EP1356075A2 - Proteingerüste für antikörpermimetika und andere bindungsproteine - Google Patents

Proteingerüste für antikörpermimetika und andere bindungsproteine

Info

Publication number
EP1356075A2
EP1356075A2 EP01981621A EP01981621A EP1356075A2 EP 1356075 A2 EP1356075 A2 EP 1356075A2 EP 01981621 A EP01981621 A EP 01981621A EP 01981621 A EP01981621 A EP 01981621A EP 1356075 A2 EP1356075 A2 EP 1356075A2
Authority
EP
European Patent Office
Prior art keywords
protein
antibody
derivative
domain
compound
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Withdrawn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
EP01981621A
Other languages
English (en)
French (fr)
Other versions
EP1356075A4 (de
Inventor
Dasa Lipovsek
Richard W. Wagner
Robert G. Kuimelis
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.)
Adnexus a Bristol Myers Squibb R&D Co
Original Assignee
Phylos Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Phylos Inc filed Critical Phylos Inc
Priority to EP09167669A priority Critical patent/EP2141243A3/de
Publication of EP1356075A2 publication Critical patent/EP1356075A2/de
Publication of EP1356075A4 publication Critical patent/EP1356075A4/de
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/24Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against cytokines, lymphokines or interferons
    • C07K16/241Tumor Necrosis Factors
    • 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
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • 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
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/525Tumour necrosis factor [TNF]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/22Immunoglobulins specific features characterized by taxonomic origin from camelids, e.g. camel, llama or dromedary
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide

Definitions

  • This invention relates to protein scaffolds useful, for example, for the generation of products having novel binding characteristics.
  • Proteins having relatively defined three-dimensional structures may be used as reagents for the design of engineered products. These scaffolds typically contain one or more regions which are amenable to specific or random sequence variation, and such sequence randomization is often carried out to produce libraries of proteins from which desired products may be selected.
  • One particular area in which such scaffolds are useful is the field of antibody design.
  • a "minibody” scaffold which is related to the immunoglobulin fold, has been designed by deleting three beta strands from a heavy chain variable domain of a monoclonal antibody (Tramontano et al., J. Mol. Recognit. 7:9, 1994).
  • This protein includes 61 residues and can be used to present two hypervariable loops. These two loops have been randomized and products selected for antigen binding, but thus far the framework appears to have somewhat limited utility due to solubility problems.
  • the present invention provides a new family of proteins capable of evolving to bind any compound of interest.
  • These proteins which generally make use of a scaffold derived from a fibronectin type III (Fn3) or Fn3-like domain, function in a manner characteristic of natural or engineered antibodies (that is, polyclonal, monoclonal, or single-chain antibodies) and, in addition, possess structural advantages.
  • the structure of these antibody mimics has been designed for optimal folding, stability, and solubility, even under conditions that normally lead to the loss of structure and function in antibodies.
  • these antibody mimics may be utilized for the purpose of designing proteins which are capable of binding to virtually any compound (for example, any protein) of interest.
  • the 10 Fn3-based molecules described herein may be used as scaffolds which are subjected to directed evolution to form a population with one or more randomized Fn3 loops that are analogous by position and structure to the complementarity-determining regions (CDRs) of an antibody variable region, and/or to randomize Fn3's other three solvent exposed loops.
  • CDRs complementarity-determining regions
  • the scaffolds described herein may be used to display defined exposed loops (for example, loops previously randomized and selected on the basis of antigen binding) in order to direct the evolution of molecules that bind to such introduced loops.
  • a selection of this type may be carried out to identify recognition molecules for any individual CDR-like loop or, alternatively, for the recognition of two or all three CDR-like loops combined into a non-linear epitope.
  • the present invention features randomized or mutated scaffold proteins.
  • the invention features a non-antibody protein including a domain having an immunoglobulin-like fold, the non- antibody protein deriving from a reference protein by having a mutated amino acid sequence, wherein the non-antibody protein binds with a Kd at least as tight as 1 ⁇ M to a compound that is not bound as tightly by the reference protein.
  • the invention features a non-antibody protein deriving from a scaffold protein including a domain having an immunoglobulin-like fold, wherein the amino acid sequence of the domain in the derived protein is more than 50% identical to the amino acid sequence of the domain in the scaffold protein.
  • the invention features a protein that includes a fibronectin type III domain having at least one randomized loop, the protein being characterized by the ability of the Fn3 domain to bind to a compound that is not bound by the corresponding naturally-occurring Fn3 domain.
  • any of these proteins of the invention bind to their target compounds with a Kd at least as tight as 500 nM, preferably, with a Kd at least as tight as 100 nM or 10 nM, and, more preferably, with a Kd at least as tight as 1 nM, 500 pM, 100 pM, or even 20 pM.
  • the protein preferably contains one, two, or three mutated loops and at least one of the loops, and preferably two or all three of the loops, contributes to the binding of the protein to the compound.
  • the reference protein preferably lacks disulfide bonds, and the derivative protein may have at least one disulfide bond.
  • the domain having an immunoglobulin-like fold preferably has a molecular mass less than 10 kD or greater than 7.5 kD, and, more preferably, has a molecular mass between 7.5-10 kD.
  • the proteins of the invention may be monomers under physiological conditions or may be multimers, for example, dimers.
  • the reference protein used to derive a mutated protein of the invention is a naturally-occurring mammalian protein (for example, a human protein); and the domain having an immunoglobulin-like fold is mutated and includes up to 50%, and preferably up to 34%, mutated amino acids as compared to the immunoglobulin-like fold of the reference protein.
  • the domain having the immunoglobulin-like fold preferably consists of approximately 50-150 amino acids, and more preferably consists of approximately 50 amino acids.
  • Derivative proteins of the invention may be derived from any appropriate reference protein including, but not limited to, the preferred proteins, fibronectin or a fibronectin dimer, tenascin, N-cadherin, E-cadherin, ICAM, titin, GCSF-receptor, cytokine receptor, glycosidase inhibitor, antibiotic chromoprotein, myelin membrane adhesion molecule P0, CD8, CD4, CD2, class I MHC, T-cell antigen receptor, CD1, C2 and I-set domains of VCAM-1, 1-set immunoglobulin domain of myosin-binding protein C, I-set immunoglobulin domain of myosin-binding protein H, I-set immunoglobulin domain of telokin, NCAM, twitchin, neuroglian, growth hormone receptor, erythropoietin receptor, prolactin receptor, interferon-gamma receptor, ⁇ -galactosidase/glucuronidase,
  • the fibronectin type III domain is a mammalian (for example, a human) fibronectin type III domain; and the protein includes the tenth module of the fibronectin type III ( 10 Fn3) domain.
  • compound binding is preferably mediated by either one, two, or three 10 Fn3 loops.
  • the second (DE) loop of 10 Fn3 may be extended in length relative to the naturally-occurring module, or the 10 Fn3 may lack an integrin-binding motif.
  • the integrin-binding motif may be replaced by an amino acid sequence in which a polar amino acid-neutral amino acid-acidic amino acid sequence (in the N-terminal to C-terminal direction) replaces the integrin-binding motif; alternatively, one preferred sequence is serine-glycine- glutamate.
  • the fibronectin type III domain- containing proteins of the invention lack disulfide bonds.
  • any of the proteins of the invention may be formulated as part of a fusion protein.
  • the fusion protein includes a heterologous protein that does not itself bind to the compound of interest.
  • the heterologous protein may, for example, be an antibody or antibody domain (such as an immunoglobulin F c domain), a complement protein, a toxin protein, or an albumin protein.
  • any of the proteins of the invention (for example, the fibronectin type III domain proteins) may be covalently bound to a nucleic acid (for example, an RNA), and the nucleic acid may encode the protein.
  • the protein may be a multimer, or, particularly if it lacks an integrin-binding motif, it may be formulated in a physiologically-acceptable carrier.
  • the present invention also features proteins that include a fibronectin type III domain having at least one mutation in a ⁇ -sheet sequence. Again, these proteins are characterized by their ability to bind to compounds that are not bound or are not bound as tightly by the corresponding naturally-occurring fibronectin domain.
  • any of the proteins of the invention may be immobilized on a solid support (for example, a bead or chip), and these proteins may be arranged in any configuration on the solid support, including an array.
  • a solid support for example, a bead or chip
  • the invention further features nucleic acids encoding any of the proteins of the invention.
  • the nucleic acid is DNA or RNA.
  • the invention also features a method for generating a protein which includes a fibronectin type III domain and which is pharmaceutically acceptable to a mammal, involving removing the integrin- binding domain of said fibronectin type III domain. This method may be applied to any of the fibronectin type III domain-containing proteins described above and is particularly useful for generating proteins for human therapeutic applications.
  • the invention also features such fibronectin type III domain- containing proteins which lack integrin-binding domains.
  • the invention features methods of obtaining derivative non-antibody proteins which bind to compounds of interest.
  • One such method involves: (a) providing a non-antibody scaffold protein including an immunoglobulin-like fold, wherein the scaffold protein does not bind to the compound with a Kd as tight as 1 ⁇ M; (b) generating mutated derivatives of the non-antibody scaffold protein, thereby producing a library of mutated proteins; (c) contacting the library with the compound; (d) selecting from the library at least one derivative protein which binds to the compound with a Kd at least as tight as 1 ⁇ M; and (e) optionally repeating steps (b) - (d) substituting for the non-antibody scaffold protein in repeated step (b) the product from the previous step (d).
  • the invention features screening methods which may be used to obtain or evolve randomized or mutated proteins of the invention capable of binding to compounds of interest, or to obtain or evolve compounds (for example, proteins) capable of binding to a particular protein containing a randomized or mutated motif.
  • the invention features screening procedures which combine these two methods, in any order, to obtain either compounds or proteins of interest.
  • a first screening method useful for the isolation or identification of randomized or mutated proteins of interest, involves: (a) contacting a compound of interest with a candidate protein, the candidate protein being a derivative non-antibody protein including a domain having an immunoglobulin-like fold, the non-antibody protein deriving from a reference protein by having a mutated amino acid sequence wherein the non-antibody protein binds with a Kd at least as tight as 1 ⁇ M to a compound that is not bound as tightly by the reference protein, wherein the contacting is carried out under conditions that allow compound-protein complex formation; and (b) obtaining, from the complex, the derivative protein that binds to the compound.
  • the second screening method is for isolating or identifying a compound which binds to a protein of the invention.
  • This method begins with a non- antibody protein including a domain having an immunoglobulin-like fold and deriving from a reference protein by having a mutated amino acid sequence, wherein the non-antibody protein binds with a Kd at least as tight as 1 ⁇ M to a compound that is not bound as tightly by the reference protein.
  • This derivative protein is then contacted with a candidate compound, wherein the contacting is carried out under conditions that allow compound-protein complex formation, and the compound which binds to the derivative protein is obtained from the complex.
  • the invention features diagnostic methods which employ the proteins of the invention (for example, fibronectin type III scaffold proteins and their derivatives). Such diagnostic methods may be carried out on a sample (for example, a biological sample) to detect one analyte or to simultaneously detect many different analytes in the sample.
  • the method may employ any of the scaffold molecules described herein.
  • the method involves (a) contacting the sample with a protein of the invention that binds to the compound analyte, the contacting being carried out under conditions that allow compound- protein complex formation; and (b) detecting the complex, and therefore the compound in the sample.
  • this method may be used to quantitate, as well as detect, compound levels in a sample.
  • the protein of the invention binds to its target compound with a Kd at least as tight as 1 ⁇ M or 500 nM, preferably, with a Kd at least as tight as 100 nM or 10 nM, and, more preferably, with a Kd at least as tight as 1 nM, 500 pM, 100 pM, or even 20 pM.
  • the protein preferably contains one, two, or three mutated loops and at least one of the loops, and preferably two or all three of the loops contributes to the binding of the protein to the compound.
  • the reference protein preferably lacks disulfide bonds, and the derivative protein may have at least one disulfide bond.
  • the domain having an immunoglobulin-like fold preferably has a molecular mass less than 10 kD or greater than 7.5 kD, and, more preferably, has a molecular mass between 7.5-10 kD.
  • the proteins of the invention may be monomers under physiological conditions or may be multimers, for example, dimers.
  • the reference protein used to derive a mutated protein of the invention is a naturally-occurring mammalian protein (for example, a human protein); and the domain having an immunoglobulin-like fold is mutated and includes up to 50%, and preferably up to 34%, mutated amino acids as compared to the immunoglobulin-like fold of the reference protein.
  • the domain having an immunoglobulin-like fold preferably consists of approximately 50-150 amino acids, and more preferably consists of approximately 50 amino acids.
  • Derivative proteins used in the methods of the invention may be derived from any appropriate reference protein including, but not limited to, the preferred proteins, fibronectin or a fibronectin dimer, tenascin, N-cadherin, E-cadherin, ICAM, titin, GCSF-receptor, cytokine receptor, glycosidase inhibitor, antibiotic chromoprotein, myelin membrane adhesion molecule P0, CD8, CD4, CD2, class I MHC, T-cell antigen receptor, CD1, C2 and I-set domains of VCAM- 1 , I-set immunoglobulin domain of myosin-binding protein C, I-set immunoglobulin domain of myosin-binding protein H, I-set immunoglobulin domain of telokin, NCAM, twitchin, neuroglian, growth hormone receptor, erythropoietin receptor, prolactin receptor, interferon-gamma receptor, ⁇ -galactosidase/
  • the steps of the selection methods described herein may be repeated with further mutation or randomization being carried out between cycles.
  • at least one loop of the fibronectin type III domain of the protein obtained in step (b) may be mutated and steps (a) and (b) repeated using the further randomized protein, or the compound obtained in step (b) may be modified and steps (a) and (b) repeated using the further modified compound.
  • the compound is preferably a protein
  • the fibronectin type III domain is preferably a mammalian (for example, a human) fibronectin type III domain.
  • the protein includes the tenth module of the fibronectin type III domain ( 10 Fn3), and binding is mediated by one, two, or three 10 Fn3 loops.
  • the second (DE) loop of 10 Fn3 may be extended in length relative to the naturally-occurring module, or 10 Fn3 may lack an integrin-binding motif.
  • the integrin-binding motif may be replaced by an amino acid sequence in which a basic amino acid-neutral amino acid-acidic amino acid sequence (in the N- terminal to C-terminal direction) replaces the integrin-binding motif; alternatively, one preferred replacement sequence is serine-glycine-glutamate.
  • any of the proteins of the invention for example, a fibronectin type III domain-containing protein.
  • any of these proteins may be formulated as part of a fusion protein with a heterologous protein (for example, an antibody or antibody domain (including an immunoglobulin F c domain) that does not itself bind the compound of interest, or a complement protein, toxin protein, or albumin protein).
  • selections and diagnostic methods may be carried out using the proteins of the invention (for example, the fibronectin type III domain proteins) covalently bound to nucleic acids (for example, RNAs or any nucleic acid which encodes the protein).
  • the selections and diagnostic methods may be carried out using these proteins (for example, the fibronectin domain-containing proteins) as monomers or as multimers, such as dimers.
  • the selections and diagnostic methods involve the immobilization of the binding target on a solid support.
  • solid supports include columns (for example, affinity columns, such as agarose-based affinity columns), microchips, or beads.
  • the proteins for example, the Fn3 domain-containing proteins
  • the proteins may be immobilized and contacted with one or more potential binding targets.
  • the compound is often a protein, but may also be any other analyte in a sample.
  • Detection may be accomplished by any standard technique including, without limitation, radiography, fluorescence detection, mass spectroscopy, or surface plasmon resonance.
  • the invention features a non-antibody protein that binds tumor necrosis factor- ⁇ (TNF- ⁇ ) with a Kd at least as tight as 1 ⁇ M, the protein having a sequence that is less than 20% identical to TNF- ⁇ receptor (for example, a naturally-occurring TNF- ⁇ receptor, such as a mammalian or human TNF- ⁇ receptor).
  • TNF- ⁇ tumor necrosis factor- ⁇
  • the protein having a sequence that is less than 20% identical to TNF- ⁇ receptor (for example, a naturally-occurring TNF- ⁇ receptor, such as a mammalian or human TNF- ⁇ receptor).
  • this protein includes a mutated fibronectin type III domain and the protein is mutated in the DE, BC, and FG loops.
  • the mutated FG loop is the same length as the wild-type FG loop.
  • the protein includes an immunoglobulin-like fold (preferably, having a molecular mass less than 10 kD, greater than 7.5 kD, or between 7.5-10 kD) that consists of approximately 50-150 amino acids, and preferably, approximately 50 amino acids.
  • the TNF- ⁇ binders according to the invention bind TNF- ⁇ with a Kd at least as tight as 1 ⁇ M, preferably, at least as tight as 500 nM, 100 nM, or 10 nM, more preferably, at least as tight as 1 nM or 500 pM, and, most preferably, at least as tight as 100 pM or even 20 pM.
  • these proteins contain one, two, or three mutated loops, and at least one, and preferably two or all three of the loops, contribute to the binding of the non-antibody protein to TNF- ⁇ .
  • the non-antibody protein has at least one disulfide bond, and the non-antibody protein is a monomer or dimer under physiological conditions.
  • the TNF- ⁇ binders may be immobilized on a solid support (for example, a chip or bead), and may be part of an array.
  • any of the TNF- ⁇ binders may be joined to a heterologous protein (for example, a heterologous protein that is an antibody or an antibody domain that does not bind TNF- ⁇ , an immunoglobulin F c domain, a complement protein, or an albumin protein).
  • the protein may include a mutated fibronectin type III domain
  • TNF- ⁇ binders preferably include a non-naturally occurring sequence in a loop of 10 Fn3 (for example, the loop sequence PW(A/G), and may include a non-naturally occurring sequence in a ⁇ -sheet of 10 Fn3. Particularly preferred TNF- ⁇ binders of the invention are shown in Figure 25 (SEQ ID NOS: 34-140).
  • the invention features nucleic acids encoding any of the TNF- ⁇ binding proteins of the invention, as well as a loop structure on any protein that includes any one of the amino acid sequences of Figure 25 (SEQ ID NOS: 34- 140).
  • non-antibody protein a protein that is not produced by the B cells of a mammal either naturally or following immunization of a mammal. This term also excludes antibody fragments of more than 100 amino acids, preferably, more than 80 amino acids, and, most preferably, more than 50 amino acids in length.
  • immunoglobulin-like fold is meant a protein domain of between about 80-150 amino acid residues that includes two layers of antiparallel beta- sheets, and in which the flat, hydrophobic faces of the two beta-sheets are packed against each other. Proteins according to the invention may include several immunoglobulin-like folds covalently bound or associated non- covalently into larger structures.
  • scaffold is meant a protein used to select or design a protein framework with specific and favorable properties, such as binding.
  • amino acid residues that are important for the framework's favorable properties are retained, while others residues may be varied.
  • Such a scaffold has less than 50% of the amino acid residues that vary between protein derivatives having different properties and greater than or equal to 50% of the residues that are constant between such derivatives. Most commonly, these constant residues confer the same overall three-dimensional fold to all the variant domains, regardless of their properties.
  • fibronectin type III domain is meant a domain having 7 or 8 beta strands which are distributed between two beta sheets, which themselves pack against each other to form the core of the protein, and further containing loops which connect the beta strands to each other and are solvent exposed. There are at least three such loops at each edge of the beta sheet sandwich, where the edge is the boundary of the protein perpendicular to the direction of the beta strands.
  • the invention further includes polymers of I0 Fn3 -related molecules, which are an extension of the use of the monomer structure, whether or not the subunits of the polyprotein are identical.
  • Naturally occurring is meant any protein that is encoded by a living organism.
  • randomized or “mutated” is meant including one or more amino acid alterations relative to a template sequence.
  • randomizing or “mutating” is meant the process of introducing, into a sequence, such an amino acid alteration. Randomization or mutation may be accomplished through intentional, blind, or spontaneous sequence variation, generally of a nucleic acid coding sequence, and may occur by any technique, for example, PCR, error-prone PCR, or chemical DNA synthesis.
  • a “corresponding, non-mutated protein” is meant a protein that is identical in sequence, except for the introduced amino acid mutations.
  • Protein By a “protein” is meant any sequence of two or more amino acids, regardless of length, post-translation modification, or function. "Protein” and “peptide” are used interchangeably herein.
  • RNA is meant a sequence of two or more covalently bonded, naturally occurring or modified ribonucleotides.
  • a modified RNA included within this term is phosphorothioate RNA.
  • DNA is meant a sequence of two or more covalently bonded, naturally occurring or modified deoxyribonucleotides.
  • nucleic acid any two or more covalently bonded nucleotides or nucleotide analogs or derivatives. As used herein, this term includes, without limitation, DNA, RNA, and PNA.
  • pharmaceutically acceptable is meant a compound or protein that may be administered to an animal (for example, a mammal) without significant adverse medical consequences.
  • physiologically acceptable carrier a carrier which does not have a significant detrimental impact on the treated host and which retains the therapeutic properties of the compound with which it is administered.
  • physiologically acceptable carrier is physiological saline.
  • Other physiologically acceptable carriers and their formulations are known to one skilled in the art and are described, for example, in Remington's Pharmaceutical Sciences. (18 th edition), ed. A. Gennaro, 1990, Mack Publishing Company, Easton, PA, incorporated herein by reference.
  • fusion protein a protein that includes (i) a scaffold protein of the invention joined to (ii) a second, different (i.e., “heterologous") protein.
  • Fusion proteins are distinguished from “nucleic acid-protein fusions” and “RNA-protein fusions” in that a “fusion protein” is composed entirely of amino acids, while both a “nucleic acid-protein fusion” and an “RNA-protein fusion” include a stretch of nucleic acids (the nucleic acid or RNA component) joined to a stretch of amino acids (the protein component).
  • a “selecting” step provides at least a 2-fold, preferably, at least a 30-fold, more preferably, at least a 100-fold, and, most preferably, at least a 1000-fold enrichment of a desired molecule relative to undesired molecules in a population following the selection step.
  • a selection step may be repeated any number of times, and different types of selection steps may be combined in a given approach.
  • binding partner as used herein, is meant any molecule which has a specific, covalent or non-covalent affinity for a portion of a desired compound (for example, protein) of interest.
  • binding partners include, without limitation, members of antigen/antibody pairs, protein inhibitor pairs, receptor/ligand pairs (for example cell surface receptor/ligand pairs, such as hormone receptor/peptide hormone pairs), enzyme/substrate pairs (for example, kinase/substrate pairs), lectin/carbohydrate pairs, oligomeric or heterooligomeric protein aggregates, DNA binding protein/DNA binding site pairs, RNA/protein pairs, and nucleic acid duplexes, heteroduplexes, or ligated strands, as well as any molecule which is capable of forming one or more covalent or non-covalent bonds (for example, disulfide bonds) with any portion of another molecule (for example, a compound or protein).
  • receptor/ligand pairs for example cell surface receptor/ligand pairs, such as hormone receptor/peptide hormone pairs
  • enzyme/substrate pairs for example, kinase/substrate pairs
  • lectin/carbohydrate pairs oligomeric or heterooligomeric protein aggregates,
  • solid support is meant, without limitation, any column (or column material), bead, test tube, microtiter dish, solid particle (for example, agarose or sepharose), microchip (for example, silicon, silicon-glass, or gold chip), or membrane (for example, an inorganic membrane, nitrocellulose, or the membrane of a liposome or vesicle) to which an antibody mimic or an affinity complex may be bound, either directly or indirectly (for example, through other binding partner intermediates such as other antibodies or Protein A), or in which an antibody mimic or an affinity complex may be embedded (for example, through a receptor or channel).
  • the present antibody mimics exhibit improved biophysical properties, such as stability under reducing conditions and solubility at high concentrations.
  • these molecules may be readily expressed and folded in prokaryotic systems, such as E. coli. in eukaryotic systems, such as yeast, and in in vitro translation systems, such as the rabbit reticulocyte lysate system.
  • prokaryotic systems such as E. coli.
  • eukaryotic systems such as yeast
  • in vitro translation systems such as the rabbit reticulocyte lysate system.
  • these molecules are extremely amenable to affinity maturation techniques involving multiple cycles of selection, including in vitro selection using RNA-protein fusion technology (Roberts and Szostak, Proc. Natl. Acad. Sci USA 94:12297, 1997; Szostak et al., U.S.S.N.
  • FIGURE 1 is a photograph showing a comparison between the structures of antibody heavy chain variable regions from camel (dark blue) and llama (light blue), in each of two orientations.
  • FIGURE 2 is a photograph showing a comparison between the structures of the camel antibody heavy chain variable region (dark blue), the llama antibody heavy chain variable region (light blue), and a fibronectin type III module number 10 ( 10 Fn3) (yellow).
  • FIGURE 3 is a photograph showing a fibronectin type III module number
  • FIGURE 4 is a graph illustrating a sequence alignment between a fibronectin type III protein domain and related protein domains.
  • FIGURE 5 is a photograph showing the structural similarities between a
  • FIGURE 6 is a photograph showing space filling models of fibronectin III modules 9 and 10, in each of two different orientations. The two modules and the integrin binding loop (RGD) are labeled. In this figure, blue indicates positively charged residues, red indicates negatively charged residues, and white indicates uncharged residues.
  • FIGURE 7 is a photograph showing space filling models of fibronectin III modules 7-10, in each of three different orientiations.
  • FIGURE 8 is a photograph illustrating the formation, under different salt conditions, of RNA-protein fusions which include fibronectin type III domains.
  • FIGURE 9 is a series of photographs illustrating the selection of fibronectin type III domain-containing RNA-protein fusions, as measured by PCR signal analysis.
  • FIGURE 10 is a graph illustrating an increase in the percent TNF- ⁇ binding during the selections described herein, as well as a comparison between RNA-protein fusion and free protein selections.
  • FIGURE 11 is a series of schematic representations showing IgG, 10 Fn3, Fn-CH r CH 2 -CH 3 , and Fn-CH 2 -CH 3 (clockwise from top left).
  • FIGURE 12 is a photograph showing a molecular model of Fn-CH r CH 2 -
  • CH 3 based on known three-dimensional structures of IgG (X-ray crystallography) and 10 Fn3 (NMR and X-ray crystallography).
  • FIGURE 13 is a graph showing the time course of an exemplary 10 Fn3-based nucleic acid-protein fusion selection of TNF- ⁇ binders. The proportion of nucleic acid-protein fusion pool (open diamonds) and free protein pool (open circles) that bound to TNF- ⁇ -Sepharose, and the proportion of free protein pool (full circles) that bound to underivatized Sepharose, are shown.
  • FIGURES 14 and 15 are graphs illustrating TNF- ⁇ binding by TNF- ⁇ Fn- binders. In particular, these figures show mass spectra data obtained from a I0 Fn3 fusion chip and non-fusion chip, respectively.
  • FIGURES 16 and 17 are the phosphorimage and fluorescence scan, respectively, of an 10 Fn3 array, illustrating TNF- ⁇ binding.
  • FIGURE 18 is a graph showing an alignment of the primary sequences of the llama V H domain and the wild-type human 10 Fn3 domain. Homologous residues between the two sequences are indicated. The 10 Fn3 residues outside the randomized loops that were found to have mutated in approximately 45% of the selected clones are marked with arrows under the wild-type 10 Fn3 sequence and with the letter that identifies the selected residue.
  • FIGURE 19 shows schematic representations of the llama V H domain and the wild-type human 10 Fn3 domain. The locations of the mutated framework residues are indicated.
  • FIGURE 20 is a graph illustrating the efficiency and specificity of binding of a free-protein pool translated from the original library (RO) and after ten rounds of selection with TNF- ⁇ (RIO). Protein pool binding to underivatized Sepharose, to TNF- ⁇ -Sepharose, to IL-l ⁇ -Sepharose, and to IL-13-Sepharose is compared.
  • FIGURE 21 is a series of IgG-like scaffolds for the display of up to three loops.
  • FIGURE 22 is a series of IgG-like scaffolds for the display of up to four, or even six, loops.
  • FIGURE 23 is a series of scaffolds, unrelated to IgG, for the display of loop structures.
  • FIGURES 24A-24D are photographic and graphic illustrations demonstrating the specific capture of a target (TNF- ⁇ ) by a mimic immobilized on a solid surface.
  • FIGURE 25 is a graph listing exemplary TNF- ⁇ binders (SEQ ID NOS: 33-140) according to the invention.
  • novel antibody mimics described herein have been designed to be superior both to antibody-derived fragments and to non-antibody frameworks, for example, those frameworks cited above.
  • antibody mimics over antibody fragments.
  • These antibody mimics are derived from whole, stable, and soluble structural scaffolds.
  • the Fn3 scaffold is found in the human body. Consequently, they exhibit better folding and thermostability properties than antibody fragments, whose creation involves the removal of parts of the antibody native fold, often exposing amino acid residues that, in an intact antibody, would be buried in a hydrophobic environment, such as an interface between variable and constant domains. Exposure of such hydrophobic residues to solvent increases the likelihood of aggregation of the antibody fragments.
  • the scaffolds described herein have no disulfide bonds, which have been reported to retard or prevent proper folding of antibody fragments under certain conditions. Since the present scaffolds do not rely on disulf ⁇ des for native fold stability, they are stable under reducing conditions, unlike antibodies and their fragments which unravel upon disulfide bond reduction.
  • these scaffolds provide the functional advantages of antibody molecules.
  • the 10 Fn3 module is not an immunoglobulin
  • its overall fold is close to that of the variable region of the IgG heavy chain ( Figure 2), making it possible to display the three fibronectin loops analogous to CDRs in relative orientations similar to those of native antibodies.
  • the present antibody mimics possess antigen binding properties that are similar in nature and affinity to those of antibodies, and a loop randomization and shuffling strategy may be employed in vitro that is similar to the process of affinity maturation of antibodies in vivo.
  • exemplary scaffolds for example, f ⁇ bronectin-based scaffolds, and their use for identifying, selecting, and evolving novel binding proteins as well as their target ligands. These examples are provided for the purpose of illustrating, and not limiting, the invention.
  • Preferred antibody mimics of the present invention are based on the structure of a fibronectin module of type III (Fn3), a common domain found in mammalian blood and structural proteins. This domain occurs more than 400 times in the protein sequence database and has been estimated to occur in 2% of the proteins sequenced to date, including fibronectins, tenascin, intracellular cytoskeletal proteins, and prokaryotic enzymes (Bork and Doolittle, Proc. Natl. Acad. Sci. USA 89:8990, 1992; Bork et al., Nature Biotech. 15:553, 1997; Meinke et al., J. Bacteriol. 175:1910, 1993; Watanabe et al., J. Biol. Chem.
  • Fn3 fibronectin module of type III
  • a particular scaffold is the tenth module of human Fn3 ( 10 Fn3), which comprises 94 amino acid residues.
  • the overall fold of this domain is closely related to that of the smallest functional antibody fragment, the variable region of the heavy chain, which comprises the entire antigen recognition unit in camel and llama IgG ( Figure 1, 2).
  • the major differences between camel and llama domains and the 10 Fn3 domain are that (i) 10 Fn3 has fewer beta strands (seven vs. nine) and (ii) the two beta sheets packed against each other are connected by a disulfide bridge in the camel and llama domains, but not in 10 Fn3.
  • the three loops of 10 Fn3 corresponding to the antigen-binding loops of the IgG heavy chain run between amino acid residues 21-31 (BC), 51 -56 (DE), and 76-88 (FG) ( Figure 3).
  • the length of the BC and DE loop, 10 and 6 residues, respectively, fall within the narrow range of the corresponding antigen-recognition loops found in antibody heavy chains, that is, 7-10 and 4-8 residues, respectively. Accordingly, once randomized and selected for high antigen affinity, these two loops may make contacts with antigens equivalent to the contacts of the corresponding loops in antibodies.
  • the FG loop of 10 Fn3 is 12 residues long, whereas the corresponding loop in antibody heavy chains ranges from 4-28 residues.
  • the length of the FG loop of 10 Fn3 is preferably randomized in length as well as in sequence to cover the CDR3 range of 4-28 residues to obtain the greatest possible flexibility and affinity in antigen binding.
  • the lengths as well as the sequences of the CDR-like loops of the antibody mimics may be randomized during in vitro or in vivo affinity maturation (as described in more detail below).
  • the tenth human fibronectin type III domain, 10 Fn3, refolds rapidly even at low temperature; its backbone conformation has been recovered within 1 second at 5°C.
  • One of the physiological roles of 10 Fn3 is as a subunit of fibronectin, a glycoprotein that exists in a soluble form in body fluids and in an insoluble form in the extracellular matrix (Dickinson et al., J. Mol. Biol. 236:1079, 1994).
  • a fibronectin monomer of 220-250 kD contains 12 type I modules, two type II modules, and 17 fibronectin type III modules (Potts and Campbell, Curr. Opin.Cell Biol. 6:648, 1994).
  • the 10 Fn3 framework possesses exposed loop sequences tolerant of randomization, facilitating the generation of diverse pools of antibody mimics. This determination was made by examining the flexibility of the 10 Fn3 sequence.
  • the human 10 Fn3 sequence was aligned with the sequences of fibronectins from other sources as well as sequences of related proteins ( Figure 4), and the results of this alignment were mapped onto the three-dimensional structure of the human 10 Fn3 domain ( Figure 5).
  • changes in the ⁇ -sheet sequences may also be used to evolve new proteins. These mutations change the scaffold and thereby indirectly alter loop structure(s). If this approach is taken, mutations should not saturate the sequence, but rather few mutations should be introduced. Preferably, no more than between 3-20 changes should be introduced to the ⁇ -sheet sequences by this approach. Sequence variation may be introduced by any technique including, for example, mutagenesis by Taq polymerase (Tindall and Kunkel, Biochemistry 27:6008 (1988)), fragment recombination, or a combination thereof.
  • an increase of the structural diversity of libraries for example, by varying the length as well as the sequence of the CDR-like loops, or by structural redesign based on the advantageous framework mutations found in selected pools, may be used to introduce further improvements in antibody mimic scaffolds.
  • the antibody mimics described herein may be fused to other protein domains.
  • these mimics may be integrated with the human immune response by fusing the constant region of an IgG (F c ) with an antibody mimic, such as an 10 Fn3 module, preferably through the C-terminus of 10 Fn3.
  • the F c in such a I0 Fn3-F c fusion molecule activates the complement component of the immune response and increases the therapeutic value of the antibody mimic.
  • a fusion between an antibody mimic, such as 10 Fn3, and a complement protein, such as Clq may be used to target cells
  • a fusion between an antibody mimic, such as 10 Fn3, and a toxin may be used to specifically destroy cells that carry a particular antigen.
  • an antibody scaffold such as 10 Fn3, in any form may be fused with albumin to increase its half-life in the bloodstream and its tissue penetration.
  • Any of these fusions may be generated by standard techniques, for example, by expression of the fusion protein from a recombinant fusion gene constructed using publically available gene sequences.
  • any of the scaffold constructs described herein may be generated as dimers or multimers of antibody mimics as a means to increase the valency and thus the avidity of antigen binding.
  • Such multimers may be generated through covalent binding.
  • individual 10 Fn3 modules may be bound by imitating the natural 8 Fn3- 9 Fn3- 10 Fn3 C-to-N-terminus binding or by imitating antibody dimers that are held together through their constant regions.
  • a 10 Fn3-Fc construct may be exploited to design dimers of the general scheme of 10 Fn3-Fc::Fc- 10 Fn3.
  • the bonds engineered into the Fc::Fc interface may be covalent or non-covalent.
  • dimerizing or multimerizing partners other than Fc can be used in hybrids, such as 10 Fn3 hybrids, to create such higher order structures.
  • covalently bonded multimers may be generated by constructing fusion genes that encode the multimer or, alternatively, by engineering codons for cysteine residues into monomer sequences and allowing disulfide bond formation to occur between the expression products.
  • Non- covalently bonded multimers may also be generated by a variety of techniques. These include the introduction, into monomer sequences, of codons corresponding to positively and/or negatively charged residues and allowing interactions between these residues in the expression products (and therefore between the monomers) to occur. This approach may be simplified by taking advantage of charged residues naturally present in a monomer subunit, for example, the negatively charged residues of fibronectin.
  • Another means for generating non-covalently bonded antibody mimics is to introduce, into the monomer gene (for example, at the amino- or carboxy-termini), the coding sequences for proteins or protein domains known to interact.
  • proteins or protein domains include coil-coil motifs, leucine zipper motifs, and any of the numerous protein subunits (or fragments thereof) known to direct formation of dimers or higher order multimers.
  • I0 Fn3 represents a preferred scaffold for the generation of antibody mimics
  • other molecules may be substituted for 10 Fn3 in the molecules described herein.
  • These include, without limitation, human fibronectin modules 'FnS ⁇ FnS and ⁇ Fn3- Fn3 as well as related Fn3 modules from non-human animals and prokaryotes.
  • Fn3 modules from other proteins with sequence homology to 10 Fn3, such as tenascins and undulins may also be used.
  • FIG. 21 Other exemplary scaffolds having immunoglobulin-like folds (but with sequences that are unrelated to the V H domain) are shown in Figure 21 and include N-cadherin, ICAM-2, titin, GCSF receptor, cytokine receptor, glycosidase inhibitor, E-cadherin, and antibiotic chromoprotein.
  • Yet further domains with related structures may be derived from myelin membrane adhesion molecule P0, CD8, CD4, CD2, class I MHC, T-cell antigen receptor, CD1, C2 and I-set domains of VCAM- 1, I-set immunoglobulin domain of myosin-binding protein C, I-set immunoglobulin domain of myosin-binding protein H, I-set immunoglobulin domain of telokin, telikin, NCAM, twitchin, neuroglian, growth hormone receptor, erythropoietin receptor, prolactin receptor, GC-SF receptor, interferon-gamma receptor, ⁇ -galactosidase/glucuronidase, ⁇ - glucuronidase, and transglutaminase.
  • any other protein that includes one or more immunoglobulin-like folds may be utilized.
  • Such proteins may be identified, for example, using the program SCOP (Murzin et al., J. Mol. Biol. 247:536 (1995); Lo Conte et al., Nucleic Acids Res. 25:257 (2000).
  • any molecule that exhibits a structural relatedness to the V H domain may be utilized as an antibody mimic.
  • Such molecules may, like fibronectin, include three loops at the N-terminal pole of the molecule and three loops at the C- terminal pole, each of which may be randomized to create diverse libraries; alternatively, larger domains may be utilized, having larger numbers of loops, as long as a number of such surface randomizable loops are positioned closely enough in space that they can participate in antigen binding.
  • Figure 22 shows examples of useful domains having more than three loops positioned close to each other.
  • T-cell antigen receptor and superoxide dismutase which each have four loops that can be randomized; and an Fn3 dimer, tissue factor domains, and cytokine receptor domains, each of which have three sets of two similar domains where three randomizable loops are part of the two domains (bringing the total number of loops to six).
  • any protein having variable loops positioned close enough in space may be utilized for candidate binding protein production.
  • large proteins having spatially related, solvent accessible loops may be used, even if unrelated structurally to an immunoglobulin-like fold.
  • Exemplary proteins include, without limitation, cytochrome F, green fluorescent protein, GroEL, and thaumatin ( Figure 23).
  • the loops displayed by these proteins may be randomized and superior binders selected from a randomized library as described herein. Because of their size, molecules may be obtained that exhibit an antigen binding surface considerably larger than that found in an antibody-antigen interaction.
  • Other useful scaffolds of this type may also be identified using the program SCOP (Murzin et al., J. Mol. Biol. 247:536 (1995)) to browse among candidate proteins having numerous loops, particularly loops positioned among parallel beta sheets or a number of alpha-helices.
  • Modules from different organisms and parent proteins may be most appropriate for different applications. For example, in designing an antibody mimic, it may be most desirable to generate that protein from a fibronectin or fibronectin-like molecule native to the organism for which a therapeutic is intended. In contrast, the organism of origin is less important or even irrelevant for antibody mimics that are to be used for in vitro applications, such as diagnostics, or as research reagents.
  • libraries may be generated and used to select binding proteins by any of the methods described herein.
  • the antibody mimics described herein may be used in any technique for evolving new or improved binding proteins.
  • the target of binding is immobilized on a solid support, such as a column resin or microtiter plate well, and the target contacted with a library of candidate scaffold-based binding proteins.
  • a library may consist of antibody mimic clones, such as 10 Fn3 clones constructed from the wild type 10 Fn3 scaffold through randomization of the sequence and/or the length of the 10 Fn3 CDR-like loops.
  • this library may be an RNA-protein fusion library generated, for example, by the techniques described in Szostak et al., U.S.S.N.
  • it may be a DNA-protein library (for example, as described in Lohse, DNA-Protein Fusions and Uses Thereof, U.S.S.N. 60/110,549, U.S.S.N. 09/459,190, and WO 00/32823).
  • the fusion library is incubated with the immobilized target, the support is washed to remove non-specific binders, and the tightest binders are eluted under very stringent conditions and subjected to PCR to recover the sequence information or to create a new library of binders which may be used to repeat the selection process, with or without further mutagenesis of the sequence. A number of rounds of selection may be performed until binders of sufficient affinity for the antigen are obtained.
  • the 10 Fn3 scaffold may be used as the selection target.
  • a protein is required that binds a specific peptide sequence presented in a ten residue loop
  • a single 10 Fn3 clone is constructed in which one of its loops has been set to the length often and to the desired sequence.
  • the new clone is expressed in vivo and purified, and then immobilized on a solid support.
  • An RNA-protein fusion library based on an appropriate scaffold is then allowed to interact with the support, which is then washed, and desired molecules eluted and re-selected as described above.
  • the scaffolds described herein may be used to find natural proteins that interact with the peptide sequence displayed by the scaffold, for example, in an 10 Fn3 loop.
  • the scaffold protein such as the 10 Fn3 protein, is immobilized as described above, and an RNA-protein fusion library is screened for binders to the displayed loop.
  • the binders are enriched through multiple rounds of selection and identified by DNA sequencing.
  • RNA-protein libraries represent exemplary libraries for directed evolution
  • any type of scaffold-based library may be used in the selection methods of the invention.
  • the antibody mimics described herein may be evolved to bind any antigen of interest. These proteins have thermodynamic properties superior to those of natural antibodies and can be evolved rapidly in vitro. Accordingly, these antibody mimics may be employed in place of antibodies in all areas in which antibodies are used, including in the research, therapeutic, and diagnostic fields. In addition, because these scaffolds possess solubility and stability properties superior to antibodies, the antibody mimics described herein may also be used under conditions which would destroy or inactivate antibody molecules. Finally, because the scaffolds of the present invention may be evolved to bind virtually any compound, these molecules provide completely novel binding proteins which also find use in the research, diagnostic, and therapeutic areas.
  • Exemplary scaffold molecules described above were generated and tested, for example, in selection protocols, as follows.
  • a complex library was constructed from three fragments, each of which contained one randomized area corresponding to a CDR-like loop.
  • the randomized residues are indicated in Figure 18 as underlined sequences, specifically, residues 23-29 of the 10 Fn3 BC loop (corresponding to CDR-Hl of the llama V H ); residues 52-55 of the 10 Fn3 DE loop (corresponding to CDR-H2 of the llama V H ); and residues 78-87 of the 10 Fn3 FG loop (corresponding to CDR-H3 of the llama V H ).
  • each of the fragments contained stretches encoding an N-terminal His 6 domain or a C-terminal FLAG peptide tag.
  • each DNA fragment contained recognition sequences for the Earl Type IIS restriction endonuclease. This restriction enzyme allowed the splicing together of adjacent fragments while removing all foreign, non- 10 Fn3, sequences. It also allowed for a recombination-like mixing of the three 10 Fn3 fragments between cycles of mutagenesis and selection.
  • the wild-type, human 10 Fn3 gene was cloned from a human liver library
  • the BC r -DE r -FG r library was obtained by randomizing the selected residues in BC, DE, and FG loops; the BC r -DE wt -FG r library was obtained by randomizing the selected residues in BC and FG loops, leaving the DE loop sequence wild-type; and the BC wt -DE wt -FG r library was obtained by randomizing the selected residues in the FG loop only.
  • the BC r , DE r , and FG r fragments were made synthetically. Each fragment was assembled from two overlapping oligonucleotides, which were first annealed, then extended to form the double-stranded DNA form of the fragment.
  • oligonucleotides that were used to construct and process the three fragments are listed below; the "Top” and “Bottom” species for each fragment are the oligonucleotides that contained the entire 10 Fn3 encoding sequence. In these oligonucleotides designations, "N” indicates A, T, C, or G; and “S” indicates C or G.
  • HfhLbcTop 5'- GG AAT TCC TAA TAC GAC TCA CTA TAG GGA CAA TTA CTA TTT ACA ATT ACA ATG CAT CAC CAT CAC CAT CAC GTT TCT GAT GTT CCG AGG GAC CTG GAA GTT GTT GCT GCG ACC CCC ACC AGC-3' (SEQ ID NO: 1)
  • HfhLbcTop an alternative N-terminus
  • HFnBC3'-flag8 5'-AGC GGA TGC CTT GTC GTC GTC CTT GTA GTC GCT CTT CCC TGT TTC TCC GTAAGT GAT CC-3' (SEQ ID NO: 4)
  • HFnFG3'-flag8 5'-AGC GGA TGC CTT GTC GTC GTC CTT GTA GTC TGT TCG GTA ATT AAT GGAAAT TGG -3' (SEQ ID NO: 10)
  • T7Tmv (introduces T7 promoter and TMV untranslated region needed for in vitro translation):
  • Unispl-s spint oligonucleotide used to ligate mRNA to the puromycin-containing linker, described by Roberts et al, 1997, supra: 5-.TTTTTTTTTNAGCGGATGC-3' (SEQ ID NO: 13)
  • the oligonucleotide pair BC Top and BC Bot . flag8 was used to construct the fragment which contains the randomized BC loop; the pair DE Top and DE Bot . flag8 was used to construct the fragment which contains the randomized DE loop; the pair BC Top and DE 3 _ Flagg was used to PCR-amplify the BC wt - DE wt fragments; and the pair FG Top and FG Bot . Flag8 was used to construct the fragment which contains the randomized FG loop.
  • the pairs of oligonucleotides (500 pmol of each) were annealed in 100 ⁇ L of 10 mM Tris 7.5, 50 mM NaCl for 10 minutes at 85°C, followed by a slow (0.5-1 hour) cooling to room temperature.
  • the annealed fragments with single-stranded overhangs were then extended using 100 U Klenow (New England Biolabs, Beverly, MA) for each 100 ⁇ L aliquot of annealed oligos, and the buffer made of 838.5 ⁇ l H 2 O, 9 ⁇ l 1 M Tris 7.5, 5 ⁇ l IM MgCl 2 , 20 ⁇ l 10 mM dNTPs, and 7.5 ⁇ l IM DTT.
  • the extension reactions proceeded for 1 hour at 25°C.
  • the randomized residues were encoded by (NNS) n , where N stands for any nucleotide and S for an equimolar mixture of C and G; only one of the three stop codons (TAG) conforms to the NNS restriction.
  • N stands for any nucleotide and S for an equimolar mixture of C and G; only one of the three stop codons (TAG) conforms to the NNS restriction.
  • the gene fragments contained the 5' Tobacco Mosaic Virus (TMV) untranslated region and the T7 promoter, as well as the sequences encoding a 5' hexahistidine protein purification tag and a 3' FLAG epitope purification tag.
  • Ear I restriction endonuclease recognition sites were engineered into the overlaps between adjacent fragments in order to facilitate the assembly of the three fragments.
  • each of the double-stranded fragments was transformed into an RNA-protein fusion (PROfusionTM) using the technique developed by Szostak et al, U.S.S.N. 09/007,005 and U.S.S.N. 09/247,190; Szostak et al., WO98/31700; and Roberts & Szostak, Proc. Natl. Acad. Sci. USA (1997) vol. 94, p. 12297-12302.
  • the fragments were transcribed using an Ambion in vitro transcription kit, T7-MEGAshortscriptTM (Ambion, Austin, TX), and the resulting mRNA was gel-purified and ligated to a 5'-phosphorylated DNA-puromycin linker, preferably, 5' dA 18 PEG 2 dCdCPur) using DNA ligase (Promega, Madison, Wl); the mRNA was aligned with the DNA linker using a DNA splint oligonucleotide (5' TTTTTTTTTNAGCGGATGC 3'; SEQ ID NO: 30) as described in Szostak (supra).
  • the mRNA-DNA-puromycin molecule was then translated using the Ambion rabbit reticulocyte lysate-based translation kit in the presence of 35 S-methionine.
  • the resulting mRNA-DNA-puromycin- protein fusion was purified using Oligo(dT) cellulose, (Type 7, Amersham Pharmacia, Piscataway, NJ) and a complementary DNA strand was synthesized using reverse transcriptase (Superscript TMII, Gibco, Life Technologies, Rockville, MD) and the RT primers described above (Unisplint-S or flagASA), following the manufacturer's instructions (preferably, a two-minute annealing at 70°C and a 40 minute reaction at 42°C).
  • RNA-protein fusion with annealing cDNA obtained for each fragment was next purified on the resin appropriate to its peptide purification tag, i.e., on Ni-NTA agarose (Qiagen, Valencia, CA) for the His 6 -tag and M2 Anti-Flag Agarose (Sigma, St. Louis, MO) for the FLAG-tag, following the procedures recommended by the manufacturers.
  • Ni-NTA agarose Qiagen, Valencia, CA
  • M2 Anti-Flag Agarose Sigma, St. Louis, MO
  • the fragment-encoding genetic information recovered by KOH elution was amplified by PCR using Pharmacia Ready-to-Go PCR Beads, 10 pmol of 5' and 3' PCR primers, and the following PCR program (Pharmacia, Piscataway, NJ): Step 1: 95°C for 3 minutes; Step 2: 95°C for 30 seconds, 58/62°C for 30 seconds, 72°C for 1 minute, 20/25/30 cycles, as required; Step 3: 72°C for 5 minutes; Step 4: 4°C until end (typically, 25 cycles).
  • the resulting DNA was cleaved by 5-6 U Earl (New England Biolabs) per ⁇ g DNA; the reaction took place in T4 DNA Ligase Buffer (New England
  • sequences of 76 randomly picked clones from the original, randomized, BC r -DE r -FG r library showed no pattern in the randomized loops (data not shown); the amino acid frequency in the library varied in proportion to the number of codons available that encoded each residue, between 1% per position (glutamic acid, methionine, tryptophan) and 14% per position (proline). In contrast, the average probability for a residue in the preserved, beta-sheet framework to have remained as wild type was found to be 99%.
  • RNA-protein fusions were obtained from the master library following the general procedure described in Szostak et al., U.S.S.N. 09/007,005 and 09/247,190; Szostak et al., WO98/31700; and Roberts & Szostak, Proc. Natl. Acad. Sci. USA (1997) vol. 94, p. 12297-12302 ( Figure 8), except that affinity purification performed in rounds three to ten used only M2- Sepharose (see below). Fusion Selections
  • the master library in the RNA-protein fusion form was subjected to selection for binding to TNF- ⁇ (Pepro Tech, Rocky Hill, NJ). Two initial protocols were employed: one in which the target was immobilized on an agarose column and one in which the target was immobilized on a BIACORE chip.
  • Two initial protocols were employed: one in which the target was immobilized on an agarose column and one in which the target was immobilized on a BIACORE chip.
  • an extensive optimization of conditions to minimize background binders to the agarose column yielded the favorable buffer conditions of 50 mM HEPES pH 7.4, 0.02% Triton, 100 ⁇ g/ml sheared salmon sperm DNA. In this buffer, the non-specific binding of the 10 Fn3-RNA fusion to TNF- ⁇ Sepharose was 0.3%.
  • the non-specific binding background of the 10 Fn3-RNA/cDNA library to TNF- ⁇ Sepharose was found to be 0.1%.
  • the library was first preincubated for an hour with underivatized Sepharose to remove any remaining non-specific binders; the flow-through from this pre-clearing was incubated for another hour with TNF- ⁇ Sepharose. The TNF- ⁇ Sepharose was washed for 3- 30 minutes.
  • the fusion pools selected in the first eight rounds of selection bound to TNF- ⁇ -Sepharose at levels close to the background ( ⁇ 0.25%) ( Figure 13).
  • the binding of fusion to TNF- ⁇ -Sepharose increased sharply to 0.7%, and, after ten rounds of selection (RIO), the binding increased further to 7% ( Figure 13).
  • These selections were carried out using TNF- ⁇ immobilized on Epoxy- Activated SepharoseTM 6B (Amersham Pharmacia) at 10 mg TNF/g Sepharose in 10 mL.
  • TNF- ⁇ -derivatized Sepharose was blocked in Binding Buffer (50 mM HEPES, pH 7.4, 0.02% Triton, 0.1 mg/mL sheared salmon sperm DNA (Ambion)), overnight, at 4°C.
  • Binding Buffer 50 mM HEPES, pH 7.4, 0.02% Triton, 0.1 mg/mL sheared salmon sperm DNA (Ambion)
  • the 10 Fn3-based master library was transcribed, ligated to the puromycin-bearing linker, translated into an mRNA-protein library in the presence of 5-10 ⁇ L/300 ⁇ L 35 S-methionine, affinity purified on Oligo(dT) Cellulose, reverse-transcribed into a DNA/mRNA-protein library, and affinity-purified on M2-Sepharose (for rounds 3-10), as described above. Forty pmol of DNA/mRNA-protein fusion library molecules, the equivalent of 20 copies of 4 x 10 12 different sequences, were recovered, then subjected to the first round (Rl) of the selection.
  • the supernatant was recovered by microcentrifugation through a Micro Bio-Spin® chromatography column (BIO-RAD, Hercules, CA), then incubated with 30 ⁇ L of TNF- ⁇ -Sepharose (6 ⁇ M) in 300 ⁇ L of the Binding Buffer for 1 hour at 4°C (during Rounds 7-10, the Binding Buffer contained an additional 1 mg/mL of BSA).
  • the TNF- ⁇ -Sepharose was recovered on a spin column, then washed with 3 x 300 ⁇ L of Binding Buffer, eluted with 100 ⁇ L of 0.1 M KOH, and finally neutralized with 1 ⁇ L of 1 M Tris 8.0, 8 ⁇ L of 1 M HCl.
  • Samples of the library, of the TNF- ⁇ -Sepharose before and after the elution, of the washes, and of the elutions were quantified by counting 35 S-methionine in the sample in a scintillation counter.
  • the next round of selection began with the formation of a new DNA/mRNA-protein pool by PCR amplification, which was transcribed, translated, and reverse-transcribed from the PCR product.
  • the DNA pools obtained from the elution after nine and after ten rounds were cloned into the TOPOTM TA®, pCR2.1 cloning vector (Invitrogen, Carlsbad, CA) and transformed into E. coli. Between 30 and 100 clones were picked and grown into plasmid minipreps (Qiagen). Thirty-eight clones from R9 and 29 clones from RIO were picked at random and sequenced (DNA Sequencing Core Facility, Massachusetts General Hospital, Dept. of Molecular Biology, Boston, MA). The program ClustalW 60 was used to align the resulting protein sequences.
  • proline is ' found at positions equivalent to the 10 Fn3 residue 52 as frequently as is the wild-type glycine; similarly, alanine, glycine, and the wild-type lysine are all common at positions equivalent to the 10 Fn3 position 54. In consequence, it appears likely that the selected residues at positions 52 and 54 are at least consistent with favorable biophysical properties. In contrast, no tryptophan is found at the position equivalent to the 10 Fn3 residue 53, which suggests that Tryptophan 53 may have been selected for a reason specific to the present selection, such as due to a contribution to TNF- ⁇ binding.
  • NRSGLQS (12/61) SEQ ID NO: 31
  • AQTGHHLHDK 6/61)
  • SEQ ID NO: 32 The sequences selected most frequently in the BC loop.
  • An NRSGLQS BC loop and an AQTGHHLHDK FG loop have not been found in the same molecule, but two clones were found which contain the most frequently isolated sequences on two of the three randomized loops. These clones, T10.06 (BC: NRSGLQS, DE: PWA) and T09.12 (DE: PWA, FG: AQTGHHLHDK), have two of the lowest four dissociation constants from TNF- ⁇ of the clones examined (Table 1).
  • Figure 18 indicates the residues in the 10 Fn3 beta sheet that had not been randomized, but nevertheless mutated during selection. This mutagenesis occurred at the frequency of 26-28 of the 61 clones; these mutations are marked with arrows under the wild-type 10 Fn3 sequence and with the letter that identifies the selected residue. In particular, 28 of the 61 clones mutated from Leucine 18 to Arginine or to Glutamine, and 26 clones mutated from Threonine 56 to Isoleucine. Figure 19 shows the location of these scaffold mutations.
  • the PCR product of the elution after the tenth round of selection was transcribed and translated in vitro, in the presence of 35 S-methionine but without forming the mRNA-protein fusion.
  • the resulting fraction of the free protein bound to TNF- ⁇ -Sepharose, to IL-l ⁇ -Sepharose, to IL-13-Sepharose at approximately 10 ⁇ M, 30 ⁇ M, and 50 ⁇ M, respectively, and to underivatized Sepharose was compared ( Figure 20), using the procedure described above for DNA/mRNA-protein fusion binding to TNF- ⁇ -Sepharose.
  • the amount of the selected pool bound to each of the targets was measured by scintillation counting of the washed beads.
  • Figure 20 shows that, whereas the binding of R0 to TNF- ⁇ , IL-l ⁇ , and IL-13 was similar (2%, 4%, and 3%, respectively), the ten rounds of selection resulted in 32% binding to the targeted TNF- ⁇ , in 3% binding to IL-l ⁇ , and in 1% binding to IL-13.
  • the absolute and the relative increase of protein binding to TNF- ⁇ demonstrates the ability of the 10 Fn3 scaffold and of the DNA mRNA-protein fusion-based selection system to select target-specific binders.
  • clone T09.12 was immobilized in a microarray format (as generally described below) and was tested for binding to soluble TNF- ⁇ . Specific binding of TNF- ⁇ to this clone was detected using fluorescence detection ( Figure 24A) and mass spectroscopy ( Figure 24B). For the mass spectroscopy results, binding assays were carried out in the presence of fetal bovine serum, an exemplary complex biological fluid containing a variety of potential interfering proteins.
  • RNA- 10 Fn3 fusion of wild-type 10 Fn3 and of the T09.12 variant (Table 1) was hybridized onto a DNA microarray on which oligonucleotides complementary to the RNA portion of the fusion molecules had been immobilized at 600 micron pitch, with 24 replicate features. After removal of unliybridized fusion by washing, the surface was exposed to biotin-TNF- ⁇ (2.6 ⁇ g/mL in TBS, 0.02% Tween-20, 0.2% BSA), washed, and air-dried.
  • RNA- 10 Fn3 fusion of the T09.12 variant ( Figure 24B) and wild-type 10 Fn3 ( Figure 24C) was treated with RNase A to generate a fusion between the protein and the DNA linker.
  • the resulting DNA-linked protein was hybridized to a glass coverslip arrayed with an immobilized oligonucleotide complementary to the DNA linker ( Figures 24B and 24C; no fusion was applied in Figure 24D).
  • Dissociation constants were determined for all the clones that were represented more than once in the two pools generated after nine and after ten rounds of selection, as well as for the only clone that originated from the BC wt -DE wt -FG r library (clone T09.08).
  • biotinylated TNF- ⁇ was prepared using the NHS-LC-LC-Biotin reagent supplied by Pierce (Rockford, IL).
  • MALDI-TOF mass spectrometry was used to estimate that more than 80% of the monomeric TNF- ⁇ , and hence more than 99% of the trimer, was biotinylated.
  • each sample was loaded on a pre-soaked, SAM R Biotin Capture Membrane (Promega, Madison, Wl) using a 96 well, Easy-TiterTM ELIFA system (Pierce). Under vacuum, each spot was washed with 200 ⁇ L of HBS pH 7.4, 1 % BSA, 0.05% Triton; next the entire membrane was rinsed in the buffer and air-dried. The membrane was exposed with a Storage Phosphor Screen (Molecular Dynamics, Sunnyvale, CA) overnight, and the intensities of the resulting individual spots were quantified using a STORM 860 phosphoimager with the ImageQuaNT densitometry program (Molecular Dynamics). The K d of the binding was determined by fitting the equilibrium equation to the resulting binding curve (KaleidaGraph, Synergy Software); the error of the experiment was estimated from 2-4 independent experiments.
  • TNF- ⁇ binding Clones Despite the duplicate clones isolated, the TNF- ⁇ -binding pools after nine and after ten rounds of selection contained numerous different clones, i.e., 38 different sequences in 61 clones sampled. Therefore, further selection, with more stringent binding requirements, was undertaken to recover a subset of these clones with superior TNF- ⁇ binding properties. Consequently, four further rounds of selections (Rl 1 -R14) were conducted in solution, where the concentration of the target was controlled more easily.
  • the concentration of TNF- ⁇ was limited to 0.5 nM and the concentration of DNA/mRNA- 10 Fn3 pool to 0.1 nM; in addition, the length and the temperature of the washes of the 10 Fn3/TNF-a complex bound to streptavidin-coated paramagnetic beads were increased.
  • DNA was eluted from the washed DNA/mRNA- 10 Fn3:TNF- ⁇ -biotin:streptavidin-bead complexes with two washes of 100 ⁇ L 0.1 M KOH, and treated as described above for the column-based selection to produce the next generation DNA/mRNA- 10 Fn3 fusion library.
  • Round 14 differed from Rl 1-R13 in that the selection was 5 performed at 30°C and in the presence of an additional 150 mM NaCl. Except for the elevated temperature, the sequence of washes was the same for R14 as for R13.
  • the selections described herein may also be combined with mutagenesis after all or a subset of the selection steps to further increase library diversity.
  • error-prone PCR was incorporated into the amplification of DNA between rounds (Cadwell and Joyce, PCR Methods Appl 2:28 (1992)). This technique was carried out beginning with the diverse DNA pool eluted after R8 above. This pool was amplified using error-prone PCR, with the pool divided into seven equal parts- and mutagenized at the target frequency of 0.8%, 1.6%, 2.4%, 3.2%, 4.0%, 4.8%, and 5.6%. The seven PCR reactions were combined, and cDNA/RNA-protein fusion was made from the mixture and subjected to a round of selection in solution.
  • M10, error-prone PCR was performed in three separate reactions, at 0.8%, 1.6%, and 2.4%.
  • the two remaining rounds, Ml 1 and M12, were performed using standard Taq PCR.
  • the selection conditions for M9-M12 were the same as for Rl 1-R14.
  • the twenty M12 clones tested showed tighter binding to TNF- ⁇ than the clones selected using the two earlier selection protocols (Table 3; full sequences in Figure 25); the tightest binding of TNF- ⁇ was seen in M12.04, and had the observed K d of 20 pM.
  • These results demonstrated that low-level, random mutagenesis late in a selection can improve both the binding affinity of selected antibody mimics (20 pM vs. 90 pM) and the speed with which they can be selected (12 rounds vs. 14 rounds).
  • the frequency of tight binders in this mutagenesis approach was observed to be about 5%, whereas the frequency is approximately 3% in other selections.
  • the randomized loops remained the length of the corresponding, wild-type 10 Fn3 loops.
  • the length of the loops as well as their sequences may be varied, to incorporate favorable mutations in the 10 Fn3 beta-sheet into the wild-type scaffold used for library construction, and to create libraries with randomized beta-sheet scaffolds which will allow selection of structures even more successful at mimicking antibodies. Selections similar to those described herein may be carried out with any other binding species target (for example, IL-1 or IL-13).
  • Wild-type 10 Fn3 contains an integrin-binding tripepetide motif, Arginine 78 - Glycine 79 - Aspartate 80 (the "RGD motif) at the tip of the FG loop.
  • Arginine 78 - Glycine 79 - Aspartate 80 the "RGD motif
  • a mutant form of 10 Fn3 was generated that contained an inert sequence, Serine 78 - Glycine 79 - Glutamate 80 (the "SGE mutant”), a sequence which is found in the closely related, wild-type u Fn3 domain.
  • This SGE mutant was expressed as an N-terminally His 6 -tagged, free protein in K coli, and purified to homogeneity on a metal chelate column followed by a size exclusion column.
  • the DNA sequence encoding His 6 - 10 Fn3(SGE) was cloned into the pET9a expression vector and transformed into BL21 DE3 pLysS cells.
  • the induced culture was further incubated, under the same conditions, overnight (14-18 hours); the bacteria were recovered by standard, low speed centrifugation.
  • the cell pellet was resuspended in 1/50 of the original culture volume of lysis buffer (50 mM Tris 8.0, 0.5 M NaCl, 5% glycerol, 0.05% Triton X-100, and 1 mM PMSF), and the cells were lysed by passing the resulting paste through a Microfluidics Corporation Microfluidizer Ml 10-EH, three times.
  • the lysate was clarified by centrifugation, and the supernatant was filtered through a 0.45 ⁇ m filter followed by filtration through a 0.2 ⁇ m filter.
  • the column was run at 0.8 mL/min, in buffer 1; all fractions that contained a protein of the expected MW were pooled, concentrated 10X as described above, then dialyzed into PBS. Endotoxin screens and animal studies were performed on the resulting sample (Toxikon; MA).
  • This 10 Fn3-F register molecule contains the -CH r CH 2 -CH 3 ( Figure 11) or -CH 2 -CH 3 domains of the IgG constant region of the host; in these constructs, the 10 Fn3 domain is grafted onto the N-terminus in place of the IgG V H domain ( Figures 11 and 12).
  • Such antibody-like constructs are to improve the pharmacokinetics of the protein as well as its ability to harness the natural immune response.
  • the -CH J -CK J -CH J region was first amplified from a mouse liver spleen cDNA library (Clontech), then ligated into the pET25b vector.
  • the primers used in the cloning were 5' Fc Nest and 3' 5 Fc Nest, and the primers used to graft the appropriate restriction sites onto the ends of the recovered insert were 5' Fc HIII and 3' Fc Nhe:
  • Mo 5PCR-NdeI 5' CATATGGTTTCTGATATTCCGAGAGATCTGGAG 3' (SEQ ID NO: 19); Mo5PCR-His-NdeI (for an alternative N-terminus with the His 6 purification tag):
  • the human equivalents of the clones are constructed using the same strategy with human oligonucleotide sequences.
  • Antibody Mimics in Protein Chip Applications Any of the antibody mimics described herein may be immobilized on a solid support, such as a microchip.
  • a solid support such as a microchip.
  • the suitability of the present scaffolds, for example, the 10 Fn3 scaffold, for protein chip applications is the consequence of (1) their ability to support many binding functions which can be selected rapidly on the bench or in an automated setup, and (2) their superior biophysical properties.
  • 10 Fn3 The versatile binding properties of 10 Fn3 are a function of the loops displayed by the Fn3 immunoglobulin-like, beta sandwich fold. As discussed above, these loops are similar to the complementarity determining regions of antibody variable domains and can cooperate in a way similar to those antibody loops in order to bind antigens.
  • 10 Fn3 loops BC for example, residues 21-30
  • DE for example, residues 51-56
  • FG for example, residues 76-87
  • binders in such libraries are then enriched based on their affinity for an immobilized or tagged target, until a small population of high affinity binders are generated.
  • error-prone PCR and recombination can be employed to facilitate affinity maturation of selected binders. Due to the rapid and efficient selection and affinity maturation protocols, binders to a large number of targets can be- selected in a short time.
  • the 10 Fn3 domain has the advantage over antibody fragments and single-chain antibodies of being smaller and easier to handle.
  • 10 Fn3 is extremely stable, with a melting temperature of 110°C, and solubility at a concentration > 16 mg/mL.
  • the 10 Fn3 scaffold also contains no disulfides or free cysteines; consequently, it is insensitive to the redox potential of its environment.
  • a further advantage of 10 Fn3 is that its antigen-binding loops and N-terminus are on the edge of the beta-sandwich opposite to the C-terminus; thus the attachment of a 10 Fn3 scaffold to a chip by its C-terminus aligns the antigen-binding loops, allowing for their greatest accessibility to the solution being assayed. Since 10 Fn3 is a single domain of only 94 amino acid residues, it is also possible to immobilize it onto a chip surface at a higher density than is used for single-chain antibodies, with their approximately 250 residues. In addition, the hydrophilicity of the 10 Fn3 scaffold, which is reflected in the high solubility of this domain, minimizes unwanted binding of 10 Fn3 to a chip surface.
  • the stability of the 10 Fn3 scaffold as well as its suitability for library formation and selection of binders are likely to be shared by the large, Fn3-like class of protein domains with an immunoglobulin-like fold, such as the domains of tenascin, N-cadherin, E-cadherin, ICAM, titin, GCSF-R, cytokine receptor, glycosidase inhibitor, and antibiotic chromoprotein.
  • any of these loops may be immobilized alone or in combination with other loops onto a solid support surface. Immobilization of Fn3-Based Antibody Mimics
  • antibody mimics such as Fn3-based antibody mimics
  • a number of exemplary techniques may be utilized.
  • such antibody mimics may be immobilized as RNA-protein fusions by Watson-Crick hybridization of the RNA moiety of the fusion to a base complementary DNA immobilized on the chip surface (as described, for example, in Addressable Protein Arrays, U.S.S.N. 60/080,686; U.S.S.N. 09/282,734; and WO 99/51773; and Methods for Encoding and Sorting In Vitro Translated Proteins, U.S.S.N. 60/151,261 and U.S.S.N. 09/648,040).
  • antibody mimics can be immobilized as free proteins directly on a chip surface.
  • Manual as well as robotic devices may be used for deposition of the antibody mimics on the chip surface.
  • Spotting robots can be used for deposition of antibody mimics with high density in an array format (for example, by the method of Lueking et al., Anal Biochem. 1999 May 15;270(1):103-11).
  • Different methods may also be utilized for anchoring the antibody mimic on the chip surface.
  • a number of standard immobilization procedures may be used including those described in Methods in Enzymology (K. Mosbach and B. Da ⁇ ielsson, eds.), vols. 135 and 136, Academic Press, Orlando, Florida, 1987; Nilsson et al., Protein Expr. Purif.
  • Oriented immobilization of antibody mimics can help to increase the binding capacity of chip-bound antibody mimics.
  • Exemplary approaches for achieving oriented coupling are described in Lu et al., The Analyst (1996), vol. 121, p. 29R-32R; and Turkova, J Chromatogr B Biomed Sci App. 1999 Feb 5;722(1-2):11-31.
  • any of the methods described herein for anchoring antibody mimics to chip surfaces can also be applied to the immobilization of antibody mimics on beads, or other supports.
  • Selected populations of scaffold-binders may be used for detection and/or quantitation of analyte targets, for example, in samples such as biological samples.
  • selected scaffold-binders to targets of interest are immobilized on an appropriate support to form multi-featured protein chips.
  • a sample is applied to the chip, and the components of the sample that associate with the binders are identified based on the target-specificity of the immobilized binders.
  • one or more components may be simultaneously identified or quantitated in a sample (for example, as a means to carry out sample profiling).
  • Methods for target detection allow measuring the levels of bound protein targets and include, without limitation, radiography, fluorescence scanning, mass spectroscopy (MS), and surface plasmon resonance (SPR).
  • Autoradiography using a phosphorimager system can be used for detection and quantification of target protein which has been radioactively labeled, e.g., using 35 S methionine.
  • Fluorescence scanning using a laser scanner may be used for detection and quantification of fluorescently labeled targets.
  • fluorescence scanning may be used for the detection of fluorescently labeled ligands which themselves bind to the target protein (e.g., fluorescently labeled target-specific antibodies or fluorescently labeled streptavidin binding to target-biotin, as described below).
  • Mass spectroscopy can be used to detect and identify bound targets based on their molecular mass.
  • Desorption of bound target protein can be achieved with laser assistance directly from the chip surface as described below. Mass detection also allows determinations, based on molecular mass, of target modifications including post-translational modifications like phosophorylation or glycosylation. Surface plasmon resonance can be used for quantification of bound protein targets where the scaffold-binder(s) are immobilized on a suitable gold-surface (for example, as obtained from Biacore, Sweden).
  • binders in this case, Fn-binders specific for the protein, TNF- ⁇
  • TNF- ⁇ binders specific for the protein
  • This example is provided for the purpose of illustrating the invention, and should not be construed as limiting.
  • an 10 Fn3-based selection was perforaied against TNF- ⁇ , using a library of human 10 Fn3 variants with randomized loops BC, DE, and FG.
  • the library was constructed from three DNA fragments, each of which contained nucleotide sequences that encoded approximately one third of human 10 Fn3, including one of the randomized loops.
  • the DNA sequences that encoded the loop residues listed above were rebuilt by oligonucleotide synthesis, so that the codons for the residues of interest were replaced by (NNS) n , where N represents any of the four deoxyribonucleotides (A, C, G, or T), and S represents either C or G.
  • N represents any of the four deoxyribonucleotides (A, C, G, or T)
  • S represents either C or G.
  • the C-terminus of each fragment contained the sequence for the FLAG purification tag.
  • each DNA fragment was transcribed, ligated to a puromycin-containing DNA linker, and translated in vitro, as described by Szostak et al. (Roberts and Szostak, Proc. Natl. Acad. Sci USA 94:12297, 1997; Szostak et al, U.S.S.N. 09/007,005 and U.S.S.N. 09/247,190; Szostak et al, WO98/31700), to generate an mRNA-peptide fusion, which was then reverse-transcribed into a DNA-mRNA-peptide fusion.
  • the binding of the FLAG-tagged peptide to M2 agarose separated full-length fusion molecules from those containing frameshifts or superfluous stop codons; the DNA associated with the purified full-length fusion was amplified by PCR, then the three DNA fragments were cut by Ear I restriction endonuclease and ligated to form the full length template.
  • the template was transcribed, ligated to puromycin-containing DNA linkers, and translated to generate a 10 Fn3- RNA/cDNA library, which was then reverse-transcribed to yield the DNA-mRNA-peptide fusion library which was subsequently used in the selection.
  • TNF- ⁇ binders were placed in 50 mM HEPES, pH 7.4, 0.02% Triton-X, 0.1 mg/mL salmon sperm DNA.
  • the PROfusionTM library was incubated with Sepharose-immobilized TNF- ⁇ ; after washing, the DNA associated with the tightest binders was eluted with 0.1 M KOH, amplified by PCR, and transcribed, ligated, translated, and reverse-transcribed into the starting material for the next round of selection.
  • an oligonucleotide capture probe was prepared with an automated DNA synthesizer (PE BioSystems Expedite 8909) using the solid-support phosphoramidite approach. All reagents were obtained from Glen Research. Synthesis was initiated with a solid support containing a disulfide bond to eventually provide a 3 '-terminal thiol functionality. The first four monomers to be added were hexaethylene oxide units, followed by 20 T monomers. The 5'-terminal DMT group was not removed.
  • the capture probe was cleaved from the solid support and deprotected with ammonium hydroxide, concentrated to dryness in a vacuum centrifuge, and purified by reverse-phase HPLC using an acetonitrile gradient in triethylammonium acetate buffer. Appropriate fractions from the HPLC were collected, evaporated to dryness in a vacuum centrifuge, and the 5 '-terminal DMT group was removed by treatment with 80% AcOH for 30 minutes. The acid was removed by evaporation, and the oligonucleotide was then treated with 100 mM DTT for 30 minutes to cleave the disulfide bond. DTT was removed by repeated extraction with EtOAc.
  • the oligonucleotide was ethanol precipitated from the remaining aqueous layer and checked for purity by reverse-phase HPLC.
  • the 3'-thiol capture probe was adjusted to 250 ⁇ M in degassed IX PBS buffer and applied as a single droplet (75 ⁇ L) to a 9x9mm gold-coated chip (Biacore) in an argon-flushed chamber containing a small amount of water. After 18 hours at room temperature, the capture probe solution was removed, and the functionalized chip was washed with 50 mL IX PBS buffer (2x for 15 minutes each) with gentle agitation, and then rinsed with 50 mL water (2x for 15 minutes each) in the same fashion. Remaining liquid was carefully removed and the functionalized chips were either used immediately or stored at 4°C under argon. About lpmol of 10 Fn3 fusion pool from the Round 10 TNF- ⁇ selection
  • RNAse A (above) was treated with RNAse A for several hours, adjusted to 5X SSC in 70 ⁇ L, and applied to a functionalized gold chip from above as a single droplet.
  • a 50 ⁇ L volume gasket device was used to seal the fusion mixture with the functionalized chip, and the apparatus was continuously rotated at 4°C. After 18 hours the apparatus was disassembled, and the gold chip was washed with 50 mL 5X SSC for 10 minutes with gentle agitation. Excess liquid was carefully removed from the chip surface, and the chip was passivated with a blocking solution (IX TBS + 0.02% Tween-20 + 0.25% BSA) for 10 minutes at 4°C.
  • a blocking solution IX TBS + 0.02% Tween-20 + 0.25% BSA
  • MALDI-TOF matrix (15 mg/mL 3,5-dimethoxy-4-hydroxycinnamic acid in 1: 1 ethanol/ 10% formic acid in water) was uniformly applied to the gold chips with a high-precision 3-axis robot (MicroGrid, BioRobotics).
  • a 16-pin tool was used to transfer the matrix from a 384-well microtiter plate to the chips, producing 200 micron diameter features with a 600 micron pitch.
  • the gold chips were individually placed on a MALDI sample stage modified to keep the level of the chip the same as the level of the stage, thus allowing proper flight distance.
  • the instrument's video monitor and motion control system were used to direct the laser beam to individual matrix features.
  • Figures 14 and 15 show the mass spectra from the 10 Fn3 fusion chip and the non-fusion chip, respectively. In each case, a small number of 200 micron features were analyzed to collect the spectra, but Figure 15 required significantly more acquisitions.
  • the signal at 17.4 kDa corresponds to TNF- ⁇ monomer.
  • silane was removed by subjecting the slides to ten 5-minute washes, using fresh portions of 95% acetone / 5% water for each wash, with gentle agitation. The slides were then cured by heating at 110°C for 20 minutes. The silane treated slides were immersed in a freshly prepared 0.2% solution of phenylene 1,4-diisothiocyanate in 90% DMF / 10% pyridine for two hours, with gentle agitation. The slides were washed sequentially with 90% DMF / 10% pyridine, methanol, and acetone. After air drying, the functionalized slides were stored at 0°C in a vacuum desiccator over anhydrous calcium sulfate.
  • Oligonucleotide capture probes were prepared with an automated DNA synthesizer (PE BioSystems Expedite 8909) using conventional phosphoramidite chemistry. All reagents were from Glen Research. Synthesis was initiated with a solid support bearing an orthogonally protected amino functionality, whereby the 3 '-terminal amine is not unmasked until final deprotection step. The first four monomers to be added were hexaethylene oxide units, followed by the standard A, G, C and T monomers.
  • the purified, amine-labeled capture oligos were adjusted to a concentration of 250 ⁇ M in 50 mM sodium carbonate buffer (pH 9.0) containing 10% glycerol.
  • the probes were spotted onto the amine-reactive glass surface at defined positions in a 5x5x6 array pattern with a 3-axis robot (MicroGrid, BioRobotics).
  • a 16-pin tool was used to transfer the liquid from 384-well microtiter plates, producing 200 micron features with a 600 micron pitch. Each sub-grid of 24 features represents a single capture probe (i.e., 24 duplicate spots).
  • the arrays were incubated at room temperature in a moisture-saturated environment for 12-18 hours.
  • the attachment reaction was terminated by immersing the chips in 2% aqueous ammonium hydroxide for five minutes with gentle agitation, followed by rinsing with distilled water (3X for 5 minutes each).
  • the array was finally soaked in 10X PBS solution for 30 minutes at room temperature, and then rinsed again for 5 minutes in distilled water.
  • Specific and thermodynamically isoenergetic sequences along the 10 Fn3 mRNA were identified to serve as capture points to self-assemble and anchor the 10 Fn3 protein.
  • the software program HybSimulator v4.0 Advanced Gene Computing Technology, Inc. facilitated the identification and analysis of potential capture probes.
  • Each of the capture probes possesses a 3'-amino terminus and four hexaethylene oxide spacer units, as described above. The following is a list of the capture probe sequences that were employed (5' ⁇ 3'):
  • CPnegl CCTGTAGGTGTCCAT (SEQ ID NO: 24)
  • CPflag CATCGTCCTTGTAGTC (SEQ ID NO: 25)
  • CPneg2 CGTCGTAGGGGTA (SEQ ID NO: 26)
  • CPneg3 CAGGTCTTCTTCAGAGA (SEQ ID NO: 27)
  • Recombinant human TNF- ⁇ (500 ⁇ g, lyophilized, from PreproTech) was taken up in 230 ⁇ L IX PBS and dialyzed against 700 mL stirred IX PBS at 4°C for 18 hours in a Microdialyzer unit (3,500 MWCO, Pierce).
  • the dialyzed TNF- ⁇ was treated with EZ-Link NHS-LC-LC biotinylation reagent (20 ⁇ g, Pierce) for 2 hours at 0°C, and again dialyzed against 700 mL stirred IX PBS at 4°C for 18 hours in a Microdialyzer unit (3,500 MWCO, Pierce).
  • the resulting conjugate was analyzed by MALDI-TOF mass spectrometry and was found to be almost completely functionalized with a single biotin moiety.
  • the protein microarray surface was passivated by treatment with IX TBS containing 0.02% Tween-20 and 0.2% BSA (200 ⁇ L) for 60 minutes.
  • Biotinylated TNF- ⁇ 100 nM concentration made up in the passivation buffer
  • the microarray was washed with IX TBS containing 0.02% Tween-20 (3X 50 mL, 5 minutes each wash).
  • Fluorescently labeled streptavidin (2.5 ⁇ g/mL Alexa 546-streptavidin conjugate from Molecular Probes, made up in the passivation buffer) was contacted with the microarray for 60 minutes.
  • the microarray was washed with IX TBS containing 0.02% Tween-20 (2X 50 mL, 5 minutes each wash) followed by a 3 minute rinse with IX TBS. Traces of liquid were removed by centrifugation, and the slide was allowed to air-dry at room temperature. Fluorescence laser scanning was performed with a GSI Lumonics ScanArray 5000 system using 10 ⁇ M pixel resolution and preset excitation and emission wavelengths for Alexa 546 dye. Phosphorimage analysis was performed with a Molecular Dynamics Storm system. Exposure time was 48 hours with direct contact between the microarray and the phosphor storage screen. Phosphorimage scanning was performed at the 50 micron resolution setting, and data was extracted with ImageQuant v.4.3 software.
  • Figures 16 and 17 are the phosphorimage and fluorescence scan, respectively, of the same array.
  • the phosphorimage shows where the 10 Fn3 fusion hybridized based on the 35 S methionine signal.
  • the fluorescence scan shows where the labeled TNF- ⁇ bound.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • General Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Zoology (AREA)
  • Toxicology (AREA)
  • Immunology (AREA)
  • Peptides Or Proteins (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
EP01981621A 2000-10-16 2001-10-16 Proteingerüste für antikörpermimetika und andere bindungsproteine Withdrawn EP1356075A4 (de)

Priority Applications (1)

Application Number Priority Date Filing Date Title
EP09167669A EP2141243A3 (de) 2000-10-16 2001-10-16 Proteingerüste für Antikörpermimetika und andere Bindungsproteine

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US68856600A 2000-10-16 2000-10-16
US688566 2000-10-16
PCT/US2001/032233 WO2002032925A2 (en) 2000-10-16 2001-10-16 Protein scaffolds for antibody mimics and other binding proteins

Related Child Applications (1)

Application Number Title Priority Date Filing Date
EP09167669A Division EP2141243A3 (de) 2000-10-16 2001-10-16 Proteingerüste für Antikörpermimetika und andere Bindungsproteine

Publications (2)

Publication Number Publication Date
EP1356075A2 true EP1356075A2 (de) 2003-10-29
EP1356075A4 EP1356075A4 (de) 2005-04-13

Family

ID=24764915

Family Applications (2)

Application Number Title Priority Date Filing Date
EP01981621A Withdrawn EP1356075A4 (de) 2000-10-16 2001-10-16 Proteingerüste für antikörpermimetika und andere bindungsproteine
EP09167669A Ceased EP2141243A3 (de) 2000-10-16 2001-10-16 Proteingerüste für Antikörpermimetika und andere Bindungsproteine

Family Applications After (1)

Application Number Title Priority Date Filing Date
EP09167669A Ceased EP2141243A3 (de) 2000-10-16 2001-10-16 Proteingerüste für Antikörpermimetika und andere Bindungsproteine

Country Status (5)

Country Link
EP (2) EP1356075A4 (de)
JP (4) JP2004526419A (de)
AU (2) AU2002213251B2 (de)
CA (1) CA2418835A1 (de)
WO (1) WO2002032925A2 (de)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9139825B2 (en) 2009-10-30 2015-09-22 Novartis Ag Universal fibronectin type III bottom-side binding domain libraries

Families Citing this family (170)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7115396B2 (en) 1998-12-10 2006-10-03 Compound Therapeutics, Inc. Protein scaffolds for antibody mimics and other binding proteins
US6818418B1 (en) * 1998-12-10 2004-11-16 Compound Therapeutics, Inc. Protein scaffolds for antibody mimics and other binding proteins
CA2416219C (en) 2000-07-11 2016-10-11 Research Corporation Technologies, Inc. Artificial antibody polypeptides
CA2418835A1 (en) * 2000-10-16 2002-04-25 Phylos, Inc. Protein scaffolds for antibody mimics and other binding proteins
CN1678634A (zh) * 2002-06-28 2005-10-05 多曼蒂斯有限公司 免疫球蛋白单个变体抗原结合区及其特异性构建体
US20050074865A1 (en) 2002-08-27 2005-04-07 Compound Therapeutics, Inc. Adzymes and uses thereof
WO2004096271A1 (en) 2003-04-30 2004-11-11 Uwe Zangemeister-Wittke Methods for treating cancer using an immunotoxin
SG149004A1 (en) 2003-12-05 2009-01-29 Bristol Myers Squibb Co Inhibitors of type 2 vascular endothelial growth factor receptors
JP2007537716A (ja) * 2003-12-16 2007-12-27 コモンウェルス サイエンティフィック アンド インダストリアル リサーチ オーガナイゼイション サイトカイン結合ドメイン
EP2330120A3 (de) * 2004-06-02 2011-11-16 AdAlta Pty Ltd Bindende Teile basierend auf Hai-IgNAR-Domänen
EA011879B1 (ru) 2004-09-24 2009-06-30 Эмджин Инк. МОЛЕКУЛЫ С МОДИФИЦИРОВАННЫМ Fc ФРАГМЕНТОМ
RS50785B (sr) * 2005-01-05 2010-08-31 F-Star Biotechnologische Forschungs-Und Entwicklungsges M.B.H. Domeni sintetskih imunoglobulina sa svojstvima vezivanja, konstruisani u svojstvima molekula, koji se razlikuju od regiona, koji određuju komplementarnost
US8008453B2 (en) 2005-08-12 2011-08-30 Amgen Inc. Modified Fc molecules
WO2007056812A1 (en) * 2005-11-16 2007-05-24 Apollo Life Sciences Limited A molecule and chimeric molecules thereof
EP3811965A1 (de) 2005-11-23 2021-04-28 Acceleron Pharma, Inc. Activin-actriia antagonisten zur verwendung in der förderung von knochenwachstum
US9181543B2 (en) 2006-05-26 2015-11-10 Obodies Limited OB fold domains
AT503889B1 (de) 2006-07-05 2011-12-15 Star Biotechnologische Forschungs Und Entwicklungsges M B H F Multivalente immunglobuline
WO2008031098A1 (en) * 2006-09-09 2008-03-13 The University Of Chicago Binary amino acid libraries for fibronectin type iii polypeptide monobodies
CN101588816B (zh) 2006-10-19 2013-06-19 Csl有限公司 白介素-13受体α1的高亲和性抗体拮抗物
AU2007325838B2 (en) 2006-11-22 2013-09-19 Bristol-Myers Squibb Company Targeted therapeutics based on engineered proteins for tyrosine kinases receptors, including IGF-IR
EP1930342B1 (de) 2006-12-04 2012-01-25 Institut Pasteur Als Gerüst verwendeter OB-Fold zur Entwicklung neuer spezifischer Bindemittel
ME02335B (de) 2006-12-18 2013-04-30 Acceleron Pharma Inc Activin-actrii-antagonisten und ihre verwendungen zur behandlung von anämie
ES2415666T3 (es) 2007-02-01 2013-07-26 Acceleron Pharma, Inc. Composiciones farmacéuticas que comprenden antagonistas de Activina-ActRIIa para uso en la prevención o el tratamiento de metástasis de cáncer de mama o pérdida ósea relacionada con el cáncer de mama
WO2008097497A2 (en) 2007-02-02 2008-08-14 Adnexus, A Bristol-Myers Squibb R & D Company Vegf pathway blockade
TW201934141A (zh) 2007-02-09 2019-09-01 美商艾瑟勒朗法瑪公司 包含ActRIIa-Fc融合蛋白的醫藥組合物;ActRIIa-Fc融合蛋白於治療或預防與癌症相關的骨質流失之用途;ActRIIa-Fc融合蛋白於治療或預防多發性骨髓瘤之用途
EP2076289B1 (de) 2007-04-13 2014-11-12 Dana-Farber Cancer Institute, Inc. Methoden zur behandlung von erbb-therapie resistenten krebs
EP2145021A2 (de) 2007-05-17 2010-01-20 Bristol-Myers Squibb Company Biomarker und verfahren zur bestimmung der sensitivität gegenüber insulinwachstumsfaktor-1-rezeptormodulatoren
JP2010527936A (ja) * 2007-05-18 2010-08-19 メディミューン,エルエルシー 炎症性疾患におけるil−33
HUE066143T2 (hu) 2007-06-26 2024-07-28 F Star Therapeutics Ltd Kötõágensek megjelenítése
US8680019B2 (en) 2007-08-10 2014-03-25 Protelica, Inc. Universal fibronectin Type III binding-domain libraries
WO2009023184A2 (en) 2007-08-10 2009-02-19 Protelix, Inc. Universal fibronectin type iii binding-domain libraries
US8470966B2 (en) 2007-08-10 2013-06-25 Protelica, Inc. Universal fibronectin type III binding-domain libraries
EP2207562B1 (de) 2007-09-18 2017-05-31 Acceleron Pharma, Inc. Activin-actriia antagonisten und verfahren zur verringerung oder inhibierung der fsh-sekretion
WO2009058379A2 (en) 2007-10-31 2009-05-07 Medimmune, Llc Protein scaffolds
EP2207568B1 (de) 2007-11-16 2017-05-31 The Rockefeller University Für die protofibrillenform des proteins beta-amyloid spezifische antikörper
KR20100111283A (ko) * 2007-12-27 2010-10-14 노파르티스 아게 개선된 피브로넥틴계 결합 분자 및 그들의 용도
US9493747B2 (en) 2008-01-03 2016-11-15 The General Hospital Corporation Engineered transglutaminase barrel proteins
MX2010008874A (es) 2008-02-14 2010-09-22 Bristol Myers Squibb Co Terapeuticos dirigidos a base de proteinas manipuladas que se unen al receptor de factor de crecimiento epidermico.
PL2275443T3 (pl) 2008-04-11 2016-05-31 Chugai Pharmaceutical Co Ltd Cząsteczka wiążąca antygen zdolna do wiązania dwóch lub więcej cząsteczek antygenu w sposób powtarzalny
EP2113255A1 (de) 2008-05-02 2009-11-04 f-star Biotechnologische Forschungs- und Entwicklungsges.m.b.H. Zytotoxisches Immunglobulin
EP2274331B1 (de) * 2008-05-02 2013-11-06 Novartis AG Verbesserte bindemoleküle auf fibronectin-basis und ihre verwendung
CN102099373A (zh) 2008-05-22 2011-06-15 百时美施贵宝公司 基于纤连蛋白的多价支架结构域蛋白
CA2729100C (en) 2008-06-26 2018-01-02 Acceleron Pharma Inc. Methods for dosing an activin-actriia antagonist and monitoring of treated patients
CA2729054C (en) 2008-06-26 2019-09-03 Acceleron Pharma Inc. Antagonists of actriib and uses for increasing red blood cell levels
US8415291B2 (en) * 2008-10-31 2013-04-09 Centocor Ortho Biotech Inc. Anti-TNF alpha fibronectin type III domain based scaffold compositions, methods and uses
DK2356269T3 (en) * 2008-10-31 2016-08-15 Janssen Biotech Inc FIBRONECTIN TYPE III DOMAIN-BASED SCAFFOLD COMPOSITIONS, PROCEDURES AND APPLICATIONS
TWI496582B (zh) 2008-11-24 2015-08-21 必治妥美雅史谷比公司 雙重專一性之egfr/igfir結合分子
AU2009327189B2 (en) 2008-12-16 2012-09-06 Novartis Ag Yeast display systems
WO2010093627A2 (en) * 2009-02-12 2010-08-19 Centocor Ortho Biotech Inc. Fibronectin type iii domain based scaffold compositions, methods and uses
CA2757095C (en) 2009-03-30 2020-04-14 Acceleron Pharma Inc. Bmp-alk3 antagonists and uses for promoting bone growth
US8067201B2 (en) 2009-04-17 2011-11-29 Bristol-Myers Squibb Company Methods for protein refolding
ES2869580T3 (es) 2009-09-09 2021-10-25 Acceleron Pharma Inc Antagonistas de ActRIIB y dosificación y usos de los mismos para tratar obesidad o diabetes tipo 2 regulando el contenido de grasa corporal
WO2011053779A2 (en) 2009-10-30 2011-05-05 Bristol-Myers Squibb Company Methods for treating cancer in patients having igf-1r inhibitor resistance
BR112012019881A2 (pt) * 2010-02-18 2017-06-27 Bristol Myers Squibb Co proteínas de domínio estrutural baseadas na fibronectina que ligam-se à il-23
TWI667257B (zh) 2010-03-30 2019-08-01 中外製藥股份有限公司 促進抗原消失之具有經修飾的FcRn親和力之抗體
CA2795325A1 (en) 2010-04-13 2011-10-20 Medimmune, Llc Fibronectin type iii domain-based multimeric scaffolds
RU2603272C2 (ru) 2010-04-30 2016-11-27 Янссен Байотек, Инк. Композиции на основе стабилизированных фибронектиновых доменов, способы и области их применения
TW201138808A (en) 2010-05-03 2011-11-16 Bristol Myers Squibb Co Serum albumin binding molecules
US9562089B2 (en) 2010-05-26 2017-02-07 Bristol-Myers Squibb Company Fibronectin based scaffold proteins having improved stability
AU2011283646B2 (en) * 2010-07-30 2015-07-09 Novartis Ag Fibronectin cradle molecules and libraries thereof
KR101398363B1 (ko) 2010-11-17 2014-05-22 추가이 세이야쿠 가부시키가이샤 혈액응고 제viii 인자의 기능을 대체하는 기능을 갖는 다중특이성 항원 결합 분자
MX365235B (es) 2010-11-30 2019-05-28 Chugai Pharmaceutical Co Ltd Molécula de unión a antígeno capaz de unir repetidamente a la pluralidad de moléculas de antígeno.
WO2012093704A1 (ja) 2011-01-07 2012-07-12 中外製薬株式会社 抗体の物性を改善させる方法
EP2679681B2 (de) 2011-02-25 2023-11-15 Chugai Seiyaku Kabushiki Kaisha FcgammaRIIB-spezifischer Fc Antikörper
DK2683708T3 (da) 2011-03-11 2018-01-29 Celgene Corp Faste former af 3-(5-amino-2-methyl-4-oxo-4h-quinazolin-3-yl)-piperidin-2,6-dion og farmaceutiske sammensætninger og anvendelser deraf
BR112013023280A2 (pt) 2011-03-11 2017-09-19 Celgene Corp uso de 3-(5-amino-2-metil-4-oxoquinazolina-3(4h)-il)-piperidina-2,6-diona em tratamento de doenças inflamatórias e relacionadas ao sistema imune
DK2698431T3 (da) 2011-03-30 2020-11-30 Chugai Pharmaceutical Co Ltd Opretholdelse af antigen-bindende molekyler i blodplasma og fremgangsmåde til modifikation af immunogenicitet
DK2697257T3 (en) 2011-04-13 2017-01-30 Bristol Myers Squibb Co FC FUSION PROTEINS INCLUDING UNKNOWN LINKERS OR EVENTS
US20140187488A1 (en) 2011-05-17 2014-07-03 Bristol-Myers Squibb Company Methods for maintaining pegylation of polypeptides
US9347058B2 (en) 2011-05-17 2016-05-24 Bristol-Myers Squibb Company Methods for the selection of binding proteins
BR112013032630B1 (pt) 2011-06-30 2022-06-14 Chugai Seiyaku Kabushiki Kaisha Polipeptídeo heterodimerizado compreendendo região fc de igg
US9200273B2 (en) * 2011-09-27 2015-12-01 Janssen Biotech, Inc. Fibronectin type III repeat based protein scaffolds with alternative binding surfaces
WO2013046722A1 (ja) 2011-09-30 2013-04-04 中外製薬株式会社 イオン濃度依存性結合分子ライブラリ
KR102143331B1 (ko) 2011-09-30 2020-08-11 추가이 세이야쿠 가부시키가이샤 항원의 소실을 촉진시키는 항원 결합 분자
US20150050269A1 (en) 2011-09-30 2015-02-19 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule promoting disappearance of antigens having plurality of biological activities
KR102239138B1 (ko) 2011-09-30 2021-04-12 추가이 세이야쿠 가부시키가이샤 표적 항원에 대한 면역응답을 유도하는 항원 결합 분자
TW201817745A (zh) 2011-09-30 2018-05-16 日商中外製藥股份有限公司 具有促進抗原清除之FcRn結合域的治療性抗原結合分子
JP6271251B2 (ja) 2011-10-05 2018-01-31 中外製薬株式会社 糖鎖受容体結合ドメインを含む抗原の血漿中からの消失を促進する抗原結合分子
DK2771022T3 (da) 2011-10-11 2020-09-28 Viela Bio Inc Cd40l-specifikke tn3-afledte skeletter (scaffolds) og fremgangsmåder til anvendelse deraf
WO2013056069A1 (en) 2011-10-13 2013-04-18 Bristol-Myers Squibb Company Methods for selecting and treating cancer in patients with igf-1r/ir inhibitors
EP2773659A2 (de) 2011-10-31 2014-09-10 Bristol-Myers Squibb Company Fibronectinbindungsdomänen mit verminderter immunogenität
CN104080909A (zh) 2011-11-30 2014-10-01 中外制药株式会社 包含进入细胞内以形成免疫复合体的搬运体(载体)的药物
SG10201704849PA (en) 2012-02-09 2017-07-28 Chugai Pharmaceutical Co Ltd Modified fc region of antibody
ES2795419T3 (es) 2012-02-24 2020-11-23 Chugai Pharmaceutical Co Ltd Molécula de unión al antígeno que promueve la desaparición del antígeno vía Fc RIIB
JPWO2013180200A1 (ja) 2012-05-30 2016-01-21 中外製薬株式会社 標的組織特異的抗原結合分子
ES2856272T3 (es) 2012-05-30 2021-09-27 Chugai Pharmaceutical Co Ltd Molécula de unión a antígenos para eliminar antígenos agregados
EP4310191A3 (de) 2012-06-14 2024-05-15 Chugai Seiyaku Kabushiki Kaisha Antigenbindendes molekül mit modifizierter fc-region
EP3741372A1 (de) 2012-08-09 2020-11-25 Celgene Corporation (s)-3-[4-(4-morphlin-4-ylmethylbenzyloxy)-1- oxo-1,3-dihydro-isoindo-2-yl]piperidine-2,6-dione zur behandlung von immunbedingten entzündungserkrankungen
CA2878954C (en) 2012-08-09 2020-12-08 Benjamin M. Cohen Salts and solid forms of (s)-3-(4-((4-(morpholinomethyl)benzyl)oxy)-1-oxoisoindolin-2-yl)piperidine-2,6-dione and compositions comprising and methods of using the same
KR102146848B1 (ko) 2012-08-09 2020-08-21 셀진 코포레이션 3-(4-((4-모르포리노메틸)벤질)옥시)-1-옥소이소인돌린-2-일)피페리딘-2,6-디온을 이용한 암의 치료방법
JP6774164B2 (ja) 2012-08-24 2020-10-21 中外製薬株式会社 マウスFcγRII特異的Fc抗体
US10919953B2 (en) 2012-08-24 2021-02-16 Chugai Seiyaku Kabushiki Kaisha FcgammaRIIB-specific Fc region variant
WO2014039775A1 (en) 2012-09-07 2014-03-13 Rutgers, The State University Of New Jersey Hiv-1 antigens with discrete conformational forms of the v1/v2 domain and methods of use thereof
PE20150954A1 (es) 2012-09-13 2015-06-20 Bristol Myers Squibb Co Proteinas del dominio de soporte basadas en fibronectina que se fijan a miostatina
TWI609083B (zh) 2012-09-28 2017-12-21 Chugai Pharmaceutical Co Ltd Blood coagulation reaction assessment method
EP3608419A1 (de) 2012-10-24 2020-02-12 Celgene Corporation Biomarker zur verwendung bei der behandlung von anämie
ES2884095T3 (es) 2012-11-02 2021-12-10 Celgene Corp Antagonistas de activina-actrii y usos para el tratamiento de trastornos óseos y otros trastornos
EP3489396B1 (de) 2012-12-27 2021-03-03 National Institute of Advanced Industrial Science and Technology Auf basis der backbone-struktur von mikroproteinen konstruierte molekülbibliothek
EP2940135B9 (de) 2012-12-27 2021-09-08 Chugai Seiyaku Kabushiki Kaisha Heterodimerisiertes polypeptid
WO2014120891A2 (en) 2013-02-01 2014-08-07 Bristol-Myers Squibb Company Fibronectin based scaffold proteins
ES2689372T3 (es) 2013-02-06 2018-11-13 Bristol-Myers Squibb Company Proteínas de dominio de fibronectina tipo III con solubilidad mejorada
ES2645634T3 (es) 2013-02-12 2017-12-07 Bristol-Myers Squibb Company Métodos de replegado de proteínas a elevado pH
EP2968587A2 (de) 2013-03-13 2016-01-20 Bristol-Myers Squibb Company Serumalbumingebundene fibronectin-gerüstdomänen oder daran bindendes element
CN113621057A (zh) 2013-04-02 2021-11-09 中外制药株式会社 Fc区变体
AU2014358191B2 (en) 2013-12-04 2020-12-24 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecules, the antigen-binding activity of which varies according to the concentration of compounds, and libraries of said molecules
CN113150117A (zh) 2014-03-20 2021-07-23 百时美施贵宝公司 新的结合血清白蛋白的纤连蛋白iii型结构域
MX371403B (es) 2014-03-20 2020-01-29 Bristol Myers Squibb Co Moleculas de andamiaje a base de fibronectina estabilizada.
TWI831106B (zh) 2014-06-20 2024-02-01 日商中外製藥股份有限公司 用於因第viii凝血因子及/或活化的第viii凝血因子的活性降低或欠缺而發病及/或進展的疾病之預防及/或治療之醫藥組成物
MA41052A (fr) 2014-10-09 2017-08-15 Celgene Corp Traitement d'une maladie cardiovasculaire à l'aide de pièges de ligands d'actrii
SI3227675T1 (sl) 2014-12-03 2023-07-31 Celgene Corporation Antagonisti aktivin-actrii in uporaba za zdravljenje mielodisplastičnega sindroma
KR102650420B1 (ko) 2014-12-19 2024-03-21 추가이 세이야쿠 가부시키가이샤 항-마이오스타틴 항체, 변이체 Fc 영역을 함유하는 폴리펩타이드, 및 사용 방법
CN106397606B (zh) * 2015-01-28 2020-11-10 中国科学院天津工业生物技术研究所 一种多肽复合物作为sst药物载体的应用、方法及其融合蛋白复合物
US9969800B2 (en) 2015-02-05 2018-05-15 Chugai Seiyaku Kabushiki Kaisha IL-8 antibodies
KR102057767B1 (ko) 2015-04-17 2019-12-19 에프. 호프만-라 로슈 아게 응고 인자 및 다중특이적 항체의 조합 요법
JP7019423B2 (ja) * 2015-05-06 2022-02-15 ヤンセン バイオテツク,インコーポレーテツド 前立腺特異的膜抗原(psma)二重特異性結合剤及びその使用
EP3292222A4 (de) * 2015-05-06 2018-10-17 Janssen Biotech, Inc. Prostataspezifisches membranantigen zur bindung von fibronectin-typ-iii-domänen
CN108350057A (zh) 2015-05-20 2018-07-31 细胞基因公司 使用II型活化素受体配体阱的用于β-地中海贫血的体外细胞培养方法
TWI814162B (zh) 2015-09-18 2023-09-01 日商中外製藥股份有限公司 Il-8結合抗體及其用途
US10766946B2 (en) 2015-09-23 2020-09-08 Bristol-Myers Squibb Company Fast-off rate serum albumin binding fibronectin type III domains
CN108884147B (zh) * 2015-09-23 2024-02-27 百时美施贵宝公司 结合磷脂酰肌醇蛋白聚糖3的基于纤连蛋白的支架分子
EA201891289A1 (ru) 2015-12-02 2018-11-30 Селджин Корпорейшн Циклическая терапия с применением 3-(5-амино-2-метил-4-оксо-4h-хиназолин-3-ил)пиперидин-2,6-диона
LT3402491T (lt) 2016-01-11 2022-02-25 Rubius Therapeutics, Inc. Kompozicijos ir būdai, susiję su daugiamodalinėmis terapinėmis ląstelių sistemomis, skirti vėžio indikacijoms
US11634705B2 (en) 2016-03-30 2023-04-25 Epsilon Molecular Engineering Inc. High-speed in vitro screening method
ES2929650T3 (es) 2016-03-30 2022-11-30 Epsilon Molecular Eng Inc Método para el cribado in vitro de alta velocidad
MX2019002510A (es) 2016-09-06 2019-06-24 Chugai Pharmaceutical Co Ltd Metodos para usar un anticuerpo biespecifico que reconoce el factor de coagulacion ix y/o el factor de coagulacion ix activado y el factor de coagulacion x y/o factor de coagulacion x activado.
EP3468991A1 (de) 2016-11-21 2019-04-17 cureab GmbH Anti-gp73-antikörper und immunkonjugate
BR112019008265A2 (pt) 2016-11-28 2019-07-09 Chugai Pharmaceutical Co Ltd molécula de ligação ao ligante cuja atividade de li-gação ao ligante é ajustável
CN110177875B (zh) 2016-11-28 2023-11-28 中外制药株式会社 包含抗原结合结构域和运送部分的多肽
JP7104703B2 (ja) 2016-12-14 2022-07-21 ヤンセン バイオテツク,インコーポレーテツド Cd8a結合フィブロネクチンiii型ドメイン
EP3554535A4 (de) 2016-12-14 2020-10-21 Janssen Biotech, Inc. Pd-l1-bindende fibronectin-typ iii-domänen
US10611823B2 (en) 2016-12-14 2020-04-07 Hanssen Biotech, Inc CD137 binding fibronectin type III domains
US20180258064A1 (en) 2017-03-07 2018-09-13 Celgene Corporation Solid forms of 3-(5-amino-2-methyl-4-oxo-4h-quinazolin-3-yl)-piperidine-2,6-dione, and their pharmaceutical compositions and uses
CN110461358A (zh) 2017-03-31 2019-11-15 公立大学法人奈良县立医科大学 可用于预防和/或治疗凝血因子ⅸ异常、包含代替凝血因子ⅷ的功能的多特异性抗原结合分子的药物组合物
US20200172868A1 (en) 2017-07-19 2020-06-04 Rubius Therapeutics, Inc. Compositions and methods related to multimodal therapeutic cell systems for infectious disease
US20200172867A1 (en) 2017-07-19 2020-06-04 Rubius Therapeutics, Inc. Compositions and methods related to multimodal therapeutic cell systems for cardiometabolic disease
SG11201810270SA (en) 2017-09-29 2019-04-29 Chugai Pharmaceutical Co Ltd Multispecific antigen-binding molecules having blood coagulation factor viii (fviii) cofactor function-substituting activity and pharmaceutical formulations containing such a molecule as an active ing
BR112020010450A2 (pt) 2017-11-28 2020-11-24 Chugai Seiyaku Kabushiki Kaisha polipeptídeo que inclui domínio de ligação a antígeno e seção transportadora
JP7266532B2 (ja) 2017-11-28 2023-04-28 中外製薬株式会社 リガンド結合活性が調整可能なリガンド結合分子
US11466077B2 (en) 2018-01-05 2022-10-11 Ac Immune Sa Misfolded TDP-43 binding molecules
JP7350756B2 (ja) 2018-02-14 2023-09-26 アバ セラピューティクス アーゲー 抗ヒトpd-l2抗体
US20210261669A1 (en) 2018-06-20 2021-08-26 Chugai Seiyaku Kabushiki Kaisha Method for activating immune response of target cell and composition therefor
WO2020032230A1 (ja) 2018-08-10 2020-02-13 中外製薬株式会社 抗cd137抗原結合分子およびその使用
EP3943108A4 (de) 2019-03-19 2023-01-04 Chugai Seiyaku Kabushiki Kaisha Antigen-bindendes molekül mit antigen-bindender domäne mit veränderlicher bindungsaktivität gegenüber antigen in abhängigkeit von mta und bibliothek zur gewinnung der antigen-bindenden domäne
CN113661238B (zh) 2019-04-02 2024-08-27 中外制药株式会社 引入靶特异性外源基因的方法
JP2022529344A (ja) 2019-04-18 2022-06-21 エイシー イミューン ソシエテ アノニム 治療及び診断のための新規分子
SG11202112453TA (en) 2019-05-23 2021-12-30 Ac Immune Sa Anti-tdp-43 binding molecules and uses thereof
EP3981428A4 (de) 2019-06-05 2023-07-12 Chugai Seiyaku Kabushiki Kaisha Proteasesubstrat und polypeptid mit proteasespaltungssequenz
BR112021023735A2 (pt) 2019-06-05 2022-01-04 Chugai Pharmaceutical Co Ltd Molécula de ligação de sítio de clivagem de anti-corpo
EP3982369A4 (de) 2019-06-07 2023-06-14 Chugai Seiyaku Kabushiki Kaisha Informationsverarbeitungssystem, informationsverarbeitungsverfahren, programm und verfahren zur herstellung eines antigen-bindenden moleküls oder proteins
CN114072177A (zh) 2019-06-18 2022-02-18 马克思—普朗克科学促进协会公司 标记物和/或载体与靶分子例如带His-标签的蛋白质通过金属配合物试剂的位点特异性动力学惰性缀合
US11781138B2 (en) 2019-10-14 2023-10-10 Aro Biotherapeutics Company FN3 domain-siRNA conjugates and uses thereof
WO2021076546A1 (en) 2019-10-14 2021-04-22 Aro Biotherapeutics Company Cd71 binding fibronectin type iii domains
JP6845973B1 (ja) 2019-11-20 2021-03-24 中外製薬株式会社 抗体含有製剤
AU2020385631A1 (en) 2019-11-20 2022-06-02 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. Archaeal peptide recombinase – a novel peptide ligating enzyme
CN115666594A (zh) 2019-11-29 2023-01-31 苏州诺沃泰医药科技有限公司 Cart细胞在制备治疗癌症的药物中的应用
EP3845553A1 (de) 2019-12-30 2021-07-07 Johann Wolfgang Goethe-Universität Trägermatrix mit dodecin-protein
TW202144395A (zh) 2020-02-12 2021-12-01 日商中外製藥股份有限公司 用於癌症之治療的抗cd137抗原結合分子
MX2023001120A (es) 2020-07-31 2023-02-22 Chugai Pharmaceutical Co Ltd Composicion farmaceutica que comprende una celula que expresa un receptor quimerico.
US20240002430A1 (en) 2020-08-07 2024-01-04 Hoffman-La Roche Inc. Method for producing protein compositions
TW202221026A (zh) 2020-08-14 2022-06-01 瑞士商Ac 免疫有限公司 人源化抗tdp-43結合分子及其用途
EP4204448A2 (de) 2020-08-27 2023-07-05 cureab GmbH Anti-golph2-antikörper zur makrophagen- und dendritischen zelldifferenzierung
CN116096411A (zh) 2020-09-01 2023-05-09 中外制药株式会社 含有作为活性成分的il-31拮抗剂的用于预防和/或治疗透析瘙痒的药物组合物
WO2022079297A1 (en) 2020-10-16 2022-04-21 Ac Immune Sa Antibodies binding to alpha-synuclein for therapy and diagnosis
CA3214552A1 (en) 2021-04-14 2022-10-20 Russell C. Addis Cd71 binding fibronectin type iii domains
WO2022244838A1 (ja) 2021-05-19 2022-11-24 中外製薬株式会社 分子のin vivo薬物動態を予測する方法
MX2024005876A (es) 2021-11-16 2024-05-29 Ac Immune Sa Moleculas novedosas para terapias y diagnostico.
AU2023221539A1 (en) 2022-02-16 2024-08-22 Ac Immune Sa Humanized anti-tdp-43 binding molecules and uses thereof
WO2023194565A1 (en) 2022-04-08 2023-10-12 Ac Immune Sa Anti-tdp-43 binding molecules
WO2024064646A1 (en) 2022-09-20 2024-03-28 Celgene Corporation Salts and solid forms of (s)- or racemic 3-(4-((4-(morpholinomethyl)benzyl)oxy)-1-oxoisoindolin-2-yl)piperidine-2,6-dione and methods of using the same
WO2024184494A1 (en) 2023-03-08 2024-09-12 Ac Immune Sa Anti-tdp-43 binding molecules and uses thereof

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000034784A1 (en) * 1998-12-10 2000-06-15 Phylos, Inc. Protein scaffolds for antibody mimics and other binding proteins
WO2001064942A1 (en) * 2000-02-29 2001-09-07 Phylos, Inc. Protein scaffolds for antibody mimics and other binding proteins

Family Cites Families (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US700598A (en) 1901-03-21 1902-05-20 Le Carbone Sa Primary battery.
US5773574A (en) * 1990-12-03 1998-06-30 The Scripps Research Institute Polypeptides for promoting cell attachment
CN1238366C (zh) 1997-01-21 2006-01-25 综合医院公司 利用rna-蛋白融合体筛选蛋白
CA2293632C (en) * 1997-06-12 2011-11-29 Research Corporation Technologies, Inc. Artificial antibody polypeptides
JP2002510505A (ja) 1998-04-03 2002-04-09 フィロス インク. 位置特定可能な蛋白質アレイ
AU775997B2 (en) 1998-12-02 2004-08-19 Bristol-Myers Squibb Company DNA-protein fusions and uses thereof
CA2418835A1 (en) * 2000-10-16 2002-04-25 Phylos, Inc. Protein scaffolds for antibody mimics and other binding proteins
US8068698B2 (en) 2007-05-29 2011-11-29 Research In Motion Limited System and method for resizing images prior to upload
US11054998B1 (en) 2019-12-12 2021-07-06 Facebook, Inc. High bandwidth memory system with distributed request broadcasting masters

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000034784A1 (en) * 1998-12-10 2000-06-15 Phylos, Inc. Protein scaffolds for antibody mimics and other binding proteins
WO2001064942A1 (en) * 2000-02-29 2001-09-07 Phylos, Inc. Protein scaffolds for antibody mimics and other binding proteins

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
KOIDE A ET AL: "THE FIBRONECTIN TYPE III DOMAIN AS A SCAFFOLD FOR NOVEL BINDING PROTEINS" JOURNAL OF MOLECULAR BIOLOGY, LONDON, GB, vol. 284, 1998, pages 1141-1151, XP002944100 ISSN: 0022-2836 *
NYGREN PER-AKE ET AL: "Scaffolds for engineering novel binding sites in proteins" CURRENT OPINION IN STRUCTURAL BIOLOGY, CURRENT BIOLOGY LTD., LONDON, GB, vol. 7, no. 4, 1997, pages 463-469, XP002196760 ISSN: 0959-440X *
See also references of WO0232925A2 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9139825B2 (en) 2009-10-30 2015-09-22 Novartis Ag Universal fibronectin type III bottom-side binding domain libraries
US10253313B2 (en) 2009-10-30 2019-04-09 Novartis Ag Universal fibronectin type III bottom-side binding domain libraries

Also Published As

Publication number Publication date
WO2002032925A3 (en) 2003-08-21
JP5460097B2 (ja) 2014-04-02
JP2013162789A (ja) 2013-08-22
AU2002213251B2 (en) 2007-06-14
EP2141243A3 (de) 2010-01-27
JP2004526419A (ja) 2004-09-02
EP2141243A2 (de) 2010-01-06
EP1356075A4 (de) 2005-04-13
JP2009183292A (ja) 2009-08-20
JP2015051023A (ja) 2015-03-19
WO2002032925A9 (en) 2003-02-06
WO2002032925A2 (en) 2002-04-25
CA2418835A1 (en) 2002-04-25
AU1325102A (en) 2002-04-29
JP5868346B2 (ja) 2016-02-24

Similar Documents

Publication Publication Date Title
US20170275342A1 (en) Protein scaffolds for antibody mimics and other binding proteins
AU2002213251B2 (en) Protein scaffolds for antibody mimics and other binding proteins
US6818418B1 (en) Protein scaffolds for antibody mimics and other binding proteins
AU2002213251A1 (en) Protein scaffolds for antibody mimics and other binding proteins
AU2001241850A1 (en) Protein scaffolds for antibody mimics and other binding proteins
EP1137941B1 (de) Proteingerüste für antikörper-nachahmer und andere bindende proteine

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

AK Designated contracting states

Kind code of ref document: A2

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

AX Request for extension of the european patent

Extension state: AL LT LV MK RO SI

17P Request for examination filed

Effective date: 20040223

RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: COMPOUND THERAPEUTICS, INC.

A4 Supplementary search report drawn up and despatched

Effective date: 20050225

RIC1 Information provided on ipc code assigned before grant

Ipc: 7C 07K 16/24 B

Ipc: 7C 07K 16/00 B

Ipc: 7C 07K 14/525 B

Ipc: 7C 07K 14/47 B

Ipc: 7C 12P 21/06 A

17Q First examination report despatched

Effective date: 20050617

RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: ADNEXUS THERAPEUTICS, INC.

RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: BRISTOL-MYERS SQUIBB COMPANY

RIN1 Information on inventor provided before grant (corrected)

Inventor name: KUIMELIS, ROBERT, G.

Inventor name: WAGNER, RICHARD, W.

Inventor name: LIPOVSEK, DASA

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

Free format text: STATUS: THE APPLICATION HAS BEEN WITHDRAWN

18W Application withdrawn

Effective date: 20090911