EP1163339A1 - Eine polypeptidstruktur zur verwendung als gerüst - Google Patents

Eine polypeptidstruktur zur verwendung als gerüst

Info

Publication number
EP1163339A1
EP1163339A1 EP99927740A EP99927740A EP1163339A1 EP 1163339 A1 EP1163339 A1 EP 1163339A1 EP 99927740 A EP99927740 A EP 99927740A EP 99927740 A EP99927740 A EP 99927740A EP 1163339 A1 EP1163339 A1 EP 1163339A1
Authority
EP
European Patent Office
Prior art keywords
seq
amino acid
sequence
loops
binding
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.)
Withdrawn
Application number
EP99927740A
Other languages
English (en)
French (fr)
Inventor
Johan Desmet
Simon Hufton
Hendricus Hoogenboom
Erwin Sablon
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.)
Fujirebio Europe NV SA
Original Assignee
Innogenetics NV SA
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Innogenetics NV SA filed Critical Innogenetics NV SA
Publication of EP1163339A1 publication Critical patent/EP1163339A1/de
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C40COMBINATORIAL TECHNOLOGY
    • C40BCOMBINATORIAL CHEMISTRY; LIBRARIES, e.g. CHEMICAL LIBRARIES
    • C40B40/00Libraries per se, e.g. arrays, mixtures
    • C40B40/02Libraries contained in or displayed by microorganisms, e.g. bacteria or animal cells; Libraries contained in or displayed by vectors, e.g. plasmids; Libraries containing only microorganisms or vectors
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/70521CD28, CD152
    • 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
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2318/00Antibody mimetics or scaffolds
    • C07K2318/20Antigen-binding scaffold molecules wherein the scaffold is not an immunoglobulin variable region or antibody mimetics
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide

Definitions

  • the present invention relates to a polypeptide chain having a ⁇ sandwich architecture which can be used as a scaffold, i.e. a supporting framework, carrying antigen- or receptor-binding fragments. More specifically, the present invention relates to a ⁇ sandwich derived from the naturally occuring extracellular domain of CTLA-4.
  • the primary amino acid sequence of a protein determines its three-dimensional (3D) structure, which in turn determines protein function.
  • 3D structure is essentially uneffected by amino acids at loci where the amino acid side chain is directed towards the solvent. Loci where limited variety is allowed have the side group directed toward other parts of the protein. This general rule, however, does not hold for those proteins whose function consists of, or relies upon binding with other biomolecules. Binding of such proteins to other molecules is non-covalently but often very tightly and specifically.
  • residues of the interacting domains that point towards the solvent becomes very important as well as the 3D-structure that they adopt, because binding results from complementarity of the surfaces that come into contact: bumps fit into holes, dipoles align and hydrophobic residues contact other hydrophobic residues.
  • Individual water molecules eventually can fill a well determined space in intermolecular interfaces, and then usually form hydrogen bonds to one or more atoms of the protein or other biomolecules or to other bound water.
  • the 3D structure can often be predicted on the basis of the primary amino acid sequence.
  • a major obstacle towards the correct prediction of the entire protein structure is the modelling of "loops". These are regions of the polypeptide chain that have a lower degree of structural and sequential regularity and where insertions and deletions often occur during evolution. Yet, this diversity does not fundamentally alter the core structure of the remaining polypeptide chain. Often such loops, which are on the outside of the protein structure, will determine the interaction with other biomolecules. The conformation of loop fragments is largely controlled by the attachment of their ends to fragments that constitute the framework of the molecule, for instance secondary structural units. This is also the basic idea when grafting such polypeptide chains onto the core of the protein structure which functions as a scaffold.
  • Immunoglobulins are multi domain proteins consisting of a heavy and a light chain.
  • the variable domain of immunoglobulins is composed of an antigen binding site formed by six loops clustered in space and carried by two domains that adopt a so called- ⁇ sandwich structure (one ⁇ domain from the heavy chain carrying 3 loops and one from the light chain carrying 3 loops) (Amit et al., 1986).
  • the high sequence variability of these loops allows immunoglobulins to recognise a variety of antigens.
  • the gross structure of the immunoglobulin variable domain's framework is hereby not altered.
  • the latter ⁇ -domains consisting of anti-parallel strands that are connected by the loops, function as a scaffold that strongly determines the three dimensional structure of the loops by constraining their structure and providing a carrier function (Amzel,L.M. and Poljak,R.J. , 1979; Williams A.F.,1987).
  • the ⁇ - sandwich architecture of both the heavy and light chains have a roughly half-cylindrical structure, with a more or less open side.
  • both domains (heavy and light chain) are packed onto eachother thereby forming a barrel-like fold.
  • minibody which consists of a fusion of the variable regions of an antibody and the CH3 domain of an immunoglobulin, all in a single polypeptide. Minibodies spontaneously dimerize to form an antibody fragment that is bivalent.
  • the minibody scaffold which contains a number of residue modifications with respect to the original immunoglobulin sequence, can be obtained both by solid- phase synthesis and by expression in bacteria (Bianchi et al, 1993; Bianchi et al ,1994). Further modifications of the latter molecule were successful in improving some important properties, for example its solubility in aqueous media (Bianchi et al ,1994). Other attempts to simplify the existing multi domain immunoglobulin molecule have resulted in recombinant fragments, made of two forms: Fv fragments and Fab's.
  • Fv fragments are heterodimers composed of a variable heavy chain (V H ) and a variable light chain (V L ) domain and are the smallest functional fragments of antibodies that maintain the binding and specificity of the whole antibody. In these constructs, the immunoglobulin domains forming the characteristic ⁇ -barrel can still interact with each other. However, Fv fragments are unstable. Stable Fv's have been produced by making recombinant molecules in which the V H and V L domains are connected by a peptide linker so that the antigen-binding site is regenerated in a single molecule. These recombinant molecules are termed single chain Fv's (scFv's; Raag and Whitlow, 1995).
  • V H -V L heterodimer can alternatively be stabilized by an interchain disulfide bond and is then termed dsFvs (Reiter et al, 1994).
  • Fab fragments are composed of the light chain and the heavy chain Fd fragment (V H and CHI) connected to each other via the interchain disulfide bond between CL and CHI.
  • each simplification of the otherwise complex multidomain immunoglobulin molecule still results in a unit wherein two immunoglobulin folds interact with each other thereby reconstituting the antigen binding site which consists of two pairs of three loops each grafted onto a separate ⁇ -sandwich framework.
  • Another drawback for using immunoglobulins to restrain randomized polypeptide sequences is the fact that the antigen-binding site of the latter molecules, which is constituted of six clustering loops, may not be able to bind small and/or cryptic sites within molecules. In other words, steric hindrance during the screening or selection for proteins that bind small and/or cryptic sites may occur when using an immunoglobulin framework.
  • the present invention relates to an alternative, simpler version of a monomeric polypeptide, compared to antibody-derived and small protein scaffolds, which can, to our surprise, efficiently be used as a scaffold.
  • both antibody-derived -and "small protein" scaffolds which are used to generate large peptide libraries using phage-display technology, are limited by the fact that phage display is not able to display active forms of proteins that require eukaryotic-specific posttranslational modifications (such -as glycosylation) for activity (Cannon et al., 1996). Also Metzler et al. (1997) indicated that glycosylation is important for binding activity, structural integrity and solubility of
  • an aim of the present invention to provide an alternative, simplier and preferably unglycosylated scaffold which is not easily cleared from the blood stream. It is also an aim of the present invention to provide a scaffold structure which is sufficiently stable to allow grafting of polypeptide chains as loops onto said scaffold without substantial alteration of the scaffold structure.
  • This aim more particularly relates to providing a scaffold structure that allows for the design of bispecific molecules wherein both binding domains are located on the opposite sides of the molecule.
  • Figure 1 shows a schematic representation of the scaffold of the present invention.
  • the six basic ⁇ -strands are named SI, S2, S3, S4, S5 and S6; the three additional or optional ⁇ -strands are named Al, A2 and A3.
  • the loops connecting the strands are indicated by a thin, black, curving line.
  • Figure 2 shows the sequence coding for SCA, which is based on the extracellular part of human CTLA-4 gene as described by Metzler et al. (1997) but without the C-terminal cysteine residue (Cys 123). ⁇ -sheets are underlined, Cysteine residues and glycosylation sites are shown. Mutations and there effect on CD80/86 binding are also indicated.
  • Figure 3 indicates the specific primers used to clone SCA as a ApaLllNotl fragment.
  • Figure 4 shows a shematic representation of the Phagemid pCESl construct.
  • Antibody genes V L -C L , variable (V) and constant (C) region of the light chain; V H -C H1 , variable and first constant region of the heavy chain; P/ ⁇ cZ, promoter; rbs, ribosome binding site; S, signal sequence; H6, six histidines stretch for IMAC purification; tag, c-myc-derived tag; amber, amber codon that allows production of soluble Fab fragments in non- suppressor strains, gUl, gene encoding one of the minor coat proteins of filamentous phage. Restriction sites used for cloning are indicated.
  • Figure 5 demonstrates the expected patterns of the BstNl fingerprints of clones with the correct sized insert.
  • Figure 6 demonstrates a strong signal showing binding to both B7.1-Ig and B7.2-Ig and not to BSA or plastic. The latter signal also decreases proportionately with the amount of phage present.
  • Figure 7 shows an SDS-page analysis with DTT (lanes 1 and 3) or without DTT (lanes 1 and 3) or without DTT (lanes 1 and 3)
  • Figure 8 indicates the specific primers used for amplification (oligol and oligo 2) and reamplification (oligo 1 and oligo 3) of the SCA.
  • Figure 9 schematic representation of the cloning strategy as described in example 6.
  • Figure 10 Diagrammic presentation of selection of phage on v ⁇ 3 integrin using a polyclonal phage ELISA (see also figure 10b for the enrichment factor of the several selection rounds).
  • FIG. 11 Diagrammic presentation of phage ELISA of selected clones on ⁇ v ⁇ 3 integrin
  • Figure 12 Alignment of the wild type CTLA-4 CDR3 loop and border sequences with the peptides obtained after selection and showing the RGD sequence and its selected borders
  • Figure 13 Diagrammic presentation of soluble ELISA of selected RGD clones on ⁇ v ⁇ 3 integrin
  • FIG. 14 Diagrammic presentation of binding of anti CTLA-4 antibodies with the scaffold derivatives
  • Figure 16 Diagrammic presentation of phage ELISA of scaffold derivatives with altered sequences in loops Al/Sl and S2/A2
  • the present invention relates to a scaffold composed of a single-chain polypeptide having the following structural properties:
  • - are connected by amino acid loops of variable conformation and lenght according to the following topology: S1-S2-S3-S4-S5-S6, and wherein at least one of said loops is different for at least three contiguous amino acid residues by way of substitution, deletion or insertion when compared to the naturally occurring CTLA-4 molecule, - form two ⁇ -sheets (one formed by S1/S4/S3 and one formed by
  • the present invention regards a scaffold as defined above which may contain a maximum of three additional ⁇ strands (Al, A2 and A3), and, which has the following topology: A1-S1-S2-A2-A3-S3-S4-S5-S6, and wherein at least one of said loops is different for at least three contiguous amino acid residues by way of substitution, deletion or insertion when compared to the naturally occurring CTLA-4 molecule, and, which possesses the following ⁇ -sheets: A1/S1/S4/S3 and S6/S5/S2/A2/A3.
  • the present invention also relates to a scaffold as defined above wherein : -Al is the amino acid sequence AQPAVVLA (SEQ ID 1) or any functionally equivalent derivative of said sequence, -SI is the amino acid sequence ASFPVEY (SEQ ID 2) or any functionally equivalent derivative of said sequence, -S2 is the amino acid sequence EVRVTVLRQA (SEQ ID 3) or any functionally equivalent derivative of said sequence,
  • -A2 is the amino acid sequence QVTEVCAA (SEQ ID 4) or any functionally equivalent derivative of said sequence,
  • -A3 is the amino acid sequence TYMMGNELTFLDDS (SED ID 5) or any functionally equivalent derivative of said sequence
  • -S3 is the amino acid sequence ICTGTSS (SEQ ID 6) or any functionally equivalent derivative of said sequence
  • -S4 is the amino acid sequence QVNLTIQ (SEQ ID 7) or any functionally equivalent derivative of said sequence
  • - S5 is the amino acid sequence GLYICKVE ( SEQ ID 8) or any functionally equivalent derivative of said sequence,
  • - S6 is the amino acid sequence GIGNGTQIY (SEQ ID 9) or any functionally equivalent derivative of said sequence.
  • the applicability of a scaffold lies in the ability to introduce diversity without destroying the tertiary structure of the protein fold and the ability to recover binding molecules from a diverse repertoire. The latter can be achieved by phage display and affinity selection on the ligand of choice.
  • properties of a scaffold are largely determined by the nature of the application, a list of important properties is shown below:
  • the scaffold should be a small, globular protein
  • -it should be a single domain (thus easier to produce, purify and engineer into multivalent or multispecific reagents), -it should fold correctly and the tertiary structure should not be perturbed by the introduction of diversity,
  • -it should be stable, preferably in vivo if that is of relevance for the chosen application, -it should have permissive loops , patches and/or surfaces for introducing diversity at a number of chosen sites wherein the nature of the required binding surface depends on the application,
  • -it should be engineerable to make monospecific/bispecific/trispecific or multispecific molecules -soluble and expressed well in e.g bacteria, -it should allow fusion at the N-and/or C-terminus,
  • -it should be preferably non-immunogenic and human if to be used therapeutically -and it should resistant to proteolysis.
  • the disadvantages when antibodies are used as scaffolds are that: -the binding surface is composed of two domains,
  • the disadvantages cited above are not encountered when the molecule of the present invention is used as a scaffold.
  • the molecule of the present invention has, in comparison to antibodies, the following advantages for use as a scaffold: -it has monomeric nature which facilitates easier engineering of therapeutic reagents and allows engineering into a multivalent and/or multispecific molecule, -it has improved stability and pharmacokinetics in comparison to recombinant antibody fragments due to the second non-Ig disulphide bridge, -it has a binding domain consisting of at most three loops (which is half the amount of loops that constitute the binding domains of human antibodies), which enables it to bind to cryptic sites that are otherwise not accessible by antibodies.
  • the latter "smaller" binding domain of the current invention also decreases the possibility of binding to other sites thereby decreasing the possibility of aspecific binding, and in addition it also helps in the design of small molecule mimetics, which is not the case for antibodies. -it allows for replacement of loops on the other side of the molecule (the opposite site of the ⁇ -sandwich of where the CDR loops are located) allowing an easy design of bispecific monomeric molecules.
  • the basic design of the molecule of the present invention is of human origin (the scaffold of the present invention can be considered to be derived from the naturally occurring extracellular domain of the molecule CTLA-4), so that therapeutic applications can be envisaged, reducing the risk of immunogenicity.
  • the molecule of the present invention also has been shown to function as a Fc molecule which suggests it can be engineered into a dimeric molecule that can recruit effector function, -
  • the molecule of the present invention is a very small molecule when compared to the bispecific molecules that have been described in the art and that have been designed to bind two different molecules or substances. This small size results in the advantage that if the molecule of the current invention is used to design a bispecific molecule that the two sites to which this molecule will bind will be brought into a very close proximity which has advantages in for instance drug targetting.
  • the scaffold of the present invention can be prepared by any method known in the art such as classical chemical synthesis, as described by Houbenweyl (1974) and Atherton & Shepard (1989), or by means of recombinant DNA techniques as described by Maniatis et al. (1982).
  • the number of amino acid per turn of the helix is 3.61.
  • the pitch (repeat distance) of the helix is 0.541 nm.
  • beta ( ⁇ ) strand refers to the conformation of a polypeptide segment where the main chain adopts an extended conformation (phi- and psi-angles around -120° and +120°, respectively) leading to a relatively linear structure.
  • a beta strand can be interrupted by one or two non-extended residues (for instance a beta bulge), while still be considered as a single strand, as long as the principal axes of the two consecutive sub- fragments do not form an angle greater than 90°.
  • beta ( ⁇ ) sandwich architecture refers to two beta-sheets that pack against eachother and in which the strands of both sheets form a so-called relative 'twist angle' of around 30° (within the range 20-50°).
  • any functionally equivalent derivative of said sequences refer to any variant or fragment of the amino acid (aa) sequences represented by SEQ ID 1 to 9 which does not result in a significant alteration of the global structural and functional properties of the scaffold of the present invention.
  • the latter terms include, post-translational modifications of the aa sequences represented by SEQ ID 1 to 9 such as glycosylation, acetylation, phosphorylation, modifications with fatty acids and the like.
  • amino acid sequences containing one or more analogues of an aa include unnatural aa's
  • amino acid sequences with substituted linkages include mutated versions of the amino acid sequences, peptides containing disulfide bonds between cysteine residues, biotinylated amino acid sequences as well as other modifications known in the art.
  • amino acid sequences containing one or more analogues of an aa including unnatural aa's
  • amino acid sequences with substituted linkages include mutated versions of the amino acid sequences, peptides containing disulfide bonds between cysteine residues, biotinylated amino acid sequences as well as other modifications known in the art.
  • amino acid sequences containing one or more analogues of an aa including unnatural aa's
  • amino acid sequences with substituted linkages include mutated versions of the amino acid sequences, peptides containing disulfide bonds between cysteine residues, biotinylated amino acid sequence
  • KQSPMLVAY (SEQ ID 15), KQLPRLVVY (SEQ ID 16), AQRPLLIVA (SEQ ID 17), TQSPAIMSA (SEQ ID 18), QESGPGLV (SEQ ID 19) and the like, -for SI, the amino acid sequences ASFPCEY (SEQ ID 20), ASFSCEY (SEQ ID 21), VSLSCRY (SEQ ID 22), VNLSCKY (SEQ ID 23), ATLVCNY (SEQ ID 24, VTMTCSA (SEQ ID 25), VTMTCKS (SEQ ID 26), VTITCKA (SEQ ID 27), VTMSCKS (SEQ ID
  • EFRASLYKGV SEQ ID 41
  • EFRASLYKGA SEQ ID 42
  • EFRASLHKGL SEQ ID 43
  • EFRASLHKGT SEQ ID 44
  • EVRMYMYQQK SEQ ID 45
  • KNYLTWYQQK SEQ ID 46
  • EVRVVWYQQK SEQ ID 47
  • NFRLAWYQQK SEQ ID 48
  • KNFLAWYQQK SEQ ID 49
  • EVRMSWVRQS SEQ ID 50
  • EVRMEWVRQP SEQ ID 51
  • EVRMSWVRHT SEQ ID 52
  • EVRMHWVRQA SEQ ID 53
  • EVRVNWVRQP SEQ ID 54
  • EVRMNWVKQR SEQ ID 55
  • EVRIEWVKQR SEQ ID 56
  • EVRVHWVRQS SEQ ID 57
  • EVRIEWVKER SEQ ID 58
  • SPRLLIYD SEQ ID 67
  • SPKLLIYW SEQ ID 68
  • PPKLLIYG SEQ ID 69
  • GLEWVAEI SEQ ID 70
  • RLEWIAAS SEQ ID 71
  • RLEWVATI SEQ ID 72
  • GLEWVAYI SEQ ID 73
  • GLEWLGMI SEQ ID 74
  • GLEWIGRI SEQ ID 75
  • GLEWIGWI SEQ ID 76
  • GLEWLGMI SEQ ID 77
  • GLEWIGEI SEQ ID 78
  • GLEWIGEI SEQ ID 79
  • TFTVKNTLGFLDDP (SEQ ID 80), TFTEKNTVGFLDYP (SEQ ID 81), TYMVEDELTFLDDS (SEQ ID 82), TYTVENELTFTDDS (SEQ ID 83), NGNFTYQPQFRSNAEF (SEQ ID 84), NGNFTYQPQFRPNVGF (SEQ ID 85), NGNFSHPHQFHSTTGF (SEQ ID 86), NGNHSHPLQSHTNKEF (SEQ ID 87), YGNYSQQLQVYSKTGF (SEQ ID 88),
  • NGNYSHQPQFYSSTGF SEQ ID 89
  • SWNMTHKTNSNSNKEF SEQ ID 90
  • TSNLASGVPV SEQ ID 91
  • ASTRESGVPD SEQ ID 92
  • ASTRHIGVPD SEQ ID 93
  • RLNSDNFATHYAESVKG SEQ ID 94
  • RNKGNKYTTEYSASVKG SEQ ID 95
  • SNGGGYTYYQDSVKG SEQ ID 96
  • SSGSSTLHYADTVKG SEQ ID 97
  • WGDGNTDYNSALKS SEQ ID 98
  • DPANGNIQYDPKFRG SEQ ID 99
  • YPGSGNTKYNEKFKG (SEQ ID 100), LPGSGSTNYNEKFKG (SEQ ID 101), WGGGSIEYNPALKS (SEQ ID 102), LPGSGRTNYREKFKG (SEQ ID 103) and the like, -for S3, the amino acid sequences FCSGTFN (SEQ ID 104), TCIGTSR (SEQ ID 105), TCTGISH (SEQ ID 106), NCDGDFD (SEQ ID 107), NCDGNFD (SEQ ID 108),
  • NCDGKLG SEQ ID 109
  • NCTVKVG SEQ ID 110
  • NCDGKLG SEQ ID 111
  • DCDGKLG SEQ ID 112
  • NCRGIHD SEQ ID 113
  • RFSGSGS SEQ ID 114
  • RFTGSGS SEQ ID 115
  • RFAGSGS SEQ ID 116
  • KFHSRD SEQ ID 117
  • RFIVSRD SEQ ID 118
  • RFTISRD SEQ ID 119
  • RFTISRD SEQ ID 120
  • RLSISFD SEQ ID 121
  • KATITAD SEQ ID 122
  • KATLTVD SEQ ID I23
  • RLSISKD SEQ ID 124
  • KATFTAD SEQ ID 125
  • the scaffold of the present invention contains at least six ⁇ - strands (SI to S6) and may, but does not have to, comprise one (Al or A2 or A3), preferably two (Al and A2, or, Al and A3, or A2 and A3), and most preferably three (Al and A2 and A3) additional ⁇ -strands (Al to A3).
  • amino acid loops of variable conformation and length refer to any aa sequence of any lenght and any conformation which is able to connect the ⁇ -strands of the current invention.
  • the latter aa loops preferably contain at least one aa fragment which binds to a receptor or antigen (see further).
  • receptor/antigen binding fragment s
  • one out of the 6 loops may contain an above-indicated fragment; preferably, two out of the 6 loops contain an above-indicated fragment and more preferably, three out of the 6 loops contain an above-indicated fragment.
  • four, five and six out of the 6 loops may contain an above-indicated fragment.
  • the scaffold of the present invention may contain sets of fragments which are able to bind to more than one, preferably two, different receptors/antigens.
  • the loops which connect S5 and S6, SI and S2, S3 and S4, and, A2 and A3 contain fragments binding to a tumor antigen or B7.1/B7.2 expressing cells and the loops connecting A3 and S3, S2 and A2, S4 and S5, and, Al and B contain fragments binding to a toxin able to kill the tumor cell expressing said tumor antigen or said B7.1 B7.2 expressing cells in a manner similar as the one described in WO91/07437 to Pfreundschuh and WO96/40260 to De Boer & De Gast, respectively.
  • SSRGV SEQ ID 183
  • SSRGV SEQ ID 184
  • SNE SEQ ID 185
  • NNE SEQ ID 186
  • DNE SEQ ID 187
  • DNA SEQ ID 188
  • SPGEK SEQ ID 189
  • QPGGS SEQ ID 190
  • SYNLLAK SEQ ID 196
  • TYNLFSK SEQ ID 197
  • SYNLFSR SEQ ID 198
  • TYNGTGK SEQ ID 199
  • ND SEQ ID 200
  • DS SEQ ID 201
  • GS SEQ ID 202
  • NS SEQ ID 203
  • PGS SEQ ID 204
  • PGQ SEQ ID 205
  • PEK SEQ ID 206
  • PGK SEQ H
  • PDK PDK
  • ES SEQ ID 209
  • GN SEQ ID 210
  • NE SEQ ID 211
  • KD SEQ ID 212
  • GT SEQ ID 213
  • DSKS SEQ ID 214
  • TSQS SEQ ID 215
  • NAKN SEQ ID 216
  • NPKN NPKN
  • - loops connecting said ⁇ -strands S4 and S5 GLRAVDT (SEQ ID 218), GLRAADT (SEQ ID 219), GLRAMDT (SEQ ID 220), GLSAMDT (SEQ ID 221), NLHVNHT (SEQ ID 222), NLDVNHT (SEQ ID 223), NLYVNQT (SEQ ID 224), DLYVNQT (SEQ ID 225), NLFVNQT (SEQ ID 226), NMSASQT (SEQ ID 227), and the like, - loops connecting said ⁇ -strands S5 and S6: LMYPPPYFV (SEQ ID 228), LMYPPPYYL (SEQ ID 229), LMYPPPYYV (SEQ ID 230), FMYPPPYLDN (SEQ ID 231), VMYPPPYLDN (SEQ ID 232), VLYPPPYIDN (SEQ ID 233), VMYPPPYIGN
  • homologous and homologous are used in the current invention as synonyms for “identical” and “identity”; this means that the amino acid sequences which are e.g. said to be 95 % homologous show 95 % identical amino acids in the same position upon alignment of the sequences.
  • CTLA-4 molecules as such, nor molecules derived from CTLA-4 by means of single amino acid substitutions with the intention to increase solubility or alter the binding affinity and/or binding specificity of a CTLA-4 molecule for B7-molecules, the latter being present on the outer surface of antigen presenting cells.
  • the structure as set out above therefore should have less then 98% homology with the amino acid sequence of the naturally occurring CTLA-4-molecule, preferably less then 97%, more preferably less then 95% and even more preferably less then 90% homology with the amino acid sequence of the naturally occurring CTLA-4-molecule.
  • the amino acid sequence of at least one of the loops is different as compared to the amino acid sequence of the naturally occurring CTLA-4 molecule, preferably two loops and even more preferably three loops.
  • a loop is considered different if at least three, more preferably four, and even more preferably five contiguous amino acid residues of the loop are absent (deletion), additionally present (insertion), or altered (substitution) when compared to the naturally occurring CTLA-4 molecule.
  • the loop which is different as compared to the naturally occurring CTLA-4 molecule is one of the loops which is not CDRl nor CDR2 nor CDR3 when one would take the naturally occurring CTLA-4 molecule as a reference.
  • This preferred embodiment therefore refers to the replacement of at least one of the loops which are connecting the Al and S 1, or S2 and A2, or A3 and S3, or S4 and S5 ⁇ -strands, or which are connecting the Al and SI, or S2 and S3, or S4 and S5 ⁇ -strands of the scaffold if the additional ⁇ -strands A2 and A3 are ommitted.
  • Replacement of these loops with randomized peptide sequences can not be considered to be analogous to the replacement of loops in Ig-like proteins as described in the art.
  • the molecule of the present invention is not only different, moreover, the loops being referred to are located on the other side of the molecule as the CDRl, CDR2 and CDR3 loops which can be related to the antigen determining region of Ig-like proteins.
  • the loop which is different as compared to the naturally occurring CTLA-4 molecule is one of the loops which is CDRl or CDR2 or CDR3 when one would take CTLA-4 as a reference.
  • This preferred embodiment more precisely refers to the replacement of at least one of the loops which are connecting the
  • the present invention further relates to a scaffold as defined above wherein said amino acid loops comprise fragments binding to a receptor or antigen.
  • fragment binding to a receptor or antigen refers to any possible aa sequence which is part of the said loops (i.e. said fragments comprise maximally as many aa's as the loop itself and comprise, more frequently, less aa's than the loop wherefrom they are derived) and which binds any receptor or antigen.
  • the scaffold of the present invention can carry any randomized aa sequence.
  • the present invention concerns a single-chain polypeptide as defined above for use as a scaffold.
  • the scaffold of the present invention can be used to generate large peptide libraries which can be screened for peptides with desired binding characteristics as described in US 5223409 to Ladner et al. and US 5571698 to Ladner et al.
  • a phage based system is used in which each randomized peptide is fused to the gene HI protein of the M13 phage.
  • non-phage based systems such as screening peptides on polysomes or on the surface of E. coli (Tuerk &
  • the scaffold of the present invention carrying carrying peptides selected from randomized aa sequences can be used for therapeutic, diagnostic, and related purposes.
  • the targets or ligands bound by the recombinant proteins of the present invention can be proteins, nucleic acids, lipids and carbohydrates, or combinations thereof. It has to be understood that 'combinations thereof ' refers to glycoproteins, lipoproteins, and the like but also refers to dimeric (homo- or heterodimeric) biomolecules that are bound to each other by non-covalent means. Therefore, the term 'antigen' as used throughout the specification, has to be interpreted in the broadest sense. Because one can use an artificial screening system, the man skilled in the art will also appreciate that recombinant proteins can also be selected or screened for binding to xenobiotic molecules, that would otherwise be highly toxic for a biological system.
  • the invention also relates to those recombinant proteins that are able to bind, as already indicated above, two entirely unrelated biomolecules or substances, by means of two binding surfaces, e.g. one surface consisting of the loops connecting the ⁇ -strands on one side of the ⁇ -sandwich and comprising the CDR loops in the natural protein, and one surface consisting of the loops that connect the ⁇ -strands on the other side of the ⁇ -sandwich.
  • two binding surfaces e.g. one surface consisting of the loops connecting the ⁇ -strands on one side of the ⁇ -sandwich and comprising the CDR loops in the natural protein, and one surface consisting of the loops that connect the ⁇ -strands on the other side of the ⁇ -sandwich.
  • both binding surfaces can operate independent from each other and selection or screening for such proteins can be achieved in two separate rounds: - in the first round, the biomolecule that binds to one partner is retrieved from a library wherein the amino acid sequence of one set of loops has been randomized while the other set of loops is untouched e.g. as in the naturally occurring protein, - in the second round, the biomolecule that binds to a second partner is retrieved from a library wherein the amino acid sequence of the other set of loops has been randomized.
  • the randomized library may already contain those loops that bind for the first partner or, alternatively, remain untouched as in the naturally occurring protein.
  • bispecific molecules provide key tools- in therapeutic and diagnostic procedures such as immunohistochemistry (see example 11) and enzyme immunoassays, radiotherapy and immunotherapy, drug targetting, and for redirecting biomolecules or certain cell types to new sites.
  • One binding partner can be used to target for certain cell types such as cancer cells inducing intracellular uptake, while the other binding partner can be a component of a life sustaining cellular mechanism of which the function is inhibited upon binding.
  • One of the binding sites can also be a cytotoxic molecule, thereby forcing uptake of the cytotoxic molecule into the targeted cells.
  • This strategy can also be used to force uptake of endogeneous molecules, such as auto-immune antibodies, toward cell types, such as liver cells.
  • One binding partner can be used to target for certain cell types, while the other binding partner can be used to target for other cell types thereby forcing both cell types to stay in close proximity, such as any lymphocytes with for instance lymphocytes of a different type or with for instance cancer cells.
  • the molecule of the current invention has the additional advantage that it is a much smaller molecule then the bispecific antibodies already described in the prior art.
  • the molecule of the present invention has been provided with a binding domain specific for a site on the targeted cells and a second CD3 domain or a CD3 like domain that recruits and activates the cytotoxic T lymphocytes.
  • one of the binding sites of the scaffold itself can also be used to function as a spacer or anchor in cases wherein immobilisation of the entire protein is desired, such as on membranes or on any carrier for chromatographic purposes, thereby allowing for a directed immobilisation.
  • One of the loops can for instance comprise a repeat of Histidine residues or a His-tag thereby allowing anchorage or immobilisation to a carrier or membrane by means of a metal-chelate or by means of covalent linkage as for instance is described in patent application PCT EP 98/03883.
  • the man skilled in the art will understand that it is not a prerequisite that the two binding domains bind to different molecules. Both binding domains might as well bind identical molecules, thereby providing for instance a device for use in agglutination tests.
  • One of the binding domains can be selected for recognizing red blood cells, while the other domain can be selected for binding to disease-specific antibodies, thereby providing a device for agglutination tests.
  • 'His-tag' is used throughout the specification, a broad definition can be given to it, meaning any sequence that has an amount of histidine residues that is sufficiently high to allow appropriate application. In principle a 10 amino acids long sequence that has two histidine residues, and is repeated at least twice should suffice. More typical, said 'His- tag' contains at least three histidines within a stretch of 6 amino acids.
  • the term 'His-tag' can also include any repetition of a histidine immediately followed by at least one residue.
  • His-tag refers to at least three consecutive histidine residues, more preferably to at least four histidine residues, or even more preferably to at least five or at least six consecutive histidine residues.
  • the present invention relates to the introduction of an RGD sequence, embedded within a randomized peptide sequence, within one of the loops of the scaffold, thereby making up a peptide library of RGD displaying scaffold molecules that can be selected for binding to integrin.
  • Integrins are involved in a number of pathological processes that involve angiogenesis e.g thrombosis, osteoporosis, rheumatoid artheritus, diabetic retinopathy, cancer and atherosclerosis.
  • ⁇ v ⁇ 3 integrins are unique markers for activated angiogenic endothelium. As such they are ideal targets for antibody-based endothelial cell destruction.
  • integrins on the tumor endothelial cell surface will have application as both imaging reagents, as antitumor reagents and targets for gene therapy vectors which can readily access the tumor via the tumor vasculature.
  • a number of peptides which can specifically target the tumor endothelium have been discovered (for example RGD peptides which bind to the v ⁇ 3 integrins).
  • RGD peptides which bind to the v ⁇ 3 integrins have been discovered (for example RGD peptides which bind to the v ⁇ 3 integrins).
  • Previous studies have clearly shown that peptides containing the RGD motif can specifically target v ⁇ 3 integrins (Pasqualini et al., 1995; Pasqualini et al.,
  • the present invention also regards, as stated above, a scaffold as defined above wherein said single-chain polypeptide is unglycosylated.
  • the scaffold of the present invention can also be glycosylated.
  • one of the major advantages of the claimed scaffolds is the fact that they remain functional without glycosylation, allowing expression in bacterial systems, the fact of being unglycosylated is not a prerequisite for falling under the protected matter.
  • Example 1 Cloning of a recombinant scaffold, named SCA, for display on the surface of filamentous phage
  • SCA The sequence coding for SCA, which is based on the extracellular part of human CTLA-4 gene as described by Metzler et al (1997) but without the C-terminal cysteine residue (Cys 123), is cloned into the phage display vector pCES-1 (figure 4).
  • the cysteine residue is not included because it may present some problems with the correct folding and presentation of the SCA- p3 fusion on filamentous phage.
  • This cloning creates a translational fusion of SCA to the N-terminus of p3 for display on filamentous phage.
  • SCA is cloned as a ApdLl/Notl fragment (neither of these restriction sites are present in human CTLA-4) using specific primers (figure 3).
  • Human CTLA-4 is amplified from
  • Vector pCESl is also digested with ApaLl and Notl. Both digested vector and digested SCA- insert are purified. Ligation is performed overnight at 16°C and is transformed into TGI. The BstNl fingerprints of clones with the correct sized insert, as assessed by PCR, demonstrate the expected patterns (Figure 5).
  • Example 2 Display of a recombinant, non-glycosylated and monomeric SCA- protein on the surface of filamentous phage
  • Phage displaying non-glycosylated SCA proteins on their surface are prepared as follows. Clones 2, 4 and 5 from figure 4 are grown to an OD of 0.5 in the presence of ampicillin at lOO ⁇ g/ml and 2% glucose. Five mis of culture is infected with helper phage
  • M13KO7 at a multiplicity of infection of 20:1 and incubated at 37°C for 30 minutes.
  • Cells are resuspended in TY medium plus Amplicillin and Kanamycin (25 ⁇ g/ml) and grown overnight at 30°C. Phages are recovered by precipitation by PEG and resuspended in PBS. Because the SCA protein is derived from the human CTLA-4 protein wherein the original CDR loops are retained, a binding is expected with B7.1 and B7.2 proteins.
  • Example 3 Soluble SCA - His6/myc expressed in the non-suppressor strain HB2151
  • the gene coding for SCA, preceded by the pelB signal sequence, is cloned into the phage display vector pCES-1 (figure 4) under control of the EPTG inducible lac- promoter.
  • the coding sequence is then fused C-terminally to a His6 followed with a myc-tag.
  • An amber stopcodon is located at the junction between this coding sequence and the sequence coding for the phage coat protein g3p.
  • Non- suppressor cells HB2151 are transformed with the His6/myc- tagged construct and grown in liquid LB medium with ampicilline for selection and 1% glucose for metabolic repression of the lac-promoter at 28 °C until saturation is reached.
  • This saturated culture is subsequently diluted 20 times in 20 ml LB+A+G and grown at 28 °C until an OD600 is reached of 0.5 to 0.6, which takes about three hours.
  • the glucose containing medium is then removed and replaced by LB+A and 0.1 mM IPTG.
  • the induced culture is further incubated for about 20 hours. The next day the OD600 is measured and the cells are fractionated.
  • a periplasmic fraction is prepared using a modified protocol as according to the osmotic shock procedure described by Neu and Heppel (1965).
  • the soluble SCA-molecule is tested for binding capacity to the human B7.1 and B7.2 molecules.
  • the EBV-transformed B cell line (RPMI 8866, 0.5-lx 10 5 cells/sample), which strongly expresses B7.1 and B7.2, is incubated for 20 min at 4°C with the soluble SCA-molecule. After washing twice in RPMI 1640 supplemented with 10% FCS, the cells are incubated for another 20 min at 4°C with mouse anti-cMyc mAb (l ⁇ g/cell pellet) conjugated to biotine. After washing twice in RPMI 1640 supplemented with 10% FCS, the cells are incubated for another 20 min at 4°C with Streptavidin conjugated to PE (Phycoerythrine).
  • the cells are then washed twice in RPMI 1640 supplemented with 10% FCS and finally suspended in PBS supplemented with 1% BSA and 0.1% NaN 3 and analyzed with a FACScan flow cytometer (Becton Dickinson).
  • the specific binding of the SCA-molecule is expressed as the mean fluorescent intensity in arbitrary units.
  • the results show that soluble, non-glycosylated SCA binds, in a dose- dependent way, to the RPMI 8866 cells which express B7.1 and B7.2 (results not shown).
  • RGD containing sequence that binds to integrins is inserted such that it replaces the CDR3 loop of CTLA-4. Sequences are also introduced such that they replace the Al/Sl loop and the S2/A2 loops of CTLA-4 (see example 16 and 17). These loops are situated at the side of the molecule opposite to the CDR sequences which mediate the binding of CTLA-4 to its natural ligands.
  • a fragment corresponding to CTLA-4 ( Figure 9) is digested from pCESl as an ApaLl /Notl fragment.
  • the template CTLA-4 DNA is then subjected to PCR with the primers oligo#l and oligo#2 ( Figure 8).
  • PCR product is then purified and reamplified with oligo#l and oligo#3. Fragment DNA is recovered and digested with ApaLl and Notl and cloned into pCESl for phage display ( Figure 9). In this way, a library of 1.3 x 10 7 independant clones can be obtained.
  • Example 7 Selection of repertoire on recombinant human ⁇ v ⁇ 3 integrin Three rounds of selection are performed on recombinant v ⁇ 3 integrin coated onto immunotubes at lO ⁇ g/ml ( Figure 10 a and b). Polyclonal phage ELISA is then performed to monitor the selection using input phage for 1 2 and 3 rounds of selection. As can be seen initially the repertoire has reactivity to B7-lIg representing the probable presence of wild-type CTLA-4 in the repertoire. As the selection progresses this disappears and gives way to a positive signal on integrin ( Figure 11). Selected clones are tested for binding to antigen in phage ELISA as in (Marks et al., 1991).
  • Phage ELISA is performed on ccv ⁇ 3 integrin (l ⁇ g/ml), B7-lIg (l ⁇ g/ml) and BSA (l ⁇ g/ml). In all cases all clones are negative on B7-lIg and BSA. ( Figure 10).
  • Positive clones are sequenced and some sequences are shown in ( Figure 12) by way of example. Also, another pointmutation is shown to be present in the CDR2 loop if the CTLA-4 molecule is taken as a reference, further demonstrating another permissive site.
  • Soluble CTLA-4 RGD fragments are prepared as in Marks et al. (1991) and tested for binding to v ⁇ 3 integrin (l ⁇ g/ml) either as monomeric reagent or cross linked via 9E10 to generate avidity. As can be seen, clones are positive in soluble ELISA ( Figure
  • a positive control is the mouse monoclonal antibody to ⁇ v ⁇ 3 integrin, LM609 (Brooks et al., 1994).
  • a negative control is a non binding clone as identified by phage ELISA.
  • an ELISA can be performed using mouse polyclonal anti CTLA-4 serum and a monoclonal antibody to CTLA-4 (IGH310).
  • This antibody is supposed to compete with B7-1/B7-2 so it has partial epitope overlap with the MYPPPY sequence of wild-type CTLA-4.
  • 5 x 10 11 phage are coated directly in PBS overnight at 4 ° C and are challenged with either mouse polyclonal anti-CTLA-4 or mouse monoclonal anti-CTLA-4.
  • clones 7B, 8H and 11C and wild-type CTLA-4 are all positive with both antibodies.
  • the signal obtained with the monoclonal antibody is greater than that obtained with the polyclonal serum. Furthermore the signal obtained with the RGD presenting clones in CDR3 is greater than that obtained with CTLA-4. This indicates that the general structure of SCA molecule remains intact after introduction of foreign sequences into CDR3 due to the positive signal obtained with the conformation dependant monoclonal anti-CTLA-4. ( Figure 14)
  • Example 11 FACS analysis of binding to HUVECS cells
  • Approximately 80,000 HUVECS cells/sample are cultured and are tested in FACS for binding. As can be seen a positive signal is seen with the positive clone . Whereas with both empty pCES 1 vector and wild-type CTLA-4 phage no shift can be observed.
  • This vector is cut with Notl and Seal and a 2183 bp fragment is isolated and ligated with a 2654 bp Notl Scal fragment originating from vector pCANTAB5E (Amersham Pharmacia, Uppsala, Sweden).
  • the resulting vector pCESlCTLA4E differs from the previously described vector pCESlCTLA4 in that the CTLA4 molecule now carries a C-terminal E-tag in stead of a double c-myc/His6 tag.
  • the correct sequence of the vector is then verified by DNA sequencing.
  • Example 13 Display of the recombinant SCA-E protein on the surface of filamentous phage
  • Phage displaying SCA-E protein on their surface are prepared as described in example 2 (Marks et al., 1991). To demonstrate binding of the SCA-E protein to B7.1Ig, B7.2Ig and an anti E-tag antibody (Amersham Pharmacia, Uppsala, Sweden), phages are titrated and a serial dilution is made to test for binding in ELISA to these molecules.
  • Example 14 Insertion of an integrin binding sequence (RGD) in the loop formed by ⁇ -sheets Al and SI in SCA-E
  • the DNA coding for the integrin binding sequence is inserted in the loop formed by ⁇ -sheets Al and SI using overlap extension PCR (Ho et al., 1989).
  • the RGD sequence is inserted between the SS and the RGI residues.
  • the RGD sequence is flanked by a G4S linker sequence. This insertion can be accomplished by performing two PCR reactions using pCESlCTLA4E plasmid DNA as a template. A first PCR, using primers Nos.
  • Example 15 Insertion of a His6 tag (HHHHHH) in the loop formed by ⁇ -sheets Al and SI in SCA-E
  • the HHHHHH sequence is inserted between the SS and the RGI residues. On both sides, the HHHHHH sequence is flanked by a G4S linker sequence.
  • This insertion can be accomplished by performing two PCR reactions using pCES 1CTLA4E plasmid DNA as a template. A first PCR, using primers Nos. 11209 and
  • PCR 11069 amplifies the 5' end of the CTLA4E gene including the HHHHHH sequence.
  • the second PCR using primers Nos. 11070 and 11210, amplifies the 3' end of the CTLA4E gene including the HHHHHH sequence.
  • Both PCR fragments are subsequently gelpurified and an additional PCR is performed on the mixture of both PCR fragments using primers Nos. 11209 and 11210.
  • the resulting PCR fragment is gelpurified, cut with ApaLl and Notl, and inserted into the ApaLI/Notl opened vector pCESlCTLA4E.
  • the resulting vector pCESlCTLA4E-Al/SlHis6 is verified by DNA sequencing.
  • No.11070 SEQ ID 241: 5'-CACCATCACCATCACCACGGTGGCGGTGGCTCTCGAGGCATCGCCAGCT
  • Example 16 Insertion of an integrin binding sequence (RGD) in the loop formed by ⁇ -sheets S2 and A2 in SCA-E
  • plasmid pCESlCTLA4E see example 3
  • the DNA coding for the integrin binding sequence (RGD) is inserted in the loop formed by ⁇ -sheets S2 and A21 using overlap extension PCR (Ho et al., 1989).
  • the S2/A2 loop consisting of residues DS (see figure 2)
  • the RGD sequence is inserted between the D and the S residue. No flanking sequences are present. This insertion can be accomplished by performing two PCR reactions using pCESlCTLA4E plasmid DNA as a template.
  • a first PCR using primers Nos. 11209 and 11079, amplifies the 5' end of the CTLA4E gene including the RGD sequence.
  • the second PCR using primers Nos. 11080 and 11210, amplifies the 3' end of the CTLA4E gene including the RGD sequence.
  • Both PCR fragments are subsequently gelpurified and an additional PCR is performed on the mixture of both PCR fragments using primers Nos. 11209 and 11210.
  • the resulting PCR fragment is gelpurified, cut with ApaLl and Notl, and inserted into the ApaLI/Notl opened vector pCESlCTLA4E.
  • the resulting vector pCESlCTLA4E-S2/A2RGD is verified by DNA sequencing.
  • Example 17 Insertion of a His6 tag (HHHHHH) in the loop formed by ⁇ -sheets S2 and Al in SCA-E
  • the HHHHHH sequence is inserted between the D and the S residue. No flanking sequences are present. This insertion can be accomplished by performing two
  • a first PCR using primers Nos. 11209 and 11075, amplifies the 5' end of the CTLA4E gene including the
  • the second PCR using primers Nos. 11077 and 11210, amplifies the 3' end of the CTLA4E gene including the HHHHHH sequence. Both PCR fragments are subsequently gelpurified and an additional PCR is performed on the mixture of both
  • PCR fragments using primers Nos. 11209 and 11210.
  • the resulting PCR fragment is gelpurified, cut with ApaLl and Notl, and inserted into the ApaLI/Notl opened vector pCESlCTLA4E.
  • the resulting vector pCESlCTLA4E-S2/A2His6 is verified by DNA sequencing.
  • Example 18 SCA-E Al/Sl His6 and SCA-E S2/A2 His6 displayed on phage are able to bind coated anti His6-tag antibodies in ELISA
  • Phage displaying SCA-E Al/Sl His6 or SCA-E S2/A2 His6 protein on their surface were prepared as described by Marks et al. (1991). To demonstrate binding of the SCA-E Al/Sl His6 or SCA-E S2/A2 His6 protein to an anti His6-tag antibody (BabCO, Richmond, CA, USA), phages are titrated and a serial dilution is made to test for binding in ELISA to these molecules. Significant signals were obtained showing binding to the anti His6-tag antibody.
  • Example 19 SCA-E Al/Sl His6 and SCA-E S2/A2 His6 displayed on phage are able to bind Ni 2+ coated plates in ELISA
  • Phage displaying SCA-E Al/Sl His6 or SCA-E S2/A2 His6 protein on their surface were prepared as described by Marks et al. (1991). To demonstrate binding of the SCA-E Al/Sl His6 or SCA-E S2/A2 His6 protein to Ni-NTA HisSorbTM Plates (Qiagen GmbH, Hilden, Germany), phages are titrated and a serial dilution is made to test for binding to these plates. Significant signals were obtained showing binding to the
  • Example 20 SCA-E Al/Sl His6 and SCA-E S2/A2 His6 displayed on phage are still able to bind coated recombinant B7.1Ig and B7.2Ig in ELISA Phage displaying SCA-E protein on their surface are prepared as described by Marks et al. (1991). To demonstrate binding of the SCA-E Al/Sl His6 or SCA-E S2/A2 His6 proteins to B7.1Ig and B7.2Ig, phages are titrated and a serial dilution is made to test for binding in ELISA to these molecules. Strong signals can be obtained demonstrating binding to both B7.1Ig and B7.2Ig.
  • Example 21 SCA-E Al/Sl RGD and SCA-E S2/A2 RGD displayed on phage are still able to bind coated recombinant B7.1Ig and B7.2Ig in ELISA
  • a phage ELISA is performed on antigens v ⁇ 3 integrin (l ⁇ g/ml), B7-lIg and B7-2Ig (l ⁇ g/ml) and BSA (l ⁇ g/ml).
  • AB' KL10 shows a positive signal on B7-lIg and B7-2Ig suggesting that despite the introduction of the sequence SGGGS into the Al/Sl loop, the functional integrity of the molecule is maintained ( Figure 14), proving the concept of the invention that the molecule of the presdent invention can function as a scaffold.
  • the integrity of these molecules is further verified as a weak binding in phage ELISA as can be seen on anti CTLA-4 ( Figure 16).
  • Integrin alpha v beta 3 antagonists promote tumor regression by inducing apoptosis of angiogenic blood vessels, Cell. 79: 1157-64 -Cannon E ., Ladner R.C., McCoy D. (1996) Phage-display technology. rVD

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Zoology (AREA)
  • Biochemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Biotechnology (AREA)
  • Immunology (AREA)
  • General Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Wood Science & Technology (AREA)
  • Medicinal Chemistry (AREA)
  • Biophysics (AREA)
  • General Health & Medical Sciences (AREA)
  • Microbiology (AREA)
  • Biomedical Technology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Crystallography & Structural Chemistry (AREA)
  • Physics & Mathematics (AREA)
  • Virology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Toxicology (AREA)
  • Cell Biology (AREA)
  • Plant Pathology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Peptides Or Proteins (AREA)
EP99927740A 1999-04-01 1999-04-01 Eine polypeptidstruktur zur verwendung als gerüst Withdrawn EP1163339A1 (de)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/EP1999/002283 WO2000060070A1 (en) 1999-04-01 1999-04-01 A polypeptide structure for use as a scaffold

Publications (1)

Publication Number Publication Date
EP1163339A1 true EP1163339A1 (de) 2001-12-19

Family

ID=8167259

Family Applications (1)

Application Number Title Priority Date Filing Date
EP99927740A Withdrawn EP1163339A1 (de) 1999-04-01 1999-04-01 Eine polypeptidstruktur zur verwendung als gerüst

Country Status (4)

Country Link
EP (1) EP1163339A1 (de)
AU (1) AU4499499A (de)
CA (1) CA2367212A1 (de)
WO (1) WO2000060070A1 (de)

Families Citing this family (39)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1318195A1 (de) * 2001-12-10 2003-06-11 CatchMabs B.V. Ein Struktur für die Präsentation erwünschter Peptidsequenzen
US9321832B2 (en) 2002-06-28 2016-04-26 Domantis Limited Ligand
US7842476B2 (en) 2002-09-06 2010-11-30 Isogenica Limited In vitro peptide expression library
WO2004101620A2 (en) * 2003-05-01 2004-11-25 Compound Therapeutics, Inc. Serum albumin scaffold-based proteins and uses thereof
US20060008844A1 (en) 2004-06-17 2006-01-12 Avidia Research Institute c-Met kinase binding proteins
US7749694B2 (en) 2004-12-31 2010-07-06 The Regents Of The University Of California C-type lectin fold as a scaffold for massive sequence variation
US7700729B2 (en) 2006-05-15 2010-04-20 Avidbiotics Corporation Modified bacteriocins and methods for their use
US8445639B2 (en) 2006-05-15 2013-05-21 Avidbiotics Corporation Recombinant bacteriophage and methods for their use
ES2387722T3 (es) 2006-05-15 2012-09-28 Avidbiotics Corporation Bacteriocinas modificadas y métodos para su uso
CN101641115B (zh) 2007-03-08 2013-04-24 卡罗拜奥斯制药公司 用于实体瘤治疗的EphA3抗体
JP5390621B2 (ja) * 2008-10-20 2014-01-15 グワンジュ・インスティテュート・オブ・サイエンス・アンド・テクノロジー 二座ペプチドバインダー
EP2403879A1 (de) 2009-03-06 2012-01-11 Kalobios Pharmaceuticals, Inc. Behandlung von leukämien und chronischen myeloproliferativen krankheiten mit antikörpern gegen epha3
EP2506861A1 (de) 2009-12-02 2012-10-10 Amgen Inc. Bindeproteine zur bindung von humanem fgfr1c, humanem b-klotho sowie humanem fgfr1c als auch humanem b-klotho
AU2017222700B2 (en) 2016-02-26 2018-09-27 Imunexus Therapeutics Limited Multi-specific molecules
CA3016563A1 (en) 2016-03-21 2017-09-28 Elstar Therapeutics, Inc. Multispecific and multifunctional molecules and uses thereof
CN117327650A (zh) 2016-04-13 2024-01-02 维维雅生物技术公司 离体bite激活的t细胞
WO2017210598A1 (en) 2016-06-03 2017-12-07 Amgen Inc. Compositions and methods for treating an articular disorder
EP3515936A1 (de) 2016-09-23 2019-07-31 Elstar Therapeutics, Inc. Multispezifische antikörpermoleküle mit lambda- und kappa- leichtketten
US20200291089A1 (en) 2017-02-16 2020-09-17 Elstar Therapeutics, Inc. Multifunctional molecules comprising a trimeric ligand and uses thereof
AU2018256406A1 (en) 2017-04-19 2019-10-17 Marengo Therapeutics, Inc. Multispecific molecules and uses thereof
EP4328241A3 (de) 2017-04-28 2024-06-05 Marengo Therapeutics, Inc. Multispezifische moleküle mit einer nichtimmunglobulin-heterodimerisierungsdomäne und verwendungen davon
WO2018222901A1 (en) 2017-05-31 2018-12-06 Elstar Therapeutics, Inc. Multispecific molecules that bind to myeloproliferative leukemia (mpl) protein and uses thereof
WO2019035938A1 (en) 2017-08-16 2019-02-21 Elstar Therapeutics, Inc. MULTISPECIFIC MOLECULES BINDING TO BCMA AND USES THEREOF
US20200377571A1 (en) 2017-12-08 2020-12-03 Elstar Therapeutics, Inc. Multispecific molecules and uses thereof
EP3765517A1 (de) 2018-03-14 2021-01-20 Elstar Therapeutics, Inc. An calreticulin bindende multifunktionsmoleküle und ihre verwendungen
EP3765516A2 (de) 2018-03-14 2021-01-20 Elstar Therapeutics, Inc. Multifunktionale moleküle und verwendungen davon
DE202019005887U1 (de) 2018-07-03 2023-06-14 Marengo Therapeutics, Inc. Anti-TCR-Antikörpermoleküle und Verwendungen davon
CN113164777A (zh) 2018-09-27 2021-07-23 马伦戈治疗公司 Csf1r/ccr2多特异性抗体
SG11202109122SA (en) 2019-02-21 2021-09-29 Marengo Therapeutics Inc Anti-tcr antibody molecules and uses thereof
GB2597851B (en) 2019-02-21 2024-05-29 Marengo Therapeutics Inc Antibody molecules that bind to NKP30 and uses thereof
JP2022522662A (ja) 2019-02-21 2022-04-20 マレンゴ・セラピューティクス,インコーポレーテッド T細胞に結合する多機能性分子および自己免疫障害を処置するためのその使用
SG11202109061YA (en) 2019-02-21 2021-09-29 Marengo Therapeutics Inc Multifunctional molecules that bind to t cell related cancer cells and uses thereof
JP2022521750A (ja) 2019-02-21 2022-04-12 マレンゴ・セラピューティクス,インコーポレーテッド カルレティキュリンに結合する多機能性分子およびその使用
EP4084821A4 (de) 2020-01-03 2024-04-24 Marengo Therapeutics Inc An cd33 bindende multifunktionsmoleküle und ihre verwendungen
CA3180321A1 (en) 2020-04-24 2021-10-28 Marengo Therapeutics, Inc. Multifunctional molecules that bind to t cell related cancer cells and uses thereof
KR20230074144A (ko) 2020-08-26 2023-05-26 마렝고 테라퓨틱스, 인크. NKp30에 결합하는 항체 분자 및 이의 용도
WO2022047046A1 (en) 2020-08-26 2022-03-03 Marengo Therapeutics, Inc. Methods of detecting trbc1 or trbc2
JP2023542080A (ja) 2020-08-26 2023-10-05 マレンゴ・セラピューティクス,インコーポレーテッド カルレティキュリンに結合する多機能性分子およびその使用
CN117597359A (zh) 2021-04-08 2024-02-23 马伦戈治疗公司 与tcr结合的多功能分子及其用途

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5773253A (en) * 1993-01-22 1998-06-30 Bristol-Myers Squibb Company MYPPPY variants of CTL A4 and uses thereof
AUPP221098A0 (en) * 1998-03-06 1998-04-02 Diatech Pty Ltd V-like domain binding molecules
EP0947582A1 (de) * 1998-03-31 1999-10-06 Innogenetics N.V. Eine Polypeptidstruktur zur Verwendung als Gerüst

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO0060070A1 *

Also Published As

Publication number Publication date
AU4499499A (en) 2000-10-23
WO2000060070A1 (en) 2000-10-12
CA2367212A1 (en) 2000-10-12

Similar Documents

Publication Publication Date Title
EP1163339A1 (de) Eine polypeptidstruktur zur verwendung als gerüst
JP3949712B2 (ja) 抗体セグメントレパートリー由来でファージに表示される抗自己抗体の産生
US7195866B2 (en) Production of anti-self antibodies from antibody segment repertoires and displayed on phage
Hufton et al. Development and application of cytotoxic T lymphocyte-associated antigen 4 as a protein scaffold for the generation of novel binding ligands
US7122646B2 (en) Multivalent and multispecific binding proteins, their manufacture and use
EP0672142B1 (de) Multivalente und multispezifische bindungsproteine, deren herstellung und verwendung
US5871907A (en) Methods for producing members of specific binding pairs
JP3507073B2 (ja) 特異的結合対の成員の製造方法
AU664155B2 (en) Methods for producing members of specific binding pairs
US5858657A (en) Methods for producing members of specific binding pairs
EP0585287B1 (de) Verfahren zur herstellung von spezifischen bindungspaargliedern
US6225447B1 (en) Methods for producing members of specific binding pairs
JP2006333825A (ja) 標的物質捕捉用タンパク質の製造方法及びその構成材料の選択方法
EP0947582A1 (de) Eine Polypeptidstruktur zur Verwendung als Gerüst
EP1696031B1 (de) Herstellung von Autoantikörpern auf Phagenoberflächen ausgehend von Antikörpersegmentbibliotheken

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20011022

AK Designated contracting states

Kind code of ref document: A1

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

AX Request for extension of the european patent

Free format text: AL PAYMENT 20011022;LT PAYMENT 20011022;LV PAYMENT 20011022;MK PAYMENT 20011022;RO PAYMENT 20011022;SI PAYMENT 20011022

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20031001