US20030186374A1 - Multi-chain eukaryotic display vectors and uses thereof - Google Patents

Multi-chain eukaryotic display vectors and uses thereof Download PDF

Info

Publication number
US20030186374A1
US20030186374A1 US10/262,646 US26264602A US2003186374A1 US 20030186374 A1 US20030186374 A1 US 20030186374A1 US 26264602 A US26264602 A US 26264602A US 2003186374 A1 US2003186374 A1 US 2003186374A1
Authority
US
United States
Prior art keywords
eukaryotic
cell
chain
vector
yeast
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US10/262,646
Other languages
English (en)
Inventor
Simon Hufton
Hendricus Hoogenboom
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Dyax Corp
Original Assignee
Dyax Corp
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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=23271723&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20030186374(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority to US10/262,646 priority Critical patent/US20030186374A1/en
Application filed by Dyax Corp filed Critical Dyax Corp
Assigned to DYAX CORPORATION reassignment DYAX CORPORATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: HOOGENBOOM, HENDRICUS R.J.M., HUFTON, SIMON E.
Publication of US20030186374A1 publication Critical patent/US20030186374A1/en
Priority to US12/625,337 priority patent/US9012181B2/en
Priority to US13/073,657 priority patent/US9005927B2/en
Priority to US13/170,448 priority patent/US9116149B2/en
Priority to US13/170,438 priority patent/US9068980B2/en
Priority to US13/170,362 priority patent/US9040258B2/en
Priority to US13/937,915 priority patent/US9034601B2/en
Priority to US14/718,656 priority patent/US9556428B2/en
Priority to US15/377,846 priority patent/US10059936B2/en
Priority to US16/042,004 priority patent/US10577599B2/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • 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
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • 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
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/543Immunoassay; Biospecific binding assay; Materials therefor with an insoluble carrier for immobilising immunochemicals
    • G01N33/554Immunoassay; Biospecific binding assay; Materials therefor with an insoluble carrier for immobilising immunochemicals the carrier being a biological cell or cell fragment, e.g. bacteria, yeast cells
    • 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
    • C12N2795/00Bacteriophages
    • C12N2795/00011Details
    • C12N2795/00041Use of virus, viral particle or viral elements as a vector
    • C12N2795/00043Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • 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
    • C12N2800/00Nucleic acids vectors
    • C12N2800/10Plasmid DNA
    • C12N2800/108Plasmid DNA episomal vectors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2830/00Vector systems having a special element relevant for transcription
    • C12N2830/001Vector systems having a special element relevant for transcription controllable enhancer/promoter combination
    • C12N2830/002Vector systems having a special element relevant for transcription controllable enhancer/promoter combination inducible enhancer/promoter combination, e.g. hypoxia, iron, transcription factor
    • 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
    • C12N2840/00Vectors comprising a special translation-regulating system
    • C12N2840/20Vectors comprising a special translation-regulating system translation of more than one cistron

Definitions

  • phage display technology whereby non-native (heterologous) polypeptides or proteins are expressed and anchored on the surface (“displayed”) of a bacteriophage, is a powerful tool for identifying molecules possessing biological activities of interest, for example, peptide ligands that bind with high specificity and/or affinity to a given target molecule.
  • Libraries of synthetic oligonucleotides can be cloned in frame into the coding sequence of genes encoding a phage surface protein, for example gene III or gene VIII of phage M13.
  • clones when expressed, are “displayed” on the phage surface as a plurality, due to the variation in sequence of the oligonucleotides used, of peptide-capsid fusion proteins.
  • These peptide display libraries are then screened for binding to target molecules, usually by affinity selection or “biopanning” (Ladner, R. et al., 1993; Kay et al., 1996; Hoogenboom, H. et al., 1997).
  • Phage display library screening is highly advantageous over other screening methods due to the vast number of different polypeptides (typically exceeding 1 ⁇ 10 9 ) that can be contained in a single phage display library. This allows for the screening of a highly diverse library in a single screening step. Display of small peptides or single chain proteins on phage is advantageous as long as intracellular processing or post-translational modification (of which phage or prokaryotic hosts are not capable) are not necessary or desired.
  • heterologous polypeptide may require various post-translational modifications, intracellular structures, and a compliment of specialized enzymes and chaperone proteins that are necessary to transport, to glycosylate, to conform, to assemble, and to anchor the display polypeptide properly on the surface of the host cell; however, none of these processes can be accomplished by bacteriophage or prokaryotic cell processes.
  • the present invention provides a eukaryotic expression vector capable of displaying a multi-chain polypeptide on the surface of a host cell such that a biological activity of the multi-chain polypeptide is exhibited at the surface of the host cell.
  • a eukaryotic expression vector capable of displaying a multi-chain polypeptide on the surface of a host cell such that a biological activity of the multi-chain polypeptide is exhibited at the surface of the host cell.
  • Such a vector allows for the display of more complex biologically active polypeptides, e.g., biologically active multi-chain polypeptides, than can be obtained via conventional phage display technology.
  • the present invention relates to the display and isolation of biologically active polypeptides. Specifically, the present invention is directed to the design and use of novel multi-chain display vectors.
  • the present invention describes and enables the successful display of a multi-chain polypeptide on the surface of a eukaryotic host cell.
  • Preferred vectors are described for expressing the chains of a multi-chain polypeptide in a host cell separately and independently (e.g., under separate vector control elements, and/or on separate expression vectors, thus forming a matched vector set).
  • the use of such matched vector sets provides a level of flexibility and versatility in the generation of display libraries, for example the ability to generate and to display multi-chain polypeptides by combining and recombining vectors that express a variety of the individual chains of a multi-chain polypeptide. Entire repertoires of novel chain combinations can be devised using such vector sets.
  • the invention further provides the ability to combine the power of phage display technology (with its ease of manipulation and magnitude of diversity) with the potential complexity and versatility of a multi-chain eukaryotic display vector (or vector set).
  • the particular methods described herein permit a practitioner to efficiently transfer sequence information of a peptide library (or selected members of the library) between phage display and eukaryotic display systems, accomplished either through the physical transfer of the sequence information from one display vector to the other (using conventional genetic engineering techniques) or through the use of a novel dual display vector, operable in both eukaryotic display systems and phage display systems (which necessarily involve prokaryotic expression).
  • the present invention is directed to a novel vector, useful in a eukaryotic host cell to display a multi-chain polypeptide on the surface of the host cell such that a biological activity of the multi-chain polypeptide is exhibited at the surface of the host cell, e.g., the binding activity of a multi-chain polypeptide.
  • a biological activity of the multi-chain polypeptide is exhibited at the surface of the host cell, e.g., the binding activity of a multi-chain polypeptide.
  • the multi-chain eukaryotic display vector can exist as a single vector or as multiple independent vectors of a vector set.
  • vector refers to either a single vector molecule or a vector set.
  • the display vector is a shuttle vector, or more precisely a dual display vector, wherein the vector is capable of displaying a biologically active multi-chain polypeptide on the surface of a eukaryotic host cell transformed with that vector, or on the surface of a bacteriophage generated as a result of prokaryotic expression.
  • the vector can exist as a vector set, wherein each chain of a multi-chain polypeptide is encoded on one of a matched pair of vectors such that when the vector pair is present in a single eukaryotic cell, the chains of the multi-chain polypeptide associate and the biological activity of the multi-chain polypeptide is exhibited at the surface of the eukaryotic cell.
  • the eukaryotic multi-chain display vector of the present invention comprises polynucleotides that encode polypeptide chains of the multi-chain polypeptide.
  • a first polynucleotide encodes a first chain of the multi-chain polypeptide linked to an anchor protein.
  • Other polynucleotides of the vector (or vector set) encode other chains of the multi-chain polypeptide. All of the polynucleotides of the display vector(s) are operably-situated in the display vector such that a host eukaryotic cell, transformed with the vector (or vector set), displays the multi-chain polypeptide on the surface of the host cell such that the biological activity of the multi-chain polypeptide is exhibited at the surface of the cell.
  • the multi-chain polypeptide encoded by the multi-chain display vector(s) of the present invention exists as either a two-, three-, four-, or multi-chain polypeptide. More preferably, the multi-chain polypeptide is a two-chain or four-chain polypeptide comprised of two different chains. More preferably, the multi-chain polypeptide is selected from a group of multi-chain polypeptides consisting of T cell receptors, MHC class I molecules, MHC class II molecules, and immunoglobulin Fab fragments. More preferably, the multi-chain polypeptide is an IgA, IgD, IgE, IgG, IgM, or biologically active fragment thereof.
  • the multi-chain polypeptide is a Fab fragment, wherein the first polynucleotide of the multi-chain display vector comprises a segment that encodes the V H and C H 1 domains of an Ig heavy chain, and a second polynucleotide comprises a segment that encodes an Ig light chain (V L and C L domains).
  • a first polynucleotide encoding a first chain of the multi-chain polypeptide is linked to an anchor protein.
  • the anchor protein is a cell surface protein of a eukaryotic cell or a functional fragment thereof. More preferably, the anchor protein is ⁇ -agglutinin, a-agglutinin, Aga1p, Aga2p, or FLO1.
  • linkage of the first chain polypeptide to an anchor protein can be achieved by a variety of molecular biology techniques.
  • the first polynucleotide encoding a first chain of the multi-chain polypeptide is expressed in a eukaryotic host cell as a first chain-anchor fusion protein; most preferably a first chain:Aga2p fusion protein.
  • one or more of the chains of the multi-chain polypeptide expressed by the vector(s) in a host cell is linked to a reporter gene or tag.
  • the tag is an epitope tag selected from the group consisting of 6 ⁇ His tag, HA tag, and myc tag.
  • each chain of the multi-chain polypeptide is linked to a different tag.
  • the multi-chain display vector(s) of the present invention provide cloning sites to facilitate transfer of the polynucleotide sequence(s) that encode the chains of the multi-chain polypeptide.
  • cloning sites comprise restriction endonuclease recognition site (i.e., restriction sites) positioned to facilitate excision and insertion of polynucleotides that encode one or more chains of a multi-chain polypeptide.
  • restriction sites are preferably located at the 5′ and 3′ ends of the polynucleotide(s) that encode the chains of the multi-chain polypeptide.
  • the vector of the present invention can contain only two restriction sites positioned at the ends of the polynucleotide segment that includes all segments encoding the chains of the multi-chain polypeptide, or, preferably, restriction sites occur at the ends of each polynucleotide segment encoding a chain of the multi-chain polypeptide (FIGS. 1 and 2).
  • each restriction endonuclease recognition site is a unique recognition site in the vector.
  • the vector (or vector set) of the present invention can be operable in a variety of eukaryotic host cells, and optionally can be operable in prokaryotic cells (e.g., bacteria).
  • the multi-chain display vector of the present invention is an animal cell display vector, a plant cell display vector, a fungus cell display vector, or a protist cell display vector.
  • the display vector is a yeast display vector.
  • the yeast display vector is operable in Saccharomyces cerevisiae.
  • the invention is directed to a method for using the vector (or vector set) described and taught herein for displaying a multi-chain polypeptide on the surface of a eukaryotic host cell, wherein the vector (or vector set) is introduced into the eukaryotic cell and the host cell is cultured under conditions suitable for expression, transportation, and association of the chains of the multi-chain polypeptide such that the biological activity of the multi-chain polypeptide is exhibited at the surface of the host cell.
  • the polynucleotides encoding the chains of the multi-chain polypeptide can be introduced into the host cell via one or more vectors.
  • the mode of introducing the vector(s) into the host cell includes any of the methods for introducing genetic material into a cell known in the art.
  • Preferred modes include such transformation techniques known in the art, including but not limited to electroporation, microinjection, viral transfer, ballistic insertion, and the like.
  • Another preferred mode for introducing eukaryotic multi-chain display vectors into a host cell includes the fusion of two haploid eukaryotic cells, each expressing at least one of the chains of the multi-chain polypeptide, to produce a diploid host cell expressing both (all) chains, such that the biological activity of the multi-chain polypeptide is exhibited at the surface of the resulting diploid host cell.
  • each of the two haptoid cells can contain one (or more) of the vectors of a vector set (as described herein), such that the biological activity of the multi-chain polypeptide is exhibited at the surface of the diploid host cell resulting from the haploid/haploid fusion.
  • the haploid host cell pair is of opposite mating types, thus facilitating the fusion (“mating”) of the two eukaryotic haploid cells.
  • Another object of the invention is directed to a eukaryotic host cell that exhibits at the surface of the cell the biological activity of a multi-chain polypeptide.
  • the eukaryotic host cell is preferably an animal cell, a plant cell, a fungus cell, or a protist cell. More preferably the eukaryotic host cell is a yeast cell.
  • the yeast host cell is selected from the genera Saccharomyces, Pichia, Hansenula, Schizosaccharomyces, Kluyveromyces, Yarrowia, and Candida.
  • the eukaryotic host cell is S. cerevisiae .
  • Eukaryotic host cells of the present invention can be of any genetic construct but are preferably haploid or diploid.
  • One embodiment of the present invention is directed to a eukaryotic haploid cell pair (preferably of opposite mating types) wherein the first haploid cell expresses at least a first polynucleotide encoding a first chain of a biologically active multi-chain polypeptide linked to an anchor protein, and the second haploid cell expresses at least a second polynucleotide encoding a second chain of the multi-chain polypeptide.
  • fusion of this haploid cell pair results in a diploid cell that exhibits the biological activity of the multi-chain polypeptide at the surface of the cell.
  • the present invention is further directed to assemblages of the various embodiments described herein, which form novel libraries of multi-chain polypeptides or of the polynucleotides that encode them.
  • Libraries of the present invention comprise a plurality of vectors that encode a multi-chain polypeptide such that the vector is operable in a eukaryotic host cell to direct expression and secretion of the chains of the multi-chain polypeptide, association of the chains such that the biological activity of the multi-chain polypeptide is constituted, and anchoring of at least one chain of the multi-chain polypeptide such that the biological activity of the multi-chain polypeptide is exhibited at the surface of the eukaryotic host cell.
  • the library of the present invention is comprised of library members that encode a multiplicity of different multi-chain polypeptides.
  • the library is comprised of library members that encode a multiplicity of variant multi-chain polypeptides (designed and produced by the variegation of a multi-chain polypeptide template).
  • Novel multi-chain library assemblages of the present invention include vector libraries, vector set libraries, host cell libraries, and host cell pair libraries as described and taught herein.
  • a related aspect of the present invention is directed to a method for transferring nucleic acid sequence information encoding a biologically active multi-chain polypeptide between a phage display vector and a eukaryotic display vector.
  • One transfer method comprises inserting polynucleotide sequences encoding the chains of a multi-chain polypeptide obtained from a phage display vector into a eukaryotic multi-chain display vector as described and taught herein. Transfer of the nucleic acid sequence information encoding the chains of a multi-chain polypeptide can occur as a single transfer event, or can occur as separate and independent transfer events of nucleic acid sequence information encoding each of the chains of the multi-chain polypeptide. Similarly, the sequence information encoding each of the chains of a multi-chain polypeptide can be transferred from one display vector or from multiple different display vectors.
  • Another method for transferring nucleic acid sequence information encoding a biologically active multi-chain polypeptide between a phage display vector and a eukaryotic display vector comprises inserting polynucleotide sequences encoding the chains of a multi-chain polypeptides obtained from a eukaryotic multi-chain display vector as described and taught herein into a phage display vector.
  • the phage display-eukaryotic display transfer process of the present invention is bidirectional, i.e., it can occur from phage display vector to eukaryotic display vector or from eukaryotic display vector to phage display vector.
  • nucleic acid sequence information between a phage display vector and the eukaryotic vector of the present invention can be achieved by a variety of genetic transfer methods known in the art (e.g., genetic engineering technology such as recombinant DNA technology).
  • Preferred modes of transfer include techniques of restriction digestion, PCR amplification, or homologous recombination (e.g., see Liu, Q. et al., 2000; Walhout, A. et al., 2000).
  • the present invention is also directed to methods for detecting and isolating multi-chain polypeptides that exhibit a biological activity of interest to the practitioner.
  • the methods of the present invention permit the detection of desirable interactions between multi-chain polypeptides and another molecular species, preferably protein-protein interactions, and more preferably interactions between multi-chain polypeptides and their ligands/substrates (i.e., target molecules).
  • the nature of this interaction comprises a non-covalent association (i.e., binding) between the molecular species, however the nature of the binding can be transient (e.g., enzyme-substrate binding) or of high affinity/avidity (e.g., as with affinity ligands useful in separations, diagnostics, and/or therapeutics).
  • the method of the present invention is useful to screen a library of multi-chain polypeptides (displayed on the surface of a eukaryotic host cell) by detecting those members of the library that exhibit a biological activity of interest to the practitioner.
  • host cells which display multi-chain polypeptides exhibiting the biological activity of interest, are isolated. Isolated host cells can then, optionally, undergo repeated rounds of screening, or otherwise be manipulated to characterize or to utilize the polypeptide sequence of the displayed multi-chain polypeptide.
  • the screening method of the present invention can be combined with a (preliminary) phage display screen and transfer of the selected phage display isolates to the eukaryotic display system described herein for eukaryotic display screening.
  • a library of multi-chain polypeptides displayed on the surface of a diploid eukaryotic host cell can be screened to detect (and optionally to isolate) multi-chain polypeptides that exhibit a biological activity of interest to the practitioner.
  • the diploid eukaryotic host cell is the product of the fusion of a haploid eukaryotic host cell pair as described and taught herein.
  • screened diploid cells displaying a multi-chain polypeptide exhibiting a biological activity of interest can be isolated and then, optionally, undergo meiosis, whereby the daughter (haploid) cells express separate chains of the selected multi-chain polypeptide.
  • Daughter cells can then, optionally, be fused with other haploid cells that express chains of a multi-chain polypeptide (e.g., other daughter cells from the same sub-population of isolated diploid cells), producing a recombination population of diploid eukaryotic host cells that display a multi-chain polypeptide on their surface.
  • Additional rounds of screening and repeat recombination of the individual chains of the selected multi-chain polypeptide can be performed, and ultimately the polypeptide sequence of the displayed multi-chain polypeptide can be characterized or utilized as discussed above.
  • Recombination of the selected haploid daughter cells can also be recombined (via cellular fusion) with other biased or non-biased eukaryotic display vectors to produce novel multi-chain display host cell libraries.
  • the eukaryotic display vector can be used to create a eukaryotic display library, such as a yeast display library, comprising a plurality of such eukaryotic display vectors.
  • a plurality of eukaryotic display vectors will encode a heterogeneous population of multi-chain polypeptides, yielding a displayed repertoire of multi-chain polypeptides, e.g., at least 10 4 , preferably at least 10 5 , more preferably at least 10 6 , more preferably at least 10 7 , more preferably at least 10 8 , most preferably at least 10 9 different polypeptides.
  • the anchor is a polypeptide operable as an anchor on the surface of a eukaryotic cell and operable as an anchor on the surface of a phage.
  • the anchor is a portion of a surface protein that anchors to the cell surface of a eukaryotic host cell and to the surface of a phage.
  • the anchor and one chain of the multi-chain polypeptide are expressed as a fusion protein.
  • the anchor and one chain of the multi-chain polypeptide become linked on expression via an indirect linkage, such as, preferably, a Jun/Fos linkage.
  • the invention is directed to a method for displaying, on the surface of a eukaryotic host cell, a biologically active multi-chain polypeptide comprising at least two polypeptide chains, comprising the steps of introducing into a eukaryotic host cell a first eukaryotic vector comprising a first polynucleotide encoding a first polypeptide chain of a biologically active multi-chain polypeptide linked to a cell surface anchor, wherein said vector is operable in a eukaryotic host cell to direct expression and secretion of said first chain; and a second eukaryotic vector comprising a second polynucleotide encoding a second polypeptide chain of said multi-chain polypeptide, wherein said vector is operable in a eukaryotic host cell to direct expression and secretion of said second chain, wherein a eukaryotic host cell transformed with said first eukaryotic vector and said second eukaryotic vector exhibits,
  • the invention is directed to a method for displaying, on the surface of a eukaryotic host cell, a biologically active multi-chain polypeptide comprising at least two polypeptide chains, comprising the steps of introducing into a eukaryotic host cell a eukaryotic display vector, a eukaryotic display vector set, or a dual display vector as described above, and culturing said host cell under conditions suitable for expression of said polynucleotides.
  • the present invention further provides a eukaryotic host cell comprising a eukaryotic display vector, a eukaryotic display vector set, or a dual display vector as described herein.
  • Suitable eukaryotic host cells can be animal cells, plant cells, or fungal cells.
  • the eukaryotic host cell will be a mammalian cell, an insect cell, and a yeast cell.
  • the eukaryotic host cell will be a yeast cell, e.g., selected from the genus Saccharomyces, Pichia, Hansenula, Schizosaccharomyces, Kluyveromyces, Yarrowia, Debaryomyces, or Candida.
  • Preferred yeast hosts include Saccharomyces cerevisiae, Hansenula polymorpha, Kluyveromyces lactis, Pichia pastoris, Schizosaccharomyces pombe , and Yarrowia lipolytica .
  • the most preferred yeast host cell is Saccharomyces cerevisiae.
  • FIG. 1 is a schematic diagram that illustrates the phage display-eukaryotic display transfer system.
  • the genetic information encoding the chains of a Fab polypeptide are transferred from a phage display vector to a multi-chain eukaryotic vector of the present invention as a single, excised nucleic acid. Unwanted intervening genetic elements (if any) are then replaced.
  • FIG. 2 is a schematic diagram that illustrates the phage display/eukaryotic display transfer system wherein the genetic information encoding the chains of a Fab polypeptide are independently and separately transferred from a phage display vector to a multi-chain eukaryotic vector of the present invention.
  • FIG. 3 is a schematic diagram of the multi-chain yeast display vector, pTQ3, according to the invention, having unique cloning sites for insertion of at least two chains of a multi-chain polypeptide (e.g., Fab light and heavy chain components), with additional elements arranged so that the two chains are independently expressed by induction of tandem GALL promoters.
  • a first chain e.g., an Ig light chain
  • ApaLI/AscI fragment is expressed as a soluble secretory protein using the Aga2p signal sequence (Aga2p/ss) and fused with an HA epitope tag.
  • a second chain (e.g., an Ig heavy chain fragment), inserted as an SfiI/NotI fragment, is expressed as a cell surface bound fusion protein using the Aga2p/ss and anchoring protein subunit (mature Aga2p).
  • the second chain is similarly fused with a myc epitope tag.
  • Other elements useful for plasmid replication e.g., pMB1-ori and Cen6/ARSH4
  • useful as selective markers i.e., ampR and TRP
  • FIGS. 4 A- 4 C are representations of data demonstrating independent expression of fusion proteins.
  • FIG. 4A shows the expression of the 45 kD Aga 2 p-V H -C H 1 fusion protein in yeast host cells EBY100 pTQ3-F2 and EBY100 pTQ3-PH1
  • FIG. 4B shows the expression of the 30 kD V L -C L chain in yeast host cells EBY100 pTQ3-F2 and EBY100 pTQ3-PH1. No fusion products were detected in either empty vector control.
  • samples were prepared both before ( ⁇ ) and after (+) galactose induction of the GAL1 promoters operable in the yeast display vectors.
  • FIG. 4 C is a representation of immunofluorescence detection of assembled Fab antibodies on the yeast cell surface.
  • (a) phase contrast (b) detection of HC (c) detection of LC
  • FIGS. 5 A-C represent a series of cytometric plots.
  • FIG. 5A depicts yeast cells transformed with pTQ3-F2 (left panel) and pTQ3-PH1 (right panel) constructs were left untreated (dotted line) or induced for 48 hours at 20° C. (light grey line). Heavy chain (a), light chain display (b) and antigen binding (c) were analyzed using flow cytometry.
  • FIG. 6 is a histogram plot illustrating whole cell ELISA of three different anti-streptavidin Fabs displayed on the surface of yeast host cells EBY100 pTQ3-F2, EBY100 pTQ3-A12, and EBY100 pTQ3-4C8. Antigen binding, LC display and HC display are indicated respectively.
  • FIG. 7 is a cytometric plot of yeast cell mix. EBY100 pTQ3-F2, EBY100 pTQ3-A12, and EBY100 pTQ3-A12/pESC were double-labeled for both antigen binding and LC display. A plot of LC display against antigen binding and a gating for normalized antigen binding are indicated.
  • FIGS. 8 A- 8 D are representations of data showing binding to yeast repertoires and individually selected yeast clones at different antigen concentrations.
  • FIG. 8A shows a series of histograms of antigen binding and Fab display are shown for the unselected library (a) and polyclonal outputs of selection round 1, 2 and 3 (b, c, d).
  • the diversified anti-streptavidin yeast repertoire was subjected to three rounds of FACS. The sorting gate used in each library selections is indicated.
  • FIG. 8B shows polyclonal FACS analysis at different antigen concentrations of a FACS affinity selection campaign of a anti-streptavidin repertoire.
  • FIG. 8C shows data obtained from yeast cells displaying the wild-type F2 (represented by “o”) and mutants R2E10 (represented by triangles), R3B 1 (represented by squares) and R3H3 (represented by diamonds) were labeled with anti-HA mAb and streptavidin-PE. The mean fluorescence for streptavidin binding was monitored over time. The dissociation rate constant is calculated from the slope of the line.
  • 8D shows a series of cytometric plots of two selection campaigns using either Kingfisher in combination with FACS (Right column) or FACS alone (right column).
  • the cytometric plots indicate the increasing percentage of antigen binding cells through unselected (a) round 1 (b) and round 2 (c) of selection.
  • FIG. 9 is a schematic diagram of the heavy chain yeast display vector, pTQ5-HC, according to the invention, having a heavy chain fragment insert under the control of an inducible GAL1 promoter.
  • the Ig heavy chain fragment is positioned as a SfiI/NotI insert fragment, and is expressed as a cell surface bound fusion protein using the Aga2p signal sequence (Aga2p/ss) and anchoring protein subunit (Aga2p protein).
  • the heavy chain fragment (HC) is fused to a myc epitope tag.
  • plasmid replication i.e., pMB 1-ori and Cen6/ARSH4
  • yeast mating i.e., Mat ⁇ terminator
  • ampR and TRP useful as selective markers
  • FIG. 10 is a representation of a western blot demonstrating expression of the 45 kD Aga2p-HC fusion product as detected with an anti c-Myc antibody in the haploid parent yeast cell EBY100 pTQ5-HC (lane 2) compared to the (control) empty vector yeast host cell EBY100 pTQ5 (lane 1) and the (standard) Fab display vector yeast host cell EBY100 pTQF2 (lane 3).
  • FIG. 11 is a series of cytometric plots showing HC display on the surface of yeast cells without the presence of a light chain at time equal to zero (i.e., background; solid black lines) and 48 hours after induction (dotted lines).
  • Yeast cells EBY100 pTQ5-HC, and control yeast cells EBY100 pTQ5 were labeled with anti-CHI and rabbit anti-mouse IgG FITC to detect the presence of the HC, and also with streptavidin FITC (strep-FITC) to detect antigen binding activity on the yeast surface.
  • HC only can be seen displayed on the yeast cell surface but does not have any antigen binding activity in the absence of a paired LC.
  • FIG. 12 is a schematic diagram of the light chain yeast expression vector, pTQ6-LC, according to the invention, having a light chain insert under the control of an inducible GAL1 promoter.
  • the Ig light chain is positioned as an ApaL1/AscI insert fragment, is expressed as a soluble protein using the Aga2p/ss.
  • the light chain fragment (LC) is also fused with a HA epitope tag.
  • Other elements useful for plasmid replication e.g., pUC1-ori and Cen6/ARSH4
  • useful as selection markers i.e., ampR and Blastocidin®
  • FIG. 13 is a representation of a western blot demonstrating expression of the 60 kD light chain polypeptide as detected in the culture supernatant with an anti-HA antibody in the haploid parent yeast cell W303 pTQ6-LC (lane S2) compared to the (control) empty vector yeast host cell W303 pYC6 (lane S1).
  • FIG. 14 is a histogram plot illustrating whole cell ELISA determination of streptavidin binding activity on the cell surface of parent haploid yeast cells (W303 pTQ6-LC and EBY10O pTQ5-HC) compared to the derived diploid yeast cell (DIPLOID LC/HC) and control empty vector yeast host cell W303 pYC6 and standard Fab display vector yeast host cell EBY100 pTQ3-F2.
  • FIGS. 15 A- 15 C are a series of FACS histograms showing antigen binding and light chain display on an anti streptavidin haploid HC parent (A) and a diploid control containing empty LC and HC expression plasmids (B) and a positive diploid expressing a streptavidin specific Fab on its surface (C).
  • FIG. 16 is a representation of a western blot demonstrating expression of the 30 kD LC polypeptide as detected with an anti-HA antibody in the diploid yeast cell formed by mating EBY100 pTQ5-HC with W303 pTQ6-LC (lane 3) compared to the (control) diploid yeast cell formed by mating EBY100 pTQ5 with W303 pYC6 (lane 2), and the parent LC vector yeast host cell W303 pTQ6-LC (lane 1).
  • FIG. 17 is an illustration of a western blot demonstrating expression of the 45 kD Aga2p-HC fusion product as detected with an anti c-Myc antibody in the diploid yeast cell formed by mating EBY100 pTQ5-HC with W303 pTQ6-LC (lane 5) compared to the (control) diploid yeast cell formed by mating EBY100 pTQ5 with W303 pYC6 (lane 4), the parent HC vector yeast host cell EBY100 pTQ5-HC (lane 3), the standard Fab display vector yeast host cell EBY100 pTQ3F2 (lane 2), and the (control) empty vector yeast host cell EBY100 pTQ5 (lane 1).
  • FIGS. 18 A- 18 C are representations of immunofluorescence detection of combinatorially assembled Fab antibodies on the surface of yeast diploid cells (A) LC display (B) HC display (C) Antigen binding. The top row shows immunofluorescence and the bottom row shows phase contrast.
  • the invention disclosed in the present application describes the first demonstration of the successful expression, transport, assembly, and immobilization (or “display”) of a functional heterologous multi-chain polypeptide (e.g., Fab antibody fragments) on the surface of a eukaryotic host cell (e.g., yeast).
  • a functional heterologous multi-chain polypeptide e.g., Fab antibody fragments
  • the present invention makes possible the construction of vector libraries and eukaryotic host cell libraries, wherein the cells display a highly variable repertoire of multi-chain polypeptides, which multi-chain polypeptides exhibit a high degree of sequence diversity within the repertoire and a consequently highly variable range of biological activities such as target (e.g., antigen) specificity.
  • target e.g., antigen
  • a “multi-chain polypeptide” refers to a functional polypeptide comprised of two or more discrete polypeptide elements (i.e., “chains”), covalently or non-covalently linked together by molecular association other than by peptide bonding.
  • the chains of a multi-chain polypeptide can be the same or different.
  • a prominent example of a multi-chain polypeptide is an immunoglobulin (e.g., IgA, IgD, IgE, IgG, and IgM), typically composed of four chains, two heavy chains and two light chains, which assemble into a multi-chain polypeptide in which the chains are linked via several disulfide (covalent) bonds.
  • Active immunoglobulin Fab fragments involving a combination of a light chain (LC) domain and a heavy chain (HC) domain, form a particularly important class of multi-chain polypeptides.
  • LC light chain
  • HC heavy chain
  • the LC and HC of a Fab are also known to effectively associate (non-covalently) in the absence of any disulfide bridge.
  • multi-chain polypeptides include, but are not limited to, the extracellular domains of T cell receptor (TCR) molecules (involving ⁇ and ⁇ chains, or ⁇ and ⁇ chains), MHC class I molecules (involving ⁇ 1, ⁇ 2, and ⁇ 3 domains, non-covalently associated to ⁇ 2 microglobulin), and MHC class II molecules (involving ⁇ and ⁇ chains).
  • TCR T cell receptor
  • MHC class I molecules involving ⁇ 1, ⁇ 2, and ⁇ 3 domains, non-covalently associated to ⁇ 2 microglobulin
  • MHC class II molecules involving ⁇ and ⁇ chains.
  • biologically active when referring, e.g., to a multi-chain polypeptide, means that the polypeptide exhibits a functionality or property that is useful as relating to some biological process, pathway or reaction.
  • Biological activity can refer to, for example, an ability to interact or associate with (e.g., bind to) another polypeptide or molecule, or it can refer to an ability to catalyze or regulate the interaction of other proteins or molecules (e.g., enzymatic reactions).
  • Biological activity can also refer to the ability to achieve a physical conformation characteristic of a naturally occurring structure, such as the four-chain conformation of naturally occurring immunoglobulin gamma (IgG) molecules, the ⁇ and ⁇ chains of a T cell receptor molecule, or the conformation of an antigen presenting structure of a major histocompatability complex (e.g., MHC peptide groove).
  • a physical conformation characteristic of a naturally occurring structure such as the four-chain conformation of naturally occurring immunoglobulin gamma (IgG) molecules, the ⁇ and ⁇ chains of a T cell receptor molecule, or the conformation of an antigen presenting structure of a major histocompatability complex (e.g., MHC peptide groove).
  • vector refers to any element capable of serving as a vehicle of genetic transfer, gene expression, or replication or integration of a foreign polynucleotide in a host cell.
  • a vector can be an artificial chromosome or plasmid, and can be integrated into the host cell genome or exist as an independent genetic element (e.g., episome, plasmid).
  • a vector can exist as a single polynucleotide or as two or more separate polynucleotides.
  • a “multi-chain display vector” of the present invention is capable, in an appropriate host, of directing expression of at least one chain of a multi-chain polypeptide and processing it for display on the surface of said host.
  • Vectors according to the present invention can be single copy vectors or multicopy vectors (indicating the number of copies of the vector typically maintained in the host cell).
  • Preferred vectors of the present invention include yeast expression vectors, particularly 2 ⁇ vectors and centromere vectors.
  • a “shuttle vector” (or bi-functional vector) is known in the art as any vector that can replicate in more than one species of organism.
  • a shuttle vector that can replicate in both Escherichia coli ( E. coli ) and Saccharomyces cerevisiae ( S. cerevisiae ) can be constructed by linking sequences from an E. coli plasmid with sequences from the yeast 2 ⁇ plasmid.
  • a particularly preferred embodiment of the present invention is a “dual display vector”, which is a shuttle vector that is capable not only of replicating in two different species but is capable of expressing and displaying heterologous polypeptides in two or more host species.
  • secretion refers to peptides having a secretion signal and are processed in the endoplasmic reticulum. If secreted peptides either contain anchor sequences or associate with the outside of the cell surface, the peptides are said to be “displayed”.
  • display and “surface display” (used interchangeably herein) refer to the phenomenon wherein a heterologous polypeptide is attached, or “anchored”, to the outer surface of a phage or host cell, whereby the anchored polypeptide is exposed to the extracellular environment.
  • the present invention is particularly directed to the display of a multi-chain polypeptide on the surface of a eukaryotic host cell, by expression of each of the chains in the host cell and the anchoring of at least one chain of the multi-chain polypeptide to the surface of the host cell.
  • a “display vector” refers to a vector that is capable of expressing a polypeptide in a host cell or phage such that the expressed polypeptide is displayed on the surface of said host cell or phage.
  • Display vectors of the present invention direct expression of multi-chain polypeptides in a host cell or phage such that the biological activity of the displayed polypeptide is exhibited at the surface of the host cell or phage.
  • Dual display vectors of this invention direct expression of multi-chain polypeptides in at least two different hosts (preferably, e.g., a prokaryotic host cell and a eukaryotic host cell) such that the biological activity of the polypeptide is exhibited at the surface of the respective hosts.
  • hosts preferably, e.g., a prokaryotic host cell and a eukaryotic host cell
  • the term “repertoire” refers to a population of diverse molecules, e.g., nucleic acid molecules differing in nucleotide sequence, or polypeptides differing in amino aid sequence.
  • a repertoire of polypeptides is preferably designed to possess a diverse population of molecules that differ in their binding sites for a target molecule.
  • the polypeptides of the repertoire are designed to have common structural elements, e.g., as with a repertoire of Fabs, having a well-recognized two-chain structure (Ig light chain associated with V H and C H 1 domains of an Ig heavy chain) but exhibiting different binding specificities, due to variation in the respective variable regions of the component chains.
  • the term “library” refers to a mixture of heterogeneous polypeptides or polynucleotides.
  • a library is composed of members that have similar polypeptide or polynucleotide sequences.
  • the library is a polynucleotide library, it encodes a repertoire of polypeptides (especially, e.g., with regard to the present invention, a repertoire of multi-chain polypeptides). Sequence differences between library members are responsible for the diversity present in the library.
  • the library can take the form of a simple mixture of polypeptides or polynucleotides, or can be in the form organisms or cells, for example bacteria, viruses, animal or plant cells and the like, that are transformed with a library of polynucleotides.
  • the library is a “display library”.
  • polynucleotides are incorporated into expression vectors, in order to allow expression of the polypeptides encoded by the polynucleotides.
  • a library can take the form of a population of host organisms, each organism containing one or more copies of an expression vector containing a single member of the library in polynucleotide from that can be expressed to produce its corresponding polypeptide member.
  • the population of host organisms has the potential to encode a large repertoire of genetically diverse polypeptide variants.
  • the present invention is directed to novel multi-chain display vectors.
  • the polynucleotides that encode the chains of the multi-chain polypeptide are present on separate (i.e., two or more) expression vectors, the compilation of which form a functional display “vector set” (the general term, “vector” encompasses vector sets).
  • the multi-chain polypeptide were a two-chain polypeptide comprised of the light chain and the heavy chain of a biologically active Fab
  • the polynucleotide encoding the LC can be incorporated into one expression vector
  • the polynucleotide encoding the HC can be incorporated into a second, separate, expression vector (most preferably expressed as a HC-anchor fusion protein).
  • each vector is capable of expressing its respective polypeptide chain; the two vectors together form a matched vector set, which set encodes the chains of a biologically active multi-chain polypeptide.
  • each transformed with the different vectors of a vector set collectively form a matched host cell set (or specifically in the case of a two-vector set, a matched “cell pair”).
  • the vectors and vector sets will preferably also include one or more selectable markers (e.g., TRP, ampR, and the like) to facilitate selection and propagation of successfully transformed hosts.
  • a “host cell” refers to any cell (prokaryote or eukaryote) transformed to contain a vector.
  • preferred host cells are bacterial cells and eukaryotic cells, including, but not limited to, protist cells, fungus cells, plant cells, and animal cells.
  • Host cells of the invention can be of any genetic construct, but are preferably haploid, diploid cells, or multiploid (e.g., as is typical of immortalized cell lines in culture).
  • Preferred host cells include insect cells (e.g., Sf9), mammalian cells (e.g., CHO cells, COS cells, SP2/0 and NS/0 myeloma cells, human embryonic kidney (HEK 293) cells, baby hamster kidney (BHK) cell, human B cells, human cell line PER.C6TM (Crucell)), seed plant cells, and Ascomycete cells (e.g., Neurospora and yeast cells; particularly yeast of the genera Saccharomyces, Pichia, Hansenula, Schizosaccharomyces, Kluyveromyces, Yarrowia, and Candida).
  • Preferred exemplar yeast species include S.
  • yeast host cell is S. cerevisiae.
  • phage refers to a “bacteriophage”, which is a bacterial virus containing a nucleic acid core and a protective proteinaceous shell.
  • bacteria and phage
  • phagemids i.e., bacteriophage the genome of which includes a plasmid that can be packaged by coinfection of a host with a helper phage.
  • the phage is an M13 phage.
  • anchor refers to a polypeptide moiety that, on expression in a host cell, becomes attached or otherwise associated with the outer surface of the host cell or, in the case of a phage display system, on the surface of a phage particle (e.g., as part of the capsid or as part of a filament).
  • An anchor polypeptide can be a coat protein moiety, a transmembrane protein moiety, or can be a polypeptide moiety otherwise linked to the cell surface (e.g., via post-translational modification, such as by a phosphatidyl-inositol or disulfide bridge).
  • Anchors include any synthetic modification or truncation of a naturally occurring anchor that still retains the ability to be attached to the surface of a host cell or phage particle.
  • Preferred anchor protein moieties are contained in, for example, cell surface proteins of a eukaryotic cell.
  • Effective anchors include portions of a cell surface protein sufficient to provide a surface anchor when fused to another polypeptide, such as a chain of a multi-chain polypeptide in accordance with this invention.
  • protein pairs that are separately encoded and expressed but associate at the surface of a cell by covalent (e.g., disulfide) or non-covalent bonds is also contemplated as a suitable anchor, and in this regard particular mention is made of the yeast ⁇ -agglutinin components, Aga1p and Aga2p, which form a glycan-immobilized, disulfide-linked complex on the surface of yeast cells.
  • Another protein pair that can be employed as an anchor are proteins that form “leucine zipper” interactions and the like, such as the nuclear proteins Jun and Fos (which form a “jun/fos linkage”).
  • a display vector can be designed according to this invention to direct the expression in a host cell of a first chain of a multi-chain polypeptide fused to the leucine zipper moiety of Jun, and a second vector can be designed to direct independent expression of the leucine zipper moiety of Fos fused to a surface protein of the host.
  • the first chain polypeptide will be associated (i.e., anchored) with the host cell surface via a jun/fos linkage, as the Jun and Fos leucine zipper forms a linkage between the first chain polypeptide and the host cell surface protein fused to the Fos part of the zipper. Any suitable protein binding pair of this sort can be used.
  • polypeptide anchors include the pIII coat protein of filamentous phage or fragments thereof (e.g., pIII anchor domain or “stump”, see U.S. Pat. No. 5,658,727) for phage display systems, and for yeast display systems FLO1 (a protein associated with the flocculation phenotype in S. cerevisiae ), ⁇ -agglutinin, and a-agglutinin (e.g., Aga1p and Aga2p subunits), and functional fragments thereof.
  • pIII coat protein of filamentous phage or fragments thereof e.g., pIII anchor domain or “stump”, see U.S. Pat. No. 5,658,727
  • FLO1 a protein associated with the flocculation phenotype in S. cerevisiae
  • ⁇ -agglutinin e.g., Aga1p and Aga2p subunits
  • functional fragments thereof e.g., Aga
  • fusion protein denotes a hybrid polypeptide comprised of amino acid sequences from more than one source, linked together to form a non-naturally occurring, unitary polypeptide. Fusion proteins are prepared, for example, by operably linking coding sequences for the component amino acid sequences in frame, such that, upon expression, they are produced as a single polypeptide. Alternatively, fusion proteins can be assembled synthetically, e.g., by creating a peptide bond between two or more separate polypeptides.
  • linked refers to a functional and structural connection between two or more elements.
  • the linked elements typically refer to an operable connection between two or more polynucleotide elements or polypeptide elements.
  • a polypeptide can be linked to an anchor protein (via a peptide bond or via peptide linker), thus forming a fusion protein.
  • the polynucleotides encoding the polypeptide and anchor protein can be linked such that the fusion protein is transcribed and translated as a unitary RNA message.
  • Polypeptides can also be indirectly linked to an anchor via an intermediate association, one example of which is the use of the high-affinity interaction of the Jun and Fos leucine zippers (i.e., a “jun/fos linkage”) to effectively link a polypeptide to the surface of a phage or host cell (Crameri, R. and Blaser, K., 1996). Any suitable heterodimeric or homodimeric pair of molecules can be used (Chang, H. et al., 1994; Moll, J. et al., 2001; Pu, W. and Struhl, K., 1993).
  • polynucleotides which encode one or more chains of a multi-chain polypeptide to be expressed and displayed in a phage display or host cell display system, can be operably linked to a promoter (to facilitate transcription), or operably linked to a signal sequence or leader peptide (to facilitate cellular processing and transport to the surface).
  • promoter to facilitate transcription
  • signal sequence or leader peptide to facilitate cellular processing and transport to the surface.
  • Preferred promoters include inducible promoters.
  • promoters for eukaryotic systems
  • promoters include those useful in yeast vectors, such as pGAL1, pGAL1-10, pGa1104, pGa110, pPGK, pCYC1, and pADH1.
  • Other preferred promoters include the LacZ promoter (for non-eukaryotic systems).
  • Particularly preferred signal sequences include the Aga2p signal sequence (for eukaryotic systems), and the pIII signal sequence (for non-eukaryotic systems).
  • markers e.g., epitope tags, reporter genes, radioisotope, fluorescent or chemiluminescent moieties, etc.
  • tags e.g., epitope tags, reporter genes, radioisotope, fluorescent or chemiluminescent moieties, etc.
  • Epitope tags e.g., peptide segments known to be recognized by particular antibodies or binding moieties
  • tags are typically placed under the same genetic controls as a gene of interest (preferably as a component of an expressed fusion protein). If and when the gene product of interest is not easily detectable, the tag provides an easily detectable, and often quantifiable, signal indicating the presence of the gene product of interest.
  • the practitioner can monitor such processes as, for example, gene expression, polypeptide trafficking, extracellular display, and protein-protein interactions (Fields, S. and Stemglanz, R., 1994; Phizicky, E. and Fields, S., 1995).
  • the chains of a multi-chain polypeptide can be optionally linked to one or more tags, either individually or jointly.
  • tags are known in the art and are commercially available (Amersham Pharmacia Biotech, Piscataway, N.J.; Applied Biosystems, Foster City, Calif.; Promega, Madison, Wis.; Roche Molecular Biochemicals, Indianapolis, Ind.; Stratagene, La Jolla, Calif.).
  • the linkage is achieved via a peptide bond (thus creating a fusion protein), wherein the polynucleotide encoding a chain of a multi-chain polypeptide is linked to a tag (e.g., an epitope tag).
  • Preferred tags include polyHis tags, HA tags, and myc tags.
  • the term “recombinant” is used to describe non-naturally altered or manipulated nucleic acids, host cells transfected with exogenous nucleic acids, or polypeptides expressed non-naturally, through manipulation of isolated DNA and transformation of host cells. “Recombinant” is a term that specifically encompasses DNA molecules that have been constructed in vitro using genetic engineering techniques, and use of the term “recombinant” as an adjective to describe a molecule, construct, vector, cell, polypeptide or polynucleotide specifically excludes naturally occurring molecules.
  • the term “transform” refers generally to any artificial (i.e., practitioncr-controlled) method of introducing genetic material into a cell or phage without limitation to the method of insertion. Numerous methods are known in the art and described by the references cited and incorporated herein. Specifically as applied to the present invention, the term “transformant” refers to a host cell that has been transformed and encompasses, for example, diploid cells, which are the product of the controlled fusion of matched haploid cell pairs (as with the controlled mating of haploid yeast spores of opposite mating type).
  • Methods for “transferring” nucleic acid sequence information from one vector to another is not limiting in the present invention and includes any of a variety of genetic engineering or recombinant DNA techniques known in the art. Once again, a vast array of methods are known in the art and described in the references cited and incorporated herein. Particularly preferred transfer techniques include, but are not limited to, restriction digestion and ligation techniques (utilizing unique cloning sites), PCR amplification protocols (utilizing specific primer sequences), and homologous recombination techniques (utilizing polynucleotide regions of homology).
  • Employing genetic engineering technology necessarily requires growing recombinant host cells (transformants) under a variety of specified conditions as determined by the requirements of the organism and the particular cellular state desired by the practitioner.
  • the organism can possess (as determined by its genetic disposition) certain nutritional requirements, or particular resistance or sensitivity to physical (e.g., temperature) and/or chemical (e.g., antibiotic) conditions.
  • specific culture conditions can be necessary to induce or repress the expression of a desired gene (e.g., the use of inducible promoters), or to initiate a particular cell state (e.g., yeast cell mating or sporulation).
  • host cells be cultured under “conditions suitable” or “conditions sufficient” to achieve or to induce particular cellular states.
  • Such desirable cellular states include, but are not limited to: cellular growth and reproduction; the expression, secretion or transport, and association of the chains of a multi-chain polypeptide such that the biological activity of the multi-chain polypeptide is exhibited at the surface of the host cell (or phage particle); the fusion of haploid cells to form a diploid cell (e.g., fertilization, zygote formation, the mating of cells of opposite mating types); and meiosis of a diploid cell to form haploid daughter cells (e.g., gametogenesis, sporulation).
  • the present invention is not limited by the physical and chemical parameters of such “suitable conditions”, but such conditions are determined by the organisms and vectors used to practice the invention, and by practitioner preference.
  • the present invention is directed to a novel genetic vector, useful in a eukaryotic cell to display a multi-chain polypeptide on the surface of the cell such that the biological activity of the multi-chain polypeptide is exhibited at the surface of the cell.
  • the multi-chain polypeptide can be encoded in a single vector, or individual chains of the multi-chain polypeptide can be encoded in a vector set.
  • the vector can exist as a vector set, wherein each chain of a multi-chain polypeptide is encoded on one of a matched pair of vectors such that when the vector set is present in a single eukaryotic cell, the chains of the multi-chain polypeptide associate at the surface of the eukaryotic cell.
  • the display vector can be a dual display vector, wherein the vector is capable of (i) expressing in a eukaryotic cell and displaying on the surface of a eukaryotic cell a biologically active multi-chain polypeptide, and (ii) expressing in a prokaryotic cell and displaying on the surface of a bacteriophage the biologically active multi-chain polypeptide.
  • the multi-chain polypeptide can be any polypeptide comprised of two or more discrete polypeptide elements, referred to as chains of the multi-chain polypeptide, which chains are covalently or non-covalently linked (other than by peptide bonding) to form a biologically active polypeptide.
  • the multi-chain polypeptide encoded by the multi-chain display vector(s) of the present invention exists as either a two-, three-, or four-chain polypeptide.
  • the chains of the polypeptide can be the same (e.g., a homo-dimer, -trimer, or -tetramer) or different (e.g., a hetero-dimer, -trimer, or -tetra-mer).
  • the multi-chain polypeptide is a two-chain or four-chain polypeptide comprised of two different chains. More preferably, the multi-chain polypeptide is selected from a group of multi-chain polypeptides consisting of T cell receptors, MHC class I molecules, MHC class II molecules, immunoglobulins and biologically active immunoglobulin fragments (e.g., Fabs). More preferably, the multi-chain polypeptide is an IgA, IgD, IgE, IgG, IgM, or biologically active fragment thereof.
  • the multi-chain polypeptide is a Fab fragment of an Ig, wherein the first polynucleotide of the multi-chain display vector comprises a segment that encodes the V H and C H 1 domains of an Ig heavy chain, and a second polynucleotide comprises a segment that encodes an Ig light chain (i.e., V L and C L domains).
  • the chains of the multi-chain polypeptide are encoded as polynucleotides (e.g., first polynucleotide, second polynucleotide, third polynucleotide, etc., respectively) in an expression vector.
  • polynucleotides e.g., first polynucleotide, second polynucleotide, third polynucleotide, etc., respectively.
  • polynucleotide encoding the light chain and heavy chain of an Ig Fab can be located on separate plasmids and transformed as such into an identical host cell for co-expression and co-processing into a functional multi-chain polypeptide.
  • sequences of the polynucleotides that encode the chains of a multi-chain polypeptide need not originate from an identical, or same source.
  • an Ig molecule can be produced having variable domains (V H and V L ) the same as those from a monoclonal antibody having a desired specificity, and constant domains (C H 1 and C L ) from a different monoclonal antibody having desired properties (e.g., to provide human compatibility or to provide a particular complement binding site).
  • heterologous polynucleotide encoding the chains of a multi-chain polypeptide can be variegated, to produce a family of polynucleotide homologs, encoding polypeptide chains that vary slightly in amino acid sequence from one another while having the same overall structure.
  • a library of multi-chain polypeptides of varied sequence are displayed, providing a peptide display library suitable for screening, e.g., to discover homologous multi-chain polypeptides having altered biological activity.
  • Such alterations in amino acid sequence can be achieved by suitable mutation or partial synthesis and replacement or partial or complete substitution of appropriate regions of the corresponding polynucleotide coding sequences.
  • Substitute constant domain portions can be obtained from compatible recombinant DNA sequences.
  • the chains of the multi-chain polypeptide will be displayed on the surface of a eukaryotic host cell.
  • the display vector itself can be constructed or modified from any of a number of genetic vectors and genetic control elements known in the art and commercially available (e.g., from InVitrogen (Carlsbad, Calif.); Stratagene (La Jolla, Calif.); American Type Culture Collection (Manassas, Va.)).
  • the vector construct of the present invention expresses the polypeptide chains for effective display of a fully assembled, multi-chain polypeptide on the surface of a eukaryotic cell transformed with the vector such that the biological activity of the multi-chain polypeptide is exhibited at the surface of the host cell.
  • the polynucleotides encoding each of the chains of the multi-chain polypeptide are, preferably, linked to a transcriptional promoter to regulate expression of the polypeptide chains.
  • the effective promoter must be functional in a eukaryotic system, and optionally (particularly in the case of a dual display vector) effective as a prokaryotic promoter as well.
  • the eukaryotic promoter(s) and the prokaryotic promoter(s) selected for regulating expression of the heterologous polypeptide chains of a multi-chain polypeptide can be the same or different promoters, as long as they are appropriately functional in the intended host organisms.
  • the eukaryotic promoter can be a constitutive promoter but is preferably an inducible promoter. In order to achieve balanced expression and to ensure simultaneous induction of expression, a vector construct that utilizes the same promoter for each chain is preferred.
  • a number of eukaryotic promoters useful in the present invention are known in the art.
  • Particularly preferred promoters include those useful in yeast expression vectors, such as galactose inducible promoters, pGAL1, pGAL1-10, pGal4, and pGa110; phosphoglycerate kinase promoter, pPGK; cytochrome c promoter, pCYC1; and alcohol dehydrogenase I promoter, pADH1.
  • each of the polynucleotides encoding a chain of a multi-chain polypeptide is also linked to a signal sequence (or a leader peptide sequence).
  • the signal sequence operates to direct transport (sometimes referred to as secretion) of a nascent polypeptide into or across a cellular membrane.
  • Chains of a multi-chain polypeptide expressed in a eukaryotic cell from a vector of the present invention are transported to the endoplasmic reticulum (ER) for assembly and transport to the cell surface for extracellular display.
  • An effective signal sequence should be functional in a eukaryotic system, and optionally (particularly in the case of a dual display vector) the signal sequence should be effective in a prokaryotic system as well.
  • Polynucleotides encoding the chains of a multi-chain polypeptide are typically directly linked, in frame (either immediately adjacent to the polynucleotide or optionally linked via a linker or spacer sequence), to a signal sequence, thus generating a polypeptide chain-signal sequence peptide fusion protein.
  • each chain of a multi-chain polypeptide is fused to a separate signal peptide.
  • the signal sequence encoding the signal peptide can be the same or different for each chain of the multi-chain polypeptide.
  • the signal sequence can be native to the host or heterologous, as long as it is operable to effect extracellular transport of the polypeptide to which it is fused.
  • signal sequences operable in the present invention are known to persons skilled in the art (e.g., Mf ⁇ 1 prepro, Mf ⁇ 1 pre, acid phosphatase Pho5, Invertase SUC2 signal sequences operable in yeast; pIII, Pe1B, OmpA, PhoA signal sequences operable in E.
  • the signal sequences are preferably derived from native secretory proteins of the host cell.
  • Particularly preferred eukaryotic signal sequences include those of ⁇ -mating factor of yeast, ⁇ -agglutinin of yeast, invertase of Saccharomyces, inulinase of Kluyveromyces, and most preferably the signal peptide of the Aga2p subunit of a-agglutinin (especially in embodiments where the anchoring polypeptide to be used is the Aga2p polypeptide).
  • the first polynucleotide comprises an Aga2p signal sequence in frame with a segment that encodes the V H and C H 1 regions of an Ig heavy chain
  • the second polynucleotide comprises an Aga2p signal sequence in frame with a segment that encodes an Ig light chain.
  • the multi-chain eukaryotic display vector of the present invention operates in a eukaryotic host cell such that the multi-chain polypeptide encoded by the vector is displayed on the surface of the host cell.
  • Anchorage (“tethering” or “display”) on the surface of the host cell is achieved by linking at least one chain of the multi-chain polypeptide to a molecular moiety attached to the host cell wall. More than one chain of a multi-chain polypeptide can be linked to an anchor, but because the fully assembled multi-chain polypeptide requires (and preferably contains) only one point of attachment to the host cell surface, only one chain of the multi-chain polypeptide need be the point of cellular attachment.
  • Display on the surface of the cell can be achieved by linking at least one of the polypeptide chains to an anchor protein or functional fragment (moiety) thereof.
  • the effective anchor should be functional in a eukaryotic system, and optionally (particularly in the case of a dual display vector) the anchor should be effective as an anchor on the surface of a bacteriophage as well.
  • the anchor is a surface-expressed protein native to the host cell, e.g., either a transmembrane protein or a protein linked to the cell surface via a glycan bridge.
  • anchor proteins operable in the present invention are known to persons skilled in the art (e.g., pIII, pVI, pVIII, LamB, PhoE, Lpp-OmpA, Flagellin (FliC), or at least the transmembrane portions thereof, operable in prokaryotes/phage; platelet-derived growth factor receptor (PDGFR) transmembrane domain, glycosylphosphatidylinositol (GPI) anchors, operable in mammalian cells; gp64 anchor in insect cells, and the like).
  • PDGFR platelet-derived growth factor receptor
  • GPI glycosylphosphatidylinositol
  • the anchor protein is ⁇ -agglutinin, a-agglutinin (having subcomponents Aga1p and Aga2p), or FLO1, which naturally form a linkage to the yeast cell surface.
  • Linkage of a polypeptide chain to an anchor can be achieved, directly or indirectly, by a variety of molecular biology techniques.
  • the present invention is not limited by the method of chain-anchor linkage, only by the functional requirement that the linked polypeptide chain is immobilized on the surface of the host cell (or optionally bacteriophage) as a result of such linkage.
  • a preferred method of chain-anchor linkage is through the construction of a chain-anchor fusion protein. Similar to, and preferably in concert with, a chain-signal peptide fusion protein, a polynucleotide encoding a chain of a multi-chain polypeptide is directly linked, in frame (either immediately adjacent to the polynucleotide or optionally linked via a linker or spacer sequence), to an anchor, thus generating a signal peptide-polypeptide chain-anchor fusion protein.
  • a chain of the multi-chain polypeptide can be indirectly linked to an anchor via an intermediate association such as the high affinity interaction of the Jun and Fos leucine zippers (jun/fos linkage) to covalently link a polypeptide chain to an anchor of a phage or host cell (Crameri, R. and Suter, M., 1993; Crameri, R. and Blaser. K., 1996).
  • the multi-chain polypeptide is an Ig Fab fragment: the first polynucleotide comprises an Aga2p signal sequence in frame with a segment that encodes an Aga2p anchor, and in frame with a segment that encodes the V H and C H 1 domains of an Ig heavy chain; and the second polynucleotide comprises an Aga2p signal sequence in frame with a segment that encodes an Ig light chain.
  • the multi-chain display vectors of the present invention provide cloning sites to facilitate transfer of the polynucleotide sequences that encode the chains of a multi-chain polypeptide.
  • Such vector cloning sites comprise at least one restriction endonuclease recognition site positioned to facilitate excision and insertion, in reading frame, of polynucleotides segments. Any of the restriction sites known in the art can be utilized in the vector construct of the present invention. Most commercially available vectors already contain multiple cloning site (MCS) or polylinker regions.
  • MCS multiple cloning site
  • genetic engineering techniques useful to incorporate new and unique restriction sites into a vector are known and routinely practiced by persons of ordinary skill in the art.
  • a cloning site can involve as few as one restriction endonuclease recognition site to allow for the insertion or excision of a single polynucleotide fragment. More typically, two or more restriction sites are employed to provide greater control of, for example, insertion (e.g., direction of insert), and greater flexibility of operation (e.g., the directed transfer of more than one polynucleotide fragment). Multiple restriction sites can be the same or different recognition sites.
  • the multi-chain eukaryotic display vector of the present invention preferably contains restriction sites positioned at the ends of the coding sequences for the chains of the multi-chain polypeptide. Restriction sites can be positioned at the extreme ends, 5′ and 3′ of the polynucleotide segment including all of the coding sequences for the chains of a multi-chain polypeptide (on a single vector); or, more preferably, restriction sites can be positioned at the 5′ and 3′ ends of each polynucleotide segment encoding a chain of the multi-chain polypeptide. Most preferably each of the restriction sites is unique in the vector and different from the other restriction sites. This particularly useful vector construct provides flexibility and control for the modular transfer of individual polynucleotide sequences encoding a chain of a multi-chain polypeptide.
  • the first polynucleotide comprises an Aga2p signal sequence in frame with a segment that encodes an Aga2p anchor, and in frame with a segment that encodes the V H and C H 1 regions of an Ig heavy chain, wherein the Ig heavy chain region is bordered by unique restriction sites (e.g., SfiI and NotI); and the second polynucleotide comprises an Aga2p signal sequence in frame with a segment that encodes an Ig light chain, wherein the Ig light chain region is bordered by unique restriction sites (e.g., ApaLI, and AscI).
  • unique restriction sites e.g., SfiI and NotI
  • one or more of the chains of the multi-chain polypeptide expressed by the vector in a host cell is linked to a molecular tag or reporter gene.
  • the linkage is a peptide bond that links a polypeptide tag to a chain of the multi-chain polypeptide.
  • One or more chains of the multi-chain polypeptide can be tagged using identical, similar or different tags.
  • Preferred tags include epitope tags (Munro, S. and Pelham, H., 1987).
  • Preferred epitope tags include polyHis tags, HA tags, and myc tags, and preferably each chain is fused to a different tag.
  • the first polynucleotide comprises an Aga2p signal sequence in frame with a segment that encodes an Aga2p anchor, in frame with a segment that encodes the V H and C H 1 regions of an Ig heavy chain, and in frame with a segment that encodes a myc tag, wherein the Ig heavy chain region is bordered by unique restriction sites (e.g., SfiI and NotI); and the second polynucleotide comprises an Aga2p signal sequence in frame with a segment that encodes a HA tag, and in frame with a segment that encodes an Ig light chain, wherein the Ig light chain region is bordered by unique restriction sites (e.g., ApaLI, and AscI).
  • unique restriction sites e.g., ApaLI, and AscI
  • the process for displaying a multi-chain polypeptide on the surface of a eukaryotic host cell comprises introducing the vector (possibly as a vector set) into a eukaryotic cell (i.e., a host cell), and culturing the host cell under conditions suitable for expression, transport, and association of the chains of the multi-chain polypeptide with the host cell surface such that the biological activity of the multi-chain polypeptide is exhibited at the surface of the host cell.
  • the mode of introduction of the vector of the present invention into a host cell is not limiting to the present invention and includes any method for introducing genetic material into a cell known in the art. Such methods include but are not limited to methods known and referred to in the art as transfection, transformation, electroporation, liposome mediated transfer, biolistic transfer, conjugation, cellular fusion, and nuclear microinjection. Transformation techniques known in the art are the preferred methods of genetic transfer.
  • Vectors of the present invention are operable in a eukaryotic host cell to effect expression and to display a multi-chain polypeptide on the surface of the eukaryotic host cell.
  • the vectors of the present invention are operable in a prokaryotic host cell as well, to effect expression in a bacterial host cell and to display a multi-chain polypeptide on the surface of a bacteriophage.
  • the eukaryotic host cell can be any eukaryotic cell, of any genotype, differentiated or undifferentiated, unicellular or multi-cellular, depending on the practitioner's particular interest and requirements.
  • eukaryotic cells include mammalian cells, plant cells, fungus cells, and protist cells.
  • the host cell is an undifferentiated, unicellular, haploid or diploid cellular organism.
  • Fungi are preferred host cells, particularly species of the phylum Ascomycota (sac fungi), because of their ease and diversity of culture conditions, the variety of biochemical and cellular mutants available, their short generation time, and their life cycle (see below).
  • Preferred fungal host cells include those of the genera Neurospora and the various yeasts, such as Saccharomyces, Pichia, Hansenula, Schizosaccharomyces, Kluyveromyces, Yarrowia, Debaryomyces, and Candida. Most preferred species is Saccharomyces cerevisiae (baker's yeast), perhaps the most well known, characterized, and utilized eukaryotic cell system in molecular biology research.
  • the eukaryotic host cells are suitable for cell fusion (see below).
  • yeast cells of opposite mating type can be “mated” to produce fused diploid cells.
  • yeast protoplasts or spheroplasts suitable for cell fusion are also suitable eukaryotic host cells for the purposes of the invention.
  • cells grown in culture e.g., mammalian cells, insect cells, etc.
  • can be fused by methods known in the art e.g., using Sendai virus or electric current).
  • the technical advancement of the present invention to display complex multi-chain polypeptides on the surface of a eukaryotic host cell can be coupled with the power of phage display technology.
  • a phage display-eukaryotic display transfer system as described herein, practitioners can, for the first time, combine the immense diversity provided by phage display libraries and phage display technology with the cellular expression, processing, assembly, and display provided by the aforementioned multi-chain eukaryotic display technology.
  • the transfer of nucleic acid sequence information between a phage display vector and the eukaryotic vector of the present invention can be achieved by a variety of genetic transfer methods known in the art (e.g., genetic engineering technology such as recombinant DNA technology). Preferred modes of transfer include techniques of restriction digestion, PCR amplification, or homologous recombination.
  • a eukaryotic/prokaryotic multi-chain display shuttle vector as described and taught herein is employed.
  • the genetic control elements of the dual display vector of the present invention provide, within a eukaryotic host cell, for the expression, processing, assembly, and display of a biologically active multi-chain polypeptide on the surface of the eukaryotic host cell transformed with the dual display vector, as well as provide, within a prokaryotic host cell, for the expression, processing, assembly, and display of a biologically active multi-chain polypeptide on the surface of a bacteriophage infected in the prokaryotic host cell.
  • the phage display-eukaryotic display transfer system is performed by inserting chain-encoding polynucleotide segments excised from a conventional phage display vector (i.e., a bacteriophage engineered to display an exogenous polypeptide on the surface of the phage particle) known in the art, into the multi-chain eukaryotic display vector of the present invention, thereby enabling expression of the chain-encoding segments, and eukaryotic processing, assembly, and display of a biologically active multi-chain polypeptide on the surface of a eukaryotic host cell transformed with the eukaryotic display vector.
  • a conventional phage display vector i.e., a bacteriophage engineered to display an exogenous polypeptide on the surface of the phage particle
  • transfer of the polynucleotide sequences from a phage display vector to a multi-chain eukaryotic display vector can be achieved by any genetic engineering technique known in the art.
  • Two particularly preferred methods include a single excision/insertion transfer method and a multiple (or modular) excision/insertion transfer method.
  • the polynucleotide segments that encode the chains of a multi-chain polypeptide are excised (e.g., via restriction digestion) from the phage display vector as a single, unitary nucleic acid, and subsequently inserted into the multi-chain display vector.
  • unwanted prokaryotic genetic control elements if any
  • This process is diagramed for an Ig Fab multi-chain polypeptide, transferred from a phage display vector to a particularly preferred multi-chain yeast display vector of the present invention in FIG. 1.
  • polynucleotide segments encoding chains of a multi-chain polypeptide are excised from the phage display vector individually, and subsequently inserted into the multi-chain display vector in a separate and independent manner.
  • This approach provides greater control and flexibility over the transfer of individual chains of a multi-chain polypeptide separately or en masse. Indeed, depending upon the practitioner's interests and needs, only select chains of the multi-chain polypeptide need to be transferred. This process is diagramed for an Ig Fab multi-chain polypeptide in FIG. 2.
  • phage display-eukaryotic display transfer system described and taught herein is equally functional whether transferring sequence information from a phage display vector to a multi-chain eukaryotic display vector, or from a multi-chain eukaryotic display vector to a phage display vector; i.e., the phage display-eukaryotic display transfer system of the present invention is effectively bi-directional.
  • a particularly preferred phage display library for use in the phage display-eukaryotic display transfer system according to the invention is a large human Fab fragment library (de Haard, H. et al., 1999).
  • Multi-chain eukaryotic display vectors of the present invention and host cells transformed with these vectors such that a biologically active multi-chain polypeptide in displayed on the host cell surface, are useful for the production of display libraries.
  • Such display libraries are, in turn, useful to screen for a variety of biological activities of interest to the practitioner; e.g., to screen against any of a variety of target molecules to identify binding polypeptides specific for that target.
  • Phage display libraries and the screening of the same, represent a powerful research and development tool. Methods for producing and screening phage display libraries are well known and used in the art (Hoogenboom, H. et al., 1997; Kay et al., 1996; Ladner, R et al., 1993).
  • the multi-chain eukaryotic display vectors of the present invention can be used to generate novel peptide libraries de novo, similar to known phage display libraries.
  • the vectors described herein provide allow for more efficient expression of properly folded, assembled, glycosylated, and displayed multi-chain polypeptides, as can only be achieved in a eukaryotic system.
  • These multi-chain eukaryotic display libraries can then be used in screening assays.
  • Persons of ordinary skill in the art will appreciate and easily adapt display library screening protocols known in the art (e.g., phage display screen assays) to the multi-chain eukaryotic display libraries of the present invention.
  • the present invention further enables the practitioner to transfer existing phage display libraries to the multi-chain eukaryotic display system disclosed and taught herein.
  • the phage display-eukaryotic display transfer system allows a phage display library to be constructed for the display of a very large repertoire of multi-chain polypeptides; for example Fabs, which have light chain and heavy chain components.
  • the phage display library which can have a diversity of >1 ⁇ 10 8 (preferably >1 ⁇ 10 9 , more preferably >1 ⁇ 1010) different multi-chain polypeptides in a library, can undergo an initial screen, producing a subpopulation of less than about 1 ⁇ 10 7 (preferably between 1 ⁇ 10 5 to 1 ⁇ 10 6 ) phage display isolates.
  • the polynucleotides encoding the chains of the multi-chain polypeptide isolates can then be “batch transferred” to a multi-chain eukaryotic display vector of the present invention for transformation into a eukaryotic host.
  • the multi-chain polypeptides displayed on eukaryotic host cells can be further screened and manipulated, taking advantage of the culture conditions and expression qualities of the eukaryotic host system as discussed earlier (e.g., protein folding, proper association of separate chains in the multi-chain protein, glycosylation, secretion, and post-translational modifications such as phosphtidyl inositol linkages to the cell membrane).
  • the multi-chain polypeptide library (or pre-selected isolates therefrom) can be further diversified (e.g., polypeptide chain recombination, re-shuffling, or re-mixing) for additional rounds of screening.
  • a M13 phage expression vector having:
  • an Ig light chain cloning site defined by an ApaLI restriction site and an AscI restriction site, and which is oriented 3′ to a signal sequence (e.g., a pIII signal sequence) and under the transcriptional control of a LacZ promoter; and
  • an Ig heavy chain fragment cloning site defined by a SfiI restriction site and a NotI restriction site, and which is oriented 3′ to a signal sequence (e.g., a pIII signal sequence), under the transcriptional control of a LacZ promoter, and 5′ to a sequence encoding mature pIII or an anchoring portion of pIII (stump).
  • a signal sequence e.g., a pIII signal sequence
  • the multi-chain eukaryotic display vector in this preferred embodiment is a yeast vector having:
  • an Ig light chain cloning site defined by an ApaLI restriction site and an AscI restriction site, and which is oriented 3′ to an Aga2p secretion signal and under the transcriptional control of a GAL promoter (preferably GAL1 or GAL1-10); and
  • an Ig heavy chain fragment cloning site defined by a SfiI restriction site and a NotI restriction site, and which is oriented 3′ to an Aga2p secretion signal, under the transcriptional control of a GAL promoter (preferably GAL1 or GAL1-10), and 3′ to a sequence encoding mature Aga2p.
  • a GAL promoter preferably GAL1 or GAL1-10
  • the yeast expression vector is used to transform a yeast host cell for expression of antibodies or Fab fragments displayed on the yeast cell surface.
  • Light and heavy chain coding sequences are excised individually (by ApaLI/AscI digestion and SfiI/NotI digestion respectively), or together (by ApaLI/NotI digestion) from the phage display vector, and inserted into the multi-chain yeast display vector by batch transfer, yielding a multiplicity of LC/HC chain pairings for expression and display in yeast.
  • a particularly preferred yeast display vector for yeast display of Fabs is pTQ3 (described below).
  • a particularly preferred phage display is a large human Fab fragment library (de Haard, H. et al., 1999).
  • the present invention can be used for the production of antibodies or antibody fragments useful for immunopurification, immunoassays, cytochemical labeling and targeting methods, and methods of diagnosis or therapy.
  • the antibody or fragment can bind to a therapeutically active protein such as interferon or a blood clotting factor such as, for example, Factor VIII, and can therefore be used to produce an affinity chromatography medium for use in the immunopurification or assay of the protein.
  • the basic life cycle of eukaryotic cells involves an alternation between diploid (two copies of an organism's chromosomes or genome per cell) and haploid (one copy of an organism's chromosomes or genome per cell) states.
  • the alternation between these two states is achieved by the fusion of two haploid cells (typically, although not necessarily, the fertilization of opposite mating types) to form a single diploid cell, and meiotic division of a diploid cell to form multiple haploid (daughter) cells.
  • Biologists appreciate that this basic life cycle (i.e., the alternation of haploid and diploid generations) provides an important natural mechanism for the biological recombination genetic information (i.e., sexual reproduction).
  • diploid state is the dominant stage of the life cycle, generated by the fusion of two haploid cells (commonly referred to as gametes) of opposite mating type; a sperm and an egg.
  • gametes commonly referred to as gametes
  • Meiotic cell division of diploid cells produces the haploid cell state for sexual reproduction.
  • the life cycle pattern of the plant kingdom provides a more general alternation of generation wherein the haploid and diploid state can exist as more distinct generations, depending on the particular plant species.
  • the generation of the haploid cell the “gametophyte” predominates (e.g., mosses, liverworts, and homworts); whereas in “higher” (i.e., more advanced) plants, the generation of the diploid cell (the “sporophyte”) predominates (e.g., ferns, conifers, and flowering plants).
  • the haploid stage of the life cycle predominates. Fertilization produces a diploid stage, which often almost immediately (depending upon environmental conditions) undergoes meiosis to form haploid cells.
  • the natural recombination and re-mixing of genetic material that results from meiosis of diploid cells to produce haploid cells, and the cellular fusion of separate haploid cells to produce diploid cell (of a new genetic admixture) is a powerful process that can be utilized in biological research. Described and taught herein for the first time, this powerful mechanism is utilized in combinatorial protein research for the generation of unique multi-chain peptide display libraries.
  • the mode for introducing eukaryotic multi-chain display vectors into a host cell includes the fusion of two eukaryotic cells, preferably haploid, each expressing at least one of the chains of the multi-chain polypeptide, such that the biological activity of the multi-chain polypeptide is exhibited at the surface of the resulting host cell, preferably diploid.
  • each of the two haploid cells can contain one of the vectors of a vector set (as described herein), such that once combined (e.g., via cellular fusion of host cells) and co-expressed in the resulting diploid host cell, the biological activity of the multi-chain polypeptide is exhibited at the surface of the host cell.
  • Such methods can be used to prepare novel multi-chain polypeptide libraries as described above (for example, antibody or Fab display libraries, including diploid host cells displaying multi-chain polypeptides having a greater diversity than the source repertoire).
  • populations of a matched vector set can be constructed such that one eukaryotic expression vector population expresses multiple (e.g., a repertoire or library) forms of an Ig Fab light chain (comprising V L and C L domains) and a second eukaryotic expression vector population expresses multiple forms of an Ig Fab heavy chain (comprising V H and C H 1 domains) fused to a yeast anchor protein (e.g., Aga2p).
  • yeast anchor protein e.g., Aga2p
  • Each of the vector populations are used to transform haploid yeast cells of opposite mating type; one vector construct in one mating type, the second vector construct in the opposite mating type.
  • the two haploid yeast populations are co-cultured under conditions sufficient to induce yeast mating (i.e. cellular fusion) of the two mating types.
  • the resulting diploid yeast host cells of the population possess both vector constructs and expresses and displays the fully formed and assembled Ig Fab.
  • any eukaryotic cell capable of cell fusion can be used in the present invention.
  • Cell fusion can occur sexually by mating, or artificially, e.g., in tissue culture or other artificial conditions.
  • any eukaryotic cell is suitable as long as it is capable of existing (no matter how briefly) in both a haploid and a diploid state.
  • cells are not limited by ploidy as they would be in the case of sexual fusion.
  • diploid mammalian cells maintained in tissue culture can be induced to fuse, thereby resulting in a tetraploid host cell.
  • the actual ploidy of the host cells to be fused does not pose a limitation so long as the cells can be fused.
  • the important features of the cells are that one cell partner contains a vector or vector set comprising a particular chain of a multi-chain polypeptide and a specific selectable marker, and the partner host cell contains a vector or a vector set comprising a second chain of a multi-chain polypeptide and a selectable marker.
  • the resultant fused cell contains vectors encoding two or more chains of a multi-chain polypeptide in a cell that is readily identified by the selectable markers.
  • Fungi especially sac fungi (ascomycetes; e.g., Neurospora and yeasts), are particularly preferred eukaryotic host cells. Sac fungi are so named because they produce the haploid spore products of meiosis in microscopic sacs, which render them easily collected, segregated, analyzed, and manipulated (Neurospora are particularly noted because the size and shape of their ascus maintains the order of the haploid cell products of meiosis). Also, these fungi, especially S.
  • haploid cells exist stably in both haploid and diploid form, either of which are easily induced and maintained (e.g., the yeast haploid state is typically induced and maintained under some form of nutritional stress, i.e., starvation).
  • yeast haploid state is typically induced and maintained under some form of nutritional stress, i.e., starvation.
  • haploid cells exist as two sexes (the ⁇ and ⁇ mating types), from which only opposite mating types fuse (mate) to form the diploid state. Under conditions manipulable in the lab by one of skill in the art, an ⁇ cell will fuse to an a cell, thereby creating a fused diploid cell.
  • the present invention is suitable for eukaryotic cells such as, for example, mammalian, insect or plant cells that are grown in culture. Additionally, yeast protoplasts or spheroplasts can be manipulated to undergo cell fusion even if they are of the same mating type. Such artificial methods for cell fusion are known in the art and would be suitable for the purposes of the present invention.
  • Multi-chain polypeptides libraries displayed on eukaryotic host cells can be screened and manipulated similar to procedures and techniques known in the art, e.g., phage display library screening, but also allow the practitioner to take advantage of culture conditions and expression qualities of a eukaryotic host system.
  • eukaryotic display screening can be prefaced with an initial round of phage display screening before transferring the display library from the phage display vector to a multi-chain eukaryotic display vector.
  • the multi-chain polypeptide library (or pre-selected isolates) can be subjected to one or more additional rounds of screening under the eukaryotic display system.
  • multi-chain eukaryotic display libraries can undergo further (biased or unbiased) diversification subsequent to any screen assay utilizing the alternation of generations characteristic of eukaryotic systems as discussed above.
  • Populations of diploid eukaryotic host cells containing a multi-chain eukaryotic display vector, wherein different chains of the multi-chain are expressed from different vectors e.g., where the diploid host cell is the product of haploid mating or cell fusions as described above
  • meiosis e.g., sporulation in yeast.
  • the haploid yeast cells (spores) can be segregated and/or selected depending on screening conditions in order to isolate different eukaryotic expression vectors with a preferred property of interest in separate haploid daughter cells.
  • the daughter cells can then be optionally:
  • homologous chains are co-expressed and displayed, homologous multi-chain polypeptides having greater affinities for the same target molecule can be selected; or
  • a multi-chain eukaryotic display vector specifically a yeast display vector effective in a host yeast cell transformed with the vector.
  • the vector is useful for expressing, transporting, assembling and displaying a biologically active multi-chain polypeptide (e.g., an Ig Fab) on the surface of the host yeast cell.
  • a biologically active multi-chain polypeptide e.g., an Ig Fab
  • pYD 1 InVitrogen, Carlsbad, Calif.
  • pYD 1 a commercially available vector designed for expression, secretion, and display of a single chain protein on the surface of S. cerevisiae cells
  • pYD1 includes: an aga2 gene encoding one of the subunits of the ⁇ -agglutinin receptor; a GAL1 promoter for regulated expression of an Aga2/polypeptide fusion; an HA epitope tag for detection of the displayed protein; a polyhistidine (6 ⁇ His) tag for purification on metal chelating resin; a CEN6/ARS4 for stable, episomal replication in yeast; and a TrpI gene for S. cerevisiae transformant selection, an ampicillin resistance gene (ampR) and the pMB 1 origin for selection and replication in E. coli.
  • an aga2 gene encoding one of the subunits of the ⁇ -agglutinin receptor
  • GAL1 promoter for regulated expression of an Aga2/polypeptide fusion
  • an HA epitope tag for detection of the displayed protein
  • a polyhistidine (6 ⁇ His) tag for purification on metal chelating resin
  • CEN6/ARS4 for stable, epi
  • the pYD1 plasmid was modified for expression of an Ig light chain and a heavy chain fragment from two tandem galactose inducible promoters, for the display of an intact Fab antibody fragment.
  • One GAL1 promoter directs expression of the light chain and the other GAL1 promoter directs expression of the heavy chain fragment fused to the C-terminus of the Aga2p yeast anchor protein.
  • restriction sites were generated as part of the vector construct.
  • the restriction endonuclease recognition sequences (i.e., restriction sites) chosen for this vector construct included ApaLI, AscI, SfiI, and NotI as the unique cloning sites for the chains of a two-chain polypeptide (in this case an Ig Fab), and NheI to facilitate phage display-eukaryotic display transfers with existing phage display libraries.
  • the vector thus modified having a unique ApaLI site immediately 3′ to the pre-existing GAL1 promoter-aga2p signal sequence-HA tag segments, followed by a AscI site, and a NheI site incorporated in the aga2p signal sequence was designated pTQ2.
  • a MAT ⁇ transcriptional terminator sequence was amplified by PCR from the pYD 1plasmid, and BamHI and PstI restriction sites were appended to facilitate cloning into plasmid pTQ2 above.
  • the MAT ⁇ terminator was then digested with BamHI and PstI and inserted into the BamHI/PstI site on plasmid pTQ2.
  • a DNA linker segment containing SfiI and NotI restriction sites and a segment coding for a myc tag were added at the 3′ end of the amplified pYD1 segment.
  • the myc tag was included to allow detection of the anchored chain (of the multi-chain polypeptide) on the yeast cell surface.
  • linker-myc segment sequences is as follows: GGA GGC GGA GGT TCT GGG GGC GGA GGA TCT GGT GGC GGA GGT TCT (SEQ ID NO:3) G G G G S G G G G S G G G G S (SEQ ID NO:4) Sfi I Not I GC G GCC CAG CCG GCC AGT CCT GAT GCG GCC GC A GAA CAA AAA CTC G G Q P G S P D A A A E Q K L Pac I ATC TCA GAA GAG GAT CTG AAT TTA ATTAA I S E E D L N
  • This linker-myc segment was inserted into an EcoRI and PacI digested pTQ2.
  • Plasmid pTQ3 is a 5810 bp multi-chain yeast display plasmid comprising, in pertinent part, the following vector sequence: ⁇ --------------------------Aga2p signal sequence---------- 435 ATG CAG TTA CTT CGC TGT TTT TCA ATA TTT TCT GTT ATT GCT (SEQ ID NO:6) M Q L L R C F S I F S V I A --------------> ⁇ -------------HA tag--------------> AGC GTT TTA GCA TAC CCA TAC GAC GTT CCA GAC TAC GCT S V L A Y P Y D V P D Y A Apa LI Asc I Bam HI A GT GCA C AG GAT TTC GTG CAA TGC GGC GCG CC A GGA TCC S A Q D F V Q C G A P G S ATG TAA M ⁇ ------------------------------------Mat ⁇ terminator---------
  • phage display Fabs were transferred from the phage display vector to a multi-chain eukaryotic display vector to demonstrate of the utility of the phage display-eukaryotic display transfer system, and the ability of the multi-chain eukaryotic vector of the present invention to express a multi-chain polypeptide.
  • the vector was then inserted into a eukaryotic host cell, and the transformed host cell grown under conditions suitable for expression of the Fabs.
  • Anti-streptavidin Fab antibodies, F2, A12, and 4C8 were each cloned from a large naive human Fab library (de Haard, H. et al., 1999) into the multi-chain yeast display vector pTQ3 constructed in Example 1 as a paired light chain (V L C L ) and heavy chain (V H C H 1). Additionally, an anti-mucin Fab antibody, PH1, was cloned from the same Fab library into the multi-chain yeast display vector pTQ3 constructed in Example 1 as a paired light chain (V L C L ) and heavy chain (V H C H 1).
  • cytotoxic T-lymphocyte associated antigen 4 CTLA-4
  • pTQ3 constructed in Example 1 as a paired light chain (V L C L ) and heavy chain (V H C H 1).
  • the chains of the Fabs were cloned into the multi-chain yeast display vector using the single excision/insertion transfer process described earlier and illustrated in FIG. 1.
  • the LC-HC polynucleotide from the Fab library was inserted as a single ApaLI/NotI fragment.
  • the unwanted prokaryotic genetic control elements intervening the coding regions of the LC and HC fragment and defined by the AscI/SfiI restriction fragment from the Fab library was replaced with the AscI/SfiI fragment derived from pTQ3.
  • pTQ3-F2 The resulting plasmids, designated pTQ3-F2, pTQ3-A12, pTQ3-4C8, pTQ3-PH1, pTQ3-E7, pTQ3-E8, pTQ3-A9 and pTQ3-A11, were separately transformed into S. cerevisiae strain EBY 100 (InVitrogen, Carlsbad, Calif.) following the method of Gietz, D. et al., 1992. EBY100 was also transformed pTQ3 containing no multi-chain insert as a control. Transformant selection was performed selecting for the vector tryptophan auxotrophic marker (synthetic defined medium minus tryptophan, 2% (w/v) glucose, 2% agar (SDCAA+GA)).
  • tryptophan auxotrophic marker synthetic defined medium minus tryptophan, 2% (w/v) glucose, 2% agar (SDCAA+GA)
  • Cell pellets were resuspended in 250 mL of SDS-PAGE buffer plus dithiothreitol (DTT). 425-600 micron glass beads (Sigma, St. Louis, Mo.) were added to just below the meniscus, and the suspension was vortexed 4 times for 1 minute. The suspension was kept on ice between vortexing. The supernatant was transferred to a fresh tube and heated to 100° C. for 5 minutes.
  • DTT dithiothreitol
  • Protein samples were separated on a SDS-PAGE gel and transferred to a nitrocellulose membrane for western blotting.
  • Detection of the light chain polypeptide was performed using an anti-HA antibody (1 ⁇ g/mL) (Dako, Carpinteria, Calif.).
  • Detection of the heavy chain-Aga2p fusion polypeptide was performed using an anti-c-Myc antibody (1 ⁇ g/mL) in conjunction with a secondary rabbit anti-mouse HRP antibody (Dako, Carpinteria, Calif.).
  • Immunodetection was by enhanced chemiluminescence (Amersham-Pharmacia, Piscataway, N.J.).
  • the LC product of approximately 30 kD and the HC-Aga2p fusion product of approximately 45 kD of the displayed Fabs were detected. No detectable LC or HC-Aga2p fusion product was detected prior to induction with galactose (see FIGS. 4A and 4B).
  • a multi-chain eukaryotic display vector was inserted into a eukaryotic host cell and the transformed host cell was grown under conditions suitable for expression and display of the Fab on the surface of the host cell.
  • Yeast clones EBY100 pTQ3-F2, EBY100 pTQ3-PHI, EBY100 pTQ3-E7, EBY100 pTQ3-E8, EBY100 pTQ3-A9 and EBY100 pTQ3-A11 were prepared, cultured, and induced for antibody expression as described in Example 2 above. Three 0.2 mL aliquots of yeast cells having an OD 600 of 1.0 were removed prior to induction with galactose, as the T 0 point.
  • yeast cells displaying an antigen-specific Fab antibody can be enriched over an excess of non relevant yeast cells
  • model selection experiments were performed using an automated magnetic bead selection device.
  • the Fab-displaying yeast cell EBY100 pTQ3-F2 (with a tryptophan auxotrophic selectable marker) were mixed with nonspecific yeast cells at various ratios.
  • the non specific yeast cells consisted of EBY100 pUR3867 (Unilever Research, VLaardingen, Netherlands), and encoding a scFv antibody specific for mucin-1 (PHI), and carrying a leucine auxotrophic selectable marker.
  • the ratio of Leu + /Trp + cells before and after selection was used to calculate the enrichment factor after 1 round of selection.
  • Yeast clones were grown and antibody expression induced with galactose as described in Example 2. The two yeast clones were mixed in the ratio indicated above, and incubated for 1 hour with 100 ⁇ L streptavidin paramagnetic beads (Dynal M280, Dynal Biotech, Oslo, Norway) in an end volume of 1 mL 2% a phosphate-buffered saline solution (e.g., 2% MARVEL-PBS or “MPBS”, Premier Brands Ltd., U.K.).
  • streptavidin paramagnetic beads Dynal Biotech, Oslo, Norway
  • a phosphate-buffered saline solution e.g., 2% MARVEL-PBS or “MPBS”, Premier Brands Ltd., U.K.
  • the cell-bead complexes were washed for 11 cycles in 2% MPBS by transferring the complexes from one well to the next well in an automated magnetic bead selection device. After the 2% MPBS washing, two more washing steps were performed with PBS. In the last well of the automated magnetic bead selection device, the cell-beads complexes were resuspended in 1 mL PBS and the titres determined by plating on SDCAA+G agar plates or with synthetic defined medium containing 2% (w/v) glucose containing leucine drop out media plus 2% agar (SD-Leu+G agar plates).
  • yeast cells were incubated for one hour at room temperature with 500 ⁇ L streptavidin microbeads (Miltenyl Biotec, Cologne, Germany) in 6 mL PBS+2 mM EDTA.
  • streptavidin microbeads Miltenyl Biotec, Cologne, Germany
  • the cell/bead mixture was loaded onto a pre-washed LS column (Miltenyi Biotec, Cologne, Germany) in the presence of a magnet, and the column was washed twice with PBS+2 mM EDTA. After the magnet was removed the bound yeast cells were eluted with 6 mL PBS buffer.
  • yeast selections using the capillary washing device using the capillary washing device (CWD) the yeast cell mixture and 100 ⁇ l streptavidin coated paramagnetic beads (Dynal M280) were blocked in 1 mL 2%MPBS for 1 hour.
  • the paramagnetic beads were resuspended in 1 mL of yeast cells suspension and gently rotated for 1 hour at room temperature in an eppendorf tube.
  • the mixture was introduced into the capillary (1 mL was used to load one capillary in five 200 ⁇ L steps) of the CWD.
  • a final wash with PBS was performed and the yeast/beads complex was collected by adjusting the magnet.
  • the enrichment factor was calculated as the ratio of specific yeast cells before and after selection divided by the ratio of the non specific yeast before and after selection.
  • Table 1 Model enrichment of Fab displaying yeast cells: Detection by magnetic bead selection.
  • TABLE 1 Model enrichment of Fab displaying yeast cells: Detection by magnetic bead selection. Ratio a Total cells b Enrichment c Recovery (%) d Kingfisher 1/100 ⁇ 10 7 288,000 12.8 1/1000 ⁇ 10 8 1,100,000 6 1/10000 ⁇ 10 9 400,000 10.7 Capillary Washing Device (CWD) 1/100 ⁇ 10 7 76,000 4.7 1/1000 ⁇ 10 8 41,000 6 1/10000 ⁇ 10 9 10,000 5.3 MACS 1/1000 10 7 100 12
  • specific yeast cells displaying a Fab antibody to streptavidin can be enriched by between 2 and 6 orders of magnitude over non relevant yeast cells by one round of selection in an automated magnetic bead selection device such as Kingfisher, capillary washing device or magnetic activated cell sorting (MACS).
  • an automated magnetic bead selection device such as Kingfisher, capillary washing device or magnetic activated cell sorting (MACS).
  • the Fab-displaying yeast cells EBY100 pTQ3-F2 (with a tryptophan auxotrophic selectable marker), were mixed with the nonspecific yeast cells, EBY 100 (pUR3867-PH1) carrying a Leu auxotrophic marker, at ratios of 1:100, 1:1000 and 1:10,000.
  • the yeast cell mixture was incubated with 1 ⁇ M streptavidin-FITC (Dako, Carpinteria, Calif.) and allowed to equilibrate for 30 minutes at room temperature.
  • a phage display Fab library prepared using techniques known in the art was transferred to the multi-chain yeast display vector pTQ3 produced as described in Example 1 above.
  • a 50 mL culture of TYAG (TY, ampicillin 100 ⁇ g/mL, glucose 2%) was inoculated with 10 ⁇ L of a glycerol stock from one round of selection on streptavidin of a naive Fab library cloned into phage (de Haard, H. et al., 1999). The culture was grown overnight at 37° C. and plasmid DNA was prepared (QIAGEN plasmid purification system, Qiagen, Valencia, Calif.).
  • the Fab antibody repertoire was digested with ApaLI and NotI and Fab antibody fragments of approximately 1.5 kb were recovered and purified by extraction from a 1.0% TBE ethidium bromide agarose gel (QIAEX gel extraction kit, Qiagen, Valencia, Calif.).
  • the multi-chain yeast display vector pTQ3 was digested with ApaLI and NotI and a fragment of approximately 4.6 kb was purified by extraction from a 1.0% TBE ethidium bromide agarose gel.
  • the purified ligation mix was transformed into E. coli strain TG1 (Netherlands Culture Collection of Bacteria, PC 4028, Utrecht, NL) by electroporation using a BioRad Pulser (BioRad, CA) at 2.5 kV, 25 mF and 200 W.
  • the library was plated on 2 ⁇ TY agar plates (16 g/L bacto-tryptone, 10 g/L yeast extract, 5 g/L NaCl, 15 g/L bacto-agar) containing ampicillin at 100 ⁇ g/mL and 2% w/v glucose (TYAG plates). After overnight growth at 37° C. the repertoire was recovered in 2 ⁇ TY medium plus ampicillin at 100 ⁇ g/mL by flooding the plates, and frozen in aliquots in 15% (w/v) glycerol.
  • the library contained 5.6 ⁇ 10 6 independent clones. 15 ⁇ L of a library suspension of 5.4 ⁇ 10 10 cells/mL was used to inoculate 100 mL of TYAG, and the culture was grown overnight at 37° C. Plasmid DNA recovered as described above.
  • the intermediate pTQ3-Fab repertoire was then digested with AscI and with SfiI. A fragment of approximately 6.1 kb was purified as described above.
  • Source vector pTQ3 was similarly digested with AscI and SfiI and a fragment of approximately 1150 bp was purified.
  • Plasmid DNA was prepared from pTQ3-Fab library and transformed into yeast strain EBY10O by the method of Gietz, D. et al., (1992) to give a final library size in yeast of 2 ⁇ 10 6 independent yeast clones.
  • yeast display Fab library can undergo selection from a population of yeast cells displaying a diverse repertoire of Fab antibodies
  • multiple selection experiments were performed using an automated magnetic bead selection device.
  • yeast repertoire prepared in Example 6 was grown at 30° C. in SDCAA+G, and antibody expression was induced with galactose (as in Example 4).
  • the pool of yeast cells was incubated for 1 hour with 100 ⁇ L streptavidin paramagnetic beads (Dynal M280, Dynal Biotech, Oslo, Norway) in an end volume of one mL 2% MPBS.
  • the cell-bead complexes were washed for 11 cycles in 2% MPBS by transferring the complexes from one well to the next well in the automated magnetic bead selection device. After the 2% MPBS washing, two more washing steps were performed with PBS. In the last well of the automated magnetic bead selection device, the cell-bead complexes were resuspended in 1 mL PBS and the yeast colony titres before and after selection were determined by plating on SDCAA+G agar plates. The selected yeast cells were then used to inoculate a fresh culture of 10 mL SDCAA+G and a second round of selection was performed as above.
  • the percentage of positive and negative clones was determined by yeast whole cell ELISA after the first round of selection and after the second round of selection. Cells were grown and induced in a 96 well plate (Corning Costar, Cambridge, Mass.) in 100 mL SDCAA plus 2% (w/v) galactose.
  • cells were washed one cycle with PBS and divided equally onto two plates for detection of antigen binding and heavy chain display.
  • the cells were resuspended in 100 ⁇ L 2% MPBS containing anti-streptavidin-HRP (0.87 ⁇ g/mL) for detecting antigen binding.
  • the cells of the second plate were resuspended in 100 ⁇ L 2% MPBS containing anti-c-Myc (1 ⁇ g/mL) for detecting heavy chain display.
  • clones EBY100 pTQ3-F2 and EBY100 pTQ3-A12/pESC contain an empty vector pESC (Stratagene, La Jolla, Calif.) carrying the Leu auxotrophic marker.
  • the anti-streptavidin antibody F2 has an affinity of 54 nM as determined by plasmon resonance (BIAcore) and the anti-streptavidin antibody A12 has an affinity of approximately 500 ⁇ M.
  • the high affinity antibody containing clone (EBY100 pTQ3-F2) and the low affinity antibody containing clone (EBY100 pTQ3-A12/pESC clones were mixed at a ratio of approximately 1:100.
  • Using the different selectable markers present in each clone allowed discrimination of EBY100 pTQ3-A12/pESC (which are able to grow on minus tryptophan, minus leucine selective agar plates) from EBY100 pTQ3-F2 (which can only grow on minus tryptophan selective agar plates).
  • the cell mixture was labeled as previously except with a serial dilution of streptavidin-FITC of 500 nM, 100 nM, 50 nM, 25 nM and 10 nM.
  • Cells were sorted by flow cytometry in an EPIC ALTRA (Beckman Coulter, Fullerton, Calif.) on the basis of both LC display and antigen binding.
  • the sorting rate was set at 2000 cells/sec and the sorting gate was set to collect 1% of the cell population with the highest ratio of FITC to PE 9typical FACS histogram is shown in FIG. 7).
  • Titre Titre (-Trp) Percent- Antigen Titre (-Trp/-Leu) ⁇ (-Trp/-Leu) age Enrich- (nM) a (-Trp) b Fab-A12 c Fab-F2 d Fab-F2 e ment f Input 3.2 ⁇ 10 7 3.7 ⁇ 10 7 4.7 ⁇ 10 5 1.3 Output 500 nM 9 ⁇ 10 3 8.9 ⁇ 10 3 100 1.1 0.9 100 nM 1.3 ⁇ 10 3 7.9 ⁇ 10 2 5.6 ⁇ 10 2 71 56 50 nM 2.4 ⁇ 10 3 1.2 ⁇ 10 3 1.2 ⁇ 10 3 102 80 25 nM 1.2 ⁇ 10 3 9.1 ⁇ 10 2 3.2 ⁇ 10 2 35 27 10 nM 1.53 ⁇ 10 3 1.49 ⁇ 10 3 45 3 2.3
  • Coli and grown on selective agar plates containing 100 ⁇ g/mL ampicillin to give a LC repertoire of 5 ⁇ 10 6 designated pTQ3F2-LCeP, a HC repertoire of 5.6 ⁇ 10 8 designated pTQ3F2-HCeP and a whole Fab repertoire of pTQ3F2-FabeP.
  • the repertoires were harvested and an inoculated of 200 mL (sufficient to encompass at least 10 times the library diversity) was made.
  • Plasmid DNA was isolated from a 200 mL culture and transformed into the yeast strain EBY100 as described in Example 2.
  • the mutation frequency at the nucleotide level was 1.5% for the LC and 0.8% for the HC.
  • the mutation frequency at the amino acid level was 3% for the LC and 1.3% for the HC.
  • yeast clones retrieved from the affinity selection of the repertoires EBY100-pTQ3F2-LC ep ; EBY100-pTQ3F2-HC ep ; EBY100-pTQ3F2-Fab ep were affinity screened to quantitate the improvement in affinity over the starting wild-type antibody.
  • the selected antibodies were also sequenced to determine the mutations that correlate with improved affinity.
  • Yeast colonies were picked and resuspended in 25 ⁇ L of lyticase solution (2.5 mg/mL; Sigma, St. Louis, Mo.) for 1 hour at 37° C. after which 2 ⁇ L was taken and used in a PCR reaction. Separate LC and HC were amplified and sequenced using and ABI-PRISM sequencer. Mutations from wild-type were determined using sequence alignment and are shown in Table 6. TABLE 6 Overview of mutated Fab antibodies selected from error prone repertoires by FACS.
  • the off rate of the selected Fabs was determined by measuring the dissociation rate in FACS as the decrease in fluorescence signal over time; the clone, R2H10 gave the greatest improvement in affinity (10.7 fold, 3.2 nM). This dissociation rate was fit to a exponential decay model and the k d calculated.
  • Yeast cells were labeled with both anti-HA to detect the LC and also for antigen with streptavidin PE. Yeast cultures were grown an induced as described Example 2 and approximately 2 ⁇ 10 7 cells were collected and washed with PBS. The cells were then incubated with 100 ⁇ L anti-HA Mab (20 ⁇ g/mL) for 1 hour and then washed with 0.5 mL of PBS.
  • the cells were then incubated with rabbit anti-mouse FITC (1:40) and streptavidin PE (1:40 dilution of 1 ⁇ g/mL stock) for 1 hour on ice.
  • the cell pellet was then resuspended in an excess of non-fluorescent ligand at room temperature.
  • the concentration of non-fluorescent label was taken so that it was 10-100 fold in excess of the molar concentration of yeast displayed Fab antibody assuming there are approximately 100,000 copies of a Fab antibody per yeast cell.
  • the decrease in fluorescence intensity was monitored for 1.5 mins. to 30 mins. by flow cytometry. Unlabeled yeast cells were used to set the background fluorescence.
  • FIG. 10c shows the off rate determination by FACS for clones wild-type F2, and mutants R2E10, R3B1 and R3H3.
  • H34R Y96H none 0.5 ⁇ 0.1 0.18 ⁇ 0.01 5.69 ⁇ 0.02 3.2 10.7 R3A9 ′′ S2P, D85V none 1.3 ⁇ 0.1 1.53 ⁇ 0.57 7.84 ⁇ 0.08 19.5 1.7
  • a yeast display vector effective in a host yeast cell transformed with the vector of expressing, transporting, and displaying an Ig heavy chain fragment was constructed as one vector of a matched vector set.
  • An HC fragment display vector was constructed by further altering the vector pTQ3 produced according to Example 1.
  • Display vector TQ3 was digested with BseRI, thus identifying a designed restriction site of the vector positioned in each of the two tandem GAL1 promoters (see Example 1, SEQ ID NO: 5 designated bases 990-995).
  • the vector backbone was re-ligated and transformed into E. coli .
  • the HC for the anti-streptavidin Fab antibody F2 was restriction digested from pTQ3-F2 as a 709 bp SfiI/NotI fragment, purified, and cloned into SfiI/NotI digested vector pTQ5.
  • the resultant HC display vector was designated “pTQ5-HC” (FIG. 9).
  • Eukaryotic Host Cell Expression of an Ig Heavy Chain Eukaryotic Display Vector HC Expression in a Haploid Yeast Cell
  • yeast display vector (of a vector set) encoding an Ig heavy chain fragment was inserted into a eukaryotic host cell, and the transformed host cell grown under conditions suitable for expression of the heavy chain component of an Ig Fab.
  • yeast strain EBY 100 (InVitrogen, Carlsbad, Calif.) was transformed with vector pTQ5-HC (of Example 13), and separately with pTQ5 as a control, following transformation procedures previously described.
  • the successful transformants designated EBY100 pTQ5-HC and EBY100 pTQ5 respectively, were cultured overnight at 30° C. in 10 mL SDCAA+G.
  • Cell pellets were resuspended in 200 mL of SDS-PAGE sample buffer plus DTT, and glass beads (425-600 micron) were applied to just below the meniscus. The cell and bead suspension was vortexed 4 times for 1 minute keeping the suspension on ice between vortexing. The supernatant was transferred to a fresh tube and heated to 100° C. for 5 minutes.
  • Eukaryotic Host Cell Display of an Ig Heavy Chain Eukaryotic Display Vector HC Display on the Surface of a Haploid Yeast Cell
  • a yeast display vector (of a vector set) encoding an Ig heavy chain fragment was inserted into a eukaryotic host cell, and the transformed host cell was grown under conditions suitable for expression and display of the heavy chain component of an Ig Fab.
  • EBY100 pTQ5-HC (from Example 14) was grown, and antibody expression was induced as above. HC expression was induced by 48 hours of growth with shaking at 20° C. Yeast samples were centrifuged and the cell pellet resuspended in PBS containing 1 mg/mL BSA. Two of the samples were again centrifuged and the cell pellets separately resuspended in either 100 ⁇ L of anti-human C H 1 (25 ⁇ g/mL; Zymed, San Francisco, Calif.) followed by incubation for 1 hour at room temperature. The cells were pelleted and washed once with 0.5 mL of PBS/1% (w/v) BSA. Cell samples were then incubated with rabbit anti-mouse FITC (1:50 dilution; Dako, Carpinteria, Calif.) for 1 hour in the dark.
  • a light chain yeast display vector was constructed to provide a multi-chain eukaryotic display vector set, i.e., when used in conjunction with the heavy chain yeast display vector described above (see Example 13, supra).
  • a LC yeast expression vector was constructed by amplifying a fragment containing the anti-streptavidin LC fused to the HA epitope tag and Aga2p signal sequence.
  • the amplification product was gel purified using a GFX PCR and Gel Band Purification Kit (Amersham-Pharmacia, Piscataway, N.J.), and digested with HindIII and Pmel.
  • the 783 bp LC fragment was purified on a 1.2% TAE-agarose gel together with the 4,323 bp vector backbone of a HindIII/Pmel-digested pYC6/CT vector (InVitrogen, Carlsbad, Calif.).
  • the LC fragment and pYC6/CT vector were ligated together and the ligation mix was transformed into E. coli strain TG1.
  • the resultant LC expression vector was designated “pTQ6-LC” (FIG. 12).
  • Eukaryotic Host Cell Expression of an Ig Light Chain Eukaryotic Display Vector Soluble LC Expression in a Haploid Yeast Cell
  • yeast display vector (of a vector set) encoding an Ig light chain fragment was inserted into a eukaryotic host cell, and the transformed host cell grown under conditions suitable for expression of a soluble light chain component of an Ig Fab.
  • Yeast strain W303-1B (a/alpha ura3-1/ura3-1 leu2-3,112/leu2-3,112 trpl-1/trpl-1 his3-11,15/his3-11,15 ade2-1/ade2-1 can1-100/can1-100), obtained from P. Slonimski, was transformed with pTQ6-LC (of Example 16) and separately pYC6/CT as a control, following transformation procedures previously described.
  • haploid yeast cells containing a vector expressing a soluble Ig light chain fragment were mated to haploid yeast cells containing a vector expressing and displaying an Ig heavy chain-anchor fusion polypeptide to produce a diploid yeast cell that displays a functional Fab polypeptide on the surface of the host cell.
  • Yeast clones W303 pTQ6-LC (from Example 17) and EBY100 pTQ5-HC (from Example 14) were grown on agar plates supplemented with either Blastocidin® (InVitrogen, Carlsbad, Calif.; 300 ⁇ g/mL; SD+G+Bls agar plates) or tryptophan drop out medium (SD-Trp+G agar plates). These plates were then replica plated onto double selective plates containing synthetic defined medium for tryptophan dropout plus 300 ⁇ g/mL Blasticidin® (SD-Trp+G+Bls). The resulting cell layer of diploid yeast cells was streaked to single colonies. Seven Trp+/Bls R colonies were selected and grown overnight with shaking at 30° C. in 100 mL SD+G-Trp+Bls in 96-wells plate.
  • Cells of the first plate were resuspended in 100 ⁇ L streptavidin-HRP (0.87 ⁇ g/mL), cells of the second plate were resuspended in 100 ⁇ L anti-c-Myc-HRP (1 ⁇ g/mL), and cells of the third plate were resuspended in 100 pL anti-HA (1 ⁇ g/mL) and additionally labeled with a rabbit anti-mouse-HRP ( ⁇ fraction (1/1000) ⁇ ).
  • Yeast whole cell ELISA was performed (as in Example 7) and FACS (as in Example 15) was performed to detect antigen binding and HC display. All diploids tested bound to streptavidin and displayed light chains in whole cell ELISA (FIG. 14) and FACS (FIGS. 15 A-C). Specifically, streptavidin binding activity was detected on diploid yeast cells displaying combinatorially assembled Fab antibody (Diploid LC/HC) on their surface whereas haploid parents expressing either LC only (W303 pTQ6-LC) or HC only (EBY100 pTQ5-HC) showed no binding activity.
  • Standard haploid yeast cells displaying a Fab antibody (EBY100 pTQ3-F2) showed streptavidin binding activity. Also (as expected) the haploid parent yeast cell expressing only LC (W303 pTQ6-LC) showed no HC display, while standard haploid yeast cells displaying a Fab antibody (EBY100 pTQ3-F2) showed HC display.
  • yeast clones were selected for overnight culture at 30° C. in 10 mL SD+G-Trp+Bls with shaking. The next day, cell cultures were centrifuged and the pelleted yeast cells resuspended in 10 mL SD-Trp+Bls plus 2% (w/v) galactose to an OD 600 of 0.4 for 24 hours to induce vector expression.
  • An alternative protocol involves resuspension in 10 mL of YP media plus Blastocidin® plus 2% (w/v) galactose.
  • Protein samples were separated on SDS-PAGE gels and transferred to a nitrocellulose membrane for western blotting.
  • Detection of the light chain polypeptide was performed using an anti-HA antibody (1 ⁇ g/mL) in combination with a rabbit anti-mouse conjugated to HRP on one membrane.
  • Detection of the heavy chain-Aga2p fusion polypeptide was performed using an anti-c-Myc antibody directly conjugated to HRP (1 ⁇ g/mL, Roche Molecular Biochemicals, Indianapolis, Ind.). Immunodetection was by enhanced chemilluminescence (Amersham-Pharmacia, Piscataway, N.J.).
  • the LC product of approximately 30 kD and the HC-Aga2p fusion product of approximately 45 kD were both detected in the diploid yeast lysate (FIGS. 16 and 17). No detectable LC or HC-Aga2p fusion product was detected in control diploid clones harboring the two empty vectors pTQ5 and pYC6/CT.
  • a first sample was further incubated with rabbit anti-FITC (1:40; Dako, Carpinteria, Calif.), and finally with swine anti-rabbit conjugated to FITC (1:20; Dako, Carpinteria, Calif.).
  • a second sample was submitted to a double labeling with swine anti-rabbit conjugated to FITC (1:20; Dako, Carpinteria, Calif.) for the light chain and rabbit anti-mouse conjugated to Tetramethylrhodamine isothiocyanate (TRITC, 1:30, Sigma, St. Louis, Mo.) for the heavy chain (FIGS. 18 A-C).
  • the diploid displayed the light and the heavy chain at the cell surface and was shown to bind streptavidin, as expected.
  • the haploid parent expressing HC only was stained only by the TRITC labeling of the heavy chain.
  • the haploid parent LC was negative in all the cases.
  • Each haploid parent EBY100 pTQ5 (from Example 14) and W303 pYC6/CT (from Example 17) was grown overnight at 30° C. in the appropriate selective medium SD+G-Trp and SD+G+Bls respectively.
  • 3 ⁇ 10 7 cells from the two fresh haploid cultures were mixed and collected on a 45 mm nitrocellulose filter (microfill device of Millipore, Bedford, Mass.). The filter was incubated for 4 hours at 30° C. on a non selective rich medium plate (YPD). Cells were then resuspended in YPD medium and titrated on the two parental selective media and on the double selective medium (which only allows the growth of the diploids) SD+G-Trp+Bls. Spontaneous reversion or resistance was assessed by processing each haploid parent separately in the same way and plating them on the double selective medium without dilution.
  • the mating efficiency of the haploid parent EBY100 pTQ5 was calculated as follows: (the number of diploids growing on SD+G ⁇ Trp+Bls minus the number of spontaneous resistant EBY100 pTQ5 growing on SD+G ⁇ Trp+Bls) divided by (the total number of cells from the mating reaction showing growth on SD+G ⁇ Trp).
  • the mating efficiency of haploid parent W303 (pYC6) the efficiency was calculated as follows: (the number of diploids on SD+G ⁇ Trp+Bls minus the number haploid cells W303pYC6/CT growing on SD+G ⁇ Trp+Bls) divided by (the number of cells on SD+G+Bls).
  • FACS fluorescent activated cell sorting
  • a non-relevant yeast diploid cell carrying the phenotypic markers ⁇ Trp+/ ⁇ Leu+ was used, and was able to grow on minus leucine and tryptophan containing selective agar plates. This diploid was designated the Leu+diploid.
  • Both positive (Bls R ) and non-relevant (Leu+) diploid yeast cells were grown overnight in media of SD plus 2%(w/v) glucose under selective conditions of ⁇ Trp/+Leu/+Bls media and ⁇ Trp/ ⁇ Leu media respectively.
  • Yeast cultures were induced in YP media containing galactose at 2% (w/v).
  • a mix of positive to non-relevant yeast cells was prepared in an approximate ratio of 1:10000.
  • FACS the yeast cell mixture was labeled with 500 nM streptavidin PE and selected as in Example 8.
  • streptavidin coated beads For Kingfisher, the yeast cell mixture was incubated with streptavidin coated beads and selected as in Example 4.
  • MACS induced diploids were incubated for one hour at room temperature with 500 ⁇ L streptavidin microbeads (Miltenyi Biotec, Cologne, Germany) in 6 mL PBS+2 mM EDTA.
  • the cell/bead mixture was loaded onto a pre-washed LS column (Miltenyi Biotec, Cologne, Germany) in the presence of a magnet and the column was again washed twice with PBS+2 mM EDTA. After the removal of the magnet, the cells retained on the column were eluted in 6 mL PBS buffer.
  • Yeast cells were recovered and titrated on selective agar plates for either the Bla R phenotype or the Leu + phenotype. The ratio of Bls R /Leu + colonies before and after selection was used to calculate the enrichment factor and the percentage recovery of positive yeast cells TABLE 8 Single pass enrichment experiments using MACS, Kingfisher and FACS. Input Output Bla R / Bla R / Leu + Bla R Leu + Leu + Bla R Leu + Re- Device diploid diploid (%) diploid diploid (%) covery Enrich.
  • a haploid yeast cell population containing a vector expressing a plurality soluble Ig light chain fragment variants is mated to a haploid yeast cell population of opposite mating type containing a vector expressing and displaying a plurality of Ig heavy chain-anchor fusion polypeptide variants (i.e., a library of HC variants) to produce a novel diploid yeast cell population that displays a plurality of functional Fab polypeptides on the surface of the host cells (i.e., a novel Fab library).
  • Fab phage display isolates pre-selected for a target molecule from a Fab repertoire, are used to provide the source heavy chain and light chain components for a batch transfer of the phage display isolate genetic information to a multi-chain yeast display vector set (as demonstrated in Example 6), using the multi-chain yeast display vector set (as described in Examples 14 and 17) to provide novel recombination of light and heavy chain isolates via host cell fusion of two haploid eukaryotic cells, each possessing a different vector from a matched multi-chain eukaryotic display vector set (as demonstrated in Example 18).
  • a phage display antibody library (de Haard, H. et al., 1999) was subjected to one round of selection on streptavidin coated magnetic particles using protocols familiar to those skilled in the art. This repertoire was used as a starting repertoire for transfer into the yeast display system. The input of the library was 5 ⁇ 10 12 phage particles and the output after one round of selection was 3.75 ⁇ 10 5 phage particles.
  • HC fragments were isolated from the round 1 selected phage display library as SfiI/NotI fragments and cloned into the Ig heavy chain yeast display vector pTQ5, which was digested with SfiI and NotI (Example 13).
  • the ligation mix was transformed into E. coli to give a library of 10 8 .
  • This library was then transformed into the yeast strain EBY100 to give a library of 4 ⁇ 10 7 and designated EBY100-pTQ5-HC rep .
  • the LC fragments were isolated from the round 1 selected phage display library as ApaL1/AscI fragments and cloned into the Ig light chain yeast display vector, pTQ6 digested with ApaL1 and AscI (Example 16).
  • the ligation mix was transformed into E. coli to give a library of 1 ⁇ 10 8 .
  • This library was then transformed into the yeast strain BJ5457 to give a library of 8 ⁇ 10 7 and was designated BJ5457-pTQ6-LC rep .
  • Both the HC and LC repertoires in yeast contained sufficient diversity to cover the starting repertoire of 3.75 ⁇ 10 5 in phage. DNA fingerprint analysis of individual clones showed diverse restriction patterns indicating that different germline segments were represented in the separate LC and HC libraries.
  • the HC repertoire was constructed by amplifying the anti-streptavidin F2 antibody under error prone conditions (Example 9). The amplified fragment was digested with SfiI and NotI, purified and cloned into the HC-only expression vector pTQ5 (Example 13), which had also been digested with SfiI and NotI. The resulting ligation mix was transformed into E. coli to give a library of 7 ⁇ 10 7 . This library was transformed into the yeast strain EBY100 to give a library of 9 ⁇ 10 6 and was designated EBY100 pTQ5-HC*.
  • the LC repertoire was constructed by amplifying the LC of the anti-streptavidin F2 antibody under error prone conditions (Example 9).
  • the amplified fragment was digested with ApaL1 and AscI and cloned into the LC only expression vector (Example 16), which had also been digested with ApaL1 and AscI.
  • the resulting ligation mix was transformed into E. coli to give a library of 4 ⁇ 10 7 , and this was subsequently transferred into the yeast strain BJ5457 to give a library of 1.8 ⁇ 10 7 (designated BJ5457 pTQ6-LC*).
  • the resulting mutation frequency at the nucleotide level was 0.8% for the HC repertoire and 1.5% for the LC repertoire. These frequencies correspond to 1.3% and 3% mutation frequency at the amino acid level, respectively.
  • the haploid cell repertoires EBY100 pTQ5-HC* and the BJ5457 pTQ6-LC* were inoculated with 10 ⁇ L and 30 ⁇ L of glycerol stock respectively so that at least 10 copies of each independent clone was represented and grown up overnight in selective media (Example 18).
  • haploid cells corresponding to BJ5457 pTQ6-LC* and 3 ⁇ 10 10 haploid cells corresponding to EBY100 pTQ5-HC* were mated (Example 19) to give a diploid repertoire of 5 ⁇ 10 9 when grown on selective media (designated EBY100 pTQ5-HC*/BJ5457 pTQ6-LC*).
  • Ten clones were picked and tested by yeast colony PCR (Example 11) for the presence of LC and HC containing vectors, and all gave the expected LC and HC product.
  • Example 7 To determine the fraction of the diploid repertoire EBY100 pTQ5-HC*/BJ5457 pTQ6-LC* that displayed a HC product and also showed binding to the antigen streptavidin, a yeast whole cell ELISA was performed (Example 7). 68% (15/22) diploids tested displayed a HC, and 18% (4/22) of diploid clones tested showed binding to streptavidin.
  • diploid repertoire was made by mating BJ5457 pTQ6-F2LC with EBY100 pTQ5-HC*.
  • This diploid repertoire has a constant LC and a variable HC. This mating resulted in 70% of diploids displaying a HC and 45% showing antigen binding activity by yeast whole cell ELISA.
  • Example 18 An overnight culture of the diploid repertoire EBY100 pTQ5-HC*/BJ5457 pTQ6-LC* (Example 22) was prepared (Example 18). The culture was induced as in Example 18 and a total of 100 cells were subjected to one round of Kingfisher selection (Example 7). The antigen binding yeast diploid cells were retrieved and subjected to FACS affinity driven selection (see Example 20). The percentage of antigen binding clones increased during selection as determined by yeast whole cell ELISA (Example 7). The percentage of antigen binding clone also increased, and the antigen mean fluorescent intensity as determined by FACS increased during selection (Table 9).
  • Plasmid DNA was prepared using a lyticase treatment (Example 11), and the DNA extract containing both pTQ5-HC* sel and pTQ6-LC* sel expression plasmids containing selected LC and HC was transformed directly into fresh EBY100 and BJ5457 cells, respectively.
  • the transformation mix was grown on selective plates so only BJ5457-pTQ6-LC* sel colonies (selective agar plates containing blastocidin) or EBY100 pTQ5-HC* sel (selective agar plates minus tryptophan) could grow.
  • Antibody expression was induced, followed by labeling with streptavidin PE at 0.5 nM and selection using FACS (Example 20). TABLE 11 Analysis of Fab antibodies.
  • Antibody HC fragments are isolated from a V gene peripheral blood lymphocyte source and isolated by antibody PCR methods known in the art.
  • the HC library is captured in a phage display vector following techniques known in the art and then transferred to pTQ5 as a SfiI/NotI fragment and transformed into E. coli , producing a library of approximately 1 ⁇ 10 8 .
  • the library is then transformed into yeast strain EBY100, producing library EBY100 pTQ5-HC* rep of approximately 1 ⁇ 10 7 .
  • Antibody LC fragments are isolated from a V gene peripheral blood lymphocyte source and isolated by antibody PCR methods known in the art.
  • the LC library is captured in a phage display vector following techniques known in the art and then transferred to pTQ6 as a ApaLI/AscI fragment and transformed into E. coli , producing a library of approximately 1 ⁇ 10 8 .
  • the library is then transformed into yeast strain W303, producing library W303 pTQ6-LC*rep of approximately 1 ⁇ 10 7 .
  • Fab diploid
  • yeast display library two (haploid) host cell populations; one population containing a repertoire of light chain fragments and the second population containing a repertoire of heavy chain fragments, are co-cultured under conditions sufficient to permit cellular fusion and the resulting diploid population grown under conditions sufficient to permit expression and display of the recombined Fab (LC/HC) library.
  • Example 18 Approximately 10 10 EBY100 pTQ5-HC* rep yeast cells (from Example 26) are mated with approximately 10 10 W303 pTQ6-LC* rep yeast cells (from Example 22) following the procedures outlined in Example 18. Ten percent mating efficiency results in an approximately 10 9 diploid repertoire (thus capturing approximately 109 LC/HC combinations of the possible maximum 10 14 combinatorial LC/HC diversity, given the starting diversity of the individual component LC and HC repertoires in the haploid parents). The diploid repertoire is cultured and expression of LC and HC induced (Example 15). The diploid repertoire is cultured and expression of LC and HC induced (see Example 15).
  • the diploid culture is incubated with streptavidin-FITC and affinity selected using flow cytometric sorting (see Example 8). Affinity variants are screened by off rate determination using flow cytometry (see Example 9) and additionally by surface plasmon resonance techniques known in the art, using soluble Fab antibodies.
  • diploid Fab-displaying yeast isolates resulting from the streptavidin selection screen as described in Example 23 are induced to sporulate by culturing the isolates under conditions of nitrogen starvation (as described in Guthrie and Fink, 1991). Sporulated diploids are harvested, treated with zymolase, sonicated, and plated out on rich plates.
  • Haploid colonies are separated into two subsamples; one subsample is grown under conditions to facilitate loss of the LC expression vector but selected for the HC display vector, the second subsample is grown under conditions to facilitate loss of the HC display vector but selected for the LC expression vector (for 2 ⁇ derived plasmids under non selective conditions, plasmid loss is between 2-6% per generation).
  • each yeast subculture is effectively purged of non-selected chain expressing vector and contains only the selected (LC or HC) expression vector, thus producing two biased (i.e., pre-selected) single chain expression haploid yeast cells, designated “HAPLOID pTQ6-LC* sel ” and “HAPLOID pTQ5-HC* sel ”. From these two haploid yeast populations, each containing either the light chain of pre-selected Fabs or the heavy chain of pre-selected Fabs, three mating regimes are established as follows:
  • the resulting diploid culture represents a biased repertoire containing unique combinations of the original HC repertoire against the preselected LC repertoire, which can be further screened by, e.g., flow cytometric sorting (Examples 8 and 11) and/or surface plasmon resonance techniques known in the art, using soluble Fab antibodies.
  • the resulting diploid culture represents a biased repertoire containing unique combinations of the original LC repertoire against the preselected HC repertoire, which can be further screened by, e.g., flow cytometric sorting (Examples 8 and 11) and/or surface plasmon resonance techniques known in the art, using soluble Fab antibodies.
  • the resulting diploid culture represents a biased recombination repertoire containing unique combinations of the preselected LC repertoire against the preselected HC repertoire, which can be further screened by, e.g., flow cytometric sorting (Examples 8 and 11) and/or surface plasmon resonance techniques known in the art, using soluble Fab antibodies.
  • a Fab antibody library is prepared from a lead target specific Fab where either the whole LC or a fragment of the HC is diversified using restriction-based cloning.
  • an antibody library constructed with restriction sites both bracketing the antibody V gene sequence and also internal to the V gene sequence is used to prepare a plurality of antibody gene fragments for cloning and thus leading to the diversification of the lead antibody.
  • Antibodies isolated from one such antibody library can be affinity matured by this approach.
  • Antibodies comprising, for example, the CJ phagemid library have a LC bracketed by a unique ApaL1 and AscI restriction site and a HC bracketed by a unique SfiI and NotI restriction site.
  • the HC also contains an internal and unique XbaI restriction site between the CDR2 and CDR3 sequence.
  • the Fab antibody gene(s) are first cloned into the yeast display vector pTQ3 as in Example 2, resulting in pTQ3-Fab.
  • a plurality of LC are isolated from a DNA preparation of the CJ phagemid library by restriction digestion with ApaL1 and AscI restriction enzymes.
  • pTQ3-Fab is also digested with ApaL1 and AscI, and the endogenous LC is replaced by a plurality of LC giving rise to a repertoire pTQ3-LC cj-rep .
  • This repertoire is then transferred into yeast strain EBY100 to give EBY 100 pTQ3-LC cj-rep .
  • V H CDR1-2 To diversify the V H CDR1-2 in either an antigen specific lead antibody or a pool of antigen specific lead antibodies first the Fab antibody gene(s) are cloned into the yeast display vector pTQ3 as in Example 2 to give pTQ3-Fab.
  • a plurality of V H CDR1-2 fragments are isolated from the CJ phagemid library by restriction digestion with SfiI and XbaI.
  • pTQ3-Fab is also digested with SfiI and XbaI, and the endogenous V H CDR1-2 fragment is replaced by a plurality of V H CDR1-2 fragments, resulting in the repertoire pTQ3-V H CDR1-2 cj-rep .
  • This repertoire is then transferred into yeast strain EBY100 to give EBY100 pTQ3-V H CDR1-2C cj-rep .
  • the cloning procedure can be performed in a number of different ways, e.g., by first constructing a repertoire of V H CDR1-2 fragments and then cloning in the antigen specific V H CDR3 or pool of V H CDR3s.
  • a culture of EBY100 pTQ3-V H CDR1-2c cj-rep and EBY100 pTQ3-LC cj-rep is prepared as in Example 2.
  • the yeast culture is then labeled for LC display and antigen binding and affinity selected by flow cytometric sorting as in Example 10. Selected clones are then analyzed for their DNA sequence and there improvement in affinity as in Example 10.
  • yeast mating can be used for combinatorial gene diversification and affinity maturation of an antigen specific lead antibody or antigen specific lead antibodies
  • a selected LC or pool of LCs is rediversified or a V H CDR1-2 fragment of a selected HC or pool of HCs is rediversified.
  • Antibodies comprising the CJ phagemid library are amenable to such an approach. They have a LC bracketed by a unique ApaL1 and AscI restriction site and a HC bracketed by a unique SfiI and NotI restriction site.
  • the HC also contains an internal and unique XbaI restriction site between the CDR2 and CDR3 sequence.
  • yeast mating is used to bring together antigen specific LC with a plurality of V H CDR1-2 fragments or antigen specific HC with a plurality of LC, thus eliminating the need for restriction-based cloning to pair a LC with a HC.
  • the component HC antibody genes are cloned into the yeast display vector pTQ5 as in Example 13 to give pTQ5-HCAg.
  • a plurality of V H CDR1-2 fragments are prepared by digestion of HC fragments from the CJ phagemid library with SfiI and XbaI restriction enzymes.
  • This plurality of V H CDR1-2 fragments is then cloned into DNA prepared from pTQ5-HCAg, which has been digested with SfiI and XbaI to remove the endogenous V H CDR1-2 fragment and to replace with the plurality of V H CDR1-2 fragments, the antigen specific V H -CDR3 being retained.
  • a plurality of LC are isolated from a DNA preparation of the CJ phagemid library by restriction digestion with ApaL1 and AscI.
  • This plurality of LC is cloned into pTQ6 to give a repertoire pTQ6-LC rep and serves as one master repertoire for affinity maturation of other antibodies specific for other targets.
  • This repertoire is then transferred into a yeast strain of the opposite mating type, BJ5457, to give BJ5457 pTQ6-LC rep .

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Organic Chemistry (AREA)
  • Biomedical Technology (AREA)
  • Molecular Biology (AREA)
  • Biotechnology (AREA)
  • Microbiology (AREA)
  • General Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Biochemistry (AREA)
  • Physics & Mathematics (AREA)
  • General Health & Medical Sciences (AREA)
  • Immunology (AREA)
  • Biophysics (AREA)
  • Plant Pathology (AREA)
  • Medicinal Chemistry (AREA)
  • Urology & Nephrology (AREA)
  • Cell Biology (AREA)
  • Hematology (AREA)
  • Virology (AREA)
  • Crystallography & Structural Chemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Mycology (AREA)
  • Food Science & Technology (AREA)
  • Analytical Chemistry (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Peptides Or Proteins (AREA)
  • Image Generation (AREA)
  • Control Of Indicators Other Than Cathode Ray Tubes (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Error Detection And Correction (AREA)
US10/262,646 2001-10-01 2002-09-30 Multi-chain eukaryotic display vectors and uses thereof Abandoned US20030186374A1 (en)

Priority Applications (10)

Application Number Priority Date Filing Date Title
US10/262,646 US20030186374A1 (en) 2001-10-01 2002-09-30 Multi-chain eukaryotic display vectors and uses thereof
US12/625,337 US9012181B2 (en) 2001-10-01 2009-11-24 Multi-chain eukaryotic display vectors and uses thereof
US13/073,657 US9005927B2 (en) 2001-10-01 2011-03-28 Multi-chain eukaryotic display vectors and uses thereof
US13/170,362 US9040258B2 (en) 2001-10-01 2011-06-28 Multi-chain eukaryotic display vectors and uses thereof
US13/170,438 US9068980B2 (en) 2001-10-01 2011-06-28 Multi-chain eukaryotic display vectors and uses thereof
US13/170,448 US9116149B2 (en) 2001-10-01 2011-06-28 Multi-chain eukaryotic display vectors and uses thereof
US13/937,915 US9034601B2 (en) 2001-10-01 2013-07-09 Multi-chain eukaryotic display vectors and uses thereof
US14/718,656 US9556428B2 (en) 2001-10-01 2015-05-21 Multi-chain eukaryotic display vectors and uses thereof
US15/377,846 US10059936B2 (en) 2001-10-01 2016-12-13 Multi-chain eukaryotic display vectors and uses thereof
US16/042,004 US10577599B2 (en) 2001-10-01 2018-07-23 Multi-chain eukaryotic display vectors and uses thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US32632001P 2001-10-01 2001-10-01
US10/262,646 US20030186374A1 (en) 2001-10-01 2002-09-30 Multi-chain eukaryotic display vectors and uses thereof

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US12/625,337 Continuation US9012181B2 (en) 2001-10-01 2009-11-24 Multi-chain eukaryotic display vectors and uses thereof

Publications (1)

Publication Number Publication Date
US20030186374A1 true US20030186374A1 (en) 2003-10-02

Family

ID=23271723

Family Applications (10)

Application Number Title Priority Date Filing Date
US10/262,646 Abandoned US20030186374A1 (en) 2001-10-01 2002-09-30 Multi-chain eukaryotic display vectors and uses thereof
US12/625,337 Active 2025-09-30 US9012181B2 (en) 2001-10-01 2009-11-24 Multi-chain eukaryotic display vectors and uses thereof
US13/073,657 Expired - Lifetime US9005927B2 (en) 2001-10-01 2011-03-28 Multi-chain eukaryotic display vectors and uses thereof
US13/170,438 Expired - Lifetime US9068980B2 (en) 2001-10-01 2011-06-28 Multi-chain eukaryotic display vectors and uses thereof
US13/170,362 Expired - Lifetime US9040258B2 (en) 2001-10-01 2011-06-28 Multi-chain eukaryotic display vectors and uses thereof
US13/170,448 Expired - Lifetime US9116149B2 (en) 2001-10-01 2011-06-28 Multi-chain eukaryotic display vectors and uses thereof
US13/937,915 Expired - Lifetime US9034601B2 (en) 2001-10-01 2013-07-09 Multi-chain eukaryotic display vectors and uses thereof
US14/718,656 Expired - Lifetime US9556428B2 (en) 2001-10-01 2015-05-21 Multi-chain eukaryotic display vectors and uses thereof
US15/377,846 Expired - Lifetime US10059936B2 (en) 2001-10-01 2016-12-13 Multi-chain eukaryotic display vectors and uses thereof
US16/042,004 Expired - Lifetime US10577599B2 (en) 2001-10-01 2018-07-23 Multi-chain eukaryotic display vectors and uses thereof

Family Applications After (9)

Application Number Title Priority Date Filing Date
US12/625,337 Active 2025-09-30 US9012181B2 (en) 2001-10-01 2009-11-24 Multi-chain eukaryotic display vectors and uses thereof
US13/073,657 Expired - Lifetime US9005927B2 (en) 2001-10-01 2011-03-28 Multi-chain eukaryotic display vectors and uses thereof
US13/170,438 Expired - Lifetime US9068980B2 (en) 2001-10-01 2011-06-28 Multi-chain eukaryotic display vectors and uses thereof
US13/170,362 Expired - Lifetime US9040258B2 (en) 2001-10-01 2011-06-28 Multi-chain eukaryotic display vectors and uses thereof
US13/170,448 Expired - Lifetime US9116149B2 (en) 2001-10-01 2011-06-28 Multi-chain eukaryotic display vectors and uses thereof
US13/937,915 Expired - Lifetime US9034601B2 (en) 2001-10-01 2013-07-09 Multi-chain eukaryotic display vectors and uses thereof
US14/718,656 Expired - Lifetime US9556428B2 (en) 2001-10-01 2015-05-21 Multi-chain eukaryotic display vectors and uses thereof
US15/377,846 Expired - Lifetime US10059936B2 (en) 2001-10-01 2016-12-13 Multi-chain eukaryotic display vectors and uses thereof
US16/042,004 Expired - Lifetime US10577599B2 (en) 2001-10-01 2018-07-23 Multi-chain eukaryotic display vectors and uses thereof

Country Status (11)

Country Link
US (10) US20030186374A1 (fr)
EP (2) EP1438400B1 (fr)
JP (2) JP4578098B2 (fr)
AT (1) ATE434040T1 (fr)
AU (2) AU2002330162B2 (fr)
CA (1) CA2462113C (fr)
DE (1) DE60232672D1 (fr)
DK (2) DK2093286T3 (fr)
ES (2) ES2327905T3 (fr)
PT (1) PT1438400E (fr)
WO (1) WO2003029456A1 (fr)

Cited By (122)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020123057A1 (en) * 2000-11-17 2002-09-05 University Of Rochester In vitro methods of producing and identifying immunoglobulin molecules in eukaryotic cells
US20030104402A1 (en) * 2001-01-23 2003-06-05 University Of Rochester Methods of producing or identifying intrabodies in eukaryotic cells
US20040146976A1 (en) * 1996-05-31 2004-07-29 Board Of Trustees Of The University Of Illinois Yeast cell surface display of proteins and uses thereof
US20040146952A1 (en) * 1998-01-20 2004-07-29 Kranz David M. High affinity TCR proteins and methods
US20050196755A1 (en) * 2000-11-17 2005-09-08 Maurice Zauderer In vitro methods of producing and identifying immunoglobulin molecules in eukaryotic cells
WO2007024715A2 (fr) 2005-08-19 2007-03-01 Abbott Laboratories Immunoglobuline a deux domaines variables et utilisations de celle-ci
US20070111260A1 (en) * 2005-10-14 2007-05-17 Medimmune, Inc. Cell display of antibody libraries
US20080171014A1 (en) * 2006-09-08 2008-07-17 Chengbin Wu Interleukin-13 binding proteins
EP2021795A2 (fr) * 2006-05-04 2009-02-11 Abmaxis, Inc. Systèmes d'expression à la surface multispécifique et hétérospécifique
US20090163379A1 (en) * 2007-11-16 2009-06-25 Kevin Caili Wang Eukaryotic cell display systems
US20090175847A1 (en) * 2007-05-30 2009-07-09 Abbott Laboratories Humanized antibodies to ab (20-42) globulomer and uses thereof
US20090215992A1 (en) * 2005-08-19 2009-08-27 Chengbin Wu Dual variable domain immunoglobulin and uses thereof
WO2009111183A1 (fr) * 2008-03-03 2009-09-11 Glycofi, Inc. Affichage en surface de protéines recombinantes dans des eucaryotes inférieurs
US20090232801A1 (en) * 2007-05-30 2009-09-17 Abbot Laboratories Humanized Antibodies Which Bind To AB (1-42) Globulomer And Uses Thereof
EP2116556A1 (fr) 2008-05-09 2009-11-11 Abbott GmbH & Co. KG Anticorps sur récepteur de produit terminal de glycosylation et utilisations associées
WO2009149185A2 (fr) 2008-06-03 2009-12-10 Abbott Laboratories Immunoglobulines à double domaine variable et leurs utilisations
US20100009866A1 (en) * 2008-07-09 2010-01-14 Bianka Prinz Surface Display of Whole Antibodies in Eukaryotes
US20100047239A1 (en) * 2005-08-19 2010-02-25 Abbott Laboratories Dual variable domain immunoglobulin and uses thereof
US20100047245A1 (en) * 2005-06-30 2010-02-25 Lacy Susan E Il-12/p40 binding proteins
US20100216720A1 (en) * 2009-02-24 2010-08-26 Abbott Laboratories Antibodies to troponin i and methods of use thereof
US20100221179A1 (en) * 2009-01-29 2010-09-02 Abbott Laboratories IL-1 Binding Proteins
US20100233079A1 (en) * 2008-12-04 2010-09-16 Abbott Laboratories Dual Variable Domain Immunoglobulins and Uses Thereof
US20100266531A1 (en) * 2009-03-05 2010-10-21 Abbott Laboratories Il-17 binding proteins
US20110009280A1 (en) * 2001-10-01 2011-01-13 Hufton Simon E Multi-chain eukaryotic display vectors and uses thereof
WO2011025964A2 (fr) 2009-08-29 2011-03-03 Abbott Laboratories Protéines thérapeutiques se liant à dll4
US20110076752A1 (en) * 2007-02-09 2011-03-31 Medimmune, Llc Antibody library display by yeast cell plasma membrane
US20110104174A1 (en) * 2009-10-31 2011-05-05 Abbott Laboratories Antibodies to receptor for advanced glycation end products (rage) and uses thereof
WO2011058087A1 (fr) 2009-11-11 2011-05-19 Gentian As Dosage immunologique pour le dosage d'analytes apparentés d'origine différente
WO2011070045A1 (fr) 2009-12-08 2011-06-16 Abbott Gmbh & Co. Kg Anticorps monoclonaux contre la protéine rgm a destinés à être utilisés dans le traitement de la dégénérescence de la couche de fibres nerveuses rétiniennes
US20110165063A1 (en) * 2009-01-29 2011-07-07 Abbott Laboratories Il-1 binding proteins
US20110183402A1 (en) * 2003-10-22 2011-07-28 Cregg James M Methods of synthesizing heteromultimeric polypeptides in yeast using a haploid mating strategy
WO2011130377A2 (fr) 2010-04-15 2011-10-20 Abbott Laboratories Protéines de liaison à la bêta amyloïde
EP2385070A1 (fr) 2003-11-12 2011-11-09 Abbott Laboratories Protéines se liant à IL-18
WO2011143562A2 (fr) 2010-05-14 2011-11-17 Abbott Laboratories Protéines liant l'il-1
WO2012006500A2 (fr) 2010-07-08 2012-01-12 Abbott Laboratories Anticorps monoclonaux contre la protéine de capside du virus de l'hépatite c
WO2012024650A2 (fr) 2010-08-19 2012-02-23 Abbott Laboratories Anticorps anti-ngf et leur utilisation
WO2012088094A2 (fr) 2010-12-21 2012-06-28 Abbott Laboratories Protéines liant l'il-1
WO2012121775A2 (fr) 2010-12-21 2012-09-13 Abbott Laboratories Immunoglobulines à double domaine variable et leurs utilisations
EP2500356A2 (fr) 2005-08-19 2012-09-19 Abbott Laboratories Immunoglobuline à double domaine variable et ses utilisations
US20130096281A1 (en) * 2010-01-21 2013-04-18 Oxyrane Uk Limited Methods and compositions for displaying a polypeptide on a yeast cell surface
WO2013063114A1 (fr) 2011-10-24 2013-05-02 Abbvie Inc. Agents de liaison immunologique dirigés contre le tnf
WO2013063095A1 (fr) 2011-10-24 2013-05-02 Abbvie Inc. Agents de liaison immunologique dirigés contre la sclérostine
WO2013078377A1 (fr) 2011-11-23 2013-05-30 Igenica, Inc. Anticorps anti-cd98 et leurs procédés d'utilisation
US8455626B2 (en) 2006-11-30 2013-06-04 Abbott Laboratories Aβ conformer selective anti-aβ globulomer monoclonal antibodies
WO2013090635A2 (fr) 2011-12-14 2013-06-20 AbbVie Deutschland GmbH & Co. KG Composition et méthode pour le diagnostic et le traitement de troubles liés au fer
WO2013090633A2 (fr) 2011-12-14 2013-06-20 AbbVie Deutschland GmbH & Co. KG Composition et méthode pour le diagnostic et le traitement de troubles liés au fer
US8497072B2 (en) 2005-11-30 2013-07-30 Abbott Laboratories Amyloid-beta globulomer antibodies
WO2013112922A1 (fr) 2012-01-27 2013-08-01 AbbVie Deutschland GmbH & Co. KG Composition et méthode pour le diagnostic et le traitement de maladies associées à la dégénérescence des neurites
US8586714B2 (en) 2009-09-01 2013-11-19 Abbvie, Inc. Dual variable domain immunoglobulins and uses thereof
US8680239B2 (en) 2000-12-22 2014-03-25 Max-Planck-Gesellschaft Zur Foerderung Der Wissenschaften E.V. Use of RGM and its modulators
US8691224B2 (en) 2005-11-30 2014-04-08 Abbvie Inc. Anti-Aβ globulomer 5F7 antibodies
WO2014059028A1 (fr) 2012-10-09 2014-04-17 Igenica, Inc. Anticorps anti-c16orf54 et leurs méthodes d'utilisation
US8716450B2 (en) 2009-10-15 2014-05-06 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
US8722855B2 (en) 2009-10-28 2014-05-13 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
US8735546B2 (en) 2010-08-03 2014-05-27 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
WO2014100542A1 (fr) 2012-12-21 2014-06-26 Abbvie, Inc. Humanisation d'anticorps à haut rendement
US8822645B2 (en) 2008-07-08 2014-09-02 Abbvie Inc. Prostaglandin E2 dual variable domain immunoglobulins and uses thereof
WO2014144355A2 (fr) 2013-03-15 2014-09-18 Abbott Laboratories Anticorps monoclonaux anti-gp73 et leurs procédés d'obtention
US8895004B2 (en) 2007-02-27 2014-11-25 AbbVie Deutschland GmbH & Co. KG Method for the treatment of amyloidoses
US8906864B2 (en) 2005-09-30 2014-12-09 AbbVie Deutschland GmbH & Co. KG Binding domains of proteins of the repulsive guidance molecule (RGM) protein family and functional fragments thereof, and their use
EP2810654A1 (fr) 2008-07-08 2014-12-10 AbbVie Inc. Protéines de liaison à la protéines de liaison à la prostaglandine E2 et leurs utilisations
US8962803B2 (en) 2008-02-29 2015-02-24 AbbVie Deutschland GmbH & Co. KG Antibodies against the RGM A protein and uses thereof
US8987418B2 (en) 2013-03-15 2015-03-24 Abbvie Inc. Dual specific binding proteins directed against IL-1β and/or IL-17
US9029508B2 (en) 2008-04-29 2015-05-12 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
US9035027B2 (en) 2008-06-03 2015-05-19 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
US9046513B2 (en) 2010-08-26 2015-06-02 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
US9045551B2 (en) 2012-11-01 2015-06-02 Abbvie Inc. Anti-DLL4/VEGF dual variable domain immunoglobulin and uses thereof
US9062101B2 (en) 2010-08-14 2015-06-23 AbbVie Deutschland GmbH & Co. KG Amyloid-beta binding proteins
WO2015120058A2 (fr) 2014-02-05 2015-08-13 Molecular Templates, Inc. Procédés de criblage, de sélection et d'identification de polypeptides de recombinaison cytotoxiques fondés sur une diminution provisoire de la ribotoxicité
US9115195B2 (en) 2010-03-02 2015-08-25 Abbvie Inc. Therapeutic DLL4 binding proteins
US9120870B2 (en) 2011-12-30 2015-09-01 Abbvie Inc. Dual specific binding proteins directed against IL-13 and IL-17
EP2921177A2 (fr) 2010-07-09 2015-09-23 AbbVie Inc. Immunoglobulines à double domaine variable et utilisations de celles-ci
US9176150B2 (en) 2003-01-31 2015-11-03 AbbVie Deutschland GmbH & Co. KG Amyloid beta(1-42) oligomers, derivatives thereof and antibodies thereto, methods of preparation thereof and use thereof
US9206408B2 (en) 2007-04-03 2015-12-08 Oxyrane Uk Limited Microorganisms genetically engineered to have modified N-glycosylation activity
US9249399B2 (en) 2012-03-15 2016-02-02 Oxyrane Uk Limited Methods and materials for treatment of pompe's disease
US9347050B2 (en) 2010-09-29 2016-05-24 Oxyrane Uk Limited Mannosidases capable of uncapping mannose-1-phospho-6-mannose linkages and demannosylating phosphorylated N-glycans and methods of facilitating mammalian cellular uptake of glycoproteins
US9371374B2 (en) 2013-03-14 2016-06-21 Abbott Laboratories HCV core lipid binding domain monoclonal antibodies
US9598682B2 (en) 2009-09-29 2017-03-21 Vib Vzw Hydrolysis of mannose-1-phospho-6-mannose linkage to phospho-6-mannose
US9645146B2 (en) 2007-03-26 2017-05-09 Agenus, Inc. Cell surface display, screening and production of proteins of interest
US9670276B2 (en) 2012-07-12 2017-06-06 Abbvie Inc. IL-1 binding proteins
US9689015B2 (en) 2010-09-29 2017-06-27 Oxyrane Uk Limited De-mannosylation of phosphorylated N-glycans
US9790478B2 (en) 2013-03-14 2017-10-17 Abbott Laboratories HCV NS3 recombinant antigens and mutants thereof for improved antibody detection
US9840554B2 (en) 2015-06-15 2017-12-12 Abbvie Inc. Antibodies against platelet-derived growth factor (PDGF)
US9841427B2 (en) 2013-03-14 2017-12-12 Abbott Laboratories HCV antigen-antibody combination assay and methods and compositions for use therein
WO2018067474A1 (fr) 2016-10-03 2018-04-12 Abbott Laboratories Procédés améliorés d'évaluation de l'état de gfap dans des échantillons de patient
WO2018175942A1 (fr) 2017-03-23 2018-09-27 Abbott Laboratories Méthodes d'aide au diagnostic et à la détermination de l'étendue d'une lésion cérébrale traumatique chez un sujet humain à l'aide du biomarqueur précoce hydrolase carboxy-terminale d'ubiquitine l1
US10093733B2 (en) 2014-12-11 2018-10-09 Abbvie Inc. LRP-8 binding dual variable domain immunoglobulin proteins
WO2018191531A1 (fr) 2017-04-15 2018-10-18 Abbott Laboratories Procédés d'aide au diagnostic hyperaigu et de détermination d'une lésion cérébrale traumatique chez un sujet humain à l'aide de biomarqueurs précoces
WO2018200823A1 (fr) 2017-04-28 2018-11-01 Abbott Laboratories Méthodes d'aide au diagnostic et à la détermination hyperaigus d'une lésion cérébrale traumatique au moyen de biomarqueurs précoces sur au moins deux échantillons provenant du même sujet humain
WO2018218169A1 (fr) 2017-05-25 2018-11-29 Abbott Laboratories Procédés d'aide à la détermination de la réalisation ou non d'une imagerie sur un sujet humain ayant subi ou susceptible d'avoir subi une lésion à la tête à l'aide de biomarqueurs précoces
WO2018222784A1 (fr) 2017-05-30 2018-12-06 Abbott Laboratories Procédés d'aide au diagnostic et à l'évaluation d'une lésion cérébrale traumatique légère chez un patient humain à l'aide de troponine cardiaque i
WO2019010131A1 (fr) 2017-07-03 2019-01-10 Abbott Laboratories Procédés améliorés de mesure de niveaux d'hydrolase à terminaison carboxy d'ubiquitine l1 dans le sang
US10183988B2 (en) 2013-06-07 2019-01-22 Duke University Anti-Complement factor H antibodies
US10287557B2 (en) 2009-11-19 2019-05-14 Oxyrane Uk Limited Yeast strains producing mammalian-like complex N-glycans
WO2019112860A1 (fr) 2017-12-09 2019-06-13 Abbott Laboratories Procédés d'aide au diagnostic et à l'évaluation d'un lésion cérébrale traumatique chez un sujet humain au moyen d'une combinaison de gfap et d'uch-l1
WO2019113525A2 (fr) 2017-12-09 2019-06-13 Abbott Laboratories Procédés d'aide au diagnostic et à l'évaluation d'un sujet qui a subi une lésion orthopédique et qui a subi ou peut avoir subi une lésion à la tête, telle qu'une lésion cérébrale traumatique légère (tbi), à l'aide d'une protéine acide fibrillaire gliale (gfap) et/ou d'hydrolase carboxy-terminale d'ubiquitine l1 (uch-l1)
US10428145B2 (en) 2015-09-29 2019-10-01 Celgene Corporation PD-1 binding proteins and methods of use thereof
EP3574919A1 (fr) 2011-07-13 2019-12-04 AbbVie Inc. Procédés et compositions de traitement de l'asthme à l'aide d'anticorps anti-il-13
EP3626744A1 (fr) 2015-05-29 2020-03-25 AbbVie Inc. Anticorps anti-cd40 et leurs utilisations
WO2020065594A1 (fr) 2018-09-28 2020-04-02 Kyowa Kirin Co., Ltd. Anticorps il-36 et leurs utilisations
US10751414B2 (en) 2016-09-19 2020-08-25 Celgene Corporation Methods of treating psoriasis using PD-1 binding antibodies
US10766958B2 (en) 2016-09-19 2020-09-08 Celgene Corporation Methods of treating vitiligo using PD-1 binding antibodies
EP3738981A1 (fr) 2014-01-24 2020-11-18 NGM Biopharmaceuticals, Inc. Anticorps liant beta-klotho 2 et leurs procédés d'utilisation
US10865238B1 (en) 2017-05-05 2020-12-15 Duke University Complement factor H antibodies
WO2021159024A1 (fr) 2020-02-05 2021-08-12 Larimar Therapeutics, Inc. Protéines de liaison au peptide tat et leurs utilisations
WO2021211331A1 (fr) 2020-04-13 2021-10-21 Abbott Point Of Care Inc. Procédés, complexes et kits pour détecter ou déterminer une quantité d'un anticorps anti-ss-coronavirus dans un échantillon
WO2022031804A1 (fr) 2020-08-04 2022-02-10 Abbott Laboratories Méthodes et kits améliorés pour détecter une protéine sars-cov-2 dans un échantillon
WO2022119841A1 (fr) 2020-12-01 2022-06-09 Abbott Laboratories Utilisation d'un ou plusieurs biomarqueurs pour déterminer un traumatisme crânien (tbi) chez un sujet soumis à un balayage de tomodensitométrie assistée par ordinateur de la tête à tbi négatif
WO2022147147A1 (fr) 2020-12-30 2022-07-07 Abbott Laboratories Procédés pour déterminer un antigène sras-cov-2 et anticorps anti-sras-cov-2 dans un échantillon
WO2022147463A2 (fr) 2020-12-31 2022-07-07 Alamar Biosciences, Inc. Molécules de liant ayant une affinité et/ou une spécificité élevées et leurs procédés de fabrication et d'utilisation
WO2022245920A1 (fr) 2021-05-18 2022-11-24 Abbott Laboratories Procédés d'évaluation d'une lésion cérébrale chez un sujet en pédiatrie
WO2022266034A1 (fr) 2021-06-14 2022-12-22 Abbott Laboratories Procédés de diagnostic ou d'aide au diagnostic d'une lésion cérébrale provoquée par de l'énergie acoustique, de l'énergie électromagnétique, une onde de surpression et/ou le souffle d'une explosion
WO2023034777A1 (fr) 2021-08-31 2023-03-09 Abbott Laboratories Méthodes et systèmes de diagnostic de lésion cérébrale
WO2023056268A1 (fr) 2021-09-30 2023-04-06 Abbott Laboratories Méthodes et systèmes de diagnostic de lésion cérébrale
WO2023102384A1 (fr) 2021-11-30 2023-06-08 Abbott Laboratories Utilisation d'un ou de plusieurs biomarqueurs pour déterminer un traumatisme crânien (tbi) chez un sujet ayant été soumis à un balayage de tomodensitométrie assistée par ordinateur de la tête ne démontrant par de tbi
WO2023114978A1 (fr) 2021-12-17 2023-06-22 Abbott Laboratories Systèmes et procédés de détermination d'uch-l1, de gfap et d'autres biomarqueurs dans des échantillons de sang
WO2023129942A1 (fr) 2021-12-28 2023-07-06 Abbott Laboratories Utilisation de biomarqueurs pour déterminer une lésion cérébrale traumatique (tbi) subaiguë chez un sujet ayant subi un tomodensitogramme (ct) de la tête négatif pour une tbi ou n'ayant pas subi de tomodensitogramme
WO2023131901A1 (fr) 2022-01-07 2023-07-13 Johnson & Johnson Enterprise Innovation Inc. Matériaux et procédés de protéines de liaison à il-1beta
WO2023150652A1 (fr) 2022-02-04 2023-08-10 Abbott Laboratories Procédés d'écoulement latéral, dosages et dispositifs de détection de la présence ou de mesure de la quantité d'ubiquitine carboxy-terminal hydrolase l1 et/ou d'une protéine gliofibrillaire acide dans un échantillon
WO2024006876A1 (fr) 2022-06-29 2024-01-04 Abbott Laboratories Systèmes et analyses magnétiques hors laboratoire pour déterminer une gfap dans des échantillons biologiques
WO2024013727A1 (fr) 2022-07-15 2024-01-18 Janssen Biotech, Inc. Matériau et procédés d'appariement amélioré par génie biologique de régions variables de liaison à l'antigène
WO2024059708A1 (fr) 2022-09-15 2024-03-21 Abbott Laboratories Biomarqueurs et méthodes de différenciation entre une lésion cérébrale traumatique légère et très légère

Families Citing this family (67)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8288322B2 (en) 2000-04-17 2012-10-16 Dyax Corp. Methods of constructing libraries comprising displayed and/or expressed members of a diverse family of peptides, polypeptides or proteins and the novel libraries
US20090137416A1 (en) 2001-01-16 2009-05-28 Regeneron Pharmaceuticals, Inc. Isolating Cells Expressing Secreted Proteins
US20040067532A1 (en) * 2002-08-12 2004-04-08 Genetastix Corporation High throughput generation and affinity maturation of humanized antibody
EP1587907B1 (fr) 2003-01-07 2011-03-02 Dyax Corporation Librairie du domaine de kunitz
US7884054B2 (en) 2003-12-22 2011-02-08 Amgen Inc. Methods for identifying functional antibodies
ES2320374T3 (es) 2005-01-05 2009-05-21 F-Star Biotechnologische Forschungs- Und Entwicklungsges.M.B.H. Dominios de inmunoglobulina sintetica con propiedades de enlace modificadas en regiones de la molecula diferentes de las regiones de determinacion de complementariedad.
US7845537B2 (en) 2006-01-31 2010-12-07 Ethicon Endo-Surgery, Inc. Surgical instrument having recording capabilities
US8992422B2 (en) 2006-03-23 2015-03-31 Ethicon Endo-Surgery, Inc. Robotically-controlled endoscopic accessory channel
AT503889B1 (de) 2006-07-05 2011-12-15 Star Biotechnologische Forschungs Und Entwicklungsges M B H F Multivalente immunglobuline
SG175602A1 (en) 2006-07-05 2011-11-28 Catalyst Biosciences Inc Protease screening methods and proteases identified thereby
WO2008070367A2 (fr) 2006-11-01 2008-06-12 Facet Biotech Corporation Banques d'affichage d'immunoglobine à base de cellule de mammifère
ES2627223T3 (es) 2007-06-26 2017-07-27 F-Star Biotechnologische Forschungs- Und Entwicklungsges.M.B.H Presentación de agentes de unión
RU2010102859A (ru) 2007-08-21 2011-09-27 Морфосис Аг (De) Улучшенные способы образования дисульфидных связей
JP5933894B2 (ja) 2007-09-14 2016-06-15 アディマブ, エルエルシー 合理的に設計された、合成抗体ライブラリおよびその使用
US8877688B2 (en) 2007-09-14 2014-11-04 Adimab, Llc Rationally designed, synthetic antibody libraries and uses therefor
WO2009059235A2 (fr) 2007-11-01 2009-05-07 Facet Biotech Corporation Vecteurs de présentation d'immunoglobulines
WO2009075773A2 (fr) 2007-12-07 2009-06-18 Goldstein Steven A Identification de ligands de toxine
EP2235181A1 (fr) * 2008-01-11 2010-10-06 MorphoSys AG Vecteurs et procédés d'affichage et leurs utilisations
EP2098536A1 (fr) 2008-03-05 2009-09-09 4-Antibody AG Isolation et identification de protéines de liaison spécifiques à des antigènes ou à des ligands
WO2009114815A1 (fr) 2008-03-13 2009-09-17 Dyax Corp Bibliothèques de boîtiers génétiques comprenant de nouvelles conceptions de hc cdr3
AU2009240481B2 (en) 2008-04-24 2015-07-30 Takeda Pharmaceutical Company Limited Libraries of genetic packages comprising novel HC CDR1, CDR2, and CDR3 and novel LC CDR1, CDR2, and CDR3 designs
AU2009238605B2 (en) 2008-04-25 2014-11-06 Biogen Idec Hemophilia Inc. Antibodies against FcRn and use thereof
EP2113255A1 (fr) 2008-05-02 2009-11-04 f-star Biotechnologische Forschungs- und Entwicklungsges.m.b.H. Immunoglobuline cytotoxique
WO2010040073A1 (fr) 2008-10-03 2010-04-08 Xoma Technology Ltd. Nouvelles séquences de triple étiquette et procédés d’utilisation de celles-ci
EP2349329A4 (fr) 2008-10-14 2012-10-31 Dyax Corp Utilisation de la liaison à l igf-ii/igf-iie pour le traitement et la prévention de la fibrose pulmonaire associée à la sclérodermie généralisée
CN102245771B (zh) * 2008-12-16 2013-04-10 诺瓦提斯公司 酵母展示系统
WO2010079189A1 (fr) * 2009-01-09 2010-07-15 Morphosys Ag Vecteurs d'exposition et procédés et utilisations associés
RU2569187C2 (ru) 2009-05-29 2015-11-20 МорфоСис АГ Коллекция и способы ее применения
EP2478136A4 (fr) 2009-09-14 2013-09-25 Dyax Corp Bibliothèques d'ensembles génétiques comprenant de nouvelles conceptions de hc cdr3
US20110189183A1 (en) 2009-09-18 2011-08-04 Robert Anthony Williamson Antibodies against candida, collections thereof and methods of use
PT2521568T (pt) 2010-01-06 2018-10-26 Dyax Corp Proteínas de ligação à calicreína plasmática
WO2011092233A1 (fr) 2010-01-29 2011-08-04 Novartis Ag Conjugaison de levures pour produire des combinaisons de liants à base de fibronectine à haute affinité
JP6266343B2 (ja) 2010-07-16 2018-01-24 アディマブ, エルエルシー 抗体ライブラリー
WO2012052391A1 (fr) 2010-10-19 2012-04-26 Glaxo Group Limited Polypeptide ayant l'activité catalytique de jmjd3
ES2696548T3 (es) 2010-11-19 2019-01-16 Morphosys Ag Una colección de secuencias de anticuerpos y su uso
BR112013030352B1 (pt) 2011-06-02 2020-05-19 Dyax Corp anticorpo anti-fcrn isolado, composição farmacêutica que compreende o dito anticorpo, ácido nucleico isolado, vetor, célula e uso terapêutico do dito anticorpo
EP2644693A1 (fr) * 2012-03-29 2013-10-02 IP Bewertungs AG (IPB) Polypeptide biologiquement actif exprimé sur la surface d'une cellule
WO2013184871A1 (fr) 2012-06-06 2013-12-12 Zoetis Llc Anticorps anti-ngf caninisés et procédés associés
TWI675044B (zh) 2012-11-14 2019-10-21 美商再生元醫藥公司 重組細胞表面捕捉蛋白質
WO2014152013A1 (fr) 2013-03-14 2014-09-25 The Trustees Of Columbia University In The City Of New York 4-phénylpipéridines, leur préparation et leur utilisation
MX2016008076A (es) 2013-12-19 2016-08-12 Novartis Ag Receptores quimericos de antigeno de mesotelina humana y uso de los mismos.
WO2015153969A1 (fr) 2014-04-04 2015-10-08 The Board Of Trustees Of The Leland Stanford Junior University Découverte de ligands pour les récepteurs de cellule t
BR112017001242A2 (pt) 2014-07-21 2017-12-05 Novartis Ag tratamento de câncer usando um receptor antigênico quimérico a cd33
CA2958200A1 (fr) 2014-08-14 2016-02-18 Novartis Ag Traitement du cancer a l'aide du recepteur d'antigene chimerique gfr alpha-4
BR112017003104A2 (pt) 2014-08-19 2017-12-05 Novartis Ag tratamento de câncer usando um receptor antigênico quimérico anti-cd123
US11384354B2 (en) 2015-12-21 2022-07-12 Zumutor Biologics Inc. Method of generating an antibody naïve library, said library and application(s) thereof
RU2683549C1 (ru) * 2015-12-29 2019-03-28 Федеральное государственное бюджетное учреждение науки институт биоорганической химии им. академиков М.М. Шемякина и Ю.А. Овчинникова Российской академии наук (ИБХ РАН) СИСТЕМА ДЛЯ ЭКСПРЕССИИ FAB-ФРАГМЕНТОВ АНТИТЕЛ В МЕТИЛОТРОФНЫХ ДРОЖЖАХ PICHIAPASTORIS, НА ОСНОВЕ РЕКОМБИНАНТНЫХ ПЛАЗМИДНЫХ ДНК Ab-HCh-HIS/pPICZ_α_A И Ab-LCh-LAMBDA/pPICZα_A, ПРЕДНАЗНАЧЕННЫХ ДЛЯ КЛОНИРОВАНИЯ ВАРИАБЕЛЬНЫХ ДОМЕНОВ ТЯЖЕЛОЙ И ЛЕГКОЙ ЦЕПЕЙ АНТИТЕЛ, СООТВЕТСТВЕННО
US10988759B2 (en) 2016-01-15 2021-04-27 University Of Washington High throughput protein-protein interaction screening in yeast liquid culture
US20170205421A1 (en) * 2016-01-15 2017-07-20 University Of Washington Synthetic yeast agglutination
EP3484455A2 (fr) 2016-07-15 2019-05-22 Novartis AG Traitement et prévention du syndrome de libération de cytokine à l'aide d'un récepteur d'antigène chimérique en combinaison avec un inhibiteur de kinase
MX2019003886A (es) 2016-10-07 2019-08-05 Novartis Ag Receptores de antigenos quimericos para el tratamiento del cancer.
US10550429B2 (en) 2016-12-22 2020-02-04 10X Genomics, Inc. Methods and systems for processing polynucleotides
CN110290811A (zh) 2017-01-13 2019-09-27 中央研究院 用以治疗脑部疾病的可重复装载的水胶系统
CN110312530A (zh) 2017-01-13 2019-10-08 中央研究院 用以治疗心肌梗塞的可重复装载的水胶系统
AU2018298884A1 (en) 2017-07-14 2020-02-27 Immatics Biotechnologies Gmbh Improved dual specificity polypeptide molecule
AU2018344859A1 (en) 2017-10-04 2020-04-30 Hesperix SA Articles and methods directed to personalized therapy of cancer
EP3752832A1 (fr) 2018-02-12 2020-12-23 10X Genomics, Inc. Procédés de caractérisation d'analytes multiples à partir de cellules individuelles ou de populations cellulaires
JP2021517461A (ja) 2018-03-12 2021-07-26 ゾエティス・サービシーズ・エルエルシー 抗ngf抗体およびその方法
JP2021524282A (ja) * 2018-05-17 2021-09-13 ザ ボード オブ トラスティーズ オブ ザ レランド スタンフォード ジュニア ユニバーシティー ホスファターゼ動員による受容体の阻害
WO2020061389A1 (fr) * 2018-09-20 2020-03-26 Washington University Microorganismes modifiés et procédés de fabrication et d'utilisation associés
WO2020216191A1 (fr) * 2019-04-22 2020-10-29 泷搌(上海)生物科技有限公司 Procédé de préparation d'une bibliothèque de phages
IL298006A (en) 2020-05-11 2023-01-01 A Alpha Bio Inc High-throughput screening methods for identifying small molecule targets
US20220251543A1 (en) 2020-06-01 2022-08-11 A-Alpha Bio, Inc. Methods for characterizing and engineering protein-protein interactions
CN112646833A (zh) * 2020-11-05 2021-04-13 杭州阿诺生物医药科技有限公司 一种全人源抗体酵母展示技术的设计与构建
JP2024518378A (ja) 2021-05-05 2024-05-01 イマティクス バイオテクノロジーズ ゲーエムベーハー Prameに特異的に結合する抗原結合タンパク質
EP4144898A1 (fr) * 2021-09-07 2023-03-08 New/era/mabs GmbH Procédé de sélection ou de dépistage des anticorps à partir d'une bibliothèque d'anticorps
ES2955461A1 (es) * 2022-05-24 2023-12-01 Univ Valencia Politecnica Levadura y plasmido para la exposicion de proteinas en superficie

Citations (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4816397A (en) * 1983-03-25 1989-03-28 Celltech, Limited Multichain polypeptides or proteins and processes for their production
US5223409A (en) * 1988-09-02 1993-06-29 Protein Engineering Corp. Directed evolution of novel binding proteins
US5354847A (en) * 1986-10-27 1994-10-11 Bristol-Myers Squibb Company Chimeric antibody with specificity to human tumor antigen
US5643745A (en) * 1994-02-03 1997-07-01 University Of Hawaii Heterologous dimeric proteins produced in heterokaryons
US5658727A (en) * 1991-04-10 1997-08-19 The Scripps Research Institute Heterodimeric receptor libraries using phagemids
US5695941A (en) * 1994-06-22 1997-12-09 The General Hospital Corporation Interaction trap systems for analysis of protein networks
US5723323A (en) * 1985-03-30 1998-03-03 Kauffman; Stuart Alan Method of identifying a stochastically-generated peptide, polypeptide, or protein having ligand binding property and compositions thereof
US5789208A (en) * 1994-01-31 1998-08-04 The Trustees Of Boston University Polyclonal antibody libraries
US5888732A (en) * 1995-06-07 1999-03-30 Life Technologies, Inc. Recombinational cloning using engineered recombination sites
US6074853A (en) * 1997-03-21 2000-06-13 Sri Sequence alterations using homologous recombination
US6132970A (en) * 1994-02-17 2000-10-17 Maxygen, Inc. Methods of shuffling polynucleotides
US6207442B1 (en) * 1997-10-16 2001-03-27 Zymogenetics, Inc. Plasmid construction by homologous recombination
US6291242B1 (en) * 1994-02-17 2001-09-18 Maxygen, Inc. Methods for generating polynucleotides having desired characteristics by iterative selection and recombination
US6300065B1 (en) * 1996-05-31 2001-10-09 Board Of Trustees Of The University Of Illinois Yeast cell surface display of proteins and uses thereof
US6461863B1 (en) * 1996-08-16 2002-10-08 University Of Wyoming Modifying insect cell gylcosylation pathways with baculovirus expression vectors

Family Cites Families (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4946778A (en) 1987-09-21 1990-08-07 Genex Corporation Single polypeptide chain binding molecules
AU685057B2 (en) 1992-07-08 1998-01-15 Unilever Plc Process for immobilizing enzymes to the cell wall of a microbial cell by producing a fusion protein
ATE253118T1 (de) 1993-02-10 2003-11-15 Unilever Nv Isolierungsverfahren verwendend immobilisierte proteine mit spezifischen bindungskapazitäten
US20010041333A1 (en) 1997-06-16 2001-11-15 Short Jay M. High throughput screening for a bioactivity or biomolecule
US6696251B1 (en) 1996-05-31 2004-02-24 Board Of Trustees Of The University Of Illinois Yeast cell surface display of proteins and uses thereof
US6699658B1 (en) 1996-05-31 2004-03-02 Board Of Trustees Of The University Of Illinois Yeast cell surface display of proteins and uses thereof
US5895941A (en) * 1996-07-01 1999-04-20 Mitsubishi Denki Kabushiki Kaisha Field effect transistor with electrode portions under T-shaped gate structure
US5869250A (en) 1996-12-02 1999-02-09 The University Of North Carolina At Chapel Hill Method for the identification of peptides that recognize specific DNA sequences
WO1999028502A1 (fr) 1997-11-28 1999-06-10 Invitrogen Corporation Reactifs de fusion d'anticorps monoclonal a chaine unique regulant une transcription in vivo
DE1068357T1 (de) 1998-03-30 2001-07-19 Northwest Biotherapeutics Inc Therapeutische und diagnostische anwendungen welche auf der rolle von cxcr-4 in der tumorgenese basieren
US6794132B2 (en) 1999-10-02 2004-09-21 Biosite, Inc. Human antibodies
US6867348B1 (en) 1999-12-16 2005-03-15 Xenogen Corporation Methods and compositions for screening for angiogenesis modulating compounds
AU2001253521A1 (en) 2000-04-13 2001-10-30 Genaissance Pharmaceuticals, Inc. Haplotypes of the cxcr4 gene
WO2001083796A2 (fr) 2000-05-03 2001-11-08 University Of Washington Vecteurs adenoviraux de recombinaison modifies destines a une expression genique specifique des tumeurs et utilisation associee
US6406863B1 (en) 2000-06-23 2002-06-18 Genetastix Corporation High throughput generation and screening of fully human antibody repertoire in yeast
US6410271B1 (en) 2000-06-23 2002-06-25 Genetastix Corporation Generation of highly diverse library of expression vectors via homologous recombination in yeast
US6410246B1 (en) 2000-06-23 2002-06-25 Genetastix Corporation Highly diverse library of yeast expression vectors
US20020026653A1 (en) 2000-07-10 2002-02-28 Allen Keith D. Transgenic mice containing serine protease gene disruptions
US6610472B1 (en) 2000-10-31 2003-08-26 Genetastix Corporation Assembly and screening of highly complex and fully human antibody repertoire in yeast
US20030165988A1 (en) 2002-02-08 2003-09-04 Shaobing Hua High throughput generation of human monoclonal antibody against peptide fragments derived from membrane proteins
US7005503B2 (en) 2002-02-08 2006-02-28 Genetastix Corporation Human monoclonal antibody against coreceptors for human immunodeficiency virus
US7138496B2 (en) 2002-02-08 2006-11-21 Genetastix Corporation Human monoclonal antibodies against human CXCR4
WO2002046400A2 (fr) 2000-12-08 2002-06-13 The Board Of Trustees Of The University Of Illinois Proteines mutantes du complexe majeur d'histocompatibilite de classe ii
DE60215626T2 (de) 2001-04-06 2007-08-30 Thomas Jefferson University Antagonist für die multimerisierung von hiv-1 vif-protein
EP1438400B1 (fr) * 2001-10-01 2009-06-17 Dyax Corp. Vecteurs d'affichage eukaryotes multichaine et leurs utilisations
AU2003240005B2 (en) 2002-06-14 2010-08-12 Takeda Pharmaceutical Company Limited Recombination of nucleic acid library members
CA2737351A1 (fr) 2008-09-26 2010-04-01 Wyeth Llc Conception d'une banque d'anticorps simple chaine

Patent Citations (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4816397A (en) * 1983-03-25 1989-03-28 Celltech, Limited Multichain polypeptides or proteins and processes for their production
US5723323A (en) * 1985-03-30 1998-03-03 Kauffman; Stuart Alan Method of identifying a stochastically-generated peptide, polypeptide, or protein having ligand binding property and compositions thereof
US5354847A (en) * 1986-10-27 1994-10-11 Bristol-Myers Squibb Company Chimeric antibody with specificity to human tumor antigen
US5223409A (en) * 1988-09-02 1993-06-29 Protein Engineering Corp. Directed evolution of novel binding proteins
US5658727A (en) * 1991-04-10 1997-08-19 The Scripps Research Institute Heterodimeric receptor libraries using phagemids
US5789208A (en) * 1994-01-31 1998-08-04 The Trustees Of Boston University Polyclonal antibody libraries
US5643745A (en) * 1994-02-03 1997-07-01 University Of Hawaii Heterologous dimeric proteins produced in heterokaryons
US6291242B1 (en) * 1994-02-17 2001-09-18 Maxygen, Inc. Methods for generating polynucleotides having desired characteristics by iterative selection and recombination
US6132970A (en) * 1994-02-17 2000-10-17 Maxygen, Inc. Methods of shuffling polynucleotides
US6297053B1 (en) * 1994-02-17 2001-10-02 Maxygen, Inc. Methods for generating polynucleotides having desired characteristics by iterative selection and recombination
US5695941A (en) * 1994-06-22 1997-12-09 The General Hospital Corporation Interaction trap systems for analysis of protein networks
US5888732A (en) * 1995-06-07 1999-03-30 Life Technologies, Inc. Recombinational cloning using engineered recombination sites
US6300065B1 (en) * 1996-05-31 2001-10-09 Board Of Trustees Of The University Of Illinois Yeast cell surface display of proteins and uses thereof
US6461863B1 (en) * 1996-08-16 2002-10-08 University Of Wyoming Modifying insect cell gylcosylation pathways with baculovirus expression vectors
US6074853A (en) * 1997-03-21 2000-06-13 Sri Sequence alterations using homologous recombination
US6207442B1 (en) * 1997-10-16 2001-03-27 Zymogenetics, Inc. Plasmid construction by homologous recombination

Cited By (275)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7465787B2 (en) 1996-05-31 2008-12-16 The Board Of Trustees Of The University Of Illinois Yeast cell surface display of proteins and uses thereof
US20040146976A1 (en) * 1996-05-31 2004-07-29 Board Of Trustees Of The University Of Illinois Yeast cell surface display of proteins and uses thereof
US9139637B2 (en) 1996-05-31 2015-09-22 Board Of Trustees Of The University Of Illinois Yeast cell surface display of proteins and uses thereof
US8372636B2 (en) 1996-05-31 2013-02-12 Board Of Trustees Of The University Of Illinois Yeast cell surface display of proteins and uses thereof
US20090280560A1 (en) * 1996-05-31 2009-11-12 Wittrup K Dane Yeast cell surface display of proteins and uses thereof
US20040146952A1 (en) * 1998-01-20 2004-07-29 Kranz David M. High affinity TCR proteins and methods
US20050196755A1 (en) * 2000-11-17 2005-09-08 Maurice Zauderer In vitro methods of producing and identifying immunoglobulin molecules in eukaryotic cells
US20020123057A1 (en) * 2000-11-17 2002-09-05 University Of Rochester In vitro methods of producing and identifying immunoglobulin molecules in eukaryotic cells
US20080167193A1 (en) * 2000-11-17 2008-07-10 University Of Rochester In Vitro methods of producing and identifying immunoglobulin molecules in eukaryotic cells
US7858559B2 (en) 2000-11-17 2010-12-28 University Of Rochester In vitro methods of producing and identifying immunoglobulin molecules in eukaryotic cells
US8741810B2 (en) 2000-11-17 2014-06-03 University Of Rochester In vitro methods of producing and identifying immunoglobulin molecules in eukaryotic cells
US8680239B2 (en) 2000-12-22 2014-03-25 Max-Planck-Gesellschaft Zur Foerderung Der Wissenschaften E.V. Use of RGM and its modulators
US20030104402A1 (en) * 2001-01-23 2003-06-05 University Of Rochester Methods of producing or identifying intrabodies in eukaryotic cells
US9005927B2 (en) 2001-10-01 2015-04-14 Dyax Corp. Multi-chain eukaryotic display vectors and uses thereof
US10059936B2 (en) 2001-10-01 2018-08-28 Dyax Corp. Multi-chain eukaryotic display vectors and uses thereof
US9116149B2 (en) * 2001-10-01 2015-08-25 Dyax Corp. Multi-chain eukaryotic display vectors and uses thereof
US20110009280A1 (en) * 2001-10-01 2011-01-13 Hufton Simon E Multi-chain eukaryotic display vectors and uses thereof
US9034601B2 (en) 2001-10-01 2015-05-19 Dyax Corp. Multi-chain eukaryotic display vectors and uses thereof
US9040258B2 (en) 2001-10-01 2015-05-26 Dyax Corp. Multi-chain eukaryotic display vectors and uses thereof
US9556428B2 (en) 2001-10-01 2017-01-31 Dyax Corp. Multi-chain eukaryotic display vectors and uses thereof
US9068980B2 (en) 2001-10-01 2015-06-30 Dyax Corp. Multi-chain eukaryotic display vectors and uses thereof
US20110281360A1 (en) * 2001-10-01 2011-11-17 Dyax Corp. Multi-chain eukaryotic display vectors and uses thereof
US10577599B2 (en) 2001-10-01 2020-03-03 Dyax Corp. Multi-chain eukaryotic display vectors and uses thereof
US9012181B2 (en) 2001-10-01 2015-04-21 Dyax Corp. Multi-chain eukaryotic display vectors and uses thereof
US10464976B2 (en) 2003-01-31 2019-11-05 AbbVie Deutschland GmbH & Co. KG Amyloid β(1-42) oligomers, derivatives thereof and antibodies thereto, methods of preparation thereof and use thereof
US9176150B2 (en) 2003-01-31 2015-11-03 AbbVie Deutschland GmbH & Co. KG Amyloid beta(1-42) oligomers, derivatives thereof and antibodies thereto, methods of preparation thereof and use thereof
US10259883B2 (en) 2003-10-22 2019-04-16 Keck Graduate Institute Methods of synthesizing heteromultimeric polypeptides in yeast using a haploid mating strategy
US9873746B2 (en) * 2003-10-22 2018-01-23 Keck Graduate Institute Methods of synthesizing heteromultimeric polypeptides in yeast using a haploid mating strategy
US20110183402A1 (en) * 2003-10-22 2011-07-28 Cregg James M Methods of synthesizing heteromultimeric polypeptides in yeast using a haploid mating strategy
US10155819B2 (en) 2003-10-22 2018-12-18 Alderbio Holdings Llc Methods of synthesizing heteromultimeric polypeptides in yeast using a haploid mating strategy
US11447560B2 (en) 2003-10-22 2022-09-20 Keck Graduate Institute Methods of synthesizing heteromultimeric polypeptides in yeast using a haploid mating strategy
EP2460829A2 (fr) 2003-11-12 2012-06-06 Abbott Laboratories Protéines se liant à IL-18
EP2385070A1 (fr) 2003-11-12 2011-11-09 Abbott Laboratories Protéines se liant à IL-18
EP2460830A2 (fr) 2003-11-12 2012-06-06 Abbott Laboratories Protéines se liant à IL-18
EP2395020A2 (fr) 2003-11-12 2011-12-14 Abbott Laboratories Protéines se liant à IL-18
US7700739B2 (en) 2005-06-30 2010-04-20 Abbott Laboratories IL-12/p40 binding proteins
US20100196315A1 (en) * 2005-06-30 2010-08-05 Abbott Laboratories IL-12/p40 BINDING PROTEINS
US8629257B2 (en) 2005-06-30 2014-01-14 Abbvie Inc. IL-12/p40 binding proteins
US20100047245A1 (en) * 2005-06-30 2010-02-25 Lacy Susan E Il-12/p40 binding proteins
EP2500352A1 (fr) 2005-08-19 2012-09-19 Abbott Laboratories Immunoglobuline à double domaine variable et ses utilisations
EP2500358A2 (fr) 2005-08-19 2012-09-19 Abbott Laboratories Immunoglobuline à double domaine variable et ses utilisations
EP2500353A2 (fr) 2005-08-19 2012-09-19 Abbott Laboratories Immunoglobuline à double domaine variable et ses utilisations
EP2500355A2 (fr) 2005-08-19 2012-09-19 Abbott Laboratories Immunoglobuline à double domaine variable et ses utilisations
EP2520588A1 (fr) 2005-08-19 2012-11-07 Abbott Laboratories Immunoglobuline à double domaine variable et ses utilisations
EP2500356A2 (fr) 2005-08-19 2012-09-19 Abbott Laboratories Immunoglobuline à double domaine variable et ses utilisations
EP2495257A2 (fr) 2005-08-19 2012-09-05 Abbott Laboratories Immunoglobuline à double domaine variable et ses utilisations
EP2500354A2 (fr) 2005-08-19 2012-09-19 Abbott Laboratories Immunoglobuline à double domaine variable et ses utilisations
US8258268B2 (en) 2005-08-19 2012-09-04 Abbott Laboratories Dual variable domain immunoglobulin and uses thereof
EP2500357A2 (fr) 2005-08-19 2012-09-19 Abbott Laboratories Immunoglobuline à double domaine variable et ses utilisations
US20100047239A1 (en) * 2005-08-19 2010-02-25 Abbott Laboratories Dual variable domain immunoglobulin and uses thereof
EP2500359A2 (fr) 2005-08-19 2012-09-19 Abbott Laboratories Immunoglobuline à double domaine variable et ses utilisations
WO2007024715A2 (fr) 2005-08-19 2007-03-01 Abbott Laboratories Immunoglobuline a deux domaines variables et utilisations de celle-ci
US20090215992A1 (en) * 2005-08-19 2009-08-27 Chengbin Wu Dual variable domain immunoglobulin and uses thereof
US8906864B2 (en) 2005-09-30 2014-12-09 AbbVie Deutschland GmbH & Co. KG Binding domains of proteins of the repulsive guidance molecule (RGM) protein family and functional fragments thereof, and their use
US20100331208A1 (en) * 2005-10-14 2010-12-30 Medimmune, Llc Cell Display Of Antibody Libraries
US20070111260A1 (en) * 2005-10-14 2007-05-17 Medimmune, Inc. Cell display of antibody libraries
US7790655B2 (en) 2005-10-14 2010-09-07 Medimmune, Llc Cell display of antibody libraries
US8409568B2 (en) 2005-10-14 2013-04-02 Medimmune, Llc Mutant antibody Fc domains and fusion proteins thereof
US9567389B2 (en) 2005-10-14 2017-02-14 Medimmune, Llc Cell display of antibody libraries
US8691224B2 (en) 2005-11-30 2014-04-08 Abbvie Inc. Anti-Aβ globulomer 5F7 antibodies
US10323084B2 (en) 2005-11-30 2019-06-18 Abbvie Inc. Monoclonal antibodies against amyloid beta protein and uses thereof
US10538581B2 (en) 2005-11-30 2020-01-21 Abbvie Inc. Anti-Aβ globulomer 4D10 antibodies
US8497072B2 (en) 2005-11-30 2013-07-30 Abbott Laboratories Amyloid-beta globulomer antibodies
US10208109B2 (en) 2005-11-30 2019-02-19 Abbvie Inc. Monoclonal antibodies against amyloid beta protein and uses thereof
US9540432B2 (en) 2005-11-30 2017-01-10 AbbVie Deutschland GmbH & Co. KG Anti-Aβ globulomer 7C6 antibodies
EP2021795A2 (fr) * 2006-05-04 2009-02-11 Abmaxis, Inc. Systèmes d'expression à la surface multispécifique et hétérospécifique
EP2420832A1 (fr) 2006-05-04 2012-02-22 Abmaxis, Inc. Systèmes d'affichage hétérospécifique et de plusieurs espèces
EP2021795A4 (fr) * 2006-05-04 2009-05-13 Abmaxis Inc Systèmes d'expression à la surface multispécifique et hétérospécifique
US20090082221A1 (en) * 2006-05-04 2009-03-26 Kevin Caili Wang Cross-Species and Multi-Species Display Systems
US9592293B2 (en) 2006-09-08 2017-03-14 Abbvie Inc. Interleukin-13 binding proteins
EP3339445A1 (fr) 2006-09-08 2018-06-27 AbbVie Bahamas Ltd. Protéines de liaison de l'interleukine -13
EP3524685A1 (fr) 2006-09-08 2019-08-14 AbbVie Bahamas Ltd. Protéines de liaison de l'interleukine -13
US20080171014A1 (en) * 2006-09-08 2008-07-17 Chengbin Wu Interleukin-13 binding proteins
EP3910065A1 (fr) 2006-09-08 2021-11-17 AbbVie Bahamas Ltd. Protéines de liaison de l'interleukine-13
US7915388B2 (en) 2006-09-08 2011-03-29 Abbott Laboratories Interleukin-13 binding proteins
US8604177B2 (en) 2006-09-08 2013-12-10 Abbvie Inc. Interleukin-13 binding proteins
US11344621B2 (en) 2006-09-08 2022-05-31 Abbvie, Inc. Interleukin-13 binding proteins
US20110165066A1 (en) * 2006-09-08 2011-07-07 Chengbin Wu Interleukin-13 binding proteins
US10086076B2 (en) 2006-09-08 2018-10-02 Abbvie Inc. Interleukin-13 binding proteins
US8455626B2 (en) 2006-11-30 2013-06-04 Abbott Laboratories Aβ conformer selective anti-aβ globulomer monoclonal antibodies
US9394360B2 (en) 2006-11-30 2016-07-19 Abbvie Inc. Aβ conformer selective anti-Aβ globulomer monoclonal antibodies
US9359430B2 (en) 2006-11-30 2016-06-07 Abbvie Inc. Abeta conformer selective anti-Abeta globulomer monoclonal antibodies
US9951125B2 (en) 2006-11-30 2018-04-24 Abbvie Inc. Aβ conformer selective anti-Aβ globulomer monoclonal antibodies
US8877190B2 (en) 2006-11-30 2014-11-04 Abbvie Inc. Aβ conformer selective anti-Aβ globulomer monoclonal antibodies
US20110076752A1 (en) * 2007-02-09 2011-03-31 Medimmune, Llc Antibody library display by yeast cell plasma membrane
US8895004B2 (en) 2007-02-27 2014-11-25 AbbVie Deutschland GmbH & Co. KG Method for the treatment of amyloidoses
US9645146B2 (en) 2007-03-26 2017-05-09 Agenus, Inc. Cell surface display, screening and production of proteins of interest
US9222083B2 (en) 2007-04-03 2015-12-29 Oxyrane Uk Limited Microorganisms genetically engineered to have modified N-glycosylation activity
US10023854B2 (en) 2007-04-03 2018-07-17 Oxyrane Uk Limited Microorganisms genetically engineered to have modified N-glycosylation activity
US9206408B2 (en) 2007-04-03 2015-12-08 Oxyrane Uk Limited Microorganisms genetically engineered to have modified N-glycosylation activity
US20090232801A1 (en) * 2007-05-30 2009-09-17 Abbot Laboratories Humanized Antibodies Which Bind To AB (1-42) Globulomer And Uses Thereof
US20090175847A1 (en) * 2007-05-30 2009-07-09 Abbott Laboratories Humanized antibodies to ab (20-42) globulomer and uses thereof
US8637435B2 (en) * 2007-11-16 2014-01-28 Merck Sharp & Dohme Corp. Eukaryotic cell display systems
US20090163379A1 (en) * 2007-11-16 2009-06-25 Kevin Caili Wang Eukaryotic cell display systems
US8962803B2 (en) 2008-02-29 2015-02-24 AbbVie Deutschland GmbH & Co. KG Antibodies against the RGM A protein and uses thereof
US9605069B2 (en) 2008-02-29 2017-03-28 AbbVie Deutschland GmbH & Co. KG Antibodies against the RGM a protein and uses thereof
EP3309173A1 (fr) 2008-02-29 2018-04-18 AbbVie Deutschland GmbH & Co KG Anticorps monoclonaux contre la protéine rgm a et ses utilisations
WO2009111183A1 (fr) * 2008-03-03 2009-09-11 Glycofi, Inc. Affichage en surface de protéines recombinantes dans des eucaryotes inférieurs
US9845464B2 (en) 2008-03-03 2017-12-19 Glycofi, Inc. Surface display of recombinant proteins in lower eukaryotes
US20100331192A1 (en) * 2008-03-03 2010-12-30 Dongxing Zha Surface display of recombinant proteins in lower eukaryotes
US8877686B2 (en) 2008-03-03 2014-11-04 Glycofi, Inc. Surface display of recombinant proteins in lower eukaryotes
EP2899209A1 (fr) 2008-04-29 2015-07-29 Abbvie Inc. Immunoglobuline à double domaine variable et ses utilisations
US9029508B2 (en) 2008-04-29 2015-05-12 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
US20100028359A1 (en) * 2008-05-09 2010-02-04 Abbott Gmbh & Co. Kg Antibodies to receptor of advanced glycation end products (rage) and uses thereof
EP2116556A1 (fr) 2008-05-09 2009-11-11 Abbott GmbH & Co. KG Anticorps sur récepteur de produit terminal de glycosylation et utilisations associées
EP2500361A1 (fr) 2008-05-09 2012-09-19 Abbott GmbH & Co. KG Anticorps sur récepteur de produit terminal de glycosylation et utilisations associées
US8323651B2 (en) 2008-05-09 2012-12-04 Abbott Laboratories Antibodies to receptor of advanced glycation end products (RAGE) and uses thereof
US9394363B2 (en) 2008-05-09 2016-07-19 AbbVie Deutschland GmbH & Co. KG Antibodies to receptor of advanced glycation end products (RAGE) and uses thereof
EP3059248A1 (fr) 2008-05-09 2016-08-24 Abbvie Deutschland GmbH & Co. KG Anticorps contre le récepteur pour produits terminaux de glycation avancée (rage) et utilisations de ceux-ci
EP3002299A1 (fr) 2008-06-03 2016-04-06 AbbVie Inc. Immunoglobulines à deux domaines variables et leurs utilisations
WO2009149185A2 (fr) 2008-06-03 2009-12-10 Abbott Laboratories Immunoglobulines à double domaine variable et leurs utilisations
US9109026B2 (en) 2008-06-03 2015-08-18 Abbvie, Inc. Dual variable domain immunoglobulins and uses thereof
US9035027B2 (en) 2008-06-03 2015-05-19 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
US8822645B2 (en) 2008-07-08 2014-09-02 Abbvie Inc. Prostaglandin E2 dual variable domain immunoglobulins and uses thereof
EP2810654A1 (fr) 2008-07-08 2014-12-10 AbbVie Inc. Protéines de liaison à la protéines de liaison à la prostaglandine E2 et leurs utilisations
US20100009866A1 (en) * 2008-07-09 2010-01-14 Bianka Prinz Surface Display of Whole Antibodies in Eukaryotes
US8067339B2 (en) 2008-07-09 2011-11-29 Merck Sharp & Dohme Corp. Surface display of whole antibodies in eukaryotes
US9260712B2 (en) 2008-07-09 2016-02-16 Merck Sharp & Dohme Corp. Surface display of whole antibodies in eukaryotes
US11046951B2 (en) 2008-07-09 2021-06-29 Merck Sharp & Dohme Corp. Surface display of whole antibodies in eukaryotes
US20100233079A1 (en) * 2008-12-04 2010-09-16 Abbott Laboratories Dual Variable Domain Immunoglobulins and Uses Thereof
US20100221179A1 (en) * 2009-01-29 2010-09-02 Abbott Laboratories IL-1 Binding Proteins
US8383778B2 (en) 2009-01-29 2013-02-26 Abbvie Inc. IL-1 binding proteins
US20110165063A1 (en) * 2009-01-29 2011-07-07 Abbott Laboratories Il-1 binding proteins
EP2853540A1 (fr) 2009-02-24 2015-04-01 Abbott Laboratories Anticorps dirigés contre la troponine I et leurs procédés d'utilisation
WO2010099079A1 (fr) 2009-02-24 2010-09-02 Abbott Laboratories Anticorps dirigés contre la troponine i et leurs procédés d'utilisation
US20100216720A1 (en) * 2009-02-24 2010-08-26 Abbott Laboratories Antibodies to troponin i and methods of use thereof
US8030026B2 (en) 2009-02-24 2011-10-04 Abbott Laboratories Antibodies to troponin I and methods of use thereof
USRE45763E1 (en) 2009-02-24 2015-10-20 Abbott Laboratories Antibodies to troponin I and methods of use thereof
EP2772269A2 (fr) 2009-03-05 2014-09-03 Abbvie Inc. Protéines se liant à un IL-17
US8835610B2 (en) 2009-03-05 2014-09-16 Abbvie Inc. IL-17 binding proteins
US9481735B2 (en) 2009-03-05 2016-11-01 Abbvie Inc. IL-17 binding proteins
US8779101B2 (en) 2009-03-05 2014-07-15 Abbvie, Inc. IL-17 binding proteins
US9481736B2 (en) 2009-03-05 2016-11-01 Abbvie, Inc. IL-17 binding proteins
US9663587B2 (en) 2009-03-05 2017-05-30 Abbvie Inc. IL-17 binding proteins
EP2810652A2 (fr) 2009-03-05 2014-12-10 AbbVie Inc. Protéines se liant à un IL-17
US20100266531A1 (en) * 2009-03-05 2010-10-21 Abbott Laboratories Il-17 binding proteins
US9132190B2 (en) 2009-08-29 2015-09-15 Abbvie Inc. Therapeutic DLL4 binding proteins
US8623358B2 (en) 2009-08-29 2014-01-07 Abbvie Inc. Therapeutic DLL4 binding proteins
US9469688B2 (en) 2009-08-29 2016-10-18 Abbvie Inc. Therapeutic DLL4 binding proteins
WO2011025964A2 (fr) 2009-08-29 2011-03-03 Abbott Laboratories Protéines thérapeutiques se liant à dll4
EP3029070A1 (fr) 2009-08-29 2016-06-08 AbbVie Inc. Protéines de liaison dll4 thérapeutiques
US20110117079A1 (en) * 2009-08-29 2011-05-19 Abbott Laboratories Therapeutic dll4 binding proteins
US8586714B2 (en) 2009-09-01 2013-11-19 Abbvie, Inc. Dual variable domain immunoglobulins and uses thereof
US10392609B2 (en) 2009-09-29 2019-08-27 Oxyrane Uk Limited Hydrolysis of mannose-1-phospho-6-mannose linkage to phospho-6-mannose
US9598682B2 (en) 2009-09-29 2017-03-21 Vib Vzw Hydrolysis of mannose-1-phospho-6-mannose linkage to phospho-6-mannose
US8716450B2 (en) 2009-10-15 2014-05-06 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
US8722855B2 (en) 2009-10-28 2014-05-13 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
US9067996B2 (en) 2009-10-31 2015-06-30 Abbvie Inc. Antibodies to receptor for advanced glycation end products (RAGE) and uses thereof
US20110104174A1 (en) * 2009-10-31 2011-05-05 Abbott Laboratories Antibodies to receptor for advanced glycation end products (rage) and uses thereof
US8420083B2 (en) 2009-10-31 2013-04-16 Abbvie Inc. Antibodies to receptor for advanced glycation end products (RAGE) and uses thereof
WO2011058087A1 (fr) 2009-11-11 2011-05-19 Gentian As Dosage immunologique pour le dosage d'analytes apparentés d'origine différente
US11225646B2 (en) 2009-11-19 2022-01-18 Oxyrane Uk Limited Yeast strains producing mammalian-like complex n-glycans
US10287557B2 (en) 2009-11-19 2019-05-14 Oxyrane Uk Limited Yeast strains producing mammalian-like complex N-glycans
WO2011070045A1 (fr) 2009-12-08 2011-06-16 Abbott Gmbh & Co. Kg Anticorps monoclonaux contre la protéine rgm a destinés à être utilisés dans le traitement de la dégénérescence de la couche de fibres nerveuses rétiniennes
US9175075B2 (en) 2009-12-08 2015-11-03 AbbVie Deutschland GmbH & Co. KG Methods of treating retinal nerve fiber layer degeneration with monoclonal antibodies against a retinal guidance molecule (RGM) protein
US20130096281A1 (en) * 2010-01-21 2013-04-18 Oxyrane Uk Limited Methods and compositions for displaying a polypeptide on a yeast cell surface
EP3680253A2 (fr) 2010-03-02 2020-07-15 AbbVie Inc. Protéines de liaison dll4 thérapeutiques
US9115195B2 (en) 2010-03-02 2015-08-25 Abbvie Inc. Therapeutic DLL4 binding proteins
EP3072904A1 (fr) 2010-03-02 2016-09-28 Abbvie Inc. Protéines de liaison dll4 thérapeutiques
US9469689B2 (en) 2010-03-02 2016-10-18 Abbvie Inc. Therapeutic DLL4 binding proteins
US9822171B2 (en) 2010-04-15 2017-11-21 AbbVie Deutschland GmbH & Co. KG Amyloid-beta binding proteins
WO2011130377A2 (fr) 2010-04-15 2011-10-20 Abbott Laboratories Protéines de liaison à la bêta amyloïde
US8987419B2 (en) 2010-04-15 2015-03-24 AbbVie Deutschland GmbH & Co. KG Amyloid-beta binding proteins
US8841417B2 (en) 2010-05-14 2014-09-23 Abbvie Inc. IL-1 binding proteins
US9409986B2 (en) 2010-05-14 2016-08-09 Abbvie Inc. IL-1 binding proteins
US9447183B2 (en) 2010-05-14 2016-09-20 Abbvie Inc. IL-1 binding proteins
US9447184B2 (en) 2010-05-14 2016-09-20 Abbvie Inc. IL-1 binding proteins
US8664367B2 (en) 2010-05-14 2014-03-04 Abbvie, Inc. IL-I binding proteins
WO2011143562A2 (fr) 2010-05-14 2011-11-17 Abbott Laboratories Protéines liant l'il-1
US9303085B2 (en) 2010-05-14 2016-04-05 Abbvie Inc. IL-1 binding proteins
US9441038B2 (en) 2010-05-14 2016-09-13 Abbvie Inc. IL-1 binding proteins
WO2012006500A2 (fr) 2010-07-08 2012-01-12 Abbott Laboratories Anticorps monoclonaux contre la protéine de capside du virus de l'hépatite c
EP2921177A2 (fr) 2010-07-09 2015-09-23 AbbVie Inc. Immunoglobulines à double domaine variable et utilisations de celles-ci
EP3252072A2 (fr) 2010-08-03 2017-12-06 AbbVie Inc. Immunoglobuline à double domaine variable et ses utilisations
US9493560B2 (en) 2010-08-03 2016-11-15 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
US8735546B2 (en) 2010-08-03 2014-05-27 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
US9062101B2 (en) 2010-08-14 2015-06-23 AbbVie Deutschland GmbH & Co. KG Amyloid-beta binding proteins
US10047121B2 (en) 2010-08-14 2018-08-14 AbbVie Deutschland GmbH & Co. KG Amyloid-beta binding proteins
EP3333188A1 (fr) 2010-08-19 2018-06-13 Zoetis Belgium S.A. Anticorps anti-ngf et leur utilisation
EP4056589A1 (fr) 2010-08-19 2022-09-14 Zoetis Belgium S.A. Anticorps anti-ngf et leur utilisation
WO2012024650A2 (fr) 2010-08-19 2012-02-23 Abbott Laboratories Anticorps anti-ngf et leur utilisation
US9046513B2 (en) 2010-08-26 2015-06-02 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
US9689015B2 (en) 2010-09-29 2017-06-27 Oxyrane Uk Limited De-mannosylation of phosphorylated N-glycans
US10344310B2 (en) 2010-09-29 2019-07-09 Oxyrane Uk Limited De-mannosylation of phosphorylated N-glycans
US9347050B2 (en) 2010-09-29 2016-05-24 Oxyrane Uk Limited Mannosidases capable of uncapping mannose-1-phospho-6-mannose linkages and demannosylating phosphorylated N-glycans and methods of facilitating mammalian cellular uptake of glycoproteins
US10011857B2 (en) 2010-09-29 2018-07-03 Oxyrane Uk Limited Mannosidases capable of uncapping mannose-1-phospho-6-mannose linkages and demannosylating phosphorylated N-glycans and methods of facilitating mammalian cellular uptake of glycoproteins
WO2012121775A2 (fr) 2010-12-21 2012-09-13 Abbott Laboratories Immunoglobulines à double domaine variable et leurs utilisations
WO2012088094A2 (fr) 2010-12-21 2012-06-28 Abbott Laboratories Protéines liant l'il-1
EP3574919A1 (fr) 2011-07-13 2019-12-04 AbbVie Inc. Procédés et compositions de traitement de l'asthme à l'aide d'anticorps anti-il-13
WO2013063095A1 (fr) 2011-10-24 2013-05-02 Abbvie Inc. Agents de liaison immunologique dirigés contre la sclérostine
WO2013063114A1 (fr) 2011-10-24 2013-05-02 Abbvie Inc. Agents de liaison immunologique dirigés contre le tnf
WO2013078377A1 (fr) 2011-11-23 2013-05-30 Igenica, Inc. Anticorps anti-cd98 et leurs procédés d'utilisation
EP3800200A1 (fr) 2011-12-14 2021-04-07 AbbVie Deutschland GmbH & Co. KG Composition et méthode pour le diagnostic et le traitement de troubles liés au fer
US10822403B2 (en) 2011-12-14 2020-11-03 AbbVie Deutschland GmbH & Co. KG Composition and method for the diagnosis and treatment of iron-related disorders
US10118958B2 (en) 2011-12-14 2018-11-06 AbbVie Deutschland GmbH & Co. KG Composition and method for the diagnosis and treatment of iron-related disorders
WO2013090633A2 (fr) 2011-12-14 2013-06-20 AbbVie Deutschland GmbH & Co. KG Composition et méthode pour le diagnostic et le traitement de troubles liés au fer
WO2013090635A2 (fr) 2011-12-14 2013-06-20 AbbVie Deutschland GmbH & Co. KG Composition et méthode pour le diagnostic et le traitement de troubles liés au fer
US9636398B2 (en) 2011-12-14 2017-05-02 AbbVie Deutschland GmbH & Co. KG Composition and method for the diagnosis and treatment of iron-related disorders
US9120870B2 (en) 2011-12-30 2015-09-01 Abbvie Inc. Dual specific binding proteins directed against IL-13 and IL-17
US10106602B2 (en) 2012-01-27 2018-10-23 AbbVie Deutschland GmbH & Co. KG Isolated monoclonal anti-repulsive guidance molecule A antibodies and uses thereof
US9365643B2 (en) 2012-01-27 2016-06-14 AbbVie Deutschland GmbH & Co. KG Antibodies that bind to repulsive guidance molecule A (RGMA)
EP3369746A1 (fr) 2012-01-27 2018-09-05 AbbVie Deutschland GmbH & Co KG Composition et procédé pour le diagnostic et le traitement de maladies associées à la dégénérescence des neurites
US9102722B2 (en) 2012-01-27 2015-08-11 AbbVie Deutschland GmbH & Co. KG Composition and method for the diagnosis and treatment of diseases associated with neurite degeneration
EP3653647A1 (fr) 2012-01-27 2020-05-20 AbbVie Deutschland GmbH & Co KG Composition et procédé pour le diagnostic et le traitement de maladies associées à la dégénérescence des neurites
WO2013112922A1 (fr) 2012-01-27 2013-08-01 AbbVie Deutschland GmbH & Co. KG Composition et méthode pour le diagnostic et le traitement de maladies associées à la dégénérescence des neurites
US9249399B2 (en) 2012-03-15 2016-02-02 Oxyrane Uk Limited Methods and materials for treatment of pompe's disease
US10648044B2 (en) 2012-03-15 2020-05-12 Oxyrane Uk Limited Methods and materials for treatment of Pompe's disease
US9670276B2 (en) 2012-07-12 2017-06-06 Abbvie Inc. IL-1 binding proteins
WO2014059028A1 (fr) 2012-10-09 2014-04-17 Igenica, Inc. Anticorps anti-c16orf54 et leurs méthodes d'utilisation
US9045551B2 (en) 2012-11-01 2015-06-02 Abbvie Inc. Anti-DLL4/VEGF dual variable domain immunoglobulin and uses thereof
US9944720B2 (en) 2012-11-01 2018-04-17 Abbvie Inc. Anti-DLL4/VEGF dual variable domain immunoglobulin and uses thereof
US9163093B2 (en) 2012-11-01 2015-10-20 Abbvie Inc. Anti-DLL4/VEGF dual variable domain immunoglobulin and uses thereof
US9550986B2 (en) 2012-12-21 2017-01-24 Abbvie Inc. High-throughput antibody humanization
WO2014100542A1 (fr) 2012-12-21 2014-06-26 Abbvie, Inc. Humanisation d'anticorps à haut rendement
EP3916103A1 (fr) 2013-03-14 2021-12-01 Abbott Laboratories Anticorps monoclonaux à domaine de liaison à un lipide du c ur de vhc
US9841427B2 (en) 2013-03-14 2017-12-12 Abbott Laboratories HCV antigen-antibody combination assay and methods and compositions for use therein
US10197573B2 (en) 2013-03-14 2019-02-05 Abbott Laboratories HCV core lipid binding domain monoclonal antibodies
EP3564384A1 (fr) 2013-03-14 2019-11-06 Abbott Laboratories Anticorps monoclonaux à domaine de liaison à un lipide du noyau de vhc
US9371374B2 (en) 2013-03-14 2016-06-21 Abbott Laboratories HCV core lipid binding domain monoclonal antibodies
US10444242B2 (en) 2013-03-14 2019-10-15 Abbott Laboratories Detection methods employing HCV core lipid and DNA binding domain monoclonal antibodies
US9790478B2 (en) 2013-03-14 2017-10-17 Abbott Laboratories HCV NS3 recombinant antigens and mutants thereof for improved antibody detection
US11428694B2 (en) 2013-03-14 2022-08-30 Abbott Laboratories Detection methods employing HCV core lipid and DNA binding domain monoclonal antibodies
US10345311B2 (en) 2013-03-14 2019-07-09 Abbott Laboratories Detection methods employing HCV core lipid and DNA binding domain monoclonal antibodies
EP3124499A1 (fr) 2013-03-15 2017-02-01 Abbott Laboratories Anticorps monoclonaux anti-gp73 et procédés d'obtention de ceux-ci
US8987418B2 (en) 2013-03-15 2015-03-24 Abbvie Inc. Dual specific binding proteins directed against IL-1β and/or IL-17
US11421023B2 (en) 2013-03-15 2022-08-23 Abbott Laboratories Anti-GP73 monoclonal antibodies and methods of obtaining the same
US9062108B2 (en) 2013-03-15 2015-06-23 Abbvie Inc. Dual specific binding proteins directed against IL-1 and/or IL-17
EP3527586A1 (fr) 2013-03-15 2019-08-21 Abbott Laboratories Anticorps monoclonaux anti-gp73 et procédés d'obtention de ceux-ci
US9469686B2 (en) 2013-03-15 2016-10-18 Abbott Laboratories Anti-GP73 monoclonal antibodies and methods of obtaining the same
US10308709B2 (en) 2013-03-15 2019-06-04 Abbott Laboratories Anti-GP73 monoclonal antibodies and methods of obtaining the same
WO2014144355A2 (fr) 2013-03-15 2014-09-18 Abbott Laboratories Anticorps monoclonaux anti-gp73 et leurs procédés d'obtention
US11897946B2 (en) 2013-06-07 2024-02-13 Duke University Methods of inhibiting complement factor H (CFH) comprising administering an antibody that binds CFH
US11136380B2 (en) 2013-06-07 2021-10-05 Duke University Anti-complement factor H antibodies
US10183988B2 (en) 2013-06-07 2019-01-22 Duke University Anti-Complement factor H antibodies
EP3632467A1 (fr) 2013-06-07 2020-04-08 Duke University Inhibiteurs de facteur h du complément
EP3738981A1 (fr) 2014-01-24 2020-11-18 NGM Biopharmaceuticals, Inc. Anticorps liant beta-klotho 2 et leurs procédés d'utilisation
WO2015120058A2 (fr) 2014-02-05 2015-08-13 Molecular Templates, Inc. Procédés de criblage, de sélection et d'identification de polypeptides de recombinaison cytotoxiques fondés sur une diminution provisoire de la ribotoxicité
US10093733B2 (en) 2014-12-11 2018-10-09 Abbvie Inc. LRP-8 binding dual variable domain immunoglobulin proteins
EP4047022A1 (fr) 2015-05-29 2022-08-24 AbbVie Inc. Anticorps anti-cd40 et leurs utilisations
EP3626744A1 (fr) 2015-05-29 2020-03-25 AbbVie Inc. Anticorps anti-cd40 et leurs utilisations
US9840554B2 (en) 2015-06-15 2017-12-12 Abbvie Inc. Antibodies against platelet-derived growth factor (PDGF)
US10428145B2 (en) 2015-09-29 2019-10-01 Celgene Corporation PD-1 binding proteins and methods of use thereof
US10766958B2 (en) 2016-09-19 2020-09-08 Celgene Corporation Methods of treating vitiligo using PD-1 binding antibodies
US10751414B2 (en) 2016-09-19 2020-08-25 Celgene Corporation Methods of treating psoriasis using PD-1 binding antibodies
WO2018067474A1 (fr) 2016-10-03 2018-04-12 Abbott Laboratories Procédés améliorés d'évaluation de l'état de gfap dans des échantillons de patient
WO2018067468A1 (fr) 2016-10-03 2018-04-12 Abbott Laboratories Procédés améliorés d'évaluation de l'état de uch-l1 dans des échantillons de patient
WO2018175942A1 (fr) 2017-03-23 2018-09-27 Abbott Laboratories Méthodes d'aide au diagnostic et à la détermination de l'étendue d'une lésion cérébrale traumatique chez un sujet humain à l'aide du biomarqueur précoce hydrolase carboxy-terminale d'ubiquitine l1
WO2018191531A1 (fr) 2017-04-15 2018-10-18 Abbott Laboratories Procédés d'aide au diagnostic hyperaigu et de détermination d'une lésion cérébrale traumatique chez un sujet humain à l'aide de biomarqueurs précoces
WO2018200823A1 (fr) 2017-04-28 2018-11-01 Abbott Laboratories Méthodes d'aide au diagnostic et à la détermination hyperaigus d'une lésion cérébrale traumatique au moyen de biomarqueurs précoces sur au moins deux échantillons provenant du même sujet humain
US10865238B1 (en) 2017-05-05 2020-12-15 Duke University Complement factor H antibodies
WO2018218169A1 (fr) 2017-05-25 2018-11-29 Abbott Laboratories Procédés d'aide à la détermination de la réalisation ou non d'une imagerie sur un sujet humain ayant subi ou susceptible d'avoir subi une lésion à la tête à l'aide de biomarqueurs précoces
WO2018222783A1 (fr) 2017-05-30 2018-12-06 Abbott Laboratories Procédés d'aide au diagnostic et à l'évaluation d'une lésion cérébrale traumatique légère chez un sujet humain au moyen de troponine i cardiaque et de biomarqueurs précoces
WO2018222784A1 (fr) 2017-05-30 2018-12-06 Abbott Laboratories Procédés d'aide au diagnostic et à l'évaluation d'une lésion cérébrale traumatique légère chez un patient humain à l'aide de troponine cardiaque i
WO2019010131A1 (fr) 2017-07-03 2019-01-10 Abbott Laboratories Procédés améliorés de mesure de niveaux d'hydrolase à terminaison carboxy d'ubiquitine l1 dans le sang
WO2019112860A1 (fr) 2017-12-09 2019-06-13 Abbott Laboratories Procédés d'aide au diagnostic et à l'évaluation d'un lésion cérébrale traumatique chez un sujet humain au moyen d'une combinaison de gfap et d'uch-l1
WO2019113525A2 (fr) 2017-12-09 2019-06-13 Abbott Laboratories Procédés d'aide au diagnostic et à l'évaluation d'un sujet qui a subi une lésion orthopédique et qui a subi ou peut avoir subi une lésion à la tête, telle qu'une lésion cérébrale traumatique légère (tbi), à l'aide d'une protéine acide fibrillaire gliale (gfap) et/ou d'hydrolase carboxy-terminale d'ubiquitine l1 (uch-l1)
WO2020065594A1 (fr) 2018-09-28 2020-04-02 Kyowa Kirin Co., Ltd. Anticorps il-36 et leurs utilisations
WO2021159024A1 (fr) 2020-02-05 2021-08-12 Larimar Therapeutics, Inc. Protéines de liaison au peptide tat et leurs utilisations
WO2021211331A1 (fr) 2020-04-13 2021-10-21 Abbott Point Of Care Inc. Procédés, complexes et kits pour détecter ou déterminer une quantité d'un anticorps anti-ss-coronavirus dans un échantillon
WO2022031804A1 (fr) 2020-08-04 2022-02-10 Abbott Laboratories Méthodes et kits améliorés pour détecter une protéine sars-cov-2 dans un échantillon
WO2022119841A1 (fr) 2020-12-01 2022-06-09 Abbott Laboratories Utilisation d'un ou plusieurs biomarqueurs pour déterminer un traumatisme crânien (tbi) chez un sujet soumis à un balayage de tomodensitométrie assistée par ordinateur de la tête à tbi négatif
WO2022147147A1 (fr) 2020-12-30 2022-07-07 Abbott Laboratories Procédés pour déterminer un antigène sras-cov-2 et anticorps anti-sras-cov-2 dans un échantillon
WO2022147463A2 (fr) 2020-12-31 2022-07-07 Alamar Biosciences, Inc. Molécules de liant ayant une affinité et/ou une spécificité élevées et leurs procédés de fabrication et d'utilisation
WO2022245920A1 (fr) 2021-05-18 2022-11-24 Abbott Laboratories Procédés d'évaluation d'une lésion cérébrale chez un sujet en pédiatrie
WO2022266034A1 (fr) 2021-06-14 2022-12-22 Abbott Laboratories Procédés de diagnostic ou d'aide au diagnostic d'une lésion cérébrale provoquée par de l'énergie acoustique, de l'énergie électromagnétique, une onde de surpression et/ou le souffle d'une explosion
WO2023034777A1 (fr) 2021-08-31 2023-03-09 Abbott Laboratories Méthodes et systèmes de diagnostic de lésion cérébrale
WO2023056268A1 (fr) 2021-09-30 2023-04-06 Abbott Laboratories Méthodes et systèmes de diagnostic de lésion cérébrale
WO2023102384A1 (fr) 2021-11-30 2023-06-08 Abbott Laboratories Utilisation d'un ou de plusieurs biomarqueurs pour déterminer un traumatisme crânien (tbi) chez un sujet ayant été soumis à un balayage de tomodensitométrie assistée par ordinateur de la tête ne démontrant par de tbi
WO2023114978A1 (fr) 2021-12-17 2023-06-22 Abbott Laboratories Systèmes et procédés de détermination d'uch-l1, de gfap et d'autres biomarqueurs dans des échantillons de sang
WO2023129942A1 (fr) 2021-12-28 2023-07-06 Abbott Laboratories Utilisation de biomarqueurs pour déterminer une lésion cérébrale traumatique (tbi) subaiguë chez un sujet ayant subi un tomodensitogramme (ct) de la tête négatif pour une tbi ou n'ayant pas subi de tomodensitogramme
WO2023131901A1 (fr) 2022-01-07 2023-07-13 Johnson & Johnson Enterprise Innovation Inc. Matériaux et procédés de protéines de liaison à il-1beta
WO2023150652A1 (fr) 2022-02-04 2023-08-10 Abbott Laboratories Procédés d'écoulement latéral, dosages et dispositifs de détection de la présence ou de mesure de la quantité d'ubiquitine carboxy-terminal hydrolase l1 et/ou d'une protéine gliofibrillaire acide dans un échantillon
WO2024006876A1 (fr) 2022-06-29 2024-01-04 Abbott Laboratories Systèmes et analyses magnétiques hors laboratoire pour déterminer une gfap dans des échantillons biologiques
WO2024013727A1 (fr) 2022-07-15 2024-01-18 Janssen Biotech, Inc. Matériau et procédés d'appariement amélioré par génie biologique de régions variables de liaison à l'antigène
WO2024059708A1 (fr) 2022-09-15 2024-03-21 Abbott Laboratories Biomarqueurs et méthodes de différenciation entre une lésion cérébrale traumatique légère et très légère

Also Published As

Publication number Publication date
EP2093286B1 (fr) 2013-02-27
PT1438400E (pt) 2009-09-10
US9040258B2 (en) 2015-05-26
DE60232672D1 (de) 2009-07-30
EP1438400A1 (fr) 2004-07-21
US20110281761A1 (en) 2011-11-17
JP2010227114A (ja) 2010-10-14
JP5111558B2 (ja) 2013-01-09
US20110190159A1 (en) 2011-08-04
US20110287971A1 (en) 2011-11-24
JP2005504541A (ja) 2005-02-17
CA2462113A1 (fr) 2003-04-10
EP1438400B1 (fr) 2009-06-17
US20170314012A1 (en) 2017-11-02
DK2093286T3 (da) 2013-05-13
AU2008203340B2 (en) 2012-01-19
US9005927B2 (en) 2015-04-14
US9116149B2 (en) 2015-08-25
US20110281360A1 (en) 2011-11-17
ES2405551T3 (es) 2013-05-31
US9012181B2 (en) 2015-04-21
EP1438400A4 (fr) 2006-01-04
US20160040157A1 (en) 2016-02-11
US9034601B2 (en) 2015-05-19
JP4578098B2 (ja) 2010-11-10
US9068980B2 (en) 2015-06-30
DK1438400T3 (da) 2009-10-05
US9556428B2 (en) 2017-01-31
CA2462113C (fr) 2013-01-29
US20110009280A1 (en) 2011-01-13
AU2008203340A1 (en) 2008-08-14
ES2327905T3 (es) 2009-11-05
ATE434040T1 (de) 2009-07-15
US20130331289A1 (en) 2013-12-12
WO2003029456A1 (fr) 2003-04-10
AU2002330162B2 (en) 2008-08-07
EP2093286A1 (fr) 2009-08-26
US20190211322A1 (en) 2019-07-11
US10059936B2 (en) 2018-08-28
US10577599B2 (en) 2020-03-03

Similar Documents

Publication Publication Date Title
US10577599B2 (en) Multi-chain eukaryotic display vectors and uses thereof
AU2002330162A1 (en) Multi-chain eukaryotic display vectors and uses thereof
Boder et al. Yeast surface display for screening combinatorial polypeptide libraries
US20220090053A1 (en) Integrated system for library construction, affinity binder screening and expression thereof
Breous-Nystrom et al. Retrocyte Display® technology: generation and screening of a high diversity cellular antibody library
AU2003240005A1 (en) Recombination of nucleic acid library members
DE69932446T2 (de) Verfahren zur herstellung von nukleinsäure- und polypeptidbanken durch in vivo rekombination und ihre verwendungen
AU2016234978B2 (en) Multi-chain eukaryotic display vectors and uses thereof
AU2012202068B2 (en) Multi-chain eukaryotic display vectors and uses thereof
AU2013203954B2 (en) Multi-chain eukaryotic display vectors and uses thereof
AU2011244861B2 (en) Recombination of nucleic acid library members

Legal Events

Date Code Title Description
AS Assignment

Owner name: DYAX CORPORATION, MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:HUFTON, SIMON E.;HOOGENBOOM, HENDRICUS R.J.M.;REEL/FRAME:013651/0740

Effective date: 20021113

STCB Information on status: application discontinuation

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