01 2020-03-24 Bionomics Inc. Methods of blocking cancer stem cell growth
EP2773667A1 (en) 2011-11-01 2014-09-10 Bionomics, Inc. Anti-gpr49 antibodies
US9220774B2 (en) 2011-11-01 2015-12-29 Bionomics Inc. Methods of treating cancer by administering anti-GPR49 antibodies
JP6182152B2 (en) 2011-11-07 2017-08-16 メディミューン,エルエルシー Combination therapy with anti-Pseudomonas Psl and PcrV binding molecules
US20140314787A1 (en) 2011-11-08 2014-10-23 Novartis Forschungsstiftung, Zweigniederlassung, Friedrich Miescher Institute Treatment for neurodegenerative diseases
EP2776838A1 (en) 2011-11-08 2014-09-17 Novartis Forschungsstiftung, Zweigniederlassung Friedrich Miescher Institute For Biomedical Research Early diagnostic of neurodegenerative diseases
SG11201401649VA (en) 2011-11-11 2014-07-30 Ucb Pharma Sa Albumin binding antibodies and binding fragments thereof
AU2012352168C1 (en) 2011-12-14 2018-01-25 AbbVie Deutschland GmbH & Co. KG Composition and method for the diagnosis and treatment of iron-related disorders
CA2855570A1 (en) 2011-12-14 2013-06-20 AbbVie Deutschland GmbH & Co. KG Composition and method for the diagnosis and treatment of iron-related disorders
CA2859755C (en) 2011-12-23 2021-04-20 Pfizer Inc. Engineered antibody constant regions for site-specific conjugation and methods and uses therefor
CN104159920A (en) 2011-12-30 2014-11-19 艾伯维公司 Dual specific binding proteins directed against il-13 and/or il-17
US20140363448A1 (en) 2012-01-02 2014-12-11 Novartis Ag Cdcp1 and breast cancer
GB201201332D0 (en) 2012-01-26 2012-03-14 Imp Innovations Ltd Method
RS57603B1 (en) 2012-01-27 2018-11-30 Abbvie Deutschland Composition and method for diagnosis and treatment of diseases associated with neurite degeneration
US10444241B2 (en) 2012-02-10 2019-10-15 Seattle Genetics, Inc. Detection and treatment of CD30+ cancers
PL2814844T3 (en) 2012-02-15 2017-12-29 Novo Nordisk A/S Antibodies that bind and block triggering receptor expressed on myeloid cells-1 (trem-1)
US9550830B2 (en) 2012-02-15 2017-01-24 Novo Nordisk A/S Antibodies that bind and block triggering receptor expressed on myeloid cells-1 (TREM-1)
ES2690786T3 (en) 2012-02-15 2018-11-22 Novo Nordisk A/S Antibodies that bind peptidoglycan recognition protein 1
GB201203051D0 (en) 2012-02-22 2012-04-04 Ucb Pharma Sa Biological products
GB201203071D0 (en) 2012-02-22 2012-04-04 Ucb Pharma Sa Biological products
US9421250B2 (en) 2012-03-23 2016-08-23 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Pathogenic phlebovirus isolates and compositions and methods of use
US20150266961A1 (en) 2012-03-29 2015-09-24 Novartis Forschungsstiftung, Zweigniederlassung, Fridrich Miescher Institute Inhibition of interleukin-8 and/or its receptor cxcr1 in the treatment of her2/her3-overexpressing breast cancer
WO2013151649A1 (en) 2012-04-04 2013-10-10 Sialix Inc Glycan-interacting compounds
EP2833911A4 (en) 2012-04-06 2016-07-13 Univ Cornell Subunit vaccine delivery platform for robust humoral and cellular immune responses
EP2849787A4 (en) 2012-05-14 2016-06-15 Biogen Ma Inc Lingo-2 antagonists for treatment of conditions involving motor neurons
MX362497B (en) 2012-05-15 2019-01-21 Eisai Inc Methods for treatment of gastric cancer.
WO2013184871A1 (en) 2012-06-06 2013-12-12 Zoetis Llc Caninized anti-ngf antibodies and methods thereof
WO2014001557A1 (en) 2012-06-28 2014-01-03 Ucb Pharma S.A. A method for identifying compounds of therapeutic interest
EP2866831A1 (en) 2012-06-29 2015-05-06 Novartis Forschungsstiftung, Zweigniederlassung Friedrich Miescher Institute For Biomedical Research Treating diseases by modulating a specific isoform of mkl1
EP2870242A1 (en) 2012-07-05 2015-05-13 Novartis Forschungsstiftung, Zweigniederlassung Friedrich Miescher Institute For Biomedical Research New treatment for neurodegenerative diseases
WO2014006115A1 (en) 2012-07-06 2014-01-09 Novartis Ag Combination of a phosphoinositide 3-kinase inhibitor and an inhibitor of the il-8/cxcr interaction
WO2014011955A2 (en) 2012-07-12 2014-01-16 Abbvie, Inc. Il-1 binding proteins
GB201213652D0 (en) 2012-08-01 2012-09-12 Oxford Biotherapeutics Ltd Therapeutic and diagnostic target
EP2904106A4 (en) 2012-10-01 2016-05-11 Univ Pennsylvania Compositions and methods for targeting stromal cells for the treatment of cancer
KR102229873B1 (en) 2012-10-12 2021-03-19 더 브리검 앤드 우먼즈 하스피털, 인크. Enhancement of the immune response
KR20180008921A (en) 2012-11-01 2018-01-24 애브비 인코포레이티드 Anti-vegf/dll4 dual variable domain immunoglobulins and uses thereof
KR20150100715A (en) 2012-12-21 2015-09-02 앰플리뮨, 인크. Anti-h7cr antibodies
GB201223276D0 (en) 2012-12-21 2013-02-06 Ucb Pharma Sa Antibodies and methods of producing same
WO2014100542A1 (en) 2012-12-21 2014-06-26 Abbvie, Inc. High-throughput antibody humanization
AU2013202668B2 (en) 2012-12-24 2014-12-18 Adelaide Research & Innovation Pty Ltd Inhibition of cancer growth and metastasis
MX2015010023A (en) 2013-02-01 2017-11-17 Transbio Ltd Anti-cd83 antibodies and use thereof.
GB201302447D0 (en) 2013-02-12 2013-03-27 Oxford Biotherapeutics Ltd Therapeutic and diagnostic target
MX2015012825A (en) 2013-03-14 2016-06-10 Abbott Lab Hcv core lipid binding domain monoclonal antibodies.
CA2906421C (en) 2013-03-14 2022-08-16 George J. Dawson Hcv antigen-antibody combination assay and methods and compositions for use therein
BR112015023355A8 (en) 2013-03-14 2018-01-30 Abbott Lab hcv ns3 recombinant antigens and mutants thereof for enhanced antibody detection.
DK2968588T3 (en) 2013-03-15 2019-04-23 Abbvie Deutschland ANTI-EGFR ANTIBODY CONJUGATE PHARMACEUTICAL FORMULATIONS
US9469686B2 (en) 2013-03-15 2016-10-18 Abbott Laboratories Anti-GP73 monoclonal antibodies and methods of obtaining the same
CA2906022A1 (en) 2013-03-15 2014-09-25 Abbvie Inc. Antibody drug conjugate (adc) purification
CN105324396A (en) 2013-03-15 2016-02-10 艾伯维公司 Dual specific binding proteins directed against il-1 beta and il-17
SG10201800800YA (en) 2013-05-06 2018-03-28 Scholar Rock Inc Compositions and methods for growth factor modulation
CA2913312A1 (en) 2013-05-24 2014-11-27 Medimmune, Llc Anti-b7-h5 antibodies and their uses
ES2891755T3 (en) 2013-06-06 2022-01-31 Pf Medicament Anti-C10orf54 antibodies and uses thereof
EP3632467B1 (en) 2013-06-07 2023-09-27 Duke University Inhibitors of complement factor h
ES2761587T3 (en) 2013-08-07 2020-05-20 Friedrich Miescher Institute For Biomedical Res New screening method for the treatment of Friedreich's ataxia
KR102362630B1 (en) 2013-08-26 2022-02-15 바이오엔테크 리서치 앤드 디벨롭먼트 인코포레이티드 Nucleic acids encoding human antibodies to sialyl-lewis a
EP3063317B1 (en) 2013-10-28 2020-06-03 DOTS Technology Corp. Allergen detection
US8986694B1 (en) 2014-07-15 2015-03-24 Kymab Limited Targeting human nav1.7 variants for treatment of pain
US9067998B1 (en) 2014-07-15 2015-06-30 Kymab Limited Targeting PD-1 variants for treatment of cancer
US9045545B1 (en) 2014-07-15 2015-06-02 Kymab Limited Precision medicine by targeting PD-L1 variants for treatment of cancer
US9914769B2 (en) 2014-07-15 2018-03-13 Kymab Limited Precision medicine for cholesterol treatment
US8992927B1 (en) 2014-07-15 2015-03-31 Kymab Limited Targeting human NAV1.7 variants for treatment of pain
AU2014366837B2 (en) 2013-12-18 2020-06-25 B-Creative Sweden Ab Method of treating wounds
GB201403775D0 (en) 2014-03-04 2014-04-16 Kymab Ltd Antibodies, uses & methods
US9738702B2 (en) 2014-03-14 2017-08-22 Janssen Biotech, Inc. Antibodies with improved half-life in ferrets
CN106536556B (en) 2014-04-04 2020-02-07 生态学有限公司 Humanized antibodies that bind LGR5
US20170267780A1 (en) 2014-05-16 2017-09-21 Medimmune, Llc Molecules with altered neonate fc receptor binding having enhanced therapeutic and diagnostic properties
LT3148579T (en) 2014-05-28 2021-05-25 Agenus Inc. Anti-gitr antibodies and methods of use thereof
GB201409558D0 (en) 2014-05-29 2014-07-16 Ucb Biopharma Sprl Method
HUE055189T2 (en) 2014-06-04 2021-11-29 Biontech Res And Development Inc Human monoclonal antibodies to ganglioside gd2
WO2015189816A1 (en) 2014-06-13 2015-12-17 Friedrich Miescher Institute For Biomedical Research New treatment against influenza virus
WO2015198202A1 (en) 2014-06-23 2015-12-30 Friedrich Miescher Institute For Biomedical Research Methods for triggering de novo formation of heterochromatin and or epigenetic silencing with small rnas
GB201411320D0 (en) 2014-06-25 2014-08-06 Ucb Biopharma Sprl Antibody construct
EP3164129A1 (en) 2014-07-01 2017-05-10 Friedrich Miescher Institute for Biomedical Research Combination of a brafv600e inhibitor and mertk inhibitor to treat melanoma
US9139648B1 (en) 2014-07-15 2015-09-22 Kymab Limited Precision medicine by targeting human NAV1.9 variants for treatment of pain
GB201412659D0 (en) 2014-07-16 2014-08-27 Ucb Biopharma Sprl Molecules
GB201412658D0 (en) 2014-07-16 2014-08-27 Ucb Biopharma Sprl Molecules
EP4332119A2 (en) 2014-07-17 2024-03-06 Novo Nordisk A/S Site directed mutagenesis of trem-1 antibodies for decreasing viscosity
EP3197557A1 (en) 2014-09-24 2017-08-02 Friedrich Miescher Institute for Biomedical Research Lats and breast cancer
RU2021125449A (en) 2014-10-01 2021-09-16 Медиммьюн Лимитед ANTIBODIES TO TICAGRELOR AND METHODS OF APPLICATION
US10870704B2 (en) 2014-10-23 2020-12-22 Kira Biotech Pty Limited CD83 binding proteins and uses thereof
CA2967595A1 (en) 2014-11-12 2016-05-19 Siamab Therapeutics, Inc. Glycan-interacting compounds and methods of use
US9879087B2 (en) 2014-11-12 2018-01-30 Siamab Therapeutics, Inc. Glycan-interacting compounds and methods of use
EP3227687A4 (en) 2014-12-05 2018-10-24 Prelude, Inc. Dcis recurrence and invasive breast cancer
ES2934940T3 (en) 2014-12-11 2023-02-28 Pf Medicament Anti-C10orf54 antibodies and uses thereof
US10093733B2 (en) 2014-12-11 2018-10-09 Abbvie Inc. LRP-8 binding dual variable domain immunoglobulin proteins
ES2862701T3 (en) 2014-12-22 2021-10-07 Univ Rockefeller Anti-MERTK Agonist Antibodies and Uses Thereof
JP2018504400A (en) 2015-01-08 2018-02-15 バイオジェン・エムエイ・インコーポレイテッドBiogen MA Inc. LINGO-1 antagonist and use for treatment of demyelinating disorders
EP3253793A4 (en) 2015-02-02 2018-11-14 i2 Pharmaceuticals, Inc. Anti-surrogate light chain antibodies
MX2017011194A (en) 2015-03-03 2018-04-10 Kymab Ltd Antibodies, uses & methods.
AU2016243124A1 (en) 2015-03-27 2017-11-02 University Of Southern California CAR T-cell therapy directed to LHR for the treatment of solid tumors
GB201506869D0 (en) 2015-04-22 2015-06-03 Ucb Biopharma Sprl Method
GB201506870D0 (en) 2015-04-22 2015-06-03 Ucb Biopharma Sprl Method
US10814012B2 (en) 2015-04-29 2020-10-27 University Of South Australia Compositions and methods for administering antibodies
MA42059A (en) 2015-05-06 2018-03-14 Janssen Biotech Inc PROSTATE-SPECIFIC MEMBRANE ANTIGEN (PSMA) BISPECIFIC BINDING AGENTS AND USES
MY189692A (en) 2015-05-07 2022-02-26 Memorial Sloan Kettering Cancer Center Anti-ox40 antibodies and methods of use thereof
MA47395A (en) 2015-05-27 2019-12-11 Ucb Biopharma Sprl METHOD FOR THE TREATMENT OF NEUROLOGICAL DISEASE
JP6518917B2 (en) 2015-05-29 2019-05-29 アッヴィ・インコーポレイテッド Anti-CD40 antibody and use thereof
CN113603784A (en) 2015-05-29 2021-11-05 艾吉纳斯公司 anti-CTLA-4 antibodies and methods of use thereof
US10711064B2 (en) 2015-06-04 2020-07-14 University Of Southern California Lym-1 and Lym-2 targeted CAR cell immunotherapy
TW201710286A (en) 2015-06-15 2017-03-16 艾伯維有限公司 Binding proteins against VEGF, PDGF, and/or their receptors
GB201510758D0 (en) 2015-06-18 2015-08-05 Ucb Biopharma Sprl Novel TNFa structure for use in therapy
GB201601077D0 (en) 2016-01-20 2016-03-02 Ucb Biopharma Sprl Antibody molecule
GB201601073D0 (en) 2016-01-20 2016-03-02 Ucb Biopharma Sprl Antibodies
GB201601075D0 (en) 2016-01-20 2016-03-02 Ucb Biopharma Sprl Antibodies molecules
BR112018003186A2 (en) 2015-09-01 2018-09-25 Agenus Inc. anti-pd-1 antibodies and their methods of use
EP3569244A1 (en) 2015-09-23 2019-11-20 CytoImmune Therapeutics, LLC Flt3 directed car cells for immunotherapy
CN108350068A (en) 2015-10-27 2018-07-31 Ucb生物制药私人有限公司 Use the therapy of anti-IL-17A/F antibody
WO2017072669A1 (en) 2015-10-28 2017-05-04 Friedrich Miescher Institute For Biomedical Research Tenascin-w and biliary tract cancers
KR20180088381A (en) 2015-11-12 2018-08-03 시아맙 쎄라퓨틱스, 인코포레이티드 Glycan-interacting compounds and methods of use
JP7090545B2 (en) 2015-12-02 2022-06-24 ストキューブ アンド シーオー., インコーポレイテッド Antibodies and molecules that immunospecifically bind to BTN1A1 and their therapeutic use
WO2017096017A1 (en) 2015-12-02 2017-06-08 Stsciences, Inc. Antibodies specific to glycosylated btla (b- and t- lymphocyte attenuator)
GB201521391D0 (en) 2015-12-03 2016-01-20 Ucb Biopharma Sprl Antibodies
GB201521383D0 (en) 2015-12-03 2016-01-20 Ucb Biopharma Sprl And Ucb Celltech Method
GB201521389D0 (en) 2015-12-03 2016-01-20 Ucb Biopharma Sprl Method
GB201521393D0 (en) 2015-12-03 2016-01-20 Ucb Biopharma Sprl Antibodies
GB201521382D0 (en) 2015-12-03 2016-01-20 Ucb Biopharma Sprl Antibodies
US10829562B2 (en) 2015-12-10 2020-11-10 Katholieke Universiteit Leuven Haemorrhagic disorder due to ventricular assist device
GB201602413D0 (en) 2016-02-10 2016-03-23 Nascient Ltd Method
KR20240017421A (en) 2016-03-10 2024-02-07 비엘라 바이오, 인크. Ilt7 binding molecules and methods of using the same
CA3018081A1 (en) 2016-03-22 2017-09-28 Bionomics Limited Administration of an anti-lgr5 monoclonal antibody
CN116333130A (en) 2016-05-24 2023-06-27 英斯梅德股份有限公司 Antibodies and methods of making the same
TWI781934B (en) 2016-05-27 2022-11-01 美商艾吉納斯公司 Anti-tim-3 antibodies and methods of use thereof
KR20190039937A (en) 2016-07-08 2019-04-16 스태튼 바이오테크놀로지 비.브이. Anti-ApoC3 antibodies and methods of use thereof
EP3515948A4 (en) 2016-09-23 2020-04-08 CSL Limited Coagulation factor binding proteins and uses thereof
GB201616596D0 (en) 2016-09-29 2016-11-16 Nascient Limited Epitope and antibodies
JP2019535015A (en) 2016-10-03 2019-12-05 アボット・ラボラトリーズAbbott Laboratories Improved method for assessing GFAP status in patient samples
TW202246349A (en) 2016-10-11 2022-12-01 美商艾吉納斯公司 Anti-lag-3 antibodies and methods of use thereof
RU2019114863A (en) 2016-11-02 2020-12-03 Иммуноджен, Инк. COMBINED TREATMENT WITH ANTIBODY-DRUG CONJUGATES AND PARP INHIBITORS
US11779604B2 (en) 2016-11-03 2023-10-10 Kymab Limited Antibodies, combinations comprising antibodies, biomarkers, uses and methods
WO2018083538A1 (en) 2016-11-07 2018-05-11 Neuracle Scienc3 Co., Ltd. Anti-family with sequence similarity 19, member a5 antibodies and method of use thereof
EP3541847A4 (en) 2016-11-17 2020-07-08 Seattle Genetics, Inc. Glycan-interacting compounds and methods of use
MD3551660T2 (en) 2016-12-07 2024-03-31 Agenus Inc Anti-CTLA-4 antibodies and methods of use thereof
BR112019011582A2 (en) 2016-12-07 2019-10-22 Agenus Inc. antibodies and their methods of use
EP3555625A1 (en) 2016-12-16 2019-10-23 Merck Patent GmbH Methods for the use of galectin 3 binding protein detected in the urine for monitoring the severity and progression of lupus nephritis
GB201621635D0 (en) 2016-12-19 2017-02-01 Ucb Biopharma Sprl Crystal structure
MA47812A (en) 2017-03-03 2021-04-14 Seagen Inc COMPOUNDS INTERACTING WITH GLYCAN AND METHODS OF USE
JP7346300B2 (en) 2017-03-23 2023-09-19 アボット・ラボラトリーズ Methods for aiding in the diagnosis and determination of the extent of traumatic brain injury in human subjects using the early biomarker ubiquitin carboxy-terminal hydrolase L1
TW201841942A (en) 2017-04-13 2018-12-01 美商艾吉納斯公司 Anti-CD137 antibodies and methods of use thereof
US10877048B2 (en) 2017-04-15 2020-12-29 Abbott Laboratories Methods for aiding in the hyperacute diagnosis and determination of traumatic brain injury in a human subject using early biomarkers
AU2018255938A1 (en) 2017-04-21 2019-10-31 Staten Biotechnology B.V. Anti-ApoC3 antibodies and methods of use thereof
WO2018200823A1 (en) 2017-04-28 2018-11-01 Abbott Laboratories Methods for aiding in the hyperacute diagnosis and determination of traumatic brain injury using early biomarkers on at least two samples from the same human subject
RS64576B1 (en) 2017-05-01 2023-10-31 Agenus Inc Anti-tigit antibodies and methods of use thereof
US10865238B1 (en) 2017-05-05 2020-12-15 Duke University Complement factor H antibodies
JOP20190256A1 (en) 2017-05-12 2019-10-28 Icahn School Med Mount Sinai Newcastle disease viruses and uses thereof
AU2018272054A1 (en) 2017-05-25 2019-09-26 Abbott Laboratories Methods for aiding in the determination of whether to perform imaging on a human subject who has sustained or may have sustained an injury to the head using early biomarkers
BR112019025313A2 (en) 2017-05-30 2020-06-23 Abbott Laboratories METHODS FOR ASSISTANCE IN DIAGNOSIS AND EVALUATION OF A LIGHT TRAUMATIC BRAIN INJURY IN A HUMAN INDIVIDUAL USING HEART TROPONIN I
KR20200015602A (en) 2017-05-31 2020-02-12 주식회사 에스티큐브앤컴퍼니 Antibodies and molecules immunospecifically binding to BTN1A1 and therapeutic uses thereof
CN111051346A (en) 2017-05-31 2020-04-21 斯特库伯株式会社 Methods of treating cancer using antibodies and molecules that immunospecifically bind to BTN1a1
WO2018226671A1 (en) 2017-06-06 2018-12-13 Stcube & Co., Inc. Methods of treating cancer using antibodies and molecules that bind to btn1a1 or btn1a1-ligands
GB201709379D0 (en) 2017-06-13 2017-07-26 Univ Leuven Kath Humanised ADAMTS13 binding antibodies
US20190062428A1 (en) 2017-06-19 2019-02-28 Surface Oncology, Inc. Combination of anti-cd47 antibodies and cell death-inducing agents, and uses thereof
CA3068041A1 (en) 2017-07-03 2019-01-10 Abbott Laboratories Improved methods for measuring ubiquitin carboxy-terminal hydrolase l1 levels in blood
AU2018301393A1 (en) 2017-07-11 2020-02-06 Compass Therapeutics Llc Agonist antibodies that bind human CD137 and uses thereof
EP3684811A2 (en) 2017-08-17 2020-07-29 Massachusetts Institute of Technology Multiple specificity binders of cxc chemokines and uses thereof
EP3672990A1 (en) 2017-08-25 2020-07-01 Five Prime Therapeutics, Inc. B7-h4 antibodies and methods of use thereof
KR20210027230A (en) 2017-10-04 2021-03-10 옵코 파마슈티칼스, 엘엘씨 Articles and methods for personalized treatment of cancer
CN111630069A (en) 2017-10-13 2020-09-04 勃林格殷格翰国际有限公司 Human antibodies to Thomsen-novell (Tn) antigens
WO2019089594A1 (en) 2017-10-31 2019-05-09 Immunogen, Inc. Combination treatment with antibody-drug conjugates and cytarabine
CA3080103A1 (en) 2017-10-31 2019-05-09 Staten Biotechnology B.V. Anti-apoc3 antibodies and methods of use thereof
WO2019089753A2 (en) 2017-10-31 2019-05-09 Compass Therapeutics Llc Cd137 antibodies and pd-1 antagonists and uses thereof
CA3082365A1 (en) 2017-11-09 2019-05-16 Pinteon Therapeutics Inc. Methods and compositions for the generation and use of humanized conformation-specific phosphorylated tau antibodies
US11851497B2 (en) 2017-11-20 2023-12-26 Compass Therapeutics Llc CD137 antibodies and tumor antigen-targeting antibodies and uses thereof
JP7165193B2 (en) 2017-11-27 2022-11-02 パーデュー、ファーマ、リミテッド、パートナーシップ Humanized antibody targeting human tissue factor
CN111094983A (en) 2017-12-09 2020-05-01 雅培实验室 Methods of using Glial Fibrillary Acidic Protein (GFAP) and/or ubiquitin carboxy-terminal hydrolase L1(UCH-L1) to aid in the diagnosis and evaluation of patients who have suffered orthopedic injury and who have suffered or may have suffered a head injury such as mild Traumatic Brain Injury (TBI)
CA3067055A1 (en) 2017-12-09 2019-06-13 Abbott Laboratories Methods for aiding in diagnosing and evaluating a traumatic brain injury in a human subject using a combination of gfap and uch-l1
GB201802486D0 (en) 2018-02-15 2018-04-04 Ucb Biopharma Sprl Methods
SG11202008105RA (en) 2018-03-02 2020-09-29 Five Prime Therapeutics Inc B7-h4 antibodies and methods of use thereof
EP3765499A1 (en) 2018-03-12 2021-01-20 Zoetis Services LLC Anti-ngf antibodies and methods thereof
EP3765524A4 (en) 2018-03-14 2021-12-22 Surface Oncology, Inc. Antibodies that bind cd39 and uses thereof
US11332524B2 (en) 2018-03-22 2022-05-17 Surface Oncology, Inc. Anti-IL-27 antibodies and uses thereof
TWI790370B (en) 2018-04-02 2023-01-21 美商必治妥美雅史谷比公司 Anti-trem-1 antibodies and uses thereof
WO2019200357A1 (en) 2018-04-12 2019-10-17 Surface Oncology, Inc. Biomarker for cd47 targeting therapeutics and uses therefor
WO2019207159A1 (en) 2018-04-27 2019-10-31 Fondazione Ebri Rita Levi-Montalcini Antibody directed against a tau-derived neurotoxic peptide and uses thereof
WO2019222130A1 (en) 2018-05-15 2019-11-21 Immunogen, Inc. Combination treatment with antibody-drug conjugates and flt3 inhibitors
WO2019226658A1 (en) 2018-05-21 2019-11-28 Compass Therapeutics Llc Multispecific antigen-binding compositions and methods of use
JP2021525243A (en) 2018-05-21 2021-09-24 コンパス セラピューティクス リミテッド ライアビリティ カンパニー Compositions and Methods for Promoting Killing of Target Cells by NK Cells
KR20210028191A (en) 2018-06-18 2021-03-11 유씨비 바이오파마 에스알엘 GREMLIN-1 antagonists for preventing and treating cancer
WO2019244107A1 (en) 2018-06-21 2019-12-26 Daiichi Sankyo Company, Limited Compositions including cd3 antigen binding fragments and uses thereof
MX2021000786A (en) 2018-07-20 2021-06-15 Pf Medicament Receptor for vista.
US20210309746A1 (en) 2018-08-09 2021-10-07 Compass Therapeutics Llc Antibodies that bind cd277 and uses thereof
WO2020033923A1 (en) 2018-08-09 2020-02-13 Compass Therapeutics Llc Antigen binding agents that bind cd277 and uses thereof
WO2020033925A2 (en) 2018-08-09 2020-02-13 Compass Therapeutics Llc Antibodies that bind cd277 and uses thereof
AU2019339508A1 (en) 2018-09-14 2021-04-15 Prelude Corporation Method of selection for treatment of subjects at risk of invasive breast cancer
CA3114295A1 (en) 2018-09-28 2020-04-02 Kyowa Kirin Co., Ltd. Il-36 antibodies and uses thereof
AU2019352017A1 (en) 2018-10-03 2021-05-06 Staten Biotechnology B.V. Antibodies specific for human and cynomolgus ApoC3 and methods of use thereof
GB201817311D0 (en) 2018-10-24 2018-12-05 Ucb Biopharma Sprl Antibodies
GB201817309D0 (en) 2018-10-24 2018-12-05 Ucb Biopharma Sprl Antibodies
US20220289857A1 (en) 2018-12-20 2022-09-15 Kyowa Kirin Co., Ltd. Fn14 antibodies and uses thereof
AU2020208397A1 (en) 2019-01-16 2021-08-12 Compass Therapeutics Llc Formulations of antibodies that bind human CD137 and uses thereof
GB201900732D0 (en) 2019-01-18 2019-03-06 Ucb Biopharma Sprl Antibodies
MA55080A (en) 2019-02-26 2022-01-05 Inspirna Inc HIGH AFFINITY ANTI-MERTK ANTIBODIES AND ASSOCIATED USES
CN113874392A (en) 2019-03-28 2021-12-31 丹尼斯科美国公司 Engineered antibodies
CN114867751A (en) 2019-08-12 2022-08-05 阿帕特夫研究和发展有限公司 4-1BB and OX40 binding proteins and related compositions and methods, anti-4-1 BB antibodies, anti-OX 40 antibodies
JP2022545741A (en) 2019-08-30 2022-10-28 アジェナス インコーポレイテッド ANTI-CD96 ANTIBODY AND METHODS OF USE THEREOF
US20220306736A1 (en) 2019-09-04 2022-09-29 Y-Biologics Inc. Anti-vsig4 antibody or antigen binding fragment and uses thereof
BR112022002351A2 (en) 2019-09-16 2022-07-19 Surface Oncology Inc ANTI-CD39 ANTIBODY COMPOSITIONS AND METHODS
EP4034559A1 (en) 2019-09-25 2022-08-03 Surface Oncology, Inc. Anti-il-27 antibodies and uses thereof
US20220411511A1 (en) 2019-09-26 2022-12-29 Stcube & Co. Antibodies specific to glycosylated ctla-4 and methods of use thereof
WO2021072277A1 (en) 2019-10-09 2021-04-15 Stcube & Co. Antibodies specific to glycosylated lag3 and methods of use thereof
WO2021080682A1 (en) 2019-10-24 2021-04-29 Massachusetts Institute Of Technology Monoclonal antibodies that bind human cd161 and uses thereof
GB201917480D0 (en) 2019-11-29 2020-01-15 Univ Oxford Innovation Ltd Antibodies
GB201919062D0 (en) 2019-12-20 2020-02-05 Ucb Biopharma Sprl Antibody
GB201919058D0 (en) 2019-12-20 2020-02-05 Ucb Biopharma Sprl Multi-specific antibodies
GB201919061D0 (en) 2019-12-20 2020-02-05 Ucb Biopharma Sprl Multi-specific antibody
TW202138388A (en) 2019-12-30 2021-10-16 美商西根公司 Methods of treating cancer with nonfucosylated anti-cd70 antibodies
KR20220122628A (en) 2020-01-06 2022-09-02 백시넥스 인코포레이티드 Anti-CCR8 antibodies and uses thereof
AU2021215936A1 (en) 2020-02-05 2022-08-25 Larimar Therapeutics, Inc. TAT peptide binding proteins and uses thereof
US20230096030A1 (en) 2020-02-13 2023-03-30 UCB Biopharma SRL Bispecific antibodies against cd9 and cd7
EP4103611B1 (en) 2020-02-13 2024-03-27 UCB Biopharma SRL Bispecific antibodies binding hvem and cd9
EP4103610A1 (en) 2020-02-13 2022-12-21 UCB Biopharma SRL Anti cd44-ctla4 bispecific antibodies
US20230151109A1 (en) 2020-02-13 2023-05-18 UCB Biopharma SRL Bispecific antibodies against cd9
US20230151108A1 (en) 2020-02-13 2023-05-18 UCB Biopharma SRL Bispecific antibodies against cd9 and cd137
US20230220074A1 (en) 2020-02-18 2023-07-13 Alector Llc Pilra antibodies and methods of use thereof
CA3174442A1 (en) 2020-03-06 2021-09-10 ONA Therapeutics S.L. Anti-cd36 antibodies and their use to treat cancer
JP2023517889A (en) 2020-03-10 2023-04-27 マサチューセッツ インスティテュート オブ テクノロジー Compositions and methods for immunotherapy of NPM1c-positive cancers
WO2021202473A2 (en) 2020-03-30 2021-10-07 Danisco Us Inc Engineered antibodies
EP4136459A1 (en) 2020-04-13 2023-02-22 Abbott Laboratories Methods, complexes and kits for detecting or determining an amount of a ss-coronavirus antibody in a sample
CR20220646A (en) 2020-05-17 2023-10-23 Astrazeneca Uk Ltd Sars-cov-2 antibodies and methods of selecting and using the same
KR20230038783A (en) 2020-07-20 2023-03-21 아스트라제네카 유케이 리미티드 SARS-COV-2 proteins, anti-SARS-COV-2 antibodies, and methods of using them
US20220043000A1 (en) 2020-08-04 2022-02-10 Abbott Laboratories Methods and kits for detecting sars-cov-2 protein in a sample
MX2023002001A (en) 2020-08-18 2023-03-21 Cephalon Llc Anti-par-2 antibodies and methods of use thereof.
US20230382978A1 (en) 2020-10-15 2023-11-30 The United States Of America, As Represented By The Secretary, Department Of Health & Human Services Antibody specific for sars-cov-2 receptor binding domain and therapeutic methods
KR20230087552A (en) 2020-10-15 2023-06-16 유씨비 바이오파마 에스알엘 Binding molecule that multimerizes CD45
WO2022087274A1 (en) 2020-10-21 2022-04-28 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Antibodies that neutralize type-i interferon (ifn) activity
WO2023102384A1 (en) 2021-11-30 2023-06-08 Abbott Laboratories Use of one or more biomarkers to determine traumatic brain injury (tbi) in a subject having received a head computerized tomography scan that is negative for a tbi
AU2021390501A1 (en) 2020-12-01 2023-06-29 Aptevo Research And Development Llc Heterodimeric psma and cd3-binding bispecific antibodies
CA3198161A1 (en) 2020-12-01 2022-06-09 Beth MCQUISTON Use of one or more biomarkers to determine traumatic brain injury (tbi) in a subject having received a head computerized tomography scan that is negative for a tbi
EP4271998A1 (en) 2020-12-30 2023-11-08 Abbott Laboratories Methods for determining sars-cov-2 antigen and anti-sars-cov-2 antibody in a sample
KR20230146522A (en) 2021-01-13 2023-10-19 메모리얼 슬로안 케터링 캔서 센터 Anti-DLL3 antibody-drug conjugate
CA3204628A1 (en) 2021-01-13 2022-07-21 John T. POIRIER Antibody-pyrrolobenzodiazepine derivative conjugate
EP4301786A1 (en) 2021-03-03 2024-01-10 Pierre Fabre Medicament Anti-vsig4 antibody or antigen binding fragment and uses thereof
EP4067381A1 (en) 2021-04-01 2022-10-05 Julius-Maximilians-Universität Würzburg Novel tnfr2 binding molecules
EP4334348A1 (en) 2021-05-07 2024-03-13 Surface Oncology, LLC Anti-il-27 antibodies and uses thereof
CA3216320A1 (en) 2021-05-18 2022-11-24 Abbott Laboratories Methods of evaluating brain injury in a pediatric subject
WO2022251446A1 (en) 2021-05-28 2022-12-01 Alexion Pharmaceuticals, Inc. Methods for detecting cm-tma biomarkers
WO2022263357A1 (en) 2021-06-14 2022-12-22 Argenx Iip Bv Anti-il-9 antibodies and methods of use thereof
CA3222291A1 (en) 2021-06-14 2022-12-22 Jaime MARINO Methods of diagnosing or aiding in diagnosis of brain injury caused by acoustic energy, electromagnetic energy, an over pressurization wave, and/or blast wind
TW202317190A (en) 2021-06-29 2023-05-01 美商思進公司 Methods of treating cancer with a combination of a nonfucosylated anti-cd70 antibody and a cd47 antagonist
WO2023285878A1 (en) 2021-07-13 2023-01-19 Aviation-Ophthalmology Methods for detecting, treating, and preventing gpr68-mediated ocular diseases, disorders, and conditions
CA3226281A1 (en) 2021-07-30 2023-02-02 ONA Therapeutics S.L. Anti-cd36 antibodies and their use to treat cancer
AU2022339759A1 (en) 2021-08-31 2024-03-07 Abbott Laboratories Methods and systems of diagnosing brain injury
WO2023056268A1 (en) 2021-09-30 2023-04-06 Abbott Laboratories Methods and systems of diagnosing brain injury
EP4177266A1 (en) 2021-11-05 2023-05-10 Katholieke Universiteit Leuven Neutralizing anti-sars-cov-2 human antibodies
WO2023114978A1 (en) 2021-12-17 2023-06-22 Abbott Laboratories Systems and methods for determining uch-l1, gfap, and other biomarkers in blood samples
US20230213536A1 (en) 2021-12-28 2023-07-06 Abbott Laboratories Use of biomarkers to determine sub-acute traumatic brain injury (tbi) in a subject having received a head computerized tomography (ct) scan that is negative for a tbi or no head ct scan
WO2023150652A1 (en) 2022-02-04 2023-08-10 Abbott Laboratories Lateral flow methods, assays, and devices for detecting the presence or measuring the amount of ubiquitin carboxy-terminal hydrolase l1 and/or glial fibrillary acidic protein in a sample
WO2023192436A1 (en) 2022-03-31 2023-10-05 Alexion Pharmaceuticals, Inc. Singleplex or multiplexed assay for complement markers in fresh biological samples
GB202205203D0 (en) 2022-04-08 2022-05-25 UCB Biopharma SRL Combination with inhibitor
GB202205200D0 (en) 2022-04-08 2022-05-25 Ucb Biopharma Sprl Combination with chemotherapy
WO2023209177A1 (en) 2022-04-29 2023-11-02 Astrazeneca Uk Limited Sars-cov-2 antibodies and methods of using the same
WO2023240124A1 (en) 2022-06-07 2023-12-14 Regeneron Pharmaceuticals, Inc. Pseudotyped viral particles for targeting tcr-expressing cells
WO2024006876A1 (en) 2022-06-29 2024-01-04 Abbott Laboratories Magnetic point-of-care systems and assays for determining gfap in biological samples
WO2024015953A1 (en) 2022-07-15 2024-01-18 Danisco Us Inc. Methods for producing monoclonal antibodies
WO2024013727A1 (en) 2022-07-15 2024-01-18 Janssen Biotech, Inc. Material and methods for improved bioengineered pairing of antigen-binding variable regions
WO2024050354A1 (en) 2022-08-31 2024-03-07 Washington University Alphavirus antigen binding antibodies and uses thereof
WO2024050524A1 (en) 2022-09-01 2024-03-07 University Of Georgia Research Foundation, Inc. Compositions and methods for directing apolipoprotein l1 to induce mammalian cell death
WO2024054436A1 (en) 2022-09-06 2024-03-14 Alexion Pharmaceuticals, Inc. Diagnostic and prognostic biomarker profiles in patients with hematopoietic stem cell transplant-associated thrombotic microangiopathy (hsct-tma)
WO2024059708A1 (en) 2022-09-15 2024-03-21 Abbott Laboratories Biomarkers and methods for differentiating between mild and supermild traumatic brain injury

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0264150A1 (en) * 1986-10-13 1988-04-20 Solvay Microorganisms comprising pili as carrier proteins, isolated pili, method for excreting proteins using these pili and their use
WO1991018980A1 (en) * 1990-06-01 1991-12-12 Cetus Corporation Compositions and methods for identifying biologically active molecules
WO1991019818A1 (en) * 1990-06-20 1991-12-26 Affymax Technologies N.V. Peptide library and screening systems
DE4122598C1 (en) * 1991-07-08 1992-07-30 Deutsches Krebsforschungszentrum Stiftung Des Oeffentlichen Rechts, 6900 Heidelberg, De
WO1993010214A1 (en) * 1991-11-15 1993-05-27 George Georgiou Expression of proteins on bacterial surface
WO1993018163A2 (en) * 1992-03-13 1993-09-16 The Rockefeller University Delivery and expression of a hybrid surface protein on the surface of gram positive bacteria
FR2699538A1 (en) * 1992-12-22 1994-06-24 Agronomique Inst Nat Rech Subunit of a CS31A protein capsule modified with at least one heterologous peptide, CS31A protein capsule and microorganisms carrying these subunits; methods of obtaining and using them.

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5223409A (en) * 1988-09-02 1993-06-29 Protein Engineering Corp. Directed evolution of novel binding proteins
CA2109602C (en) * 1990-07-10 2002-10-01 Gregory P. Winter Methods for producing members of specific binding pairs
GB9015198D0 (en) * 1990-07-10 1990-08-29 Brien Caroline J O Binding substance

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0264150A1 (en) * 1986-10-13 1988-04-20 Solvay Microorganisms comprising pili as carrier proteins, isolated pili, method for excreting proteins using these pili and their use
WO1991018980A1 (en) * 1990-06-01 1991-12-12 Cetus Corporation Compositions and methods for identifying biologically active molecules
WO1991019818A1 (en) * 1990-06-20 1991-12-26 Affymax Technologies N.V. Peptide library and screening systems
DE4122598C1 (en) * 1991-07-08 1992-07-30 Deutsches Krebsforschungszentrum Stiftung Des Oeffentlichen Rechts, 6900 Heidelberg, De
WO1993010214A1 (en) * 1991-11-15 1993-05-27 George Georgiou Expression of proteins on bacterial surface
WO1993018163A2 (en) * 1992-03-13 1993-09-16 The Rockefeller University Delivery and expression of a hybrid surface protein on the surface of gram positive bacteria
FR2699538A1 (en) * 1992-12-22 1994-06-24 Agronomique Inst Nat Rech Subunit of a CS31A protein capsule modified with at least one heterologous peptide, CS31A protein capsule and microorganisms carrying these subunits; methods of obtaining and using them.

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
J. MCCAFFERTY ET AL.: "Phage antibodies: filamentous phage displaying antibody variable domains", NATURE, vol. 348, 6 December 1990 (1990-12-06), MACMILLAN JOURNALS LTD., LONDON,UK, pages 552 - 554 *
L.S. FROST ET AL.: "Characterization and sequence analysis of pilin from F-like plasmids", J. BACTERIOL., vol. 164, no. 12, AM. SOC. MICROBIOL.,BALTIMORE,US;, pages 1238 - 1247 *
L.S. FROST ET AL.: "DNA sequence of the F traALE region that includes the gene for F pilin", J. BACTERIOL., vol. 160, no. 1, AM. SOC. MICROBIOL.,BALTIMORE,US;, pages 395 - 401 *
S.M.C. NEWTON ET AL.: "Immune response to cholera toxin epitope inserted in Salmonella flagellin", SCIENCE, vol. 244, 7 April 1989 (1989-04-07), AAAS,WASHINGTON,DC,US, pages 70 - 72 *

Cited By (34)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6040136A (en) * 1990-12-03 2000-03-21 Genentech, Inc. Enrichment method for variant proteins with altered binding properties
US6607881B1 (en) 1995-09-13 2003-08-19 Bioinvent International, Ab Combined ligand and receptor display
WO1997010330A1 (en) * 1995-09-13 1997-03-20 Bioinvent International Ab Combined ligand and receptor display
US6358742B1 (en) 1996-03-25 2002-03-19 Maxygen, Inc. Evolving conjugative transfer of DNA by recursive recombination
US6387702B1 (en) 1996-03-25 2002-05-14 Maxygen, Inc. Enhancing cell competence by recursive sequence recombination
US6482647B1 (en) 1996-03-25 2002-11-19 Maxygen, Inc. Evolving susceptibility of cellular receptors to viral infection by recursive recombination
EP0932670A4 (en) * 1996-03-25 1999-08-04
US6391552B2 (en) 1996-03-25 2002-05-21 Maxygen, Inc. Enhancing transfection efficiency of vectors by recursive recombination
EP0932670A1 (en) * 1996-03-25 1999-08-04 Maxygen, Inc. Evolving cellular dna uptake by recursive sequence recombination
US6096548A (en) * 1996-03-25 2000-08-01 Maxygen, Inc. Method for directing evolution of a virus
EP0944737A1 (en) * 1996-11-07 1999-09-29 RAMOT UNIVERSITY AUTHORITY FOR APPLIED RESEARCH & INDUSTRIAL DEVELOPMENT LTD. Representations of bimolecular interactions
EP0944737A4 (en) * 1996-11-07 2004-11-10 Univ Ramot Representations of bimolecular interactions
US6448083B1 (en) 1997-08-29 2002-09-10 Selective Genetics, Inc. Receptor-mediated gene delivery using bacteriophage vectors
US6451527B1 (en) 1997-08-29 2002-09-17 Selective Genetics, Inc. Methods using genetic package display for selecting internalizing ligands for gene delivery
WO1999010485A1 (en) * 1997-08-29 1999-03-04 Selective Genetics, Inc. Methods using phage display for selecting internalizing ligands for gene delivery
US6589730B1 (en) 1997-08-29 2003-07-08 Selective Genetics, Inc. Methods for identifying protein-protein interactions by selective transduction
US7148202B2 (en) 1997-08-29 2006-12-12 Selective Genetics, Inc. Receptor-mediated gene delivery using bacteriophage vectors
US6723512B2 (en) 1997-08-29 2004-04-20 Selective Genetics Inc. Methods using genetic package display for detecting and identifying protein-protein interactions that facilitate internalization and transgene expression and cells or tissues competent for the same and methods for evolving gene delivery vectors
US6472146B1 (en) 1997-08-29 2002-10-29 Selective Genetics, Inc. Methods for identification on internalizing ligands and identification of known and putative ligands
US6927025B1 (en) 1997-09-03 2005-08-09 Biovation Limited Methods for protein screening
WO1999011777A1 (en) * 1997-09-03 1999-03-11 Biovation Limited Methods for protein screening
US8685893B2 (en) 1998-07-27 2014-04-01 Genentech, Inc. Phage display
WO2000023465A3 (en) * 1998-10-19 2000-08-31 Mitotix Inc Methods and reagents for isolating biologically active peptides
US6420110B1 (en) 1998-10-19 2002-07-16 Gpc Biotech, Inc. Methods and reagents for isolating biologically active peptides
AU761648B2 (en) * 1998-10-19 2003-06-05 Gpc Biotech Inc. Methods and reagents for isolating biologically active peptides
WO2000023465A2 (en) * 1998-10-19 2000-04-27 Mitotix, Inc. Methods and reagents for isolating biologically active peptides
WO2000029555A1 (en) * 1998-11-17 2000-05-25 Selective Genetics, Inc. Methods using genetic package display for detecting and identifying protein-protein interactions
EP2857516A1 (en) 2000-04-11 2015-04-08 Genentech, Inc. Multivalent antibodies and uses therefor
US6686154B2 (en) 2000-06-05 2004-02-03 Zyomyx, Inc. Screening of phage displayed peptides without clearing of the cell culture
WO2001094950A3 (en) * 2000-06-05 2002-05-10 Zyomyx Inc Screening of phage displayed peptides without clearing of the cell culture
WO2001094950A2 (en) * 2000-06-05 2001-12-13 Zyomyx, Inc. Screening of phage displayed peptides without clearing of the cell culture
EP2014303A2 (en) 2000-07-27 2009-01-14 Genentech, Inc. APO-2L receptor agonist and CPT-11 synergism
WO2002046213A3 (en) * 2000-11-07 2003-03-13 Gpc Biotech Inc Methods and reagents for isolating angiogenic modulators
WO2002046213A2 (en) * 2000-11-07 2002-06-13 Gpc Biotech Inc. Methods and reagents for isolating angiogenic modulators

Also Published As

Publication number Publication date
US5516637A (en) 1996-05-14
AU697865B2 (en) 1998-10-22
AU2828295A (en) 1996-01-05
EP0722495A1 (en) 1996-07-24
CA2169358A1 (en) 1995-12-21
JPH09504181A (en) 1997-04-28

Similar Documents

Publication Publication Date Title
AU697865B2 (en) A method for displaying proteins
JP3447731B2 (en) Improved revealing phage
US5627024A (en) Lambdoid bacteriophage vectors for expression and display of foreign proteins
KR101268939B1 (en) Phage display using cotranslational translocation of fusion polypeptides
JP3210342B2 (en) Total synthetic affinity reagent
AU766253B2 (en) Modulation of polypeptide display on modified filamentous phage
AU2008290537B2 (en) pVII phage display
JP2007513602A5 (en) Expression vectors, polypeptide display libraries, and methods for their production and use
WO2000029555A1 (en) Methods using genetic package display for detecting and identifying protein-protein interactions
US20200385705A1 (en) Novel methods for displaying cyclic peptides on bacteriophage particles
US20020068272A1 (en) Methods using genetic package display for detecting and identifying protein-protein interactions that facilitate internalization and transgene expression and cells or tissues competent for the same and methods for evolving gene delivery vectors
US8969253B2 (en) Method for screening phage display libraries against each other
KR20090109193A (en) Production method of antibody phage display library, the antibody phage display library produced thereby and the phagemid vector comprising said antibody phage display library
US20030148263A1 (en) Methods and compositions using genetic package display
EP1536005A1 (en) Method of assaying interaction between proteins
AU2010217551B2 (en) Signal sequence-independent pIX phage display
KR100458083B1 (en) Method for the construction of phage display library using helper phage variants
US20050130124A1 (en) Phagemid display system
Cebe et al. Size of the ligand complex between the N-terminal domain of the gene III coat protein and the non-infectious phage strongly influences the usefulness of in vitro selective infective phage technology
EP1724347A2 (en) DNA & protein binding miniature proteins

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AU CA JP

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): AT BE CH DE DK ES FR GB GR IE IT LU MC NL PT SE

WWE Wipo information: entry into national phase

Ref document number: 2169358

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 1995923861

Country of ref document: EP

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWP Wipo information: published in national office

Ref document number: 1995923861

Country of ref document: EP

WWW Wipo information: withdrawn in national office

Ref document number: 1995923861

Country of ref document: EP