US20060008844A1 - c-Met kinase binding proteins - Google Patents

c-Met kinase binding proteins Download PDF

Info

Publication number
US20060008844A1
US20060008844A1 US10/957,351 US95735104A US2006008844A1 US 20060008844 A1 US20060008844 A1 US 20060008844A1 US 95735104 A US95735104 A US 95735104A US 2006008844 A1 US2006008844 A1 US 2006008844A1
Authority
US
United States
Prior art keywords
monomer
domain
domains
polypeptide
multimers
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US10/957,351
Other languages
English (en)
Inventor
Willem Stemmer
D. Victor Perlroth
Sanjeev Satyal
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.)
Amgen Mountain View Inc
Original Assignee
Avidia 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
Priority claimed from US10/871,602 external-priority patent/US20050089932A1/en
Application filed by Avidia Inc filed Critical Avidia Inc
Priority to US10/957,351 priority Critical patent/US20060008844A1/en
Assigned to ALLOY PARTNERS 2002, L.P., VAN VLASSELAER, DR PETER, WOODY, DR JAMES N, ALLOY VENTURES 2002, L.P., MAXYGEN, INC., STEMMER, DR WILLLEM P.C. reassignment ALLOY PARTNERS 2002, L.P. SECURITY AGREEMENT Assignors: AVIDIA RESEARCH INSTITUTE
Assigned to AVIDIA RESEARCH INSTITUTE reassignment AVIDIA RESEARCH INSTITUTE ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: SATYAL, SANJEEV, PERLROTH, D. VICTOR, STEMMER, WILLEM P.C.
Assigned to AVIDIA RESEARCH INSTITUTE reassignment AVIDIA RESEARCH INSTITUTE TERMINATION OF PATENT SECURITY INTEREST Assignors: ALLOY PARTNERS 2002, L.P., ALLOY VENTURES 2002, L.P., MAXYGEN, INC., STEMMER, DR. WILLEM P.C., VAN VLASSELAER, DR. PETER, WOODY, DR. JAMES N.
Priority to JP2007516800A priority patent/JP2008503216A/ja
Priority to PCT/US2005/021558 priority patent/WO2006009888A2/en
Priority to EP05787547A priority patent/EP1776134A4/de
Priority to EP09176476A priority patent/EP2151453A1/de
Priority to CA2569191A priority patent/CA2569191C/en
Priority to AU2005265150A priority patent/AU2005265150B2/en
Priority to KR1020077001057A priority patent/KR101238074B1/ko
Priority to MXPA06014796A priority patent/MXPA06014796A/es
Priority to US11/155,989 priority patent/US7803907B2/en
Publication of US20060008844A1 publication Critical patent/US20060008844A1/en
Assigned to AVIDIA, INC. reassignment AVIDIA, INC. CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: AVIDIA RESEARCH INSTITUTE
Assigned to AMGEN MOUNTAIN VIEW, INC. reassignment AMGEN MOUNTAIN VIEW, INC. CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: AVIDIA, INC.
Priority to US12/761,346 priority patent/US7968681B2/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/04Linear peptides containing only normal peptide links
    • C07K7/08Linear peptides containing only normal peptide links having 12 to 20 amino acids
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/543Immunoassay; Biospecific binding assay; Materials therefor with an insoluble carrier for immobilising immunochemicals
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/566Immunoassay; Biospecific binding assay; Materials therefor using specific carrier or receptor proteins as ligand binding reagents where possible specific carrier or receptor proteins are classified with their target compounds
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/475Assays involving growth factors
    • G01N2333/4753Hepatocyte growth factor; Scatter factor; Tumor cytotoxic factor II
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/82Translation products from oncogenes
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value

Definitions

  • Hepatocyte Growth Factor/Scatter Factor is a mesenchyme-derived pleiotropic factor, which regulates cell growth, cell motility, and morphogenesis of various types of cells and mediates epithelial-mesenchymal interactions responsible for morphogenic tissue interactions during embryonic development and organogenesis.
  • HGF was originally identified as a potent mitogen for hepatocytes, it has also been identified as an angiogenic growth factor.
  • Met was first identified in the 1980s as an oncogene and is the receptor for HGF.
  • the proto-oncogene c-Met was found to encode a receptor tyrosine kinase.
  • phosphorylation of receptor In response to HGF treatment a range of activities are observed: phosphorylation of receptor, docking of signaling intermediates Gab-1/Grb2, culminating in activation of kinases such as P13K, ERK1 and 2, and AKT. These activities aid in cell growth, survival, migration, and neovascularisation.
  • HGF/SF Hepatocyte Growth Factor/Scatter Factor
  • mice and human cell lines that ectopically overexpress HGF and/or Met become tumorigenic and metastatic in athymic nude mice.
  • downregulation of Met or HGF expression in human tumour cells decreases their tumorigenic potential.
  • Mouse models that express the receptor or ligand as a transgene develop various types of tumour and metastatic tumors.
  • HGF and/or Met are frequently expressed in carcinomas, in other types of human solid tumours and in their metastases, and that HGF and/or Met over- or misexpression often correlates with poor prognosis.
  • unequivocal evidence that implicates Met in human cancer is provided by the activating mutations that have been discovered in both sporadic and inherited forms of human renal papillary carcinomas.
  • the present invention provides a polypeptide comprising a monomer domain that binds to c-Met.
  • the monomer domain :
  • binding of at least one monomer domain to c-Met inhibits dimerization of Met. In some embodiments, at least one monomer domain binds to the Sema domain of c-Met, thereby preventing binding of Met ligands to c-Met.
  • the polypeptide comprises at least one and no more than six monomer domains. In some embodiments, the polypeptide comprises at least two monomer domains and the monomer domains are linked by a linker. In some embodiments, the linker is a peptide linker. In some embodiments, the linker is between 4 to 12 amino acids long.
  • the monomer domains are each between 35 to 45 amino acids.
  • the polypeptide comprises at least one monomer domain with binding specificity for a blood factor, thereby increasing the serum half-life of the polypeptide compared to a polypeptide lacking the blood factor monomer domain.
  • the blood factor is serum albumin, an immunoglobulin or an erythrocyte.
  • each monomer domain comprises two disulfide bonds. In some embodiments, each monomer domain comprises three disulfide bonds.
  • the ion is a metal ion. In some embodiments, the ion is a calcium ion.
  • At least one of the monomer domains is derived from a LDL-receptor class A domain. In some embodiments, at least one of the monomer domains is derived from an EGF-like domain.
  • the monomer comprises an amino acid sequence in which at least 10% of the amino acids in the sequence are cysteine; and/or at least 25% of the amino acids are non-naturally-occurring amino acids.
  • the present invention also provides methods for identifying a polypeptide that binds to c-Met.
  • the method comprises,
  • the selecting step comprises selecting a polypeptide that reduces HGF-mediated cell proliferation and/or migration. In some embodiments, the method further comprises selecting a polypeptide that inhibits tumor growth in an animal.
  • the monomer domain comprises an amino acid sequence in which at least 10% of the amino acids in the sequence are cysteine; and/or at least 25% of the amino acids are non-naturally-occurring amino acids.
  • the method further comprises
  • the method further comprises
  • the method further comprises a step of mutating at least one monomer domain, thereby providing a library comprising mutated monomer domains.
  • the library of monomer domains is expressed as a phage display, ribosome display or cell surface display.
  • the polypeptide comprises at least two monomer domains and the monomer domains are linked by a linker.
  • the linker is a peptide linker. In some embodiments, the linker is between 4 to 12 amino acids long.
  • the monomer domains are each between 35 to 45 amino acids.
  • each monomer domain comprises two disulfide bonds. In some embodiments, each monomer domain comprises three disulfide bonds.
  • the ion is a metal ion. In some embodiments, the ion is a calcium ion.
  • At least one of the monomer domains is derived from a LDL-receptor class A domain. In some embodiments, at least one of the monomer domains is derived from an EGF-like domain.
  • the present invention also provides polynucleotides encoding a polypeptide comprising a monomer domain that binds to c-Met, wherein the monomer domain:
  • HGF/SF Hepatocyte Growth Factor/Scatter Factor
  • phosphorylation of receptor In response to HGF treatment a range of activities are observed: phosphorylation of receptor, docking of signaling intermediates Gab-1/Grb2, culminating in activation of kinases such as P13K, ERK1 and 2, and AKT.
  • These activities aid in cell growth, survival, migration, and neovascularisation. See, e.g., Birchmeier et al., Mol. Cell Biol. 4:915-925 (2003).
  • the amino acid sequence of Met is known and is displayed in SEQ ID NO:1. See, e.g., Park et al., Proc. Natl. Acad. Sci. USA 84(18):6379 (1987).
  • monomer domain or “monomer” is used interchangeably herein refer to a discrete region found in a protein or polypeptide.
  • a monomer domain forms a native three-dimensional structure in solution in the absence of flanking native amino acid sequences.
  • Monomer domains of the invention will often bind to a target molecule.
  • a polypeptide that forms a three-dimensional structure that binds to a target molecule is a monomer domain.
  • the term “monomer domain” does not encompass the complementarity determining region (CDR) of an antibody.
  • loop refers to that portion of a monomer domain that is typically exposed to the environment by the assembly of the scaffold structure of the monomer domain protein, and which is involved in target binding.
  • the present invention provides three types of loops that are identified by specific features, such as, potential for disulfide bonding, bridging between secondary protein structures, and molecular dynamics (i.e., flexibility).
  • the three types of loop sequences are a cysteine-defined loop sequence, a structure-defined loop sequence, and a B-factor-defined loop sequence.
  • cyste-defined loop sequence refers to a subsequence of a naturally occurring monomer domain-encoding sequence that is bound at each end by a cysteine residue that is conserved with respect to at least one other naturally occurring monomer domain of the same family. Cysteine-defined loop sequences are identified by multiple sequence alignment of the naturally occurring monomer domains, followed by sequence analysis to identify conserved cysteine residues. The sequence between each consecutive pair of conserved cysteine residues is a cysteine-defined loop sequence. The cysteine-defined loop sequence does not include the cysteine residues adjacent to each terminus.
  • Monomer domains having cysteine-defined loop sequences include the LDL receptor A-domains, EGF-like domains, sushi domains, Fibronectin type 1 domains, and the like.
  • LDL receptor A-domains represented by the consensus sequence, CX 6 CX 4 CX 6 CX 5 CX 8 C (see also FIG. 9 ), wherein X 6 , X 4 , X 5 , and X 8 each represent a cysteine-defined loop sequence comprising the designated number of amino acids.
  • structure-defined loop sequence refers to a subsequence of a monomer-domain encoding sequence that is bound at each end to subsequences that each form a secondary structure.
  • Secondary structures for proteins with known three dimensional structures are identified in accordance with the algorithm STRIDE for assigning protein secondary structure as described in Frishman, D. and Argos, P. (1995) “Knowledge-based secondary structure assignment,” Proteins, 23(4):566-79 (see also //hgmp.mrc.ac.uk/Registered/Option/stride.html at the World Wide Web).
  • Secondary structures for proteins with unknown or uncharacterized three dimensional structures are identified in accordance with the algorithm described in Jones, D. T.
  • Secondary structures include, for example, pleated sheets, helices, and the like. Examples of monomer domains having structure-defined loop sequences are the C2 domains, Ig domains, Factor 5/8 C domains, Fibronectin type 3 domains, and the like.
  • B-factor-defined loop sequence refers to a subsequence of at least three amino acid residues of a monomer-domain encoding sequence in which the B-factors for the alpha carbons in the B-factor-defined loop are among the 25% highest alpha carbon B factors in the entire monomer domain. Typically the average alpha-carbon B-factor for the subsequence is at least about 65.
  • B-factor (or “temperature factor” or “Debye-Waller factor”) is derived from X-ray scattering data.
  • multimer is used herein to indicate a polypeptide comprising at least two monomer domains.
  • the separate monomer domains in a multimer can be joined together by a linker.
  • a multimer is also known as a combinatorial mosaic protein or a recombinant mosaic protein.
  • family and “family class” are used interchangeably to indicate proteins that are grouped together based on similarities in their amino acid sequences. These similar sequences are generally conserved because they are important for the function of the protein and/or the maintenance of the three dimensional structure of the protein. Examples of such families include the LDL Receptor A-domain family, the EGF-like family, and the like.
  • ligand also referred to herein as a “target molecule,” encompasses a wide variety of substances and molecules, which range from simple molecules to complex targets.
  • Target molecules can be proteins, nucleic acids, lipids, carbohydrates or any other molecule capable of recognition by a polypeptide domain.
  • a target molecule can include a chemical compound (i.e., non-biological compound such as, e.g., an organic molecule, an inorganic molecule, or a molecule having both organic and inorganic atoms, but excluding polynucleotides and proteins), a mixture of chemical compounds, an array of spatially localized compounds, a biological macromolecule, a bacteriophage peptide display library, a polysome peptide display library, an extract made from a biological materials such as bacteria, plants, fungi, or animal (e.g., mammalian) cells or tissue, a protein, a toxin, a peptide hormone, a cell, a virus, or the like.
  • a chemical compound i.e., non-biological compound such as, e.g., an organic molecule, an inorganic molecule, or a molecule having both organic and inorganic atoms, but excluding polynucleotides and proteins
  • Target molecules include, e.g., a whole cell, a whole tissue, a mixture of related or unrelated proteins, a mixture of viruses or bacterial strains or the like.
  • Target molecules can also be defined by inclusion in screening assays described herein or by enhancing or inhibiting a specific protein interaction (i.e., an agent that selectively inhibits a binding interaction between two predetermined polypeptides).
  • linker is used herein to indicate a moiety or group of moieties that joins or connects two or more discrete separate monomer domains.
  • the linker allows the discrete separate monomer domains to remain separate when joined together in a multimer.
  • the linker moiety is typically a substantially linear moiety.
  • Suitable linkers include polypeptides, polynucleic acids, peptide nucleic acids and the like. Suitable linkers also include optionally substituted alkylene moieties that have one or more oxygen atoms incorporated in the carbon backbone.
  • the molecular weight of the linker is less than about 2000 daltons. More typically, the molecular weight of the linker is less than about 1500 daltons and usually is less than about 1000 daltons.
  • the linker can be small enough to allow the discrete separate monomer domains to cooperate, e.g., where each of the discrete separate monomer domains in a multimer binds to the same target molecule via separate binding sites.
  • exemplary linkers include a polynucleotide encoding a polypeptide, or a polypeptide of amino acids or other non-naturally occurring moieties.
  • the linker can be a portion of a native sequence, a variant thereof, or a synthetic sequence. Linkers can comprise, e.g., naturally occurring, non-naturally occurring amino acids, or a combination of both.
  • a monomer domain is a separate domain in a protein because it has an independent property that can be recognized and separated from the protein. For instance, the ligand binding ability of the A-domain in the LDLR is an independent property.
  • Other examples of separate include the separate monomer domains in a multimer that remain separate independent domains even when complexed or joined together in the multimer by a linker.
  • Another example of a separate property is the separate binding sites in a multimer for a ligand.
  • directed evolution refers to a process by which polynucleotide variants are generated, expressed, and screened for an activity (e.g., a polypeptide with binding activity) in a recursive process. One or more candidates in the screen are selected and the process is then repeated using polynucleotides that encode the selected candidates to generate new variants.
  • Directed evolution involves at least two rounds of variation generation and can include 3, 4, 5, 10, 20 or more rounds of variation generation and selection. Variation can be generated by any method known to those of skill in the art, including, e.g., by error-prone PCR, gene recombination, chemical mutagenesis and the like.
  • shuffling is used herein to indicate recombination between non-identical sequences.
  • shuffling can include crossover via homologous recombination or via non-homologous recombination, such as via cre/lox and/or flp/frt systems.
  • Shuffling can be carried out by employing a variety of different formats, including for example, in vitro and in vivo shuffling formats, in silico shuffling formats, shuffling formats that utilize either double-stranded or single-stranded templates, primer based shuffling formats, nucleic acid fragmentation-based shuffling formats, and oligonucleotide-mediated shuffling formats, all of which are based on recombination events between non-identical sequences and are described in more detail or referenced herein below, as well as other similar recombination-based formats.
  • random refers to a polynucleotide sequence or an amino acid sequence composed of two or more amino acids and constructed by a stochastic or random process.
  • the random polynucleotide sequence or amino acid sequence can include framework or scaffolding motifs, which can comprise invariant sequences.
  • pseudorandom refers to a set of sequences, polynucleotide or polypeptide, that have limited variability, so that the degree of residue variability at some positions is limited, but any pseudorandom position is allowed at least some degree of residue variation.
  • polypeptide As used herein interchangeably to refer to an amino acid sequence of two or more amino acids.
  • amino acid refers to naturally occurring and synthetic amino acids, as well as amino acid analogs and amino acid mimetics that function in a manner similar to the naturally occurring amino acids.
  • Naturally occurring amino acids are those encoded by the genetic code, as well as those amino acids that are later modified, e.g., hydroxyproline, ⁇ -carboxyglutamate, and O-phosphoserine.
  • Amino acid analogs refers to compounds that have the same basic chemical structure as a naturally occurring amino acid, i.e., an ⁇ carbon that is bound to a hydrogen, a carboxyl group, an amino group, and an R group, e.g., homoserine, norleucine, methionine sulfoxide, methionine methyl sulfonium. Such analogs have modified R groups (e.g., norleucine) or modified peptide backbones, but retain the same basic chemical structure as a naturally occurring amino acid.
  • Amino acid mimetics refers to chemical compounds that have a structure that is different from the general chemical structure of an amino acid, but that functions in a manner similar to a naturally occurring amino acid.
  • Constant amino acid substitution refers to the interchangeability of residues having similar side chains.
  • a group of amino acids having aliphatic side chains is glycine, alanine, valine, leucine, and isoleucine
  • a group of amino acids having aliphatic-hydroxyl side chains is serine and threonine
  • a group of amino acids having amide-containing side chains is asparagine and glutamine
  • a group of amino acids having aromatic side chains is phenylalanine, tyrosine, and tryptophan
  • a group of amino acids having basic side chains is lysine, arginine, and histidine
  • a group of amino acids having sulfur-containing side chains is cysteine and methionine.
  • Preferred conservative amino acids substitution groups are: valine-leucine-isoleucine, phenylalanine-tyrosine, lysine-arginine, alanine-valine, and asparagine-glutamine.
  • nucleic acid sequence refers to a single or double-stranded polymer of deoxyribonucleotide or ribonucleotide bases read from the 5′ to the 3′ end. It includes chromosomal DNA, self-replicating plasmids and DNA or RNA that performs a primarily structural role.
  • encoding refers to a polynucleotide sequence encoding one or more amino acids. The term does not require a start or stop codon. An amino acid sequence can be encoded in any one of six different reading frames provided by a polynucleotide sequence.
  • promoter refers to regions or sequence located upstream and/or downstream from the start of transcription that are involved in recognition and binding of RNA polymerase and other proteins to initiate transcription.
  • a “vector” refers to a polynucleotide, which when independent of the host chromosome, is capable of replication in a host organism.
  • vectors include plasmids.
  • Vectors typically have an origin of replication.
  • Vectors can comprise, e.g., transcription and translation terminators, transcription and translation initiation sequences, and promoters useful for regulation of the expression of the particular nucleic acid.
  • recombinant when used with reference, e.g., to a cell, or nucleic acid, protein, or vector, indicates that the cell, nucleic acid, protein or vector, has been modified by the introduction of a heterologous nucleic acid or protein or the alteration of a native nucleic acid or protein, or that the cell is derived from a cell so modified.
  • recombinant cells express genes that are not found within the native (nonrecombinant) form of the cell or express native genes that are otherwise abnormally expressed, under-expressed or not expressed at all.
  • the phrase “specifically (or selectively) binds” to a polypeptide when referring to a monomer or multimer, refers to a binding reaction that can be determinative of the presence of the polypeptide in a heterogeneous population of proteins (e.g., a cell or tissue lysate) and other biologics.
  • the specified monomer or multimer binds to a particular target molecule above background (e.g., 2 ⁇ , 5 ⁇ , 10 ⁇ or more above background) and does not bind in a significant amount to other molecules present in the sample.
  • nucleic acids or polypeptide sequences refer to two or more sequences or subsequences that are the same. “Substantially identical” refers to two or more nucleic acids or polypeptide sequences having a specified percentage of amino acid residues or nucleotides that are the same (i.e., 60% identity, optionally 65%, 70%, 75%, 80%, 85%, 90%, or 95% identity over a specified region, or, when not specified, over the entire sequence), when compared and aligned for maximum correspondence over a comparison window, or designated region as measured using one of the following sequence comparison algorithms or by manual alignment and visual inspection.
  • identity or substantial identity exists over a region that is at least about 50 nucleotides in length, or more preferably over a region that is 100 to 500 or 1000 or more nucleotides or amino acids in length.
  • a polynucleotide or amino acid sequence is “heterologous to” a second sequence if the two sequences are not linked in the same manner as found in naturally-occurring sequences.
  • a promoter operably linked to a heterologous coding sequence refers to a coding sequence which is different from any naturally-occurring allelic variants.
  • heterologous linker when used in reference to a multimer, indicates that the multimer comprises a linker and a monomer that are not found in the same relationship to each other in nature (e.g., they form a non-naturally occurring fusion protein).
  • non-naturally-occurring amino acid in a protein sequence refers to any amino acid other than the amino acid that occurs in the corresponding position in an alignment with a naturally-occurring polypeptide with the lowest smallest sum probability where the comparison window is the length of the monomer domain queried and when compared to the non-redundant (“nr”) database of Genbank using BLAST 2.0 as described herein.
  • Percentage of sequence identity is determined by comparing two optimally aligned sequences over a comparison window, wherein the portion of the polynucleotide sequence in the comparison window may comprise additions or deletions (i.e., gaps) as compared to the reference sequence (which does not comprise additions or deletions) for optimal alignment of the two sequences. The percentage is calculated by determining the number of positions at which the identical nucleic acid base or amino acid residue occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the window of comparison and multiplying the result by 100 to yield the percentage of sequence identity.
  • nucleic acids or polypeptide sequences refer to two or more sequences or subsequences that are the same or have a specified percentage of amino acid residues or nucleotides that are the same, when compared and aligned for maximum correspondence over a comparison window, or designated region as measured using one of the following sequence comparison algorithms or by manual alignment and visual inspection. Such sequences are then said to be “substantially identical.” This definition also refers to the complement of a test sequence.
  • identity exists over a region that is at least about 50 amino acids or nucleotides in length, or more preferably over a region that is 75-100 amino acids or nucleotides in length.
  • sequence comparison typically one sequence acts as a reference sequence, to which test sequences are compared.
  • test and reference sequences are entered into a computer, subsequence coordinates are designated, if necessary, and sequence algorithm program parameters are designated. Default program parameters can be used, or alternative parameters can be designated.
  • sequence comparison algorithm then calculates the percent sequence identities for the test sequences relative to the reference sequence, based on the program parameters.
  • a “comparison window”, as used herein, includes reference to a segment of any one of the number of contiguous positions selected from the group consisting of from 20 to 600, usually about 50 to about 200, more usually about 100 to about 150 in which a sequence may be compared to a reference sequence of the same number of contiguous positions after the two sequences are optimally aligned.
  • Methods of alignment of sequences for comparison are well-known in the art.
  • Optimal alignment of sequences for comparison can be conducted, e.g., by the local homology algorithm of Smith and Waterman (1970) Adv. Appl. Math. 2:482c, by the homology alignment algorithm of Needleman and Wunsch (1970) J. Mol. Biol.
  • HSPs high scoring sequence pairs
  • the word hits are extended in both directions along each sequence for as far as the cumulative alignment score can be increased. Cumulative scores are calculated using, for nucleotide sequences, the parameters M (reward score for a pair of matching residues; always >0) and N (penalty score for mismatching residues; always ⁇ 0). For amino acid sequences, a scoring matrix is used to calculate the cumulative score. Extension of the word hits in each direction are halted when the cumulative alignment score falls off by the quantity X from its maximum achieved value; the cumulative score goes to zero or below, due to the accumulation of one or more negative-scoring residue alignments; or the end of either sequence is reached.
  • the BLAST algorithm parameters W, T, and X determine the sensitivity and speed of the alignment.
  • W wordlength
  • E expectation
  • the BLAST algorithm also performs a statistical analysis of the similarity between two sequences (see, e.g., Karlin and Altschul (1993) Proc. Natl. Acad. Sci. USA 90:5873-5787).
  • One measure of similarity provided by the BLAST algorithm is the smallest sum probability (P(N)), which provides an indication of the probability by which a match between two nucleotide or amino acid sequences would occur by chance.
  • P(N) the smallest sum probability
  • a nucleic acid is considered similar to a reference sequence if the smallest sum probability in a comparison of the test nucleic acid to the reference nucleic acid is less than about 0.2, more preferably less than about 0.01, and most preferably less than about 0.001.
  • FIG. 1 schematically illustrates the type, number and order of monomer domains found in members of the LDL-receptor family. These monomer domains include ⁇ -Propeller domains, EGF-like domains and LDL receptor class A-domains.
  • the members shown include low-density lipoprotein receptor (LDLR), ApoE Receptor 2 (ApoER2), very-low-density lipoprotein receptor (VLDLR), LDLR-related protein 2 (LRP2) and LDLR-related protein 1 (LRP1).
  • LDLR low-density lipoprotein receptor
  • ApoER2 ApoE Receptor 2
  • VLDLR very-low-density lipoprotein receptor
  • LRP2 LDLR-related protein 2
  • LRP1 LDLR-related protein 1
  • FIG. 2 schematically illustrates the alignment of partial amino acid sequence from a variety of the LDL-receptor class A-domains (SEQ ID NOS: 103, 100, 65, 117, 128, 21, 29, 39, 30, 77, 58, 50, and 14, respectively in order of appearance) to demonstrate the conserved cysteines.
  • FIG. 3 panel A schematically illustrates an example of an A-domain.
  • Panel A schematically illustrates conserved amino acids in an A-domain of about 40 amino acids long. The conserved cysteine residues are indicated by C, and the negatively charged amino acids are indicated by a circle with a minus (“ ⁇ ”) sign. Circles with an “H” indicate hydrophobic residues.
  • Panel B schematically illustrates two folded A-domains connected via a linker. Panel B also indicates two calcium binding sites, dark circles with Ca +2 , and three disulfide bonds within each folded A-domain for a total of 6 disulfide bonds.
  • FIG. 5 schematically illustrates a general scheme for identifying monomer domains that bind to a ligand, isolating the selected monomer domains, creating multimers of the selected monomer domains by joining the selected monomer domains in various combinations and screening the multimers to identify multimers comprising more than one monomer that binds to a ligand.
  • FIG. 8 depicts common amino acids in each position of the A domain.
  • the percentages above the amino acid positions refer to the percentage of naturally-occurring A domains with the inter-cysteine spacing displayed. Potential amino acid residues in bold depicted under each amino acid position represent common residues at that position.
  • the final six amino acids, depicted as lighter-colored circles, represent linker sequences.
  • FIG. 10 depicts an alignment of A domains (SEQ ID NO: 1-197). At the top and the bottom of the figure, small letters (a-q) indicate conserved residues.
  • FIG. 11 depicts linkage of domains via partial linkers.
  • FIG. 12 is a graphical representation of the regions of sequence identity between the sequences of two different selected clones and known human sequences from a database.
  • the horizontal bars indicate areas of sequence identity between the sequence of the selected clone and the human sequence and the numbers indicate the exact amino acid numbers that define the region of identity.
  • the vertical arrow depicts an acceptable crossover sequence.
  • FIG. 13 illustrates screening a library of monomer domains against multiple ligands displayed on a cell.
  • FIG. 15 illustrates an embodiment for identifying polynucleotides encoding ligands and monomer domains.
  • FIG. 20 illustrates a possible sequence of multimer optimization steps in which optimal monomers and then multimers are selected followed by optimization of monomers, optimization of linkers and then optimization of multimers.
  • FIG. 22 depicts a possible conformation of a multimer of the invention comprising at least one monomer domain that binds to a half-life extending molecule and other monomer domains binding to two other different molecules.
  • two monomer domains bind to a first target molecule and a separate monomer domain binds to a second target molecule.
  • the present invention provides for non-naturally-occurring proteins that bind to c-Met.
  • the proteins of the present invention comprise a domain that binds to c-Met. These domains may be readily identified using a variety of polypeptide scaffolds to generate a plurality of polypeptide variants and then selecting a variant that binds to c-Met.
  • the present invention therefore also provides for selecting a protein that binds to c-Met. Proteins that bind c-Met are useful, e.g., for treating individuals with solid tumors that express c-Met.
  • the polypeptides of the invention are also useful to detect tissues in which Met is expressed and can be used to target molecules to those tissues.
  • c-Met is inactive in its resting monomer state and dimer formation results in receptor activation (often even in absence of ligand binding).
  • the mature form of the receptor consists of a solely extracellular ⁇ chain and a longer ⁇ chain encompassing the remainder of the extracellular domain, a transmembrane domain and a cytoplasmic tail.
  • the cytoplasmic tail contains the juxtamembrane domain, a kinase domain and docking sites for signaling intermediates.
  • the ⁇ chain and the first 212 amino acids of the ⁇ also known as the Sema domain (Kong-Beltran, et al., Cancer Cell 6:75-84 (2004), are sufficient for binding to HGF.
  • the polypeptides of the invention comprise at least one monomer domain that inhibits dimerization of c-Met ⁇ and ⁇ chains and/or functions as an antagonist to prevent ligands of c-Met from binding and/or activating c-Met.
  • Monomer domains can be polypeptide chains of any size.
  • monomer domains have about 25 to about 500, about 30 to about 200, about 30 to about 100, about 35 to about 50, about 35 to about 100, about 90 to about 200, about 30 to about 250, about 30 to about 60, about 9 to about 150, about 100 to about 150, about 25 to about 50, or about 30 to about 150 amino acids.
  • a monomer domain of the present invention can comprise, e.g., from about 30 to about 200 amino acids; from about 25 to about 180 amino acids; from about 40 to about 150 amino acids; from about 50 to about 130 amino acids; or from about 75 to about 125 amino acids.
  • Monomer domains can typically maintain a stable conformation in solution, and are often heat stable, e.g., stable at 95° C. for at least 10 minutes without losing binding affinity. Sometimes, monomer domains can fold independently into a stable conformation.
  • the stable conformation is stabilized by ions (e.g., such as metal or calcium ions).
  • the stable conformation can optionally contain disulfide bonds (e.g., at least one, two, or three or more disulfide bonds). The disulfide bonds can optionally be formed between two cysteine residues.
  • monomer domains, or monomer domain variants are substantially identical to the sequences exemplified (e.g., LDL A domains, EGF domains, etc., or otherwise referenced herein.
  • Monomer domains that are particularly suitable for use in the practice of the present invention are (1) ⁇ sandwich domains; (2) ⁇ -barrel domains; or (3) cysteine-rich domains comprising disulfide bonds.
  • Cysteine-rich domains employed in the practice of the present invention typically do not form an ⁇ helix, a ⁇ sheet, or a ⁇ -barrel structure.
  • the disulfide bonds promote folding of the domain into a three-dimensional structure.
  • cysteine-rich domains have at least two disulfide bonds, more typically at least three disulfide bonds. In some embodiments, at least 5, 10, 15 or 20% of the amino acids in a monomer domain are cysteines.
  • Domains can have any number of characteristics. For example, in some embodiments, the domains have low or no immunogenicity in an animal (e.g., a human). Domains can have a small size. In some embodiments, the domains are small enough to penetrate skin or other tissues. Domains can have a range of in vivo half-lives or stabilities.
  • Illustrative monomer domains suitable for use in the practice of the present invention include, e.g., an EGF-like domain, a Kringle-domain, a fibronectin type I domain, a fibronectin type II domain, a fibronectin type III domain, a PAN domain, a Gla domain, a SRCR domain, a Kunitz/Bovine pancreatic trypsin Inhibitor domain, a Kazal-type serine protease inhibitor domain, a Trefoil (P-type) domain, a von Willebrand factor type C domain, an Anaphylatoxin-like domain, a CUB domain, a thyroglobulin type I repeat, LDL-receptor class A domain, a Sushi domain, a Link domain, a Thrombospondin type I domain, an Immunoglobulin-like domain, a C-type lectin domain, a MAM domain, a von Willebrand factor type A domain, a Somato
  • monomer domains of the present invention include domains other than a fibronectin type III domain, an anticalin domain and a Ig-like domain from CTLA-4.
  • Some aspects of these domains are described in WO 01/64942 entitled “Protein scaffolds for antibody mimics and other binding proteins” by Lipovsek et al., published on Sep. 7, 2001, WO99/16873 entitled “Anticalins” by Beste et al., published Apr. 8, 1999 and WO 00/60070 entitled “A polypeptide structure for use as a scaffold” by Desmet, et al., published on Oct. 12, 2000.
  • monomer domains are optionally cysteine rich.
  • Suitable cysteine rich monomer domains include, e.g., the LDL receptor class A domain (“A-domain”) or the EGF-like domain.
  • the monomer domains can also have a cluster of negatively charged residues.
  • the monomer domains contain a repeated sequence, such as YWTD (SEQ ID NO: 198) as found in the ⁇ -Propeller domain.
  • monomer domains can include the ability to bind ligands (e.g., as in the LDL receptor class A domain, or the CUB domain (complement C1r/C1s, Uegf, and bone morphogenic protein-1 domain)), the ability to participate in endocytosis or internalization (e.g., as in the cytoplasmic tail of the LDL receptor or the cytoplasmic tail of Megalin), the ability to bind an ion (e.g., Ca 2+ binding by the LDL receptor A-domain), and/or the ability to be involved in cell adhesion (e.g., as in the EGF-like domain).
  • ligands e.g., as in the LDL receptor class A domain, or the CUB domain (complement C1r/C1s, Uegf, and bone morphogenic protein-1 domain)
  • endocytosis or internalization e.g., as in the cytoplasmic tail of the LDL receptor or the cytoplasmic tail of Megalin
  • Monomer domains that bind ions to maintain their secondary structure include, e.g., A domain, EGF domain, EF Hand (e.g., such as those found in present in calmodulin and troponin C), Cadherin domain, C-type lectin, C2 domain, Annexin, Gla-domain, Trombospondin type 3 domain, all of which bind calcium, and zinc fingers (e.g., C2H2 type C3HC4 type (RING finger), Integrase Zinc binding domain, PHD finger, GATA zinc finger, FYVE zinc finger, B-box zinc finger), which bind zinc.
  • ion-binding provides stability of secondary structure while providing sufficient flexibility to allow for numerous binding conformations depending on primary sequence.
  • Characteristics of a monomer domain can include the ability to fold independently and the ability to form a stable structure.
  • the structure of the monomer domain is often conserved, although the polynucleotide sequence encoding the monomer need not be conserved.
  • the A-domain structure is conserved among the members of the A-domain family, while the A-domain nucleic acid sequence is not.
  • a monomer domain is classified as an A-domain by its cysteine residues and its affinity for calcium, not necessarily by its nucleic acid sequence. See, FIG. 2 .
  • monomer domains may be selected for the ability to bind to targets other than the target that a homologous naturally occurring domain may bind.
  • the invention provides monomer domains (and multimers comprising such monomers) that do not bind to the target or the class or family of target proteins that a homologous naturally occurring domain may bind.
  • the A-domains (sometimes called “complement-type repeats”) contain about 30-50 or 30-65 amino acids. In some embodiments, the domains comprise about 35-45 amino acids and in some cases about 40 amino acids. Within the 30-50 amino acids, there are about 6 cysteine residues. Of the six cysteines, disulfide bonds typically are found between the following cysteines: C1 and C3, C2 and C5, C4 and C6. The cysteine residues of the domain are disulfide linked to form a compact, stable, functionally independent moiety. See, FIG. 3 . Clusters of these repeats make up a ligand binding domain, and differential clustering can impart specificity with respect to the ligand binding.
  • FIGS. 2, 3 and 8 Exemplary A domain sequences and consensus sequences are depicted in FIGS. 2, 3 and 8 .
  • FIG. 9 displays location and occurrence of residues in A domains with the following spacing between cysteines.
  • FIG. 10 depicts a number of A domains and provides a listing of conserved amino acids.
  • One typical consensus sequence useful to identify A domains is the following: C-[VILMA]-X (5) —C-[DNH]-X (3) -[DENQHT]—C—X (3,4) -[STADE]-[DEH]-[DE]-X (1,5) —C (SEQ ID NO: 200), where the residues in brackets indicate possible residues at one position. “X (#) ” indicates number of residues.
  • a domain variants comprise sequences substantially identical to any of the above-described sequences.
  • Additional exemplary A domains include the following sequence:
  • the monomer domain is an LDL receptor class A domain monomer comprising the following sequence:
  • Exemplary proteins containing A-domains include, e.g., complement components (e.g., C6, C7, C8, C9, and Factor I), serine proteases (e.g., enteropeptidase, matriptase, and corin), transmembrane proteins (e.g., ST7, LRP3, LRP5 and LRP6) and endocytic receptors (e.g., Sortilin-related receptor, LDL-receptor, VLDLR, LRP1, LRP2, and ApoER2).
  • complement components e.g., C6, C7, C8, C9, and Factor I
  • serine proteases e.g., enteropeptidase, matriptase, and corin
  • transmembrane proteins e.g., ST7, LRP3, LRP5 and LRP6
  • endocytic receptors e.g., Sortilin-related receptor, LDL-receptor, VLDLR, LRP1,
  • a domains and A domain variants can be readily employed in the practice of the present invention as monomer domains and variants thereof. Further description of A domains can be found in the following publications and references cited therein: Howell and Hertz, The LDL receptor gene family: signaling functions during development , (2001) Current Opinion in Neurobiology 11:74-81; Herz (2001), supra; Krieger, The “best” of cholesterols, the “worst” of cholesterols: A tale of two receptors , (1998) PNAS 95: 4077-4080; Goldstein and Brown, The Cholesterol quartet , (2001) Science, 292: 1310-1312; and, Moestrup and Verroust, Megalin - and Cubilin - Mediated Endocytosis of Protein - Bound Vitamins, Lipids, and Hormones in Polarized Epithelia , (2001) Ann. Rev. Nutr. 21:407-28.
  • monomer domains can be found in the protein Cubilin, which contains EGF-type repeats and CUB domains.
  • the CUB domains are involved in ligand binding, e.g., some ligands include intrinsic factor (IF)-vitamin B12, receptor associated protein (RAP), Apo A-I, Transferrin, Albumin, Ig light chains and calcium. See, Moestrup and Verroust, supra.
  • Megalin also contains multiple monomer domains. Specifically, megalin possesses LDL-receptor type A-domain, EGF-type repeat, a transmembrane segment and a cytoplasmic tail. Megalin binds a diverse set of ligands, e.g., ApoB, ApoE, ApoJ, clusterin, ApopH/Beta2-glycoprotein-I, PTH, Transthyretin, Thyroglobulin, Insulin, Aminoglycosides, Polymyxin B, Aprotinin, Trichosanthin, PAI-1, PAI-1-urokinase, PAI-1-tPA, Pro-urokinase, Lipoprotein lipase, alpha-Amylase, Albumin, RAP, Ig light chains, calcium, C1q, Lactoferrin, beta2-microglobulin, EGF, Prolactin, Lysozyme, Cytochrome c, PAP-1, Odorant
  • Exemplary EGF monomer domains include the sequence:
  • the monomer domain is an EGF domain monomer comprising the following sequence:
  • Polynucleotides also referred to as nucleic acids
  • Nucleic acids that encode monomer domains can be derived from a variety of different sources.
  • Libraries of monomer domains can be prepared by expressing a plurality of different nucleic acids encoding naturally occurring monomer domains, altered monomer domains (i.e., monomer domain variants), or a combinations thereof.
  • libraries may be designed in which a scaffold of amino acids remain constant (e.g., an LDL A receptor domain, EGF domain) while the intervening amino acids in the scaffold comprise randomly generated amino acids.
  • the invention provides methods of identifying monomer domains that bind to a selected or desired ligand or mixture of ligands.
  • monomer domains are identified or selected for a desired property (e.g., binding affinity) and then the monomer domains are formed into multimers. See, e.g., FIG. 5 .
  • a desired property e.g., binding affinity
  • any method resulting in selection of domains with a desired property e.g., a specific binding property
  • the methods can comprise providing a plurality of different nucleic acids, each nucleic acid encoding a monomer domain; translating the plurality of different nucleic acids, thereby providing a plurality of different monomer domains; screening the plurality of different monomer domains for binding of the desired ligand or a mixture of ligands; and, identifying members of the plurality of different monomer domains that bind the desired ligand or mixture of ligands.
  • monomer domains can be naturally-occurring or altered (non-natural variants).
  • naturally occurring is used herein to indicate that an object can be found in nature.
  • natural monomer domains can include human monomer domains or optionally, domains derived from different species or sources, e.g., mammals, primates, rodents, fish, birds, reptiles, plants, etc.
  • the natural occurring monomer domains can be obtained by a number of methods, e.g., by PCR amplification of genomic DNA or cDNA.
  • Monomer domains of the present invention can be naturally-occurring domains or non-naturally occurring variants.
  • Libraries of monomer domains employed in the practice of the present invention may contain naturally-occurring monomer domain, non-naturally occurring monomer domain variants, or a combination thereof.
  • Monomer domain variants can include ancestral domains, chimeric domains, randomized domains, mutated domains, and the like.
  • ancestral domains can be based on phylogenetic analysis.
  • Chimeric domains are domains in which one or more regions are replaced by corresponding regions from other domains of the same family.
  • chimeric domains can be constructed by combining loop sequences from multiple related domains of the same family to form novel domains with potentially lowered immunogenicity.
  • Those of skill in the art will recognized the immunologic benefit of constructing modified binding domain monomers by combining loop regions from various related domains of the same family rather than creating random amino acid sequences.
  • the present invention provides a method for generating a library of chimeric monomer domains derived from human proteins, the method comprising: providing loop sequences corresponding to at least one loop from each of at least two different naturally occurring variants of a human protein, wherein the loop sequences are polynucleotide or polypeptide sequences; and covalently combining loop sequences to generate a library of at least two different chimeric sequences, wherein each chimeric sequence encodes a chimeric monomer domain having at least two loops.
  • the chimeric domain has at least four loops, and usually at least six loops.
  • the present invention provides three types of loops that are identified by specific features, such as, potential for disulfide bonding, bridging between secondary protein structures, and molecular dynamics (i.e., flexibility).
  • the three types of loop sequences are a cysteine-defined loop sequence, a structure-defined loop sequence, and a B-factor-defined loop sequence.
  • Randomized domains are domains in which one or more regions are randomized.
  • the randomization can be based on full randomization, or optionally, partial randomization based on natural distribution of sequence diversity.
  • compositions of nucleic acids and polypeptides are included in the present invention.
  • the present invention provides a plurality of different nucleic acids wherein each nucleic acid encodes at least one monomer domain or immuno-domain.
  • the present invention also provides recombinant nucleic acids encoding one or more polypeptides comprising a plurality of monomer domains and/or immuno-domains, which monomer domains are altered in order or sequence as compared to a naturally occuring polypeptide.
  • the naturally occuring polypeptide can be selected from the group consisting of: an EGF-like domain, a Kringle-domain, a fibronectin type I domain, a fibronectin type II domain, a fibronectin type III domain, a PAN domain, a Gla domain, a SRCR domain, a Kunitz/Bovine pancreatic trypsin Inhibitor domain, a Kazal-type serine protease inhibitor domain, a Trefoil (P-type) domain, a von Willebrand factor type C domain, an Anaphylatoxin-like domain, a CUB domain, a thyroglobulin type I repeat, LDL-receptor class A domain, a Sushi domain, a Link domain, a Thrombospondin type I domain, an Immunoglobulin-like domain, a C-type lectin domain, a MAM domain, a von Willebrand factor type A domain, a Somatomedin B domain, a
  • Multimers comprise at least two monomer domains.
  • multimers of the invention can comprise from 2 to about 10 monomer domains, from 2 and about 8 monomer domains, from about 3 and about 10 monomer domains, about 7 monomer domains, about 6 monomer domains, about 5 monomer domains, or about 4 monomer domains.
  • the multimer comprises 3 or at least 3 monomer domains.
  • the multimers have no more than 2, 3, 4, 5, 6, 7, or 8 monomer domains.
  • each monomer domain specifically binds to one target molecule (e.g., Met). In some of these embodiments, each monomer binds to a different position (analogous to an epitope) on a target molecule. Multiple monomer domains that bind to the same target molecule results in an avidity effect resulting in improved avidity of the multimer for the target molecule compared to each individual monomer. In some embodiments, the multimer has an avidity of at least about 1.5, 2, 3, 4, 5, 10, 20, 50 or 100 times the avidity of a monomer domain alone. In some embodiments, at least one, two, three, four or more (e.g., all) monomers of a multimer bind an ion such as calcium or another ion.
  • an ion such as calcium or another ion.
  • Multimers can comprise a variety of combinations of monomer domains.
  • the selected monomer domains can be identical or different.
  • the selected monomer domains can comprise various different monomer domains from the same monomer domain family, or various monomer domains from different domain families, or optionally, a combination of both.
  • Multimers that are generated in the practice of the present invention may be any of the following:
  • the multimer comprises monomer domains with specificities for different target molecules.
  • the multimers of the invention comprises 1, 2, 3, or more monomer domains that bind to Met and at least one monomer domain that binds to a second target molecule.
  • target molecules include, e.g., a serum molecule that extends the serum half-life of the multimer, EGFR gene family members, VEGF receptors, PDGF receptor, other receptor tyrosine kinases, integrins, other molecules implicated in tumorigenesis, or markers of tumor tissue.
  • Exemplary molecule that extends the serum half-life of a multimer include, e.g., red blood cells, IgG, and serum albumin such as HSA.
  • Multimers of the present invention may have at least one immuno-domain such as a minibody, a single-domain antibody, a single chain variable fragment (ScFv), or a Fab fragment; and at least one monomer domain, such as, for example, an EGF-like domain, a Kringle-domain, a fibronectin type I domain, a fibronectin type II domain, a fibronectin type III domain, a PAN domain, a Gla domain, a SRCR domain, a Kunitz/Bovine pancreatic trypsin Inhibitor domain, a Kazal-type serine protease inhibitor domain, a Trefoil (P-type) domain, a von Willebrand factor type C domain, an Anaphylatoxin-like domain, a CUB domain,
  • Domains need not be selected before the domains are linked to form multimers.
  • the domains can be selected for the ability to bind to a target molecule before being linked into multimers.
  • a multimer can comprise two domains that bind to one target molecule and a third domain that binds to a second target molecule.
  • the multimers of the present invention may have the following qualities: multivalent, multispecific, single chain, heat stable, extended serum and/or shelf half-life. Moreover, at least one, more than one or all of the monomer domains may bind an ion (e.g., a metal ion or a calcium ion), at least one, more than one or all monomer domains may be derived from LDL receptor A domains and/or EGF-like domains, at least one, more than one or all of the monomer domains may be non-naturally occurring, and/or at least one, more than one or all of the monomer domains may comprise 1, 2, 3, or 4 disulfide bonds per monomer domain.
  • an ion e.g., a metal ion or a calcium ion
  • at least one, more than one or all monomer domains may be derived from LDL receptor A domains and/or EGF-like domains
  • at least one, more than one or all of the monomer domains may be non-natural
  • the multimers comprise at least 4 monomer domains, wherein at least one monomer domain is non-naturally occurring, and wherein:
  • the multimers comprise at least two monomer domains, wherein at least one monomer domain is non-naturally occurring, and wherein each domain is:
  • the multimers comprise at least two monomer domains, wherein at least one monomer domain is non-naturally-occurring and:
  • a significant advantage of the present invention is that known ligands, or unknown ligands can be used to select the monomer domains and/or multimers. No prior information regarding ligand structure is required to isolate the monomer domains of interest or the multimers of interest.
  • the monomer domains and/or multimers identified can have biological activity, which is meant to include at least specific binding affinity for a selected or desired ligand, and, in some instances, will further include the ability to block the binding of other compounds, to stimulate or inhibit metabolic pathways, to act as a signal or messenger, to stimulate or inhibit cellular activity, and the like.
  • Monomer domains can be generated to function as ligands for receptors where the natural ligand for the receptor has not yet been identified (orphan receptors). These orphan ligands can be created to either block or activate the receptor top which they bind.
  • a single ligand can be used, or optionally a variety of ligands can be used to select the monomer domains and/or multimers.
  • a monomer domain of the present invention can bind a single ligand or a variety of ligands.
  • a multimer of the present invention can have multiple discrete binding sites for a single ligand, or optionally, can have multiple binding sites for a variety of ligands.
  • the multimer comprises monomer domains with specificities for different proteins.
  • the different proteins can be related or unrelated. Examples of related proteins including members of a protein family or different serotypes of a virus.
  • the monomer domains of a multimer can target different molecules in a physiological pathway (e.g., different blood coagulation proteins).
  • monomer domains bind to proteins in unrelated pathways (e.g., two domains bind to blood factors, two other domains bind to inflammation-related proteins and a fifth binds to serum albumin).
  • a multimer is comprised of monomer domains that bind to different pathogens or contaminants of interest. Such multimers are useful as a single detection agent capable of detecting for the possibility of any of a number of pathogens or contaminants.
  • the multimers of the invention bind to the same or other multimers to form aggregates.
  • Aggregation can be mediated, for example, by the presence of hydrophobic domains on two monomer domains, resulting in the formation of non-covalent interactions between two monomer domains.
  • aggregation may be facilitated by one or more monomer domains in a multimer having binding specificity for a monomer domain in another multimer.
  • Aggregates can also form due to the presence of affinity peptides on the monomer domains or multimers. Aggregates can contain more target molecule binding domains than a single multimer.
  • Multimers with affinity for both a cell surface target and a second target may provide for increased avidity effects.
  • membrane fluidity can be more flexible than protein linkers in optimizing (by self-assembly) the spacing and valency of the interactions.
  • multimers will bind to two different targets, each on a different cell or one on a cell and another on a molecule with multiple binding sites. See. e.g., FIGS. 16 and 17 .
  • the monomer or multimer domain is linked to a molecule (e.g., a protein, nucleic acid, organic small molecule, etc.) useful as a pharmaceutical.
  • a molecule e.g., a protein, nucleic acid, organic small molecule, etc.
  • exemplary pharmaceutical proteins include, e.g., cytokines, antibodies, chemokines, growth factors, interleukins, cell-surface proteins, extracellular domains, cell surface receptors, cytotoxins, etc.
  • Exemplary small molecule pharmaceuticals include small molecule toxins or therapeutic agents.
  • the monomer or multimers are selected to bind to a tissue- or disease-specific target protein.
  • Tissue-specific proteins are proteins that are expressed exclusively, or at a significantly higher level, in one or several particular tissue(s) compared to other tissues in an animal.
  • monomer domains that bind to Met may be used to target other molecules, including other monomer domains, to the liver. This may be used to target liver-specific diseases, for example, by targeting therapeutic or toxic molecules to the liver.
  • An example of a liver disease that can be treated is hepatocellular carcinoma.
  • disease-specific proteins are proteins that are expressed exclusively, or at a significantly higher level, in one or several diseased cells or tissues compared to other non-diseased cells or tissues in an animal.
  • the monomers or multimers that bind to the target protein are linked to the pharmaceutical protein or small molecule such that the resulting complex or fusion is targeted to the specific tissue or disease-related cell(s) where the target protein (e.g., Met) is expressed.
  • Monomers or multimers for use in such complexes or fusions can be initially selected for binding to the target protein and may be subsequently selected by negative selection against other cells or tissue (e.g., to avoid targeting bone marrow or other tissues that set the lower limit of drug toxicity) where it is desired that binding be reduced or eliminated in other non-target cells or tissues.
  • the therapeutic window is increased so that a higher dose may be administered safely.
  • in vivo panning can be performed in animals by injecting a library of monomers or multimers into an animal and then isolating the monomers or multimers that bind to a particular tissue or cell of interest.
  • the fusion proteins described above may also include a linker peptide between the pharmaceutical protein and the monomer or multimers.
  • a peptide linker sequence may be employed to separate, for example, the polypeptide components by a distance sufficient to ensure that each polypeptide folds into its secondary and tertiary structures.
  • Fusion proteins may generally be prepared using standard techniques, including chemical conjugation. Fusion proteins can also be expressed as recombinant proteins in an expression system by standard techniques.
  • Multimers or monomer domains of the invention can be produced according to any methods known in the art.
  • E. coli comprising a pET-derived plasmid encoding the polypeptides are induced to express the protein. After harvesting the bacteria, they may be lysed and clarified by centrifugation.
  • the polypeptides may be purified using Ni—NTA agarose elution and refolded by dialysis. Misfolded proteins may be neutralized by capping free sulfhydrils with iodoacetic acid.
  • Q sepharose elution, butyl sepharose FT, SP sepharose elution, Q sepharose elution, and/or SP sepharose elution may be used to purify the polypeptides.
  • Monomer domains can be joined by a linker to form a multimer.
  • a linker may be positioned between each separate discrete monomer domain in a multimer.
  • Joining the selected monomer domains via a linker can be accomplished using a variety of techniques known in the art. For example, combinatorial assembly of polynucleotides encoding selected monomer domains can be achieved by restriction digestion and re-ligation, by PCR-based, self-priming overlap reactions, or other recombinant methods.
  • the linker can be attached to a monomer before the monomer is identified for its ability to bind to a target multimer or after the monomer has been selected for the ability to bind to a target multimer.
  • the linker can be naturally-occurring, synthetic or a combination of both.
  • the synthetic linker can be a randomized linker, e.g., both in sequence and size.
  • the randomized linker can comprise a fully randomized sequence, or optionally, the randomized linker can be based on natural linker sequences.
  • the linker can comprise, e.g,. a non-polypeptide moiety, a polynucleotide, a polypeptide or the like.
  • a linker can be rigid, or flexible, or a combination of both.
  • Linker flexibility can be a function of the composition of both the linker and the monomer domains that the linker interacts with.
  • the linker joins two selected monomer domain, and maintains the monomer domains as separate discrete monomer domains.
  • the linker can allow the separate discrete monomer domains to cooperate yet maintain separate properties such as multiple separate binding sites for the same ligand in a multimer, or e.g., multiple separate binding sites for different ligands in a multimer.
  • Choosing a suitable linker for a specific case where two or more monomer domains (i.e. polypeptide chains) are to be connected may depend on a variety of parameters including, e.g. the nature of the monomer domains, the structure and nature of the target to which the polypeptide multimer should bind and/or the stability of the peptide linker towards proteolysis and oxidation.
  • the present invention provides methods for optimizing the choice of linker once the desired monomer domains/variants have been identified.
  • libraries of multimers having a composition that is fixed with regard to monomer domain composition, but variable in linker composition and length can be readily prepared and screened as described above.
  • the linker polypeptide may predominantly include amino acid residues selected from the group consisting of Gly, Ser, Ala and Thr.
  • the peptide linker may contain at least 75% (calculated on the basis of the total number of residues present in the peptide linker), such as at least 80%, e.g. at least 85% or at least 90% of amino acid residues selected from the group consisting of Gly, Ser, Ala and Thr.
  • the peptide linker may also consist of Gly, Ser, Ala and/or Thr residues only.
  • the linker polypeptide should have a length, which is adequate to link two monomer domains in such a way that they assume the correct conformation relative to one another so that they retain the desired activity, for example as antagonists of a given receptor.
  • a suitable length for this purpose is a length of at least one and typically fewer than about 50 amino acid residues, such as 2-25 amino acid residues, 5-20 amino acid residues, 5-15 amino acid residues, 8-12 amino acid residues or 11 residues.
  • the polypeptide encoding a linker can range in size, e.g., from about 2 to about 15 amino acids, from about 3 to about 15, from about 4 to about 12, about 10, about 8, or about 6 amino acids.
  • the polynucleotide containing the linker sequence can be, e.g., between about 6 nucleotides and about 45 nucleotides, between about 9 nucleotides and about 45 nucleotides, between about 12 nucleotides and about 36 nucleotides, about 30 nucleotides, about 24 nucleotides, or about 18 nucleotides.
  • the amino acid residues selected for inclusion in the linker polypeptide should exhibit properties that do not interfere significantly with the activity or function of the polypeptide multimer.
  • the peptide linker should on the whole not exhibit a charge which would be inconsistent with the activity or function of the polypeptide multimer, or interfere with internal folding, or form bonds or other interactions with amino acid residues in one or more of the monomer domains which would seriously impede the binding of the polypeptide multimer to the target in question.
  • the peptide linker is selected from a library where the amino acid residues in the peptide linker are randomized for a specific set of monomer domains in a particular polypeptide multimer.
  • a flexible linker could be used to find suitable combinations of monomer domains, which is then optimized using this random library of variable linkers to obtain linkers with optimal length and geometry.
  • the optimal linkers may contain the minimal number of amino acid residues of the right type that participate in the binding to the target and restrict the movement of the monomer domains relative to each other in the polypeptide multimer when not bound to the target.
  • peptide linkers are widely used for production of single-chain antibodies where the variable regions of a light chain (V L ) and a heavy chain (V H ) are joined through an artificial linker, and a large number of publications exist within this particular field.
  • a widely used peptide linker is a 15mer consisting of three repeats of a Gly-Gly-Gly-Gly-Ser (SEQ ID NO: 240) amino acid sequence ((Gly 4 Ser) 3 ).
  • Other linkers have been used, and phage display technology, as well as, selective infective phage technology has been used to diversify and select appropriate linker sequences (Tang et al. (1996), J. Biol. Chem.
  • Peptide linkers have been used to connect individual chains in hetero- and homo-dimeric proteins such as the T-cell receptor, the lambda Cro repressor, the P22 phage Arc repressor, IL-12, TSH, FSH, 1L-5, and interferon- ⁇ . Peptide linkers have also been used to create fusion polypeptides. Various linkers have been used and in the case of the Arc repressor phage display has been used to optimize the linker length and composition for increased stability of the single-chain protein (Robinson and Sauer (1998), Proc. Natl. Acad. Sci. USA 95, 5929-5934).
  • intein i.e. a peptide stretch which is expressed with the single-chain polypeptide, but removed post-translationally by protein splicing.
  • inteins The use of inteins is reviewed by F. S. Gimble in Chemistry and Biology, 1998, Vol 5, No. 10 pp. 251-256.
  • Still another way of obtaining a suitable linker is by optimizing a simple linker, e.g. (Gly 4 Ser) n , (SEQ ID NO: 240), through random mutagenesis.
  • a simple linker e.g. (Gly 4 Ser) n , (SEQ ID NO: 240)
  • the peptide linker possess at least some flexibility. Accordingly, in some embodiments, the peptide linker contains 1-25 glycine residues, 5-20 glycine residues, 5-15 glycine residues or 8-12 glycine residues. The peptide linker will typically contain at least 50% glycine residues, such as at least 75% glycine residues. In some embodiments of the invention, the peptide linker comprises glycine residues only.
  • the peptide linker may, in addition to the glycine residues, comprise other residues, in particular residues selected from the group consisting of Ser, Ala and Thr, in particular Ser.
  • a specific peptide linker includes a peptide linker having the amino acid sequence Gly x -Xaa-Gly y -Xaa-Gly z (SEQ ID NO: 203), wherein each Xaa is independently selected from the group consisting Ala, Val, Leu, Ile, Met, Phe, Trp, Pro, Gly, Ser, Thr, Cys, Tyr, Asn, Gln, Lys, Arg, His, Asp and Glu, and wherein x, y and z are each integers in the range from 1-5.
  • each Xaa is independently selected from the group consisting of Ser, Ala and Thr, in particular Ser. More particularly, the peptide linker has the amino acid sequence Gly-Gly-Gly-Xaa-Gly-Gly-Gly-Xaa-Gly-Gly-Gly-Gly-Gly-Gly-Gly (SEQ ID NO: 204), wherein each Xaa is independently selected from the group consisting Ala, Val, Leu, Ile, Met, Phe, Trp, Pro, Gly, Ser, Thr, Cys, Tyr, Asn, Gln, Lys, Arg, His, Asp and Glu. In some embodiments, each Xaa is independently selected from the group consisting of Ser, Ala and Thr, in particular Ser.
  • the peptide linker comprises at least one proline residue in the amino acid sequence of the peptide linker.
  • the peptide linker has an amino acid sequence, wherein at least 25%, such as at least 50%, e.g. at least 75%, of the amino acid residues are proline residues.
  • the peptide linker comprises proline residues only.
  • the peptide linker is modified in such a way that an amino acid residue comprising an attachment group for a non-polypeptide moiety is introduced.
  • amino acid residues may be a cysteine residue (to which the non-polypeptide moiety is then subsequently attached) or the amino acid sequence may include an in vivo N-glycosylation site (thereby attaching a sugar moiety (in vivo) to the peptide linker).
  • the peptide linker comprises at least one cysteine residue, such as one cysteine residue.
  • the peptide linker comprises amino acid residues selected from the group consisting of Gly, Ser, Ala, Thr and Cys. In some embodiments, such a peptide linker comprises one cysteine residue only.
  • the peptide linker comprises glycine residues and cysteine residue, such as glycine residues and cysteine residues only. Typically, only one cysteine residue will be included per peptide linker.
  • a specific peptide linker comprising a cysteine residue includes a peptide linker having the amino acid sequence Gly n -Cys-Gly m (SEQ ID NO: 205), wherein n and m are each integers from 1-12, e.g., from 3-9, from 4-8, or from 4-7. More particularly, the peptide linker may have the amino acid sequence GGGGG-C-GGGGG (SEQ ID NO: 206).
  • the peptide linker may comprise proline and cysteine residues, such as proline and cysteine residues only.
  • An example of a specific proline-containing peptide linker comprising a cysteine residue includes a peptide linker having the amino acid sequence Pro n -Cys-Pro m (SEQ ID NO: 207), wherein n and m are each integers from 1-12, preferably from 3-9, such as from 4-8 or from 4-7. More particularly, the peptide linker may have the amino acid sequence PPPPP—C—PPPPP (SEQ ID NO: 208).
  • the purpose of introducing an amino acid residue, such as a cysteine residue, comprising an attachment group for a non-polypeptide moiety is to subsequently attach a non-polypeptide moiety to said residue.
  • non-polypeptide moieties can improve the serum half-life of the polypeptide multimer.
  • the cysteine residue can be covalently attached to a non-polypeptide moiety.
  • Preferred examples of non-polypeptide moieties include polymer molecules, such as PEG or mPEG, in particular mPEG as well as non-polypeptide therapeutic agents.
  • amino acid residues other than cysteine may be used for attaching a non-polypeptide to the peptide linker.
  • One particular example of such other residue includes coupling the non-polypeptide moiety to a lysine residue.
  • Another possibility of introducing a site-specific attachment group for a non-polypeptide moiety in the peptide linker is to introduce an in vivo N-glycosylation site, such as one in vivo N-glycosylation site, in the peptide linker.
  • an in vivo N-glycosylation site may be introduced in a peptide linker comprising amino acid residues selected from the group consisting of Gly, Ser, Ala and Thr.
  • the nucleotide sequence encoding the polypeptide multimer must be inserted in a glycosylating, eukaryotic expression host.
  • a specific example of a peptide linker comprising an in vivo N-glycosylation site is a peptide linker having the amino acid sequence Gly n -Asn-Xaa-Ser/Thr-Gly m (SEQ ID NO: 209), preferably Gly n -Asn-Xaa-Thr-Gly m (SEQ ID NO: 210), wherein Xaa is any amino acid residue except proline, and wherein n and m are each integers in the range from 1-8, preferably in the range from 2-5.
  • a linker can be a native or synthetic linker sequence.
  • An exemplary native linker includes, e.g., the sequence between the last cysteine of a first LDL receptor A domain and the first cysteine of a second LDL receptor A domain can be used as a linker sequence.
  • Analysis of various A domain linkages reveals that native linkers range from at least 3 amino acids to fewer than 20 amino acids, e.g., 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or 18 amino acids long. However, those of skill in the art will recognize that longer or shorter linker sequences can be used.
  • An exemplary A domain linker sequence is depicted in FIG. 8 .
  • the linker is a 6-mer of the following sequence A 1 A 2 A 3 A 4 A 5 A 6 (SEQ ID NO: 244), wherein A1 is selected from the amino acids A, P, T, Q, E and K; A 2 and A 3 are any amino acid except C, F, Y, W, or M; A 4 is selected from the amino acids S, G and R; A 5 is selected from the amino acids H, P, and R; and A 6 is the amino acid, T.
  • Methods for generating multimers from monomer domains can include joining the selected domains with at least one linker to generate at least one multimer, e.g., the multimer can comprise at least two of the monomer domains and the linker.
  • the multimer(s) is then screened for an improved avidity or affinity or altered specificity for the desired ligand or mixture of ligands as compared to the selected monomer domains.
  • a composition of the multimer produced by the method is included in the present invention.
  • the selected multimer domains are joined with at least one linker to generate at least two multimers, wherein the two multimers comprise two or more of the selected monomer domains and the linker.
  • the two or more multimers are screened for an improved avidity or affinity or altered specificity for the desired ligand or mixture of ligands as compared to the selected monomer domains.
  • Compositions of two or more multimers produced by the above method are also features of the invention.
  • multimers of the present invention are a single discrete polypeptide.
  • Multimers of partial linker-domain-partial linker moieties are an association of multiple polypeptides, each corresponding to a partial linker-domain-partial linker moiety.
  • Partial linkers can contain terminal amino acid sequences that specifically bind to a defined heterologous amino acid sequence.
  • An example of such an amino acid sequence is the Hydra neuropeptide head activator as described in Bodenmuller et al., The neuropeptide head activator loses its biological activity by dimerization , (1986) EMBO J. 5(8):1825-1829. See, e.g., U.S. Pat. No.
  • partial linkers allow the multimer to be produced first as monomer-partial linker units or partial linker-monomer-partial linker units that are then mixed together and allowed to assemble into the ideal order based on the binding specificities of each partial linker.
  • monomers linked to partial linkers can be contacted to a surface, such as a cell, in which multiple monomers can associate to form higher avidity complexes via partial linkers. In some cases, the association will form via random Brownian motion.
  • each monomer domain has an upstream and a downstream partial linker (i.e., Lp-domain-Lp, where “Lp” is a representation of a partial linker) that contains a DNA binding protein with exclusively unique DNA binding specificity.
  • Lp-domain-Lp a partial linker
  • These domains can be produced individually and then assembled into a specific multimer by the mixing of the domains with DNA fragments containing the proper nucleotide sequences (i.e., the specific recognition sites for the DNA binding proteins of the partial linkers of the two desired domains) so as to join the domains in the desired order. Additionally, the same domains may be assembled into many different multimers by the addition of DNA sequences containing various combinations of DNA binding protein recognition sites. Further randomization of the combinations of DNA binding protein recognition sites in the DNA fragments can allow the assembly of libraries of multimers.
  • the DNA can be synthesized with backbone analogs to prevent degradation in vivo.
  • Such an amino acid sequence is the Hydra neuropeptide head activator as described in Bodenmuller et al., The neuropeptide head activator loses its biological activity by dimerization , (1986) EMBO J. 5(8):1825-1829. See, e.g., U.S. Pat. No. 5,491,074 and WO 94/28173.
  • These partial linkers allow the multimer to be produced first as monomer-partial linker units or partial linker-monomer-partial linker units that are then mixed together and allowed to assemble into the ideal order based on the binding specificities of each partial linker.
  • monomers linked to partial linkers can be contacted to a surface, such as a cell, in which multiple monomers can associate to form higher avidity complexes via partial linkers. In some cases, the association will form via random Brownian motion.
  • any method resulting in selection of domains with a desired property can be used.
  • the methods can comprise providing a plurality of different nucleic acids, each nucleic acid encoding a monomer domain; translating the plurality of different nucleic acids, thereby providing a plurality of different monomer domains; screening the plurality of different monomer domains for binding of the desired ligand or a mixture of ligands; and, identifying members of the plurality of different monomer domains that bind the desired ligand or mixture of ligands.
  • any method of mutagenesis such as site-directed mutagenesis and random mutagenesis (e.g., chemical mutagenesis) can be used to produce monomer domains, e.g., for a monomer domain library.
  • error-prone PCR is employed to create variants. Additional methods include aligning a plurality of naturally occurring monomer domains by aligning conserved amino acids in the plurality of naturally occurring monomer domains; and, designing the non-naturally occurring monomer domain by maintaining the conserved amino acids and inserting, deleting or altering amino acids around the conserved amino acids to generate the non-naturally occurring monomer domain.
  • the conserved amino acids comprise cysteines.
  • the inserting step uses random amino acids, or optionally, the inserting step uses portions of the naturally occurring monomer domains.
  • the portions could ideally encode loops from domains from the same family.
  • Amino acids are inserted or exchanged using synthetic oligonucleotides, or by shuffling, or by restriction enzyme based recombination.
  • Human chimeric domains of the present invention are useful for therapeutic applications where minimal immunogenicity is desired.
  • the present invention provides methods for generating libraries of human chimeric domains.
  • Human chimeric monomer domain libraries can be constructed by combining loop sequences from different variants of a human monomer domain, as described above.
  • the loop sequences that are combined may be sequence-defined loops, structure-defined loops, B-factor-defined loops, or a combination of any two or more thereof.
  • Libraries of human chimeric monomer domains can be employed to identify human chimeric monomer domains that bind to a target of interest by: screening the library of human chimeric monomer domains for binding to a target molecule, and identifying a human chimeric monomer domain that binds to the target molecule.
  • Suitable naturally-occurring human monomer domain sequences employed in the initial sequence alignment step include those corresponding to any of the naturally-occurring monomer domains described herein.
  • monomer domains of the invention are screened for potential immunogenicity by:
  • the method is particularly useful in determining whether a crossover sequence in a chimeric protein, such as, for example, a chimeric monomer domain, is likely to cause an immunogenic event. If the crossover sequence corresponds to a portion of a sequence found in the database of human protein sequences, it is believed that the crossover sequence is less likely to cause an immunogenic event.
  • FIG. 12 An example of the comparison step is depicted in FIG. 12 , which shows a comparison of two candidate protein sequences to human sequences from a database. The horizontal lines indicate where the human protein sequences from the database are identical to the candidate protein sequence.
  • Information pertaining to portions of human protein sequences from the database can be used to design a protein library of human-like chimeric proteins.
  • Such library can be generated by using information pertaining to “crossover sequences” that exist in naturally occurring human proteins.
  • the term “crossover sequence” refers herein to a sequence that is found in its entirety in at least one naturally occurring human protein, in which portions of the sequence are found in two or more naturally occurring proteins. Thus, recombination of the latter two or more naturally occurring proteins would generate a chimeric protein in which the chimeric portion of the sequence actually corresponds to a sequence found in another naturally occurring protein.
  • the crossover sequence contains a chimeric junction of two consecutive amino acid residue positions in which the first amino acid position is occupied by an amino acid residue identical in type and position found in a first and second naturally occurring human protein sequence, but not a third naturally occurring human protein sequence.
  • the second amino acid position is occupied by an amino acid residue identical in type and position found in a second and third naturally occurring human protein sequence, but not the first naturally occurring human protein sequence.
  • the “second” naturally occurring human protein sequence corresponds to the naturally occurring human protein in which the crossover sequence appears in its entirety, as described above.
  • the chimeric junction is C-D.
  • the first naturally-occurring human protein sequence is D-E-F-G, and the second is B-C-D-E-F, and the third is A-B-C-D, then the chimeric junction is D-E.
  • Human-like chimeric protein molecules can be generated in a variety of ways.
  • oligonucleotides comprising sequences encoding the chimeric junctions can be recombined with oligonucleotides corresponding in sequence to two or more subsequences from the above-described set of subsequences to generate a human-like chimeric protein, and libraries thereof.
  • the reference sequence used to align the naturally occurring human proteins is a sequence from the same family of naturally occurring human proteins, or a chimera or other variant of proteins in the family.
  • Nucleic acids encoding fragments of naturally-occurring monomer domains can also be mixed and/or recombined (e.g., by using chemically or enzymatically-produced fragments) to generate full-length, modified monomer domains.
  • the fragments and the monomer domain can also be recombined by manipulating nucleic acids encoding domains or fragments thereof. For example, ligating a nucleic acid construct encoding fragments of the monomer domain can be used to generate an altered monomer domain.
  • Altered monomer domains can also be generated by providing a collection of synthetic oligonucleotides (e.g., overlapping oligonucleotides) encoding conserved, random, pseudorandom, or a defined sequence of peptide sequences that are then inserted by ligation into a predetermined site in a polynucleotide encoding a monomer domain.
  • sequence diversity of one or more monomer domains can be expanded by mutating the monomer domain(s) with site-directed mutagenesis, random mutation, pseudorandom mutation, defined kernal mutation, codon-based mutation, and the like.
  • the resultant nucleic acid molecules can be propagated in a host for cloning and amplification. In some embodiments, the nucleic acids are shuffled.
  • the present invention also provides a method for recombining a plurality of nucleic acids encoding monomer domains and screening the resulting library for monomer domains that bind to the desired ligand or mixture of ligands or the like.
  • Selected monomer domain nucleic acids can also be back-crossed by shuffling with polynucleotide sequences encoding neutral sequences (i.e., having insubstantial functional effect on binding), such as for example, by back-crossing with a wild-type or naturally-occurring sequence substantially identical to a selected sequence to produce native-like functional monomer domains.
  • subsequent selection is applied to retain the property, e.g., binding to the ligand.
  • the monomer library is prepared by shuffling.
  • monomer domains are isolated and shuffled to combinatorially recombine the nucleic acid sequences that encode the monomer domains (recombination can occur between or within monomer domains, or both).
  • the first step involves identifying a monomer domain having the desired property, e.g., affinity for a certain ligand. While maintaining the conserved amino acids during the recombination, the nucleic acid sequences encoding the monomer domains can be recombined, or recombined and joined into multimers.
  • a significant advantage of the present invention is that known ligands, or unknown ligands can be used to select the monomer domains and/or multimers. No prior information regarding ligand structure is required to isolate the monomer domains of interest or the multimers of interest.
  • the monomer domains and/or multimers identified can have biological activity, which is meant to include at least specific binding affinity for a selected or desired ligand, and, in some instances, will further include the ability to block the binding of other compounds, to stimulate or inhibit metabolic pathways, to act as a signal or messenger, to stimulate or inhibit cellular activity, and the like.
  • Monomer domains can be generated to function as ligands for receptors where the natural ligand for the receptor has not yet been identified (orphan receptors). These orphan ligands can be created to either block or activate the receptor to which they bind.
  • a single ligand can be used, or optionally a variety of ligands can be used to select the monomer domains and/or multimers.
  • a monomer domain of the present invention can bind a single ligand or a variety of ligands.
  • a multimer of the present invention can have multiple discrete binding sites for a single ligand, or optionally, can have multiple binding sites for a variety of ligands.
  • the invention also includes compositions that are produced by methods of the present invention.
  • the present invention includes monomer domains selected or identified from a library and/or libraries comprising monomer domains produced by the methods of the present invention.
  • the present invention also provides libraries of monomer domains and libraries of nucleic acids that encode monomer domains.
  • the libraries can include, e.g., about 100, 250, 500 or more nucleic acids encoding monomer domains, or the library can include, e.g., about 100, 250, 500 or more polypeptides that encode monomer domains.
  • Libraries can include monomer domains containing the same cysteine frame, e.g., A-domains or EGF-like domains.
  • variants are generated by recombining two or more different sequences from the same family of monomer domains (e.g., the LDL receptor class A domain).
  • two or more different monomer domains from different families can be combined to form a multimer.
  • the multimers are formed from monomers or monomer variants of at least one of the following family classes: an EGF-like domain, a Kringle-domain, a fibronectin type I domain, a fibronectin type II domain, a fibronectin type III domain, a PAN domain, a Gla domain, a SRCR domain, a Kunitz/Bovine pancreatic trypsin Inhibitor domain, a Kazal-type serine protease inhibitor domain, a Trefoil (P-type) domain, a von Willebrand factor type C domain, an Anaphylatoxin-like domain, a CUB domain, a thyroglobulin type I repeat, LDL-receptor class A domain, a Sushi domain, a Link domain, a Thrombospondin type I domain, an Immunoglobulin-like domain, a C-type lectin domain, a MAM domain, a von Willebrand factor type A domain, a S
  • the monomer domain and the different monomer domain can include one or more domains found in the Pfam database and/or the SMART database.
  • Libraries produced by the methods above, one or more cell(s) comprising one or more members of the library, and one or more displays comprising one or more members of the library are also included in the present invention.
  • a data set of nucleic acid character strings encoding monomer domains can be generated e.g., by mixing a first character string encoding a monomer domain, with one or more character string encoding a different monomer domain, thereby producing a data set of nucleic acids character strings encoding monomer domains, including those described herein.
  • the monomer domain and the different monomer domain can include one or more domains found in the Pfam database and/or the SMART database.
  • the methods can further comprise inserting the first character string encoding the monomer domain and the one or more second character string encoding the different monomer domain in a computer and generating a multimer character string(s) or library(s), thereof in the computer.
  • the libraries can be screened for a desired property such as binding of a desired ligand or mixture of ligands.
  • members of the library of monomer domains can be displayed and prescreened for binding to a known or unknown ligand or a mixture of ligands.
  • the monomer domain sequences can then be mutagenized (e.g., recombined, chemically altered, etc.) or otherwise altered and the new monomer domains can be screened again for binding to the ligand or the mixture of ligands with an improved affinity.
  • the selected monomer domains can be combined or joined to form multimers, which can then be screened for an improved affinity or avidity or altered specificity for the ligand or the mixture of ligands.
  • Altered specificity can mean that the specificity is broadened, e.g., binding of multiple related viruses, or optionally, altered specificity can mean that the specificity is narrowed, e.g., binding within a specific region of a ligand.
  • Those of skill in the art will recognize that there are a number of methods available to calculate avidity. See, e.g., Mammen et al., Angew Chem Int. Ed. 37:2754-2794 (1998); Muller et al., Anal. Biochem. 261:149-158 (1998).
  • Preliminary screens can be conducted by screening for agents capable of binding to c-Met, as at least some of the agents so identified are likely c-Met modulators (e.g., antagonists or agonists).
  • the binding assays usually involve contacting a c-Met protein (or a fragment thereof such as a fragment comprising the SEMA rdomain or the ⁇ chain) with one or more test agents (i.e., monomers or multimers of the invention) and allowing sufficient time for the protein and test agents to form a binding complex. Any binding complexes formed can be detected using any of a number of established analytical techniques. Protein binding assays include, but are not limited to, immunohistochemical binding assays, flow cytometry or other assays. The c-Met protein utilized in such assays can be naturally expressed, cloned or synthesized.
  • the screening methods of the invention can be performed as in vitro or cell-based assays.
  • Cell based assays can be performed in any cells in which c-Met is expressed.
  • Cell-based assays may involve whole cells or cell fractions containing a c-Met receptor to screen for agent binding or modulation of activity of c-Met by the agent.
  • Exemplary cell types that can be used according to the methods of the invention include, e.g., any mammalian cells, as well as fungal cells, including yeast, and bacterial cells.
  • Cells can be primary cells or tumor cells or other types of immortal cell lines.
  • c-Met can be expressed in cells that do not endogenously contain c-Met.
  • c-Met activity assays may also be used to identify a modulator (antagonist or agonist) of c-Met.
  • one or more test agents are contacted to a cell expressing c-Met and then tested for an activity of c-Met.
  • Exemplary c-Met activities include HGF-dependent or constitutive kinase activity. See, e.g., Christensen et al., Cancer Res. 63:7345-7355 (2003).
  • down stream molecular events can also be monitored to determine signaling activity. For example, c-Met induces cell growth (proliferation and survival), cell motility, invasion and morphology changes.
  • c-Met indirectly mediates phosphorylation of Gab-1, Akt, signal transducer and activator of transcription 3, phospholipase C ⁇ , and focal adhesions kinase, among others. See, e.g., Christensen et al., Cancer Res. 63:7345-7355 (2003).
  • activity assays are also used to confirm that identified antagonist monomers or multimers (i.e., that compete with HGF) lack agonist activity (i.e., that they do not activate c-Met in the absence of HGF or another agonist).
  • Agents that are initially identified by any of the foregoing screening methods can be further tested to validate the apparent activity.
  • Such studies may be conducted with suitable animal models.
  • the basic format of such methods involves administering a lead compound identified during an initial screen to an animal that serves as a model for humans and then determining if c-Met is in fact modulated and/or the disease or condition is ameliorated.
  • the animal models utilized in validation studies generally are mammals of any kind. Specific examples of suitable animals include, but are not limited to, primates, mice and rats.
  • Selection of monomer domains that bind c-Met from a library of domains can be accomplished by a variety of procedures.
  • one method of identifying monomer domains which have a desired property involves translating a plurality of nucleic acids, where each nucleic acid encodes a monomer domain, screening the polypeptides encoded by the plurality of nucleic acids, and identifying those monomer domains that, e.g., bind to a desired ligand or mixture of ligands, thereby producing a selected monomer domain.
  • the monomer domains expressed by each of the nucleic acids can be tested for their ability to bind to the ligand by methods known in the art (i.e. panning, affinity chromatography, FACS analysis).
  • selection of monomer domains can be based on binding to a ligand such as c-Met, or a fragment therof or other target molecule (e.g., lipid, carbohydrate, nucleic acid and the like).
  • a ligand such as c-Met
  • target molecule e.g., lipid, carbohydrate, nucleic acid and the like.
  • Other molecules can optionally be included in the methods along with the target, e.g., ions such as Ca +2 .
  • the selection basis can include selection based on a slow dissociation rate, which is usually predictive of high affinity.
  • the valency of the ligand can also be varied to control the average binding affinity of selected monomer domains.
  • the ligand can be bound to a surface or substrate at varying densities, such as by including a competitor compound, by dilution, or by other method known to those in the art.
  • High density (valency) of predetermined ligand can be used to enrich for monomer domains that have relatively low affinity, whereas a low density (valency) can preferentially enrich for higher affinity monomer domains.
  • a variety of reporting display vectors or systems can be used to express nucleic acids encoding the monomer domains and/or multimers of the present invention and to test for a desired activity.
  • a phage display system is a system in which monomer domains are expressed as fusion proteins on the phage surface (Pharmacia, Milwaukee Wis.).
  • Phage display can involve the presentation of a polypeptide sequence encoding monomer domains on the surface of a filamentous bacteriophage, typically as a fusion with a bacteriophage coat protein.
  • each phage particle or cell serves as an individual library member displaying a single species of displayed polypeptide in addition to the natural phage or cell protein sequences.
  • the nucleic acids are cloned into the phage DNA at a site which results in the transcription of a fusion protein, a portion of which is encoded by the plurality of the nucleic acids.
  • the phage containing a nucleic acid molecule undergoes replication and transcription in the cell.
  • the leader sequence of the fusion protein directs the transport of the fusion protein to the tip of the phage particle.
  • the fusion protein that is partially encoded by the nucleic acid is displayed on the phage particle for detection and selection by the methods described above and below.
  • the phage library can be incubated with a predetermined ligand such as c-Met or a fragment thereof, so that phage particles which present a fusion protein sequence that binds to the ligand can be differentially partitioned from those that do not present polypeptide sequences that bind to the predetermined ligand.
  • the separation can be provided by immobilizing the predetermined ligand.
  • the phage particles i.e., library members
  • which are bound to the immobilized ligand are then recovered and replicated to amplify the selected phage subpopulation for a subsequent round of affinity enrichment and phage replication.
  • the phage library members that are thus selected are isolated and the nucleotide sequence encoding the displayed polypeptide sequence is determined, thereby identifying the sequence(s) of polypeptides that bind to the predetermined ligand.
  • Such methods are further described in PCT patent publication Nos. 91/17271, 91/18980, and 91/19818 and 93/08278.
  • Examples of other display systems include ribosome displays, a nucleotide-linked display (see, e.g., U.S. Pat. Nos. 6,281,344; 6,194,550, 6,207,446, 6,214,553, and 6,258,558), polysome display, cell surface displays and the like.
  • the cell surface displays include a variety of cells, e.g., E. coli , yeast and/or mammalian cells.
  • the nucleic acids e.g., obtained by PCR amplification followed by digestion, are introduced into the cell and translated.
  • polypeptides encoding the monomer domains or the multimers of the present invention can be introduced, e.g., by injection, into the cell.
  • the monomer and multimer libraries of the invention can be screened for a desired property such as binding of a desired ligand (e.g., c-Met) or mixture of ligands.
  • a desired ligand e.g., c-Met
  • members of the library of monomer domains can be displayed and prescreened for binding to a known or unknown ligand or a mixture of ligands.
  • the monomer domain sequences can then be mutagenized (e.g., recombined, chemically altered, etc.) or otherwise altered and the new monomer domains can be screened again for binding to the ligand or the mixture of ligands with an improved affinity.
  • the selected monomer domains can be combined or joined to form multimers, which can then be screened for an improved affinity or avidity or altered specificity for the ligand or the mixture of ligands.
  • Altered specificity can mean that the specificity is broadened, e.g., binding of multiple related ligands, or optionally, altered specificity can mean that the specificity is narrowed, e.g., binding within a specific region of a ligand.
  • Those of skill in the art will recognize that there are a number of methods available to calculate avidity. See, e.g., Mammen et al., Angew Chem Int. Ed. 37:2754-2794 (1998); Muller et al., Anal. Biochem. 261:149-158 (1998).
  • a first screening of a library can be performed at relatively lower stringency, thereby selected as many particles associated with a target molecule as possible.
  • the selected particles can then be isolated and the polynucleotides encoding the monomer or multimer can be isolated from the particles. Additional variations can then be generated from these sequences and subsequently screened at higher affinity.
  • FIG. 7 illustrates a generic cycle of selection and generation of variation.
  • compositions of the present invention e.g., monomer domains as well as multimers and libraries thereof can be optionally bound to a matrix of an affinity material.
  • affinity material include beads, a column, a solid support, a microarray, other pools of reagent-supports, and the like.
  • the “walking” monomer selection method provides a way to assemble a multimer that is composed of monomers that can act additively or even synergistically with each other given the restraints of linker length. This walking technique is very useful when selecting for and assembling multimers that are able to bind large target proteins with high affinity. The walking method can be repeated to add more monomers thereby resulting in a multimer comprising 2, 3, 4, 5, 6, 7, 8 or more monomers linked together.
  • the selected multimer comprises more than two domains.
  • Such multimers can be generated in a step fashion, e.g., where the addition of each new domain is tested individually and the effect of the domains is tested in a sequential fashion. See, e.g., FIG. 6 .
  • domains are linked to form multimers comprising more than two domains and selected for binding without prior knowledge of how smaller multimers, or alternatively, how each domain, bind.
  • Variation can be introduced into either monomers or multimers.
  • An example of improving monomers includes intra-domain recombination in which two or more (e.g., three, four, five, or more) portions of the monomer are amplified separately under conditions to introduce variation (for example by shuffling or other recombination method) in the resulting amplification products, thereby synthesizing a library of variants for different portions of the monomer.
  • variation for example by shuffling or other recombination method
  • the middle primers By locating the 5′ ends of the middle primers in a “middle” or ‘overlap’ sequence that both of the PCR fragments have in common, the resulting “left” side and “right” side libraries may be combined by overlap PCR to generate novel variants of the original pool of monomers.
  • the “middle” primer(s) may be selected to correspond to any segment of the monomer, and will typically be based on the scaffold or one or more concensus amino acids within the monomer (e.g., cysteines such as those found in A domains).
  • multimers may be created by introducing variation at the monomer level and then recombining monomer variant libraries.
  • multimers single or pools
  • desired properties may be recombined to form longer multimers.
  • variation is introduced (typically synthetically) into the monomers or into the linkers to form libraries. This may be achieved, e.g., with two different multimers that bind to two different targets, thereby eventually selecting a multimer with a portion that binds to one target and a portion that binds a second target. See, e.g., FIG. 21 .
  • Additional variation can be introduced by inserting linkers of different length and composition between domains. This allows for the selection of optimal linkers between domains.
  • optimal length and composition of linkers will allow for optimal binding of domains.
  • the domains with a particular binding affinity(s) are linked via different linkers and optimal linkers are selected in a binding assay. For example, domains are selected for desired binding properties and then formed into a library comprising a variety of linkers. The library can then be screened to identify optimal linkers. Alternatively, multimer libraries can be formed where the effect of domain or linker on target molecule binding is not known.
  • Methods of the present invention also include generating one or more selected multimers by providing a plurality of monomer domains.
  • the plurality of monomer domains is screened for binding of a desired ligand or mixture of ligands.
  • Members of the plurality of domains that bind the desired ligand or mixture of ligands are identified, thereby providing domains with a desired affinity.
  • the identified domains are joined with at least one linker to generate the multimers, wherein each multimer comprises at least two of the selected domains and the at least one linker; and, the multimers are screened for an improved affinity or avidity or altered specificity for the desired ligand or mixture of ligands as compared to the selected domains, thereby identifying the one or more selected multimers.
  • Multimer libraries may be generated, in some embodiments, by combining two or more libraries or monomers or multimers in a recombinase-based approach, where each library member comprises as recombination site (e.g., a lox site).
  • a recombination site e.g., a lox site.
  • libraries are constructed in phage vectors, which may be transformed into E. coli , library size (10 9 -10 10 ) is limited by the transformation efficiency of E. coli .
  • a recombinase/recombination site system e.g., the Cre-loxP system
  • in vivo recombination can be exploited to generate libraries that are not limited in size by the transformation efficiency of E. coli.
  • the Cre-loxP system may be used to generate dimer libraries with 10 10 , 10 11 , 10 12 , 10 13 , or greater diversity.
  • E. coli as a host for one na ⁇ ve monomer library and a filamentous phage that carries a second na ⁇ ve monomer library are used.
  • Selection of multimers can be accomplished using a variety of techniques including those mentioned above for identifying monomer domains.
  • Other selection methods include, e.g., a selection based on an improved affinity or avidity or altered specificity for the ligand compared to selected monomer domains.
  • a selection can be based on selective binding to specific cell types, or to a set of related cells or protein types (e.g., different virus serotypes).
  • Optimization of the property selected for, e.g., avidity of a ligand can then be achieved by recombining the domains, as well as manipulating amino acid sequence of the individual monomer domains or the linker domain or the nucleotide sequence encoding such domains, as mentioned in the present invention.
  • One method for identifying multimers can be accomplished by displaying the multimers.
  • the multimers are optionally expressed or displayed on a variety of display systems, e.g., phage display, ribosome display, polysome display, nucleotide-linked display (see, e.g., U.S. Pat. Nos. 6,281,344; 6,194,550, 6,207,446, 6,214,553, and 6,258,558) and/or cell surface display, as described above.
  • Cell surface displays can include but are not limited to E. coli , yeast or mammalian cells.
  • display libraries of multimers with multiple binding sites can be panned for avidity or affinity or altered specificity for a ligand or for multiple ligands.
  • Monomers or multimers can be screened for target binding activity in yeast cells using a two-hybrid screening assay.
  • the monomer or multimer library to be screened is cloned into a vector that directs the formation of a fusion protein between each monomer or multimer of the library and a yeast transcriptional activator fragment (i.e., Gal4).
  • Sequences encoding the “target” protein are cloned into a vector that results in the production of a fusion protein between the target and the remainder of the Gal4 protein (the DNA binding domain).
  • a third plasmid contains a reporter gene downstream of the DNA sequence of the Gal4 binding site.
  • a monomer that can bind to the target protein brings with it the Gal4 activation domain, thus reconstituting a functional Gal4 protein.
  • This functional Gal4 protein bound to the binding site upstream of the reporter gene results in the expression of the reporter gene and selection of the monomer or multimer as a target binding protein.
  • Using a two-hybrid system for library screening is further described in U.S. Pat. No. 5,811,238 (see also Silver S. C. and Hunt S. W. (1993) Mol. Biol. Rep.
  • affinity agents Libraries of either monomer domains or multimers (referred in the following discussion for convenience as “affinity agents”) can be screened (i.e., panned) simultaneously against multiple ligands in a number of different formats. For example, multiple ligands can be screened in a simple mixture, in an array, displayed on a cell or tissue (e.g., a cell or tissue provides numerous molecules that can be bound by the monomer domains or multimers of the invention), and/or immobilized.
  • the libraries of affinity agents can optionally be displayed on yeast or phage display systems. Similarly, if desired, the ligands (e.g., encoded in a cDNA library) can be displayed in a yeast or phage display system.
  • the affinity agent library is panned against the multiple ligands.
  • the resulting “hits” are panned against the ligands one or more times to enrich the resulting population of affinity agents. See, e.g., FIG. 13 .
  • affinity agents are displayed on phage.
  • Affinity agents identified as binding in the initial screen are divided into a first and second portion. See, e.g., FIG. 14 .
  • the first portion is infected into bacteria, resulting in either plaques or bacterial colonies, depending on the type of phage used.
  • the expressed phage are immobilized and then probed with ligands displayed in phage selected as described below.
  • the second portion are coupled to beads or otherwise immobilized and a phage display library containing at least some of the ligands in the original mixture is contacted to the immobilized second portion. Those phage that bind to the second portion are subsequently eluted and contacted to the immobilized phage described in the paragraph above. Phage-phage interactions are detected (e.g., using a monoclonal antibody specific for the ligand-expressing phage) and the resulting phage polynucleotides can be isolated.
  • the identity of an affinity agent-ligand pair is determined. For example, when both the affinity agent and the ligand are displayed on a phage or yeast, the DNA from the pair can be isolated and sequenced.
  • polynucleotides specific for the ligand and affinity agent are amplified.
  • Amplification primers for each reaction can include 5′ sequences that are complementary such that the resulting amplification products are fused, thereby forming a hybrid polynucleotide comprising a polynucleotide encoding at least a portion of the affinity agent and at least a portion of the ligand.
  • the resulting hybrid can be used to probe affinity agent or ligand (e.g., cDNA-encoded) polynucleotide libraries to identify both affinity agent and ligand. See, e.g., FIG. 15 .
  • a first library of affinity agents (monomer domains or multimers) are panned against multiple ligands and the eluted affinity agents are linked to the first or a second library of affinity agents to form a library of multimeric affinity agents (e.g., comprising 2, 3, 4, 5, 6, 7, 8, 9, or more monomer), which are subsequently panned against the multiple ligands.
  • This method can be repeated to continue to generate larger multimeric affinity agents. Increasing the number of monomer domains may result in increased affinity and avidity for a particular target.
  • trimers of monomer domains that bind CD28 have a higher affinity than dimmers, which in turn have a higher affinity than single CD28-binding monomer domains alone.
  • the panning is optionally repeated to enrich for significant binders.
  • walking will be facilitated by inserting recombination sites (e.g., lox sites) at the ends of monomers and recombining monomer libraries by a recombinase-mediated event.
  • the selected multimers of the above methods can be further manipulated, e.g., by recombining or shuffling the selected multimers (recombination can occur between or within multimers or both), mutating the selected multimers, and the like. This results in altered multimers which then can be screened and selected for members that have an enhanced property compared to the selected multimer, thereby producing selected altered multimers.
  • Naturally or non-naturally occurring monomer domains may be recombined or variants may be formed.
  • the domains initially or later are selected for those sequences that are less likely to be immunogenic in the host for which they are intended.
  • a phage library comprising the recombined domains is panned for a desired affinity.
  • Monomer domains or multimers expressed by the phage may be screened for IC 50 for a target.
  • Hetero- or homo-meric multimers may be selected.
  • the selected polypeptides may be selected for their affinity to any target, including, e.g., hetero- or homo-multimeric targets.
  • Linkers, multimers or selected multimers produced by the methods indicated above and below are features of the present invention.
  • Libraries comprising multimers e.g, a library comprising about 100, 250, 500 or more members produced by the methods of the present invention or selected by the methods of the present invention are provided.
  • one or more cell comprising members of the libraries are also included.
  • Libraries of the recombinant polypeptides are also a feature of the present invention, e.g., a library comprising about 100, 250, 500 or more different recombinant polypetides.
  • compositions of the present invention can be bound to a matrix of an affinity material, e.g., the recombinant polypeptides.
  • affinity material include, e.g., beads, a column, a solid support, and/or the like.
  • the present invention also includes methods of therapeutically or prophylactically treating a disease or disorder by administering in vivo or ex vivo one or more nucleic acids or polypeptides of the invention described above (or compositions comprising a pharmaceutically acceptable excipient and one or more such nucleic acids or polypeptides) to a subject, including, e.g., a mammal, including a human, primate, mouse, pig, cow, goat, rabbit, rat, guinea pig, hamster, horse, sheep; or a non-mammalian vertebrate such as a bird (e.g., a chicken or duck), fish, or invertebrate.
  • a mammal including a human, primate, mouse, pig, cow, goat, rabbit, rat, guinea pig, hamster, horse, sheep; or a non-mammalian vertebrate such as a bird (e.g., a chicken or duck), fish, or inverteb
  • Antagonists of c-Met are useful treating cancers in which Met is expressed.
  • Exemplary cancers include bladder, breast, cervical, colorectal, oesophageal, gastric, head and neck, kidney, liver, lung, nasopharyngeal, ovarian, pancreatic, gall bladder, prostate or thyroid cancer, osteosarcoma, synovial sarcoma, rhabdomosarcoma, MFH/fibrosarcoma, Kaposi's sarcoma, multiple myeloma, lymphomas, adult T-cell leukemia, glioblastomas, astrocytomas, melanoma, mesothelioma, and Wilm's tumor.
  • one or more cells or a population of cells of interest of the subject e.g., tumor cells, tumor tissue sample, organ cells, blood cells, cells of the skin, lung, heart, muscle, brain, mucosae, liver, intestine, spleen, stomach, lymphatic system, cervix, vagina, prostate, mouth, tongue, etc.
  • a selected monomer domain and/or multimer of the invention that is effective in prophylactically or therapeutically treating the disease, disorder, or other condition.
  • the contacted cells are then returned or delivered to the subject to the site from which they were obtained or to another site (e.g., including those defined above) of interest in the subject to be treated.
  • the contacted cells can be grafted onto a tissue, organ, or system site (including all described above) of interest in the subject using standard and well-known grafting techniques or, e.g., delivered to the blood or lymph system using standard delivery or transfusion techniques.
  • the invention also provides in vivo methods in which one or more cells or a population of cells of interest of the subject are contacted directly or indirectly with an amount of a selected monomer domain and/or multimer of the invention effective in prophylactically or therapeutically treating the disease, disorder, or other condition.
  • the selected monomer domain and/or multimer is typically administered or transferred directly to the cells to be treated or to the tissue site of interest (e.g., tumor cells, tumor tissue sample, organ cells, blood cells, cells of the skin, lung, heart, muscle, brain, mucosae, liver, intestine, spleen, stomach, lymphatic system, cervix, vagina, prostate, mouth, tongue, etc.) by any of a variety of formats, including topical administration, injection (e.g., by using a needle or syringe), or vaccine or gene gun delivery, pushing into a tissue, organ, or skin site.
  • the tissue site of interest e.g., tumor cells, tumor tissue sample, organ cells, blood cells, cells of the skin, lung, heart, muscle, brain, mucosae, liver, intestine, spleen, stomach, lymphatic system, cervix, vagina, prostate, mouth, tongue, etc.
  • the selected monomer domain and/or multimer can be delivered, for example, intramuscularly, intradermally, subdermally, subcutaneously, orally, intraperitoneally, intrathecally, intravenously, or placed within a cavity of the body (including, e.g., during surgery), or by inhalation or vaginal or rectal administration.
  • the selected monomer domain and/or multimer is typically administered or transferred indirectly to the cells to be treated or to the tissue site of interest, including those described above (such as, e.g., skin cells, organ systems, lymphatic system, or blood cell system, etc.), by contacting or administering the polypeptide of the invention directly to one or more cells or population of cells from which treatment can be facilitated.
  • tumor cells within the body of the subject can be treated by contacting cells of the blood or lymphatic system, skin, or an organ with a sufficient amount of the selected monomer domain and/or multimer such that delivery of the selected monomer domain and/or multimer to the site of interest (e.g., tissue, organ, or cells of interest or blood or lymphatic system within the body) occurs and effective prophylactic or therapeutic treatment results.
  • site of interest e.g., tissue, organ, or cells of interest or blood or lymphatic system within the body
  • Such contact, administration, or transfer is typically made by using one or more of the routes or modes of administration described above.
  • the invention provides ex vivo methods in which one or more cells of interest or a population of cells of interest of the subject (e.g., tumor cells, tumor tissue sample, organ cells, blood cells, cells of the skin, lung, heart, muscle, brain, mucosae, liver, intestine, spleen, stomach, lymphatic system, cervix, vagina, prostate, mouth, tongue, etc.) are obtained or removed from the subject and transformed by contacting said one or more cells or population of cells with a polynucleotide construct comprising a nucleic acid sequence of the invention that encodes a biologically active polypeptide of interest (e.g., a selected monomer domain and/or multimer) that is effective in prophylactically or therapeutically treating the disease, disorder, or other condition.
  • a polynucleotide construct comprising a nucleic acid sequence of the invention that encodes a biologically active polypeptide of interest (e.g., a selected monomer domain and/or multimer) that is effective in
  • the one or more cells or population of cells is contacted with a sufficient amount of the polynucleotide construct and a promoter controlling expression of said nucleic acid sequence such that uptake of the polynucleotide construct (and promoter) into the cell(s) occurs and sufficient expression of the target nucleic acid sequence of the invention results to produce an amount of the biologically active polypeptide, encoding a selected monomer domain and/or multimer, effective to prophylactically or therapeutically treat the disease, disorder, or condition.
  • the transformed cells are returned, delivered, or transferred to the subject to the tissue site or system from which they were obtained or to another site (e.g., tumor cells, tumor tissue sample, organ cells, blood cells, cells of the skin, lung, heart, muscle, brain, mucosae, liver, intestine, spleen, stomach, lymphatic system, cervix, vagina, prostate, mouth, tongue, etc.) to be treated in the subject.
  • the cells can be grafted onto a tissue, skin, organ, or body system of interest in the subject using standard and well-known grafting techniques or delivered to the blood or lymphatic system using standard delivery or transfusion techniques.
  • Such delivery, administration, or transfer of transformed cells is typically made by using one or more of the routes or modes of administration described above.
  • Expression of the target nucleic acid occurs naturally or can be induced (as described in greater detail below) and an amount of the encoded polypeptide is expressed sufficient and effective to treat the disease or condition at the site or tissue system.
  • the polynucleotide construct can include a promoter sequence (e.g., CMV promoter sequence) that controls expression of the nucleic acid sequence and/or, if desired, one or more additional nucleotide sequences encoding at least one or more of another polypeptide of the invention, a cytokine, adjuvant, or co-stimulatory molecule, or other polypeptide of interest.
  • a promoter sequence e.g., CMV promoter sequence
  • additional nucleotide sequences encoding at least one or more of another polypeptide of the invention, a cytokine, adjuvant, or co-stimulatory molecule, or other polypeptide of interest.
  • compositions comprising an excipient and the polypeptide or nucleic acid of the invention can be administered or delivered.
  • a composition comprising a pharmaceutically acceptable excipient and a polypeptide or nucleic acid of the invention is administered or delivered to the subject as described above in an amount effective to treat the disease or disorder.
  • the amount of polynucleotide administered to the cell(s) or subject can be an amount such that uptake of said polynucleotide into one or more cells of the subject occurs and sufficient expression of said nucleic acid sequence results to produce an amount of a biologically active polypeptide effective to enhance an immune response in the subject, including an immune response induced by an immunogen (e.g., antigen).
  • an immunogen e.g., antigen
  • the amount of polypeptide administered to cell(s) or subject can be an amount sufficient to enhance an immune response in the subject, including that induced by an immunogen (e.g., antigen).
  • expression of the target nucleic of the polynucleotide construct can be regulated in a precise, reversible, and quantitative manner.
  • Gene expression of the target nucleic acid can be induced, for example, after the stable transfected cells containing the polynucleotide construct comprising the target nucleic acid are delivered or transferred to or made to contact the tissue site, organ or system of interest.
  • Such systems are of particular benefit in treatment methods and formats in which it is advantageous to delay or precisely control expression of the target nucleic acid (e.g., to allow time for completion of surgery and/or healing following surgery; to allow time for the polynucleotide construct comprising the target nucleic acid to reach the site, cells, system, or tissue to be treated; to allow time for the graft containing cells transformed with the construct to become incorporated into the tissue or organ onto or into which it has been spliced or attached, etc.).
  • the target nucleic acid e.g., to allow time for completion of surgery and/or healing following surgery; to allow time for the polynucleotide construct comprising the target nucleic acid to reach the site, cells, system, or tissue to be treated; to allow time for the graft containing cells transformed with the construct to become incorporated into the tissue or organ onto or into which it has been spliced or attached, etc.
  • This complex formation results in the half-life extension protecting the multimer and/or bound protein(s) from proteolytic degradation and/or other removal of the multimer and/or protein(s) and thereby extending the half-life of the protein and/or multimer.
  • One variation of this use of the invention includes the half-life extender-binding monomer covalently linked to the protein of interest.
  • the protein of interest may include a monomer domain, a multimer of monomer domains, or a synthetic drug.
  • monomers that bind to either immunoglobulins or erythrocytes could be generated using the above method and could be used for half-life extension.
  • the half-life extender-binding multimers are typically multimers of at least two domains, chimeric domains, or mutagenized domains (i.e., one that binds to Met and one that binds to the blood-carried molecule or cell). Suitable domains include all of those described herein, that are further screened and selected for binding to a half-life extender.
  • the half-life extender-binding multimers are generated in accordance with the methods for making multimers described herein, using, for example, monomer domains pre-screened for half-life extender-binding activity. For example, some half-life extender-binding LDL receptor class A-domain monomers are described in Example 2 below.
  • the serum half-life of a molecule can be extended to be, e.g., at least 1, 2, 3, 4, 5, 10, 20, 30, 40, 50, 60, 70 80, 90, 100, 150, 200, 250, 400, 500 or more hours.
  • a pair of monomers or multimers are selected to bind to the same target (i.e., for use in sandwich-based assays).
  • a matched monomer or multimer pair two different monomers or multimers typically are able to bind the target protein simultaneously.
  • One approach to identify such pairs involves the following:
  • monomer domains or multimers are selected against the ability to bind components other than a target in a mixture.
  • the general approach can include performing the affinity selection under conditions that closely resemble the conditions of the assay, including mimicking the composition of a sample during the assay.
  • a step of selection could include contacting a monomer domain or multimer to a mixture not including the target ligand and selecting against any monomer domains or multimers that bind to the mixture.
  • the mixtures (absent the target ligand, which could be depleted using an antibody, monomer domain or multimer) representing the sample in an assay (serum, blood, tissue, cells, urine, semen, etc) can be used as a blocking agent.
  • an assay serum, blood, tissue, cells, urine, semen, etc.
  • the invention can be used in the application for creating antagonists, where the selected monomer domains or multimers block the interaction between two proteins, e.g., the ⁇ and ⁇ chains of Met and/or between Met and HGF.
  • the invention can generate agonists.
  • multimers binding two different proteins, e.g., enzyme and substrate can enhance protein function, including, for example, enzymatic activity and/or substrate conversion.
  • the monomer domains are used for ligand inhibition, ligand clearance or ligand stimulation.
  • Possible ligands in these methods include, e.g., HGF.
  • Ligand clearance refers to modulating the half-life of a soluble ligand in, bodily fluid. For example, most monomer domains, absent a half-life extender, have a short half-life. Thus, binding of a monomer domain to the ligand will reduce the half-life of the ligand, thereby reducing ligand concentration by clearing the ligand through the kidney so long as the complex is no larger than the maximum size able to pass through the kidney (less than about 50 or 40 kD).
  • a multimer comprising a first monomer domain that binds to the ligand and a second monomer domain that binds to the receptor can be used to increase the effective affinity of the ligand for the receptor.
  • multimers comprising at least two monomers that bind to receptors are used to bring two receptors into proximity by both binding the multimer, thereby activating the receptors.
  • monomer domains, and multimers thereof that are capable of drug binding (e.g., binding radionucleotides for targeting, pharmaceutical binding for half-life extension of drugs, controlled substance binding for overdose treatment and addiction therapy), immune function modulating (e.g., immunogenicity blocking by binding such receptors as CTLA-4, immunogenicity enhancing by binding such receptors as CD80, or complement activation by Fc type binding), and specialized delivery (e.g., slow release by linker cleavage, electrotransport domains, dimerization domains, or specific binding to: cell entry domains, clearance receptors such as FcR, oral delivery receptors such as plgR for trans-mucosal transport, and blood-brain transfer receptors such as transferring).
  • drug binding e.g., binding radionucleotides for targeting, pharmaceutical binding for half-life extension of drugs, controlled substance binding for overdose treatment and addiction therapy
  • immune function modulating e.g., immunogenicity blocking by binding such receptors as CTLA-4, immunogenicity
  • Monomers or multimers of the invention that bind to Met may also be used in diagnostic and predictive applications in which is is useful to detect Met. For example, detection of Met can be used to predict prognosis of breast cancer, wherein higher abundance of Met than in a normal tissue indicates a poor prognosis. See, e.g., U.S. Pat. No. 6,673,559.
  • polypeptide of the present invention can be altered. Descriptions of a variety of diversity generating procedures for generating modified or altered nucleic acid sequences encoding these polypeptides are described herein and the references cited therein.
  • Another aspect of the present invention includes the cloning and expression of monomer domains, selected monomer domains, multimers and/or selected multimers coding nucleic acids.
  • multimer domains can be synthesized as a single protein using expression systems well known in the art.
  • General texts which describe molecular biological techniques useful herein, including the use of vectors, promoters and many other topics relevant to expressing nucleic acids such as monomer domains, selected monomer domains, multimers and/or selected multimers include Berger and Kimmel, Guide to Molecular Cloning Techniques, Methods in Enzymology volume 152 Academic Press, Inc., San Diego, Calif. (Berger); Sambrook et al., Molecular Cloning—A Laboratory Manual (2nd Ed.), Vol.
  • the present invention also relates to the introduction of vectors of the invention into host cells, and the production of monomer domains, selected monomer domains, multimers and/or selected multimers of the invention by recombinant techniques.
  • Host cells are genetically engineered (i.e., transduced, transformed or transfected) with the vectors of this invention, which can be, for example, a cloning vector or an expression vector.
  • the vector can be, for example, in the form of a plasmid, a viral particle, a phage, etc.
  • the engineered host cells can be cultured in conventional nutrient media modified as appropriate for activating promoters, selecting transformants, or amplifying the monomer domain, selected monomer domain, multimer and/or selected multimer gene(s) of interest.
  • the culture conditions such as temperature, pH and the like, are those previously used with the host cell selected for expression, and will be apparent to those skilled in the art and in the references cited herein, including, e.g., Freshney (1994) Culture of Animal Cells, a Manual of Basic Technique , third edition, Wiley-Liss, New York and the references cited therein.
  • polypeptides of the invention can also be produced in non-animal cells such as plants, yeast, fungi, bacteria and the like. Indeed, as noted throughout, phage display is an especially relevant technique for producing such polypeptides. In addition to Sambrook, Berger and Ausubel, details regarding cell culture can be found in Payne et al. (1992) Plant Cell and Tissue Culture in Liquid Systems John Wiley & Sons, Inc.
  • the present invention also includes alterations of monomer domains, immuno-domains and/or multimers to improve pharmacological properties, to reduce immunogenicity, or to facilitate the transport of the multimer and/or monomer domain into a cell or tissue (e.g., through the blood-brain barrier, or through the skin).
  • These types of alterations include a variety of modifications (e.g., the addition of sugar-groups or glycosylation), the addition of PEG, the addition of protein domains that bind a certain protein (e.g., HSA or other serum protein), the addition of proteins fragments or sequences that signal movement or transport into, out of and through a cell.
  • Additional components can also be added to a multimer and/or monomer domain to manipulate the properties of the multimer and/or monomer domain.
  • a variety of components can also be added including, e.g., a domain that binds a known receptor (e.g., a Fc-region protein domain that binds a Fc receptor), a toxin(s) or part of a toxin, a prodomain that can be optionally cleaved off to activate the multimer or monomer domain, a reporter molecule (e.g., green fluorescent protein), a component that bind a reporter molecule (such as a radionuclide for radiotherapy, biotin or avidin) or a combination of modifications.
  • a domain that binds a known receptor e.g., a Fc-region protein domain that binds a Fc receptor
  • a prodomain that can be optionally cleaved off to activate the multimer or monomer domain
  • a reporter molecule e.g
  • Another aspect of the invention is the development of specific non-human animal models in which to test the immunogenicity of the monomer or multimer domains.
  • the method of producing such non-human animal model comprises: introducing into at least some cells of a recipient non-human animal, vectors comprising genes encoding a plurality of human proteins from the same family of proteins, wherein the genes are each operably linked to a promoter that is functional in at least some of the cells into which the vectors are introduced such that a genetically modified non-human animal is obtained that can express the plurality of human proteins from the same family of proteins.
  • Suitable non-human animals employed in the practice of the present invention include all vertebrate animals, except humans (e.g., mouse, rat, rabbit, sheep, and the like).
  • the plurality of members of a family of proteins includes at least two members of that family, and usually at least ten family members.
  • the plurality includes all known members of the family of proteins.
  • Exemplary genes that can be used include those encoding monomer domains, such as, for example, members of the LDL receptor class A-domain family, the EGF-like domain family, as well as the other domain families described herein.
  • the non-human animal models of the present invention can be used to screen for immunogenicity of a monomer or multimer domain that is derived from the same family of proteins expressed by the non-human animal model.
  • the present invention includes the non-human animal model made in accordance with the method described above, as well as transgenic non-human animals whose somatic and germ cells contain and express DNA molecules encoding a plurality of human proteins from the same family of proteins (such as the monomer domains described herein), wherein the DNA molecules have been introduced into the transgenic non-human animal at an embryonic stage, and wherein the DNA molecules are each operably linked to a promoter in at least some of the cells in which the DNA molecules have been introduced.
  • mice model useful for screening LDL receptor class A-domain derived binding proteins is described as follows. Gene clusters encoding the wild type human LDL receptor class A-domain monomers are amplified from human cells using PCR. Almost all of the 200 different A-domains can be amplified with only three separate PCR amplification reactions of about 7 kb each. These fragments are then used to generate transgenic mice according to the method described above. The transgenic mice will recognize the human A-domains as “self”, thus mimicking the “selfness” of a human with regard to A-domains. Individual A-domain-derived monomers or multimers are tested in these mice by injecting the A-domain-derived monomers or multimers into the mice, then analyzing the immune response (or lack of response) generated. The mice are tested to determine if they have developed a mouse anti-human response (MAHR). Monomers and multimers that do not result in the generation of a MAHR are likely to be non-immunogenic when administered to humans.
  • MAHR mouse anti-human response
  • MAHR test in transgenic mice is used to test individual proteins in mice that are transgenic for that single protein.
  • the above described method provides a non-human animal model that recognizes an entire family of human proteins as “self,” and that can be used to evaluate a huge number of variant proteins that each are capable of vastly varied binding activities and uses.
  • Kits comprising the components needed in the methods (typically in an unmixed form) and kit components (packaging materials, instructions for using the components and/or the methods, one or more containers (reaction tubes, columns, etc.)) for holding the components are a feature of the present invention.
  • Kits of the present invention may contain a multimer library, or a single type of monomer or multimer. Kits can also include reagents suitable for promoting target molecule binding, such as buffers or reagents that facilitate detection, including detectably-labeled molecules. Standards for calibrating a ligand binding to a monomer domain or the like, can also be included in the kits of the invention.
  • the present invention also provides commercially valuable binding assays and kits to practice the assays.
  • one or more ligand is employed to detect binding of a monomer domain, immuno-domains and/or multimer.
  • Such assays are based on any known method in the art, e.g., flow cytometry, fluorescent microscopy, plasmon resonance, and the like, to detect binding of a ligand(s) to the monomer domain and/or multimer.
  • kits based on the assay are also provided.
  • the kits typically include a container, and one or more ligand.
  • the kits optionally comprise directions for performing the assays, additional detection reagents, buffers, or instructions for the use of any of these components, or the like.
  • kits can include cells, vectors, (e.g., expression vectors, secretion vectors comprising a polypeptide of the invention), for the expression of a monomer domain and/or a multimer of the invention.
  • the present invention provides for the use of any composition, monomer domain, immuno-domain, multimer, cell, cell culture, apparatus, apparatus component or kit herein, for the practice of any method or assay herein, and/or for the use of any apparatus or kit to practice any assay or method herein and/or for the use of cells, cell cultures, compositions or other features herein as a therapeutic formulation.
  • manufacture of all components herein as therapeutic formulations for the treatments described herein is also provided.
  • the present invention provides computers, computer readable media and integrated systems comprising character strings corresponding to monomer domains, selected monomer domains, multimers and/or selected multimers and nucleic acids encoding such polypeptides. These sequences can be manipulated by in silico recombination methods, or by standard sequence alignment or word processing software.
  • BLAST is described in Altschul et al., (1990) J. Mol. Biol. 215:403-410.
  • Software for performing BLAST analyses is publicly available through the National Center for Biotechnology Information (available on the World Wide Web at ncbi.nlm.nih.gov).
  • HSPs high scoring sequence pairs
  • T some positive-valued threshold score
  • the word hits are then extended in both directions along each sequence for as far as the cumulative alignment score can be increased. Cumulative scores are calculated using, for nucleotide sequences, the parameters M (reward score for a pair of matching residues; always >0) and N (penalty score for mismatching residues; always ⁇ 0). For amino acid sequences, a scoring matrix is used to calculate the cumulative score. Extension of the word hits in each direction are halted when: the cumulative alignment score falls off by the quantity X from its maximum achieved value; the cumulative score goes to zero or below, due to the accumulation of one or more negative-scoring residue alignments; or the end of either sequence is reached.
  • the BLAST algorithm parameters W, T, and X determine the sensitivity and speed of the alignment.
  • W wordlength
  • E expectation
  • BLOSUM62 scoring matrix see Henikoff & Henikoff (1989) Proc. Natl. Acad. Sci. USA 89:10915.
  • PILEUP creates a multiple sequence alignment from a group of related sequences using progressive, pairwise alignments. It can also plot a tree showing the clustering relationships used to create the alignment. PILEUP uses a simplification of the progressive alignment method of Feng & Doolittle, (1987) J. Mol. Evol. 35:351-360. The method used is similar to the method described by Higgins & Sharp, (1989) CABIOS 5:151-153. The program can align, e.g., up to 300 sequences of a maximum length of 5,000 letters. The multiple alignment procedure begins with the pairwise alignment of the two most similar sequences, producing a cluster of two aligned sequences.
  • This cluster can then be aligned to the next most related sequence or cluster of aligned sequences.
  • Two clusters of sequences can be aligned by a simple extension of the pairwise alignment of two individual sequences. The final alignment is achieved by a series of progressive, pairwise alignments.
  • the program can also be used to plot a dendogram or tree representation of clustering relationships. The program is run by designating specific sequences and their amino acid or nucleotide coordinates for regions of sequence comparison. For example, in order to determine conserved amino acids in a monomer domain family or to compare the sequences of monomer domains in a family, the sequence of the invention, or coding nucleic acids, are aligned to provide structure-function information.
  • the computer system is used to perform “in silico” sequence recombination or shuffling of character strings corresponding to the monomer domains.
  • Methods for Making Character Strings, Polynucleotides & Polypeptides Having Desired Characteristics are set forth in “Methods For Making Character Strings, Polynucleotides & Polypeptides Having Desired Characteristics” by Selifonov and Stemmer, filed Feb. 5, 1999 (U.S. Ser. No. 60/118,854) and “Methods For Making Character Strings, Polynucleotides & Polypeptides Having Desired Characteristics” by Selifonov and Stemmer, filed Oct. 12, 1999 (U.S. Ser. No. 09/416,375).
  • genetic operators are used in genetic algorithms to change given sequences, e.g., by mimicking genetic events such as mutation, recombination, death and the like.
  • Multi-dimensional analysis to optimize sequences can be also be performed in the computer system, e.g., as described in the '375 application.
  • a digital system can also instruct an oligonucleotide synthesizer to synthesize oligonucleotides, e.g., used for gene reconstruction or recombination, or to order oligonucleotides from commercial sources (e.g., by printing appropriate order forms or by linking to an order form on the Internet).
  • an oligonucleotide synthesizer to synthesize oligonucleotides, e.g., used for gene reconstruction or recombination, or to order oligonucleotides from commercial sources (e.g., by printing appropriate order forms or by linking to an order form on the Internet).
  • the digital system can also include output elements for controlling nucleic acid synthesis (e.g., based upon a sequence or an alignment of a recombinant, e.g., recombined, monomer domain as herein), i.e., an integrated system of the invention optionally includes an oligonucleotide synthesizer or an oligonucleotide synthesis controller.
  • the system can include other operations that occur downstream from an alignment or other operation performed using a character string corresponding to a sequence herein, e.g., as noted above with reference to assays.
  • This example describes selection of monomer domains and the creation of multimers.
  • Starting materials for identifying monomer domains and creating multimers from the selected monomer domains and procedures can be derived from any of a variety of human and/or non-human sequences.
  • one or more monomer domain gene(s) are selected from a family of monomer domains that bind to a certain ligand.
  • the nucleic acid sequences encoding the one or more monomer domain gene can be obtained by PCR amplification of genomic DNA or cDNA, or optionally, can be produced synthetically using overlapping oligonucleotides.
  • sequences are then cloned into a cell surface display format (i.e., bacterial, yeast, or mammalian (COS) cell surface display; phage display) for expression and screening.
  • a cell surface display format i.e., bacterial, yeast, or mammalian (COS) cell surface display; phage display
  • the recombinant sequences are transfected (transduced or transformed) into the appropriate host cell where they are expressed and displayed on the cell surface.
  • the cells can be stained with a labeled (e.g., fluorescently labeled), desired ligand.
  • the stained cells are sorted by flow cytometry, and the selected monomer domains encoding genes are recovered (e.g., by plasmid isolation, PCR or expansion and cloning) from the positive cells.
  • the selected monomer domain encoding genes recovered from the desired ligand or mixture of ligands binding cells can be optionally recombined according to any of the methods described herein or in the cited references.
  • the recombinant sequences produced in this round of diversification are then screened by the same or a different method to identify recombinant genes with improved affinity for the desired or target ligand.
  • the diversification and selection process is optionally repeated until a desired affinity is obtained.
  • the selected monomer domain nucleic acids selected by the methods can be joined together via a linker sequence to create multimers, e.g., by the combinatorial assembly of nucleic acid sequences encoding selected monomer domains by DNA ligation, or optionally, PCR-based, self-priming overlap reactions.
  • the nucleic acid sequences encoding the multimers are then cloned into a cell surface display format (i.e., bacterial, yeast, or mammalian (COS) cell surface display; phage display) for expression and screening.
  • COS mammalian
  • the cells can be stained with a labeled, e.g., fluorescently labeled, desired ligand or mixture of ligands.
  • the stained cells are sorted by flow cytometry, and the selected multimers encoding genes are recovered (e.g., by PCR or expansion and cloning) from the positive cells.
  • Positive cells include multimers with an improved avidity or affinity or altered specificity to the desired ligand or mixture of ligands compared to the selected monomer domain(s).
  • the process of staining and sorting can be repeated multiple times (e.g., using progressively decreasing concentrations of the desired ligand or mixture of ligands until a desired level of enrichment is obtained).
  • any screening or detection method known in the art that can be used to identify cells that bind the desired ligand or mixture of ligands can be employed.
  • the selected multimer encoding genes recovered from the desired ligand or mixture of ligands binding cells can be optionally recombined according to any of the methods described herein or in the cited references.
  • the recombinant sequences produced in this round of diversification are then screened by the same or a different method to identify recombinant genes with improved avidity or affinity or altered specificity for the desired or target ligand.
  • the diversification and selection process is optionally repeated until a desired avidity or affinity or altered specificity is obtained.
  • This example describes the selection of monomer domains that are capable of binding to Human Serum Albumin (HSA).
  • HSA Human Serum Albumin
  • E. coli DH10B cells (Invitrogen) were transformed with phage vectors encoding a library of LDL receptor class A-domain variants as a fusions to the pIII phage protein.
  • the electroporation system MicroPulser Bio-Rad
  • the DNA solution was mixed with 100 ⁇ l of the cell suspension, incubated on ice and transferred into the cuvette (electrode gap 1 mm).
  • 160 ml of the culture containing the cells which were transformed with the phage vectors encoding the library of the A-domain variant phages, were grown for 24 h at 22 C, 250 rpm and afterwards transferred in sterile centrifuge tubes. The cells were sedimented by centrifugation (15 minutes, 5000 g, 4° C.). The supernatant containing the phage particles was mixed with 1/5 volumes 20% w/v PEG 8000, 15% w/v NaCl, and was incubated for several hours at 4° C.
  • the precipitated phage particles were dissolved in 2 ml of cold TBS (50 mM Tris, 100 mM NaCl, pH 8.0) containing 2 mM CaCl 2 .
  • the solution was incubated on ice for 30 minutes and was distributed into two 1.5 ml reaction vessels.
  • the supernatants were transferred to a new reaction vessel. Phage were reprecipitated by adding 1/5 volumes 20% w/v PEG 8000, 15% w/v NaCl and incubation for 60 minutes on ice.
  • the precipitated phage particles were dissolved in a total of 1 ml TBS containing 2 mM CaCl 2 . After incubation for 30 minutes on ice the solution was centrifuged as described above. The supernatant containing the phage particles was used directly for the affinity enrichment.
  • phage solution containing approximately 5 ⁇ 10 11 phage particles
  • 80 ⁇ l TBST containing 3% BSA and 2 mM CaCl 2 for 1 hour at RT.
  • the wells were washed 5 times for 1 min using 130 ⁇ l TBST containing 2 mM CaCl 2 .
  • Phage bound to the well surface were eluted either by incubation for 15 minutes with 130 ⁇ l 0.1 M glycine/HCl pH 2.2 or in a competitive manner by adding 130 ⁇ l of 500 ⁇ g/ml HSA in TBS.
  • the pH of the elution fraction was immediately neutralized after removal from the well by mixing the eluate with 30 ⁇ l 1 M Tris/HCl pH 8.0.
  • the eluate was used to infect E. coli K91BluKan cells (F + ).
  • 50 ⁇ l of the eluted phage solution were mixed with 50 ⁇ l of a preparation of cells and incubated for 10 minutes at RT.
  • 20 ml LB medium containing 20 ⁇ g/ml tetracycline were added and the infected cells were grown for 36 h at 22 C, 250 rpm.
  • the cells were sedimented (10 minutes, 5000 g, 4° C.). Phage were recovered from the supernatant by precipitation as described above.
  • For the repeated affinity enrichment of phage particles the same procedure as described in this example was used. After two subsequent rounds of panning against HSA, random colonies were picked and tested for their binding properties against the used target protein.
  • This example describes the determination of biological activity of monomer domains that are capable of binding to HSA.
  • an A-domain which was evolved for binding HSA (see Example 1) was fused on the genetic level with a streptavidin binding A-domain multimer using standard molecular biology methods (see Maniatis et al.).
  • the resulting genetic construct coding for an A-domain multimer as well as a hexahistidine tag and a HA tag, were used to produce protein in E. coli .
  • affinity tag mediated purification the proteins were dialysed several times against 150 mM NaCl, 5 mM Tris pH 8.0, 100 ⁇ M CaCl 2 and sterile filtered (0.45 ⁇ M).
  • Two sets of animal experiments were performed. In a first set, 1 ml of each prepared protein solution with a concentration of 2.5 ⁇ M were injected into the tail vein of separate mice and serum samples were taken 2, 5 and 10 minutes after injection. In a second set, the protein solution described before was supplemented with 50 mg/ml human serum albumin. As described above, 1 ml of each solution was injected per animal. In case of the injected streptavidin binding A-domain dimer, serum samples were taken 2, 5 and 10 minutes after injection, while in case of the trimer, serum samples were taken after 10, 30 and 120 minutes. All experiments were performed as duplicates and individual animals were assayed per time point.
  • an enzyme linked immunosorbent assay was performed. Therefore, wells of a maxisorp 96 well microtiter plate (NUNC, Denmark) were coated with each 1 ⁇ g anti-His 6 -antibody in TBS containing 2 mM CaCl 2 for 1 h at 4 C. After blocking remaining binding sites with casein (Sigma) solution for 1 h, wells were washed three times with TBS containing 0.1% Tween and 2 mM CaCl 2 . Serial concentration dilutions of the serum samples were prepared and incubated in the wells for 2 h in order to capture the a-domain proteins.
  • ELISA enzyme linked immunosorbent assay
  • HRP horse radish peroxidase
  • This example describes experiments demonstrating extension of half-life of proteins in blood.
  • Anti-6 ⁇ His antibody was immobilized by hydrophobic interaction to a 96-well plate (Nunc). Serial dilutions of serum from each blood sample were incubated with the immobilized antibody for 3 hours. Plates were washed to remove unbound protein and probed with ⁇ -HA-HRP to detect monomer.
  • Monomers identified as having long half-lives in dialysis experiments were constructed to contain either an HA, FLAG, E-Tag, or myc epitope tag. Four monomers were pooled, containing one protein for each tag, to make two pools.
  • One monkey was injected subcutaneously per pool, at a dose of 0.25 mg/kg/monomer in 2.5 mL total volume in saline. Blood samples were drawn at 24, 48, 96, and 120 hours. Anti-6 ⁇ His antibody was immobilized by hydrophobic interaction to a 96-well plate (Nunc). Serial dilutions of serum from each blood sample were incubated with the immobilized antibody for 3 hours. Plates were washed to remove unbound protein and separately probed with ⁇ -HA-HRP, ⁇ -FLAG-HRP, ⁇ -ETag-HRP, and ⁇ -myc-HRP to detect the monomer.
  • This example describes in vivo intra-protein recombination to generate libraries of greater diversity.
  • a monomer-encoding plasmid vector (pCK-derived vector; see below), flanked by orthologous loxP sites, was recombined in a Cre-dependent manner with a phage vector via its compatible loxP sites.
  • the recombinant phage vectors were detected by PCR using primers specific for the recombinant construct. DNA sequencing indicated that the correct recombinant product was generated.
  • pCK-cre-lox-Mb-loxP This vector has two particularly relevant features. First, it carries the cre gene, encoding the site-specific DNA recombinase Cre, under the control of P lac . Cre was PCR-amplified from p705-cre (from GeneBridges) with cre-specific primers that incorporated XbaI (5′) and SfiI (3′) at the ends of the PCR product. This product was digested with XbaI and SfiI and cloned into the identical sites of pCK, a bla ⁇ , Cm R derivative of pCK110919-HC-Bla (pACYC ori), yielding pCK-cre.
  • Cre was PCR-amplified from p705-cre (from GeneBridges) with cre-specific primers that incorporated XbaI (5′) and SfiI (3′) at the ends of the PCR product. This product was digested with XbaI and S
  • the second feature is the na ⁇ ve A domain library flanked by two orthologous loxP sites, loxP(wild-type) and loxP(FAS), which are required for the site-specific DNA recombination catalyzed by Cre. See, e.g., Siegel, R. W., et al. FEBS Letters 505:467-473 (2001). These sites rarely recombine with another. loxP sites were built into pCK-cre sequentially.
  • the resulting plasmid was digested with EcoRI and SphI and ligated to the hybridized, 5′-phosphorylated oligos loxP(L) and loxP (L_rc), which carry loxP(FAS) and EcoRI and SphI-compatible overhangs.
  • a large-scale purification Qiagen MAXI prep
  • Qiagen MAXI prep Qiagen MAXI prep
  • the Qiagen-purified plasmid was subjected to CsC1 gradient centrifugation for further purification.
  • This construct was then digested with SphI and BglII and ligated to digested na ⁇ ve A domain library insert, which was obtained via a PCR-amplification of a preexisting A domain library pool.
  • the loxP sites and Mb are in-frame, which generates Mbs with loxP-encoded linkers.
  • This library was utilized in the in vivo recombination procedure as detailed below.
  • fUSE5HA-Mb-lox-lox vector The vector is a derivative of fUSE5 from George Smith's laboratory (University of Missouri). It was subsequently modified to carry an HA tag for immunodetection assays. loxP sites were built into fUSE5HA sequentially. 5′phosphorylated oligonucleotides loxP(I) and loxP(I)_rc, carrying loxP(WT), a string of stop codons and XmaI and SfiI-compatible overhangs, were hybridized together and ligated to XmaI- and SfiI-digested fUSE5HA in a standard ligation reaction (New England Biolabs T4 ligase; overnight at 16 C).
  • the resulting phage vector was next digested with XmaI and SphI and ligated to the hybridized oligos loxP(J) and loxP(J)_rc, which carry loxP(FAS) and overhangs compatible with XmaI and SphI.
  • This construct was digested with XmaI/SfiI and then ligated to pre-cut (XmaI/SfiI) na ⁇ ve A domain library insert (PCR product).
  • the stop codons are located between the loxP sites, preventing expression of gIII and consequently, the production of infectious phage.
  • the ligated vector/library was subsequently transformed into an E. coli host bearing a gIII-expressing plasmid that allows the rescue of the fUSE5HA-Mb-lox-lox phage, as detailed below.
  • pCK-gIII This plasmid carries gIII under the control of its native promoter. It was constructed by PCR-amplifying gIII and its promoter from VCSM13 helper phage (Stratagene) with primers gIIIPromoter_EcoRI and gIIIPromoter_HinDIII. This product was digested with EcoRI and HinDIII and cloned into the same sites of pCK110919-HC-Bla. As gIII is under the control of its own promoter, gIII expression is presumably constitutive. pCK-gIII was transformed into E. coli EC100 (Epicentre).
  • the procedure involves the following key steps: a) Production of infective (i.e. rescue) of fUSE5HA-Mb-lox-lox library with an E. coli host expressing gIII from a plasmid; b) Cloning of 2 nd library (pCK) and transformation into F + TG1 E. coli ; c) Infection of the culture carrying the 2 nd library with the rescued fUSE5HA-Mb-lox-lox phage library.
  • Electrocompetent cells carrying pCK-gIII were prepared by a standard protocol. These cells had a transformation frequency of 4 ⁇ 10 8 / ⁇ g DNA and were electroporated with large-scale ligations ( ⁇ 5 ⁇ g vector DNA) of fUSE5HA-lox-lox vector and the na ⁇ ve A domain library insert. After individual electroporations (100 ng DNA/electroporation) with ⁇ 70 ⁇ L cells/cuvette, 930 ⁇ L warm SOC media were added, and the cells were allowed to recover with shaking at 37 C for 1 hour. Next, tetracycline was added to a final concentration of 0.2 ⁇ g/mL, and the cells were shaken for ⁇ 45 minutes at 37 C.
  • E. coli Infection of the culture carrying the 2 nd library with the rescued fUSE5HA-Mb-lox-lox phage library. To maximize the generation of recombinants, a high infection rate (>50%) of E. coli within a culture is desirable. The infectivity of E. coli depends on a number of factors, including the expression of the F pilus and growth conditions. E. coli backgrounds TG1 (carrying an F') and K91 (an Hfr strain) were hosts for the recombination system.
  • Oligonucleotides loxP(K) [P-5′ agcttataacttcgacigaaaggtatatacgaagttatagatctcgtgctgcatgcggtgcg] loxP(K_rc) [P-5′ aattcgcaccgcatgcagcacgagatctataacttcgtatatacctttctatacgaagttataagct] loxP(L) [P-5′ ataacttcgtatagcatacattatacgaagttatcgag] loxP(L_rc) [P-5′ ctcgataacttcgtataatgtatgctatacgaagttatg] loxP(I) [P-5′ ccgggagcagggcatgctaagtgagtaataagtgagtaataacttcgtatatacctttctatacg
  • This example describes construction of an EGF-based monomer library.
  • the CaEGF domain library, E3 encodes a protein domain of 36-43 amino acids having the following pattern:
  • the oligonucleotides used in this PCR reaction are in two groups, 1 and 2. They are: Group 1: 1. 5′-AAAAGGCCTCGAGGGCCTGGGTGGCAATGGT-3′ 2. 5′-CCTGAACCACCACAKHKACCGYKSNBGCACGGAYYCGRCRMACATTC ATYAAYATCTDYACCATTGCCACCC-3′ 3. 5′-CCTGAACCACCACAKNTGSCGYYGYKMHSGCACGGAYYCGRCRMACATTC ATYAAYATCTDYACCATTGCCACCC-3′ 4.
  • the Group 1 PCR fragments were digested with BpmI and group 2 PCR fragments were digested with BsrDI. Digestion products were purified using Qiagen Qiaquick columns and then ligated together. The ligated DNA was then amplified in a PCR using two primers. These are: 5′-AAAAGGCCTCGAGGGCCTGGGTGGCAATGGT-3′ 5′-AAAAGGCCCCAGAGGCCCCTGAACCACCACA-3′
  • the PCR products were purified with Qiagen Qiaquick columns and digested with SfiI.
  • the digested product was purified with Qiagen Qiaquick columns.
  • the DNA fragments were ligated into the SfiI restriction sites of phage display vector fuse5-HA(G4S)4, a derivative of fuse5 carrying an in-frame HA-epitope and a glycine, serine flexible linker.
  • the amino acids which comprise the flexible linker are: S-G-G-G-G-S-G-G-G-G-S-G-G-G-G-G-S-G-G-G-G-G-G-G-G-G-G-G.G.
  • the ligation mixture was electroporated into TransforMaxTM EC100TM electrocompetent E. coli cells.
  • This example describes construction of an EGF-based monomer library.
  • Another application of this process is to create multiple separate, na ⁇ ve (meaning unpanned) libraries in each of which only one of the intercysteine loops is randomized, to randomize a different loop in each library. After panning of these libraries separately against the target, the selected clones are then recombined. From each panned library only the randomized segment is amplified by PCR and multiple randomized segments are then combined into a single domain, creating a shuffled library which is panned and/or screened for increased potency. This process can also be used to shuffle a small number of clones of known sequence.
  • cysteine residues are logical places for the crossover.
  • there are other ways to determine optimal crossover sites such as computer modeling.
  • residues with highest entropy, or the least number of intramolecular contacts may also be good sites for crossovers.
  • the library was constructed by creating a library of DNA sequences, containing tyrosine codons (TAT) or variable non-conserved codons (NNK), by assembly PCR as described in Stemmer et al., Gene 164:49-53 (1995).
  • TAT tyrosine codons
  • NNK variable non-conserved codons
  • the rate of tyrosine residues was increased by including tyrosine codons in the oligonucleotides, because tyrosines were found to be overrepresented in antibody binding sites, presumably because of the large number of different contacts that tyrosine can make.
  • the oligonucleotides used in this PCR reaction are: 1. 5′-ATATCCCGGGTCTGGAGGCGTCTGGTGGTTCGTGTNNKNNKNNKNNKGAATTCCGA-3′ 2. 5′-ATATCCCGGGTCTGGAGGCGTCTGGTGGTTCGTGTNNKNNKNNKNNKNNKGAATTCCGA-3′ 3.
  • the library was constructed though an initial round of 10 cycles of PCR amplification using a mixture of 4 pools of oligonucleotides, each pool containing 400 pmols of DNA.
  • Pool 1 contained oligonucleotides 1-9
  • pool 2 contained 10-17
  • pool 3 contained only 18
  • pool 4 contained 19-27.
  • the fully assembled library was obtained through an additional 8 cycles of PCR using pool 1 and 4.
  • the library fragments were digested with XmaI and SfiI.
  • the DNA fragments were ligated into the corresponding restriction sites of phage display vector fuse5-HA, a derivative of fuse5 carrying an in-frame HA-epitope.
  • the ligation mixture was electroporated into TransforMaxTM EC100TM electrocompetent E.
  • E. coli cells resulting in a library of 2 ⁇ 10 9 individual clones.
  • Transformed E. coli cells were grown overnight at 37° C. in 2 ⁇ YT medium containing 20 ⁇ g/ml tetracycline. Phage particles were purified from the culture medium by PEG-precipitation and a titer of 1.1 ⁇ 10 13 /ml was determined. Sequences of 24 clones were determined and were consistent with the expectations of the library design.
  • This example describes optimization of multimers by optimizing monomers and/or linkers for binding to a target.
  • FIG. 20 illustrates an approach for optimizing multimer binding to targets, as exemplified with a trimeric multimer.
  • a library of monomers is panned for binding to the target (e.g., Met).
  • the target e.g., Met
  • some of the monomers may bind at locations on the target that are far away from each other, such that the domains that bind to these sites cannot be connected by a linker peptide. It is therefore useful to create and screen a large library of homo- or heterotrimers from these monomers before optimization of the monomers.
  • trimer libraries can be screened, e.g., on phage (typical for heterotrimers created from a large pool of monomers) or made and assayed separately (e.g., for homotrimers). By this method, the best trimer is identified.
  • the assays may include binding assays to a target or agonist or antagonist potency determination of the multimer in functional protein- or cell-based assays.
  • the monomeric domain(s) of the single best trimer are then optimized as a second step. Homomultimers are easiest to optimize, since only one domain sequence exists, though heteromultimers may also be synthesized. For homomultimers, an increase in binding by the multimer compared to the monomer is an avidity effect.
  • Linker libraries may be formed, e.g., from linkers with an NNK composition and/or variable sequence length.
  • the best clones (e.g., determined by potency in the inhibition or other functional assay) are converted into multimers composed of multiple (e.g., two, three, four, five, six, seven, eight, etc.) sequence-optimized domains and length- and sequence-optimized linkers.
  • This example describes a structural analysis of A domains.
  • half-domain sequences selected against different targets can be combined into a single sequence which would be able to bind simultaneously or alternatively to the selected targets.
  • creating binding sites that occupy only half a domain allows for the creation of molecules that are half as large and would have half the number of immunogenic epitopes, reducing the risk of immunogenicity.
  • This example describes screening for monomers or multimers that bind c-Met.
  • Phage libraries are panned through several rounds either on solid support (i.e. Maxisorp plates) or in solution (i.e. Dynal Streptavidin beads). The panning is repeated on each enriched library several times in order to select for a pool with the enrichment and maximal diversity.
  • A. Coating plates Six wells/library are coated with c-Met (approximately 0.5 ⁇ g/well) using 100 ⁇ L/well of 5 ⁇ g/mL pure c-Met diluted in TBS[pH 7.5]/2 mM CaCl 2 . In addition, one negative control well/library is coated with TBS[pH 7.5]/2 mM CaCl 2 only. The plates are then incubated for 1.5 hrs. at room temperature with shaking.
  • Blocking Plates Coating solution is removed and wells are washed one time with 200 ⁇ L/well of TBS[pH 7.5]/2 mM CaCl 2 . 250 ⁇ l/well of 1% BSA (protease free) in TBS[pH 7.5]/2 mM CaCl 2 is added and incubated for 1 hr. at room temperature with shaking.
  • Alternative reagents can be used to block if BSA contains proteins closely related to the target of interest (for example reagents such as casein or milk).
  • Blocking e Beads Coating solution is removed and beads are washed twice with TBS [pH 7.5]/2 mM CaCl 2 ; add 500 ⁇ l 1% BSA (protease free) in TBS[pH 7.5]/2 mM CaCl 2 and rotated for 1 hr. at room temperature.
  • Wash Plates Wells are washed three times with 200 ⁇ L/well of TBS[pH 7.5]/2 mM CaCl 2 to remove excess target.
  • B. Wash Beads Beads are washed three times with 1000 ⁇ L of TBS[pH 7.5]/2 mM CaCl 2 to remove excess target. Beads are allowed to collect on a magnet for a few minutes after each wash to avoid bead loss.
  • Phaze addition to Plates About 1000 library equivalents are added in 1% nonfat dry milk/0.2% BSA (protease free) (or appropriate blocking agent) in TBS [pH 7.5]/2 mM CaCl 2 and incubated at room temperature for 2 hours with shaking. In rounds 2-3, 100 ⁇ L total of harvested phage is added to 7 wells (6 target+1 negative control) diluted in phage addition buffer.
  • Phage addition to Beads About 1000 library equivalents are added in 500 ⁇ l 1% non-fat dry milk+100 ⁇ l 1% BSA (protease free) in TBS [pH 7.5]/2 mM CaCl 2 and incubated with rotation at room temperature for 2 hr. In rounds 2-3, 100 ⁇ L total of harvested phage are added to beads.
  • Washing Plates The plates are washed eight times with 200 ⁇ l/well of TBS [pH 7.5]/2 mM CaCl 2 /0.1% Tween-20 over a period of 10 minutes.
  • Phage are incubated with 100 ⁇ L/well of 50 ⁇ g/mL or 5 ⁇ g/well (10 ⁇ over target) of c-Met competitor in TBS [pH 7.5]/2 mM CaCl 2 for 1 hr. at room temperature, with shaking.
  • the eluted phage is mixed with 5 mL of BlueKan K91 culture and incubated at 37° C. for 25 minutes without shaking, covering sterile conical tubes with AirPore tape in order to allow for aeration.
  • Tetracycline is added to a final concentration of 0.2 ⁇ g/mL and shaken for 15 minutes at 37° C.
  • Infected cultures are diluted ⁇ 10-fold into 50 mL 2 ⁇ YT/20 ⁇ g/mL tetracycline and incubated with shaking at 30° C. overnight to saturation.
  • Phage are incubated at 37 C for 25 minutes without shaking, with the plates covered with AirPore tape to allow for aeration.
  • Tetracycline is added to a final concentration of 0.2 ⁇ g/mL and the plate is shaen for 15 minutes at 37° C.
  • a standard PEG/NaCl precipitation protocol is performed by adding 1/5 volume of a 20% PEG/15% NaCl stock to culture supernatant; mixing and incubating on ice for 45 minutes ⁇ 1 hour.
  • the culture is then centrifuged at 7000 rpm for 40 minutes to pellet phage and the supernatant is removed.
  • the phage pellet is resuspended in 1 mL TBS [pH 7.5]/2 mM CaCl 2 and transfered to an Eppendorf tube and centrifuged at 13 K rpm for at least 2 minutes to pellet insoluble material.
  • the supernatant is then centrifuged at 13 K rpm for at least 2 minutes, removed and purified phage is resuspended in up to 1 mL TBS [pH 7.5]/2 mM CaCl 2 and stored at 4° C.
  • the 2 nd and 3 rd round panning conditions are generally the same as round 1 described above, except the coated target (i.e., c-Met) amount is decreased 2 to 4 fold for each next round, and the plates (and beads) are washed more times for each next round of panning.
  • coated target i.e., c-Met
  • the final pool of monomer panning is optionally recombined by dividing the sequences of monomers into at least two fragments and mixing the two fragments. Two sets of oligos are used to PCR each fragment of the monomer, and then the entire monomer is fused by overlapping PCR and amplified. Once the fragments are fused together and amplified the DNAs are then cut by a restriction enzyme to allow for cloning and ligated into cut fuse5 phage vector. Recombined monomer libraries are then panned two more rounds.
  • Phage ELISAs For each output to be analyzed (typically Rounds 2, 3 and 4, if applicable), independent clones ae inoculated in 1 mL (2xYT/20 ⁇ g/mL tetracycline) cultures grown in costar 96-well polypropylene deep-well plates, leaving inoculating tips in and shaken overnight at 37° C. Cells are pelleted by centrifugation at 3600 rpm for 15 minutes. Culture supernatants are retained and ELISAs are performed as discussed below.
  • 96-well Maxisorp plate are coated with 50 ⁇ L/well of 50 ⁇ g/mL (2.5 ⁇ g/well). Streptavidin is diluted in TBS [pH 7.5]/2 mM CaCl 2 and the plate is incubated at 37° C. for 1 hr with shaking. Plates are washed three times with 200 ⁇ L/well of TBS [pH 7.5]/2 mM CaCl 2 . Wells are blocked with 200 ⁇ L/well of 1% BSA (fraction V) and the covered plate is incubated at RT for 1 hr with shaking. The plate is washed three times with TBS [pH 7.5]/2 mM CaCl 2 .
  • the 96-well Maxisorp plate is coated with 100 ⁇ L/well of 1 ⁇ g/mL (0.1 ⁇ g/well) c-Met diluted in TBS [pH 7.5]/2 mM CaCl 2 or 100 ⁇ L/well buffer only (negative control). The plate is incubated at RT for 1 hr. with shaking. Plates are washed three times with TBS [pH 7.5]/2 mM CaCl 2 . Phage is added to wells.
  • Selected phage are amplified overnight by infection of K91 cells.
  • the phage are then purified by PEG/NaCl precipitation of culture supernatants.
  • PEG/NaCl precipitation of culture supernatants To clone the monomer or multimer sequences into the expression vector, pEve, approximately 10 10 phage are amplified by 20 cycles of PCR as follows:
  • the PCR is then run on a 3% Agarose gel, the approximately 200 bp product is purified with a QIAquick column (Qiagen), digested with Sfi I (NEB), purified again with a QIAquick column and then ligated with T4 DNA Ligase (NEB) to the Sfi I digested vector, pEve.
  • the ligation is used to transform electro-competent BL21 (DE3) E. coli and plated onto 2 ⁇ YT plates containing Kanamycin at 40 ⁇ g/ml. Following growth, approximately 6000 individual clones are picked into 1 ⁇ YT/Kan and grown overnight;—included on the palte are wells that will serve as positive and negative controls.
  • Monomers may be purified in parallel in E. coli BL21DE3 (transformed with monomer-variants cloned in pETHC) in a parallel manner and subsequently purified by boiling.
  • the selected pEve/monomer clone plasmid DNAs are digested in separate reactions with BsrDI and BpmI (NEB).
  • BsrDI and BpmI The 10.1 kb BsrDI and 2.9 kb BpmI bands are isolated from 1% Agarose gels and purified with QIAquick columns.
  • the DNAs are ligated together with T4 DNA Ligase (NEB).
  • the ligation is purified with a QIAquick column and amplified as described for Phage Sub-cloning.
  • the 400 bp product is then purified, ligated and transformed as described for Phage Sub-cloning. Protein can be expressed and purified from these and screened using the same methods described above.
  • walking libraries are generated by ligating the selected monomers with the full representation of random domains (generated using the same methods used to generate the monomer libraries). These “walking libraries” are then panned and screened as described above.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Medicinal Chemistry (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Hematology (AREA)
  • Biochemistry (AREA)
  • Biomedical Technology (AREA)
  • Urology & Nephrology (AREA)
  • Cell Biology (AREA)
  • Microbiology (AREA)
  • Biotechnology (AREA)
  • Pathology (AREA)
  • General Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • Physics & Mathematics (AREA)
  • Food Science & Technology (AREA)
  • Biophysics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Genetics & Genomics (AREA)
  • Zoology (AREA)
  • Toxicology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Oncology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Hospice & Palliative Care (AREA)
  • General Chemical & Material Sciences (AREA)
  • Peptides Or Proteins (AREA)
  • Investigating Or Analysing Biological Materials (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
US10/957,351 2004-06-17 2004-09-30 c-Met kinase binding proteins Abandoned US20060008844A1 (en)

Priority Applications (11)

Application Number Priority Date Filing Date Title
US10/957,351 US20060008844A1 (en) 2004-06-17 2004-09-30 c-Met kinase binding proteins
AU2005265150A AU2005265150B2 (en) 2004-06-17 2005-06-17 C-MET kinase binding proteins
KR1020077001057A KR101238074B1 (ko) 2004-06-17 2005-06-17 c-MET 키나제 결합 단백질
US11/155,989 US7803907B2 (en) 2004-06-17 2005-06-17 c-MET kinase binding proteins
PCT/US2005/021558 WO2006009888A2 (en) 2004-06-17 2005-06-17 C-met kinase binding proteins
CA2569191A CA2569191C (en) 2004-06-17 2005-06-17 C-met kinase binding proteins
MXPA06014796A MXPA06014796A (es) 2004-06-17 2005-06-17 Proteinas de enlace cinasa c-met.
EP05787547A EP1776134A4 (de) 2004-06-17 2005-06-17 C-met-kinasebindende proteine
EP09176476A EP2151453A1 (de) 2004-06-17 2005-06-17 C-met-kinasebindende Proteine
JP2007516800A JP2008503216A (ja) 2004-06-17 2005-06-17 c−METキナーゼ結合タンパク質
US12/761,346 US7968681B2 (en) 2004-06-17 2010-04-15 c-MET kinase binding proteins

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US10/871,602 US20050089932A1 (en) 2001-04-26 2004-06-17 Novel proteins with targeted binding
US10/957,351 US20060008844A1 (en) 2004-06-17 2004-09-30 c-Met kinase binding proteins

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US10/871,602 Continuation-In-Part US20050089932A1 (en) 2001-04-26 2004-06-17 Novel proteins with targeted binding

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US11/155,989 Continuation-In-Part US7803907B2 (en) 2004-06-17 2005-06-17 c-MET kinase binding proteins

Publications (1)

Publication Number Publication Date
US20060008844A1 true US20060008844A1 (en) 2006-01-12

Family

ID=35785714

Family Applications (3)

Application Number Title Priority Date Filing Date
US10/957,351 Abandoned US20060008844A1 (en) 2004-06-17 2004-09-30 c-Met kinase binding proteins
US11/155,989 Expired - Fee Related US7803907B2 (en) 2004-06-17 2005-06-17 c-MET kinase binding proteins
US12/761,346 Expired - Fee Related US7968681B2 (en) 2004-06-17 2010-04-15 c-MET kinase binding proteins

Family Applications After (2)

Application Number Title Priority Date Filing Date
US11/155,989 Expired - Fee Related US7803907B2 (en) 2004-06-17 2005-06-17 c-MET kinase binding proteins
US12/761,346 Expired - Fee Related US7968681B2 (en) 2004-06-17 2010-04-15 c-MET kinase binding proteins

Country Status (8)

Country Link
US (3) US20060008844A1 (de)
EP (2) EP2151453A1 (de)
JP (1) JP2008503216A (de)
KR (1) KR101238074B1 (de)
AU (1) AU2005265150B2 (de)
CA (1) CA2569191C (de)
MX (1) MXPA06014796A (de)
WO (1) WO2006009888A2 (de)

Cited By (110)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040161752A1 (en) * 2000-07-21 2004-08-19 Jarrell Kevin A. Modular vector systems
US20040175756A1 (en) * 2001-04-26 2004-09-09 Avidia Research Institute Methods for using combinatorial libraries of monomer domains
US20050048512A1 (en) * 2001-04-26 2005-03-03 Avidia Research Institute Combinatorial libraries of monomer domains
US20050053973A1 (en) * 2001-04-26 2005-03-10 Avidia Research Institute Novel proteins with targeted binding
US20050191623A1 (en) * 1999-01-05 2005-09-01 Jarrell Kevin A. Nucleic acid cloning
US20060223081A1 (en) * 2004-11-11 2006-10-05 Kevin Jarrell Ladder assembly and system for generating diversity
US20070031943A1 (en) * 1999-01-05 2007-02-08 Kevin Jarrell System for manipulating nucleic acids
US20080003611A1 (en) * 2005-07-13 2008-01-03 Avidia, Inc Heat lysis production of proteins
WO2008030611A2 (en) 2006-09-05 2008-03-13 Medarex, Inc. Antibodies to bone morphogenic proteins and receptors therefor and methods for their use
WO2008070569A2 (en) 2006-12-01 2008-06-12 Medarex, Inc. Human antibodies that bind cd22 and uses thereof
WO2008153926A2 (en) 2007-06-05 2008-12-18 Yale University Inhibitors of receptor tyrosine kinases and methods of use thereof
WO2009054863A2 (en) 2006-12-13 2009-04-30 Medarex, Inc. Human antibodies that bind cd19 and uses thereof
US20090192048A1 (en) * 2005-12-20 2009-07-30 Michael A Reeve Method of producing a multimeric capture agent for binding a ligand
JP2009291191A (ja) * 2008-05-14 2009-12-17 Metheresis Translational Research Sa Hgfrの高親和結合部位およびその拮抗剤の同定方法
US20090312192A1 (en) * 2005-12-20 2009-12-17 Reeve Michael A Method for functionalising a hydrophobic substrate
WO2010011944A2 (en) 2008-07-25 2010-01-28 Wagner Richard W Protein screeing methods
WO2010015608A1 (en) 2008-08-05 2010-02-11 Novartis Ag Compositions and methods for antibodies targeting complement protein c5
US20100105567A1 (en) * 2005-12-20 2010-04-29 Reeve Michael A Novel capture agents for binding a ligand
WO2010069913A1 (en) 2008-12-16 2010-06-24 Novartis Ag Yeast display systems
WO2010084408A2 (en) 2009-01-21 2010-07-29 Oxford Biotherapeutics Ltd. Pta089 protein
US20100216663A1 (en) * 2001-04-26 2010-08-26 Amgen Mountain View Inc. Novel proteins with targeted binding
WO2010102175A1 (en) 2009-03-05 2010-09-10 Medarex, Inc. Fully human antibodies specific to cadm1
WO2010125003A1 (en) 2009-04-27 2010-11-04 Novartis Ag Compositions and methods for increasing muscle growth
WO2011018421A1 (en) 2009-08-10 2011-02-17 Morphosys Ag Novel screening strategies for the identification of binders
WO2011047083A1 (en) 2009-10-13 2011-04-21 Oxford Biotherapeutics Ltd. Antibodies against epha10
WO2011054007A1 (en) 2009-11-02 2011-05-05 Oxford Biotherapeutics Ltd. Ror1 as therapeutic and diagnostic target
WO2011054893A2 (en) 2009-11-05 2011-05-12 Novartis Ag Biomarkers predictive of progression of fibrosis
WO2011080050A2 (en) 2009-12-11 2011-07-07 Novartis Ag Binding molecules
WO2011098449A1 (en) 2010-02-10 2011-08-18 Novartis Ag Methods and compounds for muscle growth
WO2011138392A1 (en) 2010-05-06 2011-11-10 Novartis Ag Compositions and methods of use for therapeutic low density lipoprotein -related protein 6 (lrp6) antibodies
WO2011138391A1 (en) 2010-05-06 2011-11-10 Novartis Ag Compositions and methods of use for therapeutic low density lipoprotein - related protein 6 (lrp6) multivalent antibodies
WO2012022814A1 (en) 2010-08-20 2012-02-23 Novartis Ag Antibodies for epidermal growth factor receptor 3 (her3)
WO2012045703A1 (en) 2010-10-05 2012-04-12 Novartis Ag Anti-il12rbeta1 antibodies and their use in treating autoimmune and inflammatory disorders
EP2474556A2 (de) 2007-03-14 2012-07-11 Novartis AG APCDD1-Inhibitoren zur Behandlung, Diagnose und Erkennung von Krebs
EP2486941A1 (de) 2006-10-02 2012-08-15 Medarex, Inc. CXCR4-bindende humane Antikörper und deren Verwendungen
WO2012172495A1 (en) 2011-06-14 2012-12-20 Novartis Ag Compositions and methods for antibodies targeting tem8
WO2013001369A2 (en) 2011-06-28 2013-01-03 Oxford Biotherapeutics Ltd. Therapeutic and diagnostic target
WO2013003625A2 (en) 2011-06-28 2013-01-03 Oxford Biotherapeutics Ltd. Antibodies
WO2013006437A1 (en) 2011-07-01 2013-01-10 Novartis Ag Method for treating metabolic disorders
WO2013010955A1 (en) 2011-07-15 2013-01-24 Morphosys Ag Antibodies that are cross-reactive for macrophage migration inhibitory factor (mif) and d-dopachrome tautomerase (d-dt)
WO2013054307A2 (en) 2011-10-14 2013-04-18 Novartis Ag Antibodies and methods for wnt pathway-related diseases
EP2586796A1 (de) 2007-10-12 2013-05-01 Novartis AG Zusammensetzungen und Verfahren zur Verwendung für Sclerostin-Antikörper
WO2013074840A1 (en) 2011-11-15 2013-05-23 Allergan, Inc. Treatment of dry age related macular degeneration
WO2013084148A2 (en) 2011-12-05 2013-06-13 Novartis Ag Antibodies for epidermal growth factor receptor 3 (her3) directed to domain ii of her3
WO2013084147A2 (en) 2011-12-05 2013-06-13 Novartis Ag Antibodies for epidermal growth factor receptor 3 (her3)
WO2013093762A1 (en) 2011-12-21 2013-06-27 Novartis Ag Compositions and methods for antibodies targeting factor p
WO2014020331A1 (en) 2012-08-01 2014-02-06 Oxford Biotherapeutics Ltd. Therapeutic and diagnostic target
WO2014037899A2 (en) 2012-09-07 2014-03-13 Novartis Ag Il-18 binding molecules
WO2014084859A1 (en) 2012-11-30 2014-06-05 Novartis Ag Molecules and methods for modulating tmem16a activities
WO2014089111A1 (en) 2012-12-05 2014-06-12 Novartis Ag Compositions and methods for antibodies targeting epo
WO2014122613A1 (en) 2013-02-08 2014-08-14 Novartis Ag Anti-il-17a antibodies and their use in treating autoimmune and inflammatory disorders
WO2014133855A1 (en) 2013-02-28 2014-09-04 Caprion Proteomics Inc. Tuberculosis biomarkers and uses thereof
WO2014141064A1 (en) 2013-03-13 2014-09-18 Novartis Ag Notch2 binding molecules for treating respiratory diseases
WO2014159239A2 (en) 2013-03-14 2014-10-02 Novartis Ag Antibodies against notch 3
WO2014205302A2 (en) 2013-06-21 2014-12-24 Novartis Ag Lectin-like oxidized ldl receptor1 antibodies and methods of use
WO2014205300A2 (en) 2013-06-21 2014-12-24 Novartis Ag Lectin-like oxidized ldl receptor1 antibodies and methods of use
WO2015015401A2 (en) 2013-08-02 2015-02-05 Pfizer Inc. Anti-cxcr4 antibodies and antibody-drug conjugates
WO2015022658A2 (en) 2013-08-14 2015-02-19 Novartis Ag Methods of treating sporadic inclusion body myositis
WO2015069459A1 (en) 2013-11-05 2015-05-14 Novartis Ag Organic compounds
WO2015090229A1 (en) 2013-12-20 2015-06-25 Novartis Ag Regulatable chimeric antigen receptor
WO2015142661A1 (en) 2014-03-15 2015-09-24 Novartis Ag Regulatable chimeric antigen receptor
WO2015162590A1 (en) 2014-04-24 2015-10-29 Novartis Ag Methods of improving or accelerating physical recovery after surgery for hip fracture
WO2015198217A2 (en) 2013-02-08 2015-12-30 Novartis Ag Compositions and methods for long-acting antibodies targeting il-17
WO2016014553A1 (en) 2014-07-21 2016-01-28 Novartis Ag Sortase synthesized chimeric antigen receptors
WO2016020882A2 (en) 2014-08-07 2016-02-11 Novartis Ag Angiopoetin-like 4 (angptl4) antibodies and methods of use
WO2016020880A2 (en) 2014-08-07 2016-02-11 Novartis Ag Angiopoietin-like 4 antibodies and methods of use
WO2016044605A1 (en) 2014-09-17 2016-03-24 Beatty, Gregory Targeting cytotoxic cells with chimeric receptors for adoptive immunotherapy
WO2016057846A1 (en) 2014-10-08 2016-04-14 Novartis Ag Compositions and methods of use for augmented immune response and cancer therapy
EP3009454A2 (de) 2009-04-20 2016-04-20 Oxford Bio Therapeutics Limited Antikörper spezifisch gegen cadherin-17
WO2016098079A2 (en) 2014-12-19 2016-06-23 Novartis Ag Compositions and methods for antibodies targeting bmp6
WO2016164731A2 (en) 2015-04-08 2016-10-13 Novartis Ag Cd20 therapies, cd22 therapies, and combination therapies with a cd19 chimeric antigen receptor (car) - expressing cell
WO2016193872A2 (en) 2015-06-05 2016-12-08 Novartis Ag Antibodies targeting bone morphogenetic protein 9 (bmp9) and methods therefor
WO2016207858A1 (en) 2015-06-26 2016-12-29 Novartis Ag Factor xi antibodies and methods of use
WO2017021893A1 (en) 2015-08-03 2017-02-09 Novartis Ag Methods of treating fgf21-associated disorders
WO2017042701A1 (en) 2015-09-09 2017-03-16 Novartis Ag Thymic stromal lymphopoietin (tslp)-binding antibodies and methods of using the antibodies
WO2017103895A1 (en) 2015-12-18 2017-06-22 Novartis Ag Antibodies targeting cd32b and methods of use thereof
WO2017189724A1 (en) 2016-04-27 2017-11-02 Novartis Ag Antibodies against growth differentiation factor 15 and uses thereof
WO2017203450A1 (en) 2016-05-25 2017-11-30 Novartis Ag Reversal binding agents for anti-factor xi/xia antibodies and uses thereof
WO2017216724A1 (en) 2016-06-15 2017-12-21 Novartis Ag Methods for treating disease using inhibitors of bone morphogenetic protein 6 (bmp6)
WO2018116255A1 (en) 2016-12-23 2018-06-28 Novartis Ag Factor xi antibodies and methods of use
WO2018146594A1 (en) 2017-02-08 2018-08-16 Novartis Ag Fgf21 mimetic antibodies and uses thereof
WO2018158398A1 (en) 2017-03-02 2018-09-07 INSERM (Institut National de la Santé et de la Recherche Médicale) Antibodies having specificity to nectin-4 and uses thereof
WO2018175460A1 (en) 2017-03-24 2018-09-27 Novartis Ag Methods for preventing and treating heart disease
WO2018229715A1 (en) 2017-06-16 2018-12-20 Novartis Ag Compositions comprising anti-cd32b antibodies and methods of use thereof
WO2019003104A1 (en) 2017-06-28 2019-01-03 Novartis Ag METHOD FOR PREVENTING AND TREATING URINARY INCONTINENCE
GB201903233D0 (en) 2019-03-08 2019-04-24 Oxford Genetics Ltd Method of selecting for antibodies
WO2019081983A1 (en) 2017-10-25 2019-05-02 Novartis Ag CD32B TARGETING ANTIBODIES AND METHODS OF USE
WO2019099440A1 (en) 2017-11-14 2019-05-23 Arcellx, Inc. Multifunctional immune cell therapies
WO2019102353A1 (en) 2017-11-22 2019-05-31 Novartis Ag Reversal binding agents for anti-factor xi/xia antibodies and uses thereof
WO2019229658A1 (en) 2018-05-30 2019-12-05 Novartis Ag Entpd2 antibodies, combination therapies, and methods of using the antibodies and combination therapies
WO2020010250A2 (en) 2018-07-03 2020-01-09 Elstar Therapeutics, Inc. Anti-tcr antibody molecules and uses thereof
EP3623380A1 (de) 2013-03-15 2020-03-18 Michael C. Milone Targeting von zytotoxischen zellen mit chimären rezeptoren zur adoptiven immuntherapie
WO2020069409A1 (en) 2018-09-28 2020-04-02 Novartis Ag Cd19 chimeric antigen receptor (car) and cd22 car combination therapies
WO2020069405A1 (en) 2018-09-28 2020-04-02 Novartis Ag Cd22 chimeric antigen receptor (car) therapies
EP3660042A1 (de) 2014-07-31 2020-06-03 Novartis AG T-zellen mit teilmengenoptimierten chimären antigenrezeptoren
WO2020131935A1 (en) 2018-12-18 2020-06-25 Novartis Ag Reversal binding agents for anti-factor xi/xia antibodies and uses thereof
WO2020183133A1 (en) 2019-03-08 2020-09-17 Oxford Genetics Limited Method of selecting for antibodies
EP3736293A1 (de) 2013-02-12 2020-11-11 Boehringer Ingelheim International Gmbh Therapeutisches und diagnostisches target für krebs mit dll3-bindenden reagenzien
WO2021026310A1 (en) 2019-08-06 2021-02-11 L.E.A.F. Holdings Group Llc Processes of preparing polyglutamated antifolates and uses of their compositions
WO2021053559A1 (en) 2019-09-18 2021-03-25 Novartis Ag Entpd2 antibodies, combination therapies, and methods of using the antibodies and combination therapies
WO2021053560A1 (en) 2019-09-18 2021-03-25 Novartis Ag Combination therapy with entpd2 and cd73 antibodies
WO2021116119A1 (en) 2019-12-09 2021-06-17 INSERM (Institut National de la Santé et de la Recherche Médicale) Antibodies having specificity to her4 and uses thereof
EP3842457A1 (de) 2015-09-09 2021-06-30 Novartis AG Moleküle zur bindung von thymus-stroma-lymphopoietin (tslp) und verfahren zur verwendung der moleküle
WO2021228956A1 (en) 2020-05-12 2021-11-18 INSERM (Institut National de la Santé et de la Recherche Médicale) New method to treat cutaneous t-cell lymphomas and tfh derived lymphomas
WO2022190018A1 (en) 2021-03-09 2022-09-15 Molecular Partners Ag Novel darpin based cd123 engagers
WO2022190010A1 (en) 2021-03-09 2022-09-15 Molecular Partners Ag Novel darpin based cd33 engagers
WO2022216993A2 (en) 2021-04-08 2022-10-13 Marengo Therapeutics, Inc. Multifuntional molecules binding to tcr and uses thereof
WO2023170296A1 (en) 2022-03-11 2023-09-14 Inserm (Institut National De La Sante Et De La Recherche Medicale) Nucleic acid system to specifically reprogram b and t cells and uses thereof
WO2023217904A1 (en) 2022-05-10 2023-11-16 Institut National de la Santé et de la Recherche Médicale Syncitin-1 fusion proteins and uses thereof for cargo delivery into target cells
WO2024052503A1 (en) 2022-09-08 2024-03-14 Institut National de la Santé et de la Recherche Médicale Antibodies having specificity to ltbp2 and uses thereof

Families Citing this family (33)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060008844A1 (en) * 2004-06-17 2006-01-12 Avidia Research Institute c-Met kinase binding proteins
EP2567973B1 (de) 2005-11-28 2014-05-14 Zymogenetics, Inc. IL-21-Antagonisten
AU2007247176A1 (en) * 2006-05-05 2007-11-15 Universiteit Maastricht / Carim Peptides for use in diagnosing the presence of ruptured atherosclerotic lesions in an individual
DK2217268T3 (en) 2007-12-07 2016-08-15 Zymogenetics Inc MONOCLONAL ANTI-HUMAN IL-21 ANTIBODIES
US8937047B2 (en) 2007-12-19 2015-01-20 General Electric Company Biokinetics of fast-clearing polypeptides
US8124725B2 (en) * 2007-12-19 2012-02-28 General Electric Company PDGF-Rβ binders
US20140348819A1 (en) 2011-06-24 2014-11-27 Dana-Farber Cancer Institute, Inc. Methods of Treating Cancer
US10208125B2 (en) 2013-07-15 2019-02-19 University of Pittsburgh—of the Commonwealth System of Higher Education Anti-mucin 1 binding agents and uses thereof
KR102291465B1 (ko) * 2014-01-24 2021-08-18 삼성전자주식회사 c-Met 저해제의 효능 예측을 위한 바이오마커 TFF1
EP3237003B1 (de) 2014-12-24 2021-08-11 NexImmune, Inc Nanopartikelzusammensetzungen und verfahren für die immuntherapie
CA3003969A1 (en) 2015-11-06 2017-05-11 Orionis Biosciences Nv Bi-functional chimeric proteins and uses thereof
US20180339024A1 (en) 2015-11-19 2018-11-29 Asclepix Therapeutics, Llc Peptides with anti-angiogenic, anti-lymphangiogenic, and anti-edemic properties and nanoparticle formulations
MY196837A (en) 2015-12-11 2023-05-03 Immatics Biotechnologies Gmbh Novel peptides and combination of peptides for use in immunotherapy against various cancers
GB201521894D0 (en) 2015-12-11 2016-01-27 Immatics Biotechnologies Gmbh Novel peptides and combination of peptides for use in immunotherapy against various cancers
EP4059957A1 (de) 2016-02-05 2022-09-21 Orionis Biosciences BV Bispezifische signalisierungsmittel und verwendungen davon
WO2017153402A1 (en) 2016-03-07 2017-09-14 Vib Vzw Cd20 binding single domain antibodies
EP3455245A2 (de) 2016-05-13 2019-03-20 Orionis Biosciences NV Therapeutisches targeting von nichtzellulären strukturen
JP7105200B2 (ja) 2016-05-13 2022-07-22 オリオニス バイオサイエンシズ ビーブイ 標的突然変異体インターフェロン-ベータおよびその使用
US20190264197A1 (en) * 2016-07-27 2019-08-29 Protagonist Therapeutics, Inc. Disulfide-rich peptide libraries and methods of use thereof
CN110177563A (zh) 2016-10-04 2019-08-27 阿斯克雷佩西治疗公司 用于激活tie2信号传导的化合物和方法
US11084859B2 (en) 2016-10-24 2021-08-10 Orionis Biosciences BV Targeted mutant interferon-gamma and uses thereof
WO2018144999A1 (en) 2017-02-06 2018-08-09 Orionis Biosciences, Inc. Targeted engineered interferon and uses thereof
CA3052523A1 (en) 2017-02-06 2018-08-09 Orionis Biosciences Nv Targeted chimeric proteins and uses thereof
EP3580230A1 (de) 2017-02-07 2019-12-18 VIB vzw Auf immunzellen gerichtete bispezifische chimäre proteine und verwendungen davon
US11674959B2 (en) 2017-08-03 2023-06-13 The Johns Hopkins University Methods for identifying and preparing pharmaceutical agents for activating Tie1 and/or Tie2 receptors
US11896643B2 (en) 2018-02-05 2024-02-13 Orionis Biosciences, Inc. Fibroblast binding agents and use thereof
JP2020080784A (ja) * 2018-11-29 2020-06-04 シスメックス株式会社 融合ポリペプチド、融合ポリペプチドの製造方法、及び融合ポリペプチドをコードするdna
JP2022520632A (ja) 2019-02-15 2022-03-31 インテグラル・モレキュラー・インコーポレイテッド 共通軽鎖を含む抗体及びその使用
CN113767113A (zh) 2019-02-15 2021-12-07 因特格尔莫来库乐有限公司 密封蛋白6抗体及其用途
US20230174627A1 (en) * 2019-05-14 2023-06-08 Jeffrey W. FROUDE Antibodies to sudan and ebola virus
GB202004514D0 (en) 2020-03-27 2020-05-13 Inst De Medicina Molecular Joaeo Lobo Antunes Treatment of Immunosuppressive Cancer
WO2024008755A1 (en) 2022-07-04 2024-01-11 Vib Vzw Blood-cerebrospinal fluid barrier crossing antibodies
WO2024077118A2 (en) 2022-10-06 2024-04-11 Bicara Therapeutics Inc. Multispecific proteins and related methods

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050048512A1 (en) * 2001-04-26 2005-03-03 Avidia Research Institute Combinatorial libraries of monomer domains
US20060177831A1 (en) * 2004-06-17 2006-08-10 Avidia Research Institute c-MET kinase binding proteins

Family Cites Families (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4683202A (en) 1985-03-28 1987-07-28 Cetus Corporation Process for amplifying nucleic acid sequences
US6673559B1 (en) 1989-12-27 2004-01-06 The Government Of The United States Of America As Represented By The Department Of Health And Human Services Met proto-oncogene and a method for predicting breast cancer progression
US5427908A (en) 1990-05-01 1995-06-27 Affymax Technologies N.V. Recombinant library screening methods
DE69128362T2 (de) 1990-06-01 1998-05-20 Chiron Corp Zusammensetzungen und verfahren zur identifizierung von molekülen mit biologischer wirksamkeit
US5723286A (en) 1990-06-20 1998-03-03 Affymax Technologies N.V. Peptide library and screening systems
US5843701A (en) 1990-08-02 1998-12-01 Nexstar Pharmaceticals, Inc. Systematic polypeptide evolution by reverse translation
US5270170A (en) 1991-10-16 1993-12-14 Affymax Technologies N.V. Peptide library and screening method
US5426039A (en) 1993-09-08 1995-06-20 Bio-Rad Laboratories, Inc. Direct molecular cloning of primer extended DNA containing an alkane diol
US5605793A (en) 1994-02-17 1997-02-25 Affymax Technologies N.V. Methods for in vitro recombination
RU2233878C2 (ru) 1997-01-21 2004-08-10 Дзе Дженерал Хоспитал Корпорейшн Способ отбора желательного белка и нуклеиновой кислоты, средства для его осуществления
DE19742706B4 (de) 1997-09-26 2013-07-25 Pieris Proteolab Ag Lipocalinmuteine
US6818418B1 (en) 1998-12-10 2004-11-16 Compound Therapeutics, Inc. Protein scaffolds for antibody mimics and other binding proteins
US6436675B1 (en) 1999-09-28 2002-08-20 Maxygen, Inc. Use of codon-varied oligonucleotide synthesis for synthetic shuffling
US6703491B1 (en) * 1999-03-17 2004-03-09 Exelixis, Inc. Drosophila sequences
EP1163339A1 (de) 1999-04-01 2001-12-19 Innogenetics N.V. Eine polypeptidstruktur zur verwendung als gerüst
WO2002002593A2 (en) * 2000-06-29 2002-01-10 North Shore-Long Island Jewish Health System Modulators of cellular proliferation and angiogenesis, methods for use and identification thereof
EP1332212A2 (de) * 2000-08-25 2003-08-06 Curagen Corporation Proteine und dafür kodierende nukleinsäuren
WO2002068649A2 (en) * 2001-01-31 2002-09-06 Curagen Corporation Proteins and nucleic acids encoding same
DK2128246T3 (da) * 2001-04-19 2014-05-12 Univ California Fremgangsmåder og sammensætninger til fremstilling af ortogonale tRNA-aminoacyl-tRNA-syntetasepar.
US20050053973A1 (en) * 2001-04-26 2005-03-10 Avidia Research Institute Novel proteins with targeted binding
US20040175756A1 (en) * 2001-04-26 2004-09-09 Avidia Research Institute Methods for using combinatorial libraries of monomer domains
US20030157561A1 (en) * 2001-11-19 2003-08-21 Kolkman Joost A. Combinatorial libraries of monomer domains
US20050089932A1 (en) * 2001-04-26 2005-04-28 Avidia Research Institute Novel proteins with targeted binding
US20030082630A1 (en) * 2001-04-26 2003-05-01 Maxygen, Inc. Combinatorial libraries of monomer domains
US6867005B2 (en) * 2001-10-24 2005-03-15 Beckman Coulter, Inc. Method and apparatus for increasing the dynamic range and accuracy of binding assays
ES2557286T3 (es) 2003-03-03 2016-01-25 Dyax Corp. Usos de péptidos que se unen específicamente al receptor del HGF (cMet)
AU2004284090A1 (en) * 2003-10-24 2005-05-06 Avidia, Inc. LDL receptor class A and EGF domain monomers and multimers
US11624505B2 (en) 2020-03-17 2023-04-11 Weber-Stephen Products Llc Ignition-based protocols for pellet grills

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050048512A1 (en) * 2001-04-26 2005-03-03 Avidia Research Institute Combinatorial libraries of monomer domains
US20060177831A1 (en) * 2004-06-17 2006-08-10 Avidia Research Institute c-MET kinase binding proteins

Cited By (143)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050191623A1 (en) * 1999-01-05 2005-09-01 Jarrell Kevin A. Nucleic acid cloning
US20070031943A1 (en) * 1999-01-05 2007-02-08 Kevin Jarrell System for manipulating nucleic acids
US8129119B2 (en) 1999-01-05 2012-03-06 Modular Genetics, Inc. System for manipulating nucleic acids
US7439021B2 (en) 1999-01-05 2008-10-21 Trustees Of Boston University Nucleic acid cloning
US20040161752A1 (en) * 2000-07-21 2004-08-19 Jarrell Kevin A. Modular vector systems
US7435562B2 (en) 2000-07-21 2008-10-14 Modular Genetics, Inc. Modular vector systems
US20040175756A1 (en) * 2001-04-26 2004-09-09 Avidia Research Institute Methods for using combinatorial libraries of monomer domains
US20050048512A1 (en) * 2001-04-26 2005-03-03 Avidia Research Institute Combinatorial libraries of monomer domains
US20050053973A1 (en) * 2001-04-26 2005-03-10 Avidia Research Institute Novel proteins with targeted binding
US20100216663A1 (en) * 2001-04-26 2010-08-26 Amgen Mountain View Inc. Novel proteins with targeted binding
US7485423B2 (en) 2004-11-11 2009-02-03 Modular Genetics, Inc. Ladder assembly and system for generating diversity
US20060223081A1 (en) * 2004-11-11 2006-10-05 Kevin Jarrell Ladder assembly and system for generating diversity
US7786262B2 (en) 2005-07-13 2010-08-31 Amgen Mountain View Inc. IL-6 binding proteins
US8603782B2 (en) * 2005-07-13 2013-12-10 Amgen Inc. Heat lysis production of proteins
US20080281076A1 (en) * 2005-07-13 2008-11-13 Avidia, Inc. IL-6 binding proteins
US20080003611A1 (en) * 2005-07-13 2008-01-03 Avidia, Inc Heat lysis production of proteins
US20100105567A1 (en) * 2005-12-20 2010-04-29 Reeve Michael A Novel capture agents for binding a ligand
US20090312192A1 (en) * 2005-12-20 2009-12-17 Reeve Michael A Method for functionalising a hydrophobic substrate
US20090192048A1 (en) * 2005-12-20 2009-07-30 Michael A Reeve Method of producing a multimeric capture agent for binding a ligand
WO2008030611A2 (en) 2006-09-05 2008-03-13 Medarex, Inc. Antibodies to bone morphogenic proteins and receptors therefor and methods for their use
EP2486941A1 (de) 2006-10-02 2012-08-15 Medarex, Inc. CXCR4-bindende humane Antikörper und deren Verwendungen
WO2008070569A2 (en) 2006-12-01 2008-06-12 Medarex, Inc. Human antibodies that bind cd22 and uses thereof
WO2009054863A2 (en) 2006-12-13 2009-04-30 Medarex, Inc. Human antibodies that bind cd19 and uses thereof
EP2474556A2 (de) 2007-03-14 2012-07-11 Novartis AG APCDD1-Inhibitoren zur Behandlung, Diagnose und Erkennung von Krebs
WO2008153926A2 (en) 2007-06-05 2008-12-18 Yale University Inhibitors of receptor tyrosine kinases and methods of use thereof
EP2586796A1 (de) 2007-10-12 2013-05-01 Novartis AG Zusammensetzungen und Verfahren zur Verwendung für Sclerostin-Antikörper
JP2009291191A (ja) * 2008-05-14 2009-12-17 Metheresis Translational Research Sa Hgfrの高親和結合部位およびその拮抗剤の同定方法
EP3629022A1 (de) 2008-07-25 2020-04-01 Richard W. Wagner Proteinscreeningverfahren
WO2010011944A2 (en) 2008-07-25 2010-01-28 Wagner Richard W Protein screeing methods
EP2815766A1 (de) 2008-08-05 2014-12-24 Novartis AG Zusammensetzungen und Verfahren für auf das Komplementprotein C5 gerichtete Antikörper
EP2837388A1 (de) 2008-08-05 2015-02-18 Novartis AG Zusammensetzungen und Verfahren für gegen das Komplementprotein C5 gerichtete Antikörper
WO2010015608A1 (en) 2008-08-05 2010-02-11 Novartis Ag Compositions and methods for antibodies targeting complement protein c5
WO2010069913A1 (en) 2008-12-16 2010-06-24 Novartis Ag Yeast display systems
WO2010084408A2 (en) 2009-01-21 2010-07-29 Oxford Biotherapeutics Ltd. Pta089 protein
WO2010102175A1 (en) 2009-03-05 2010-09-10 Medarex, Inc. Fully human antibodies specific to cadm1
EP3009454A2 (de) 2009-04-20 2016-04-20 Oxford Bio Therapeutics Limited Antikörper spezifisch gegen cadherin-17
EP3275900A1 (de) 2009-04-27 2018-01-31 Novartis AG Zusammensetzungen und verfahren zur förderung des muskelwachstums
WO2010125003A1 (en) 2009-04-27 2010-11-04 Novartis Ag Compositions and methods for increasing muscle growth
WO2011018421A1 (en) 2009-08-10 2011-02-17 Morphosys Ag Novel screening strategies for the identification of binders
WO2011047083A1 (en) 2009-10-13 2011-04-21 Oxford Biotherapeutics Ltd. Antibodies against epha10
WO2011054007A1 (en) 2009-11-02 2011-05-05 Oxford Biotherapeutics Ltd. Ror1 as therapeutic and diagnostic target
WO2011054893A2 (en) 2009-11-05 2011-05-12 Novartis Ag Biomarkers predictive of progression of fibrosis
WO2011080050A2 (en) 2009-12-11 2011-07-07 Novartis Ag Binding molecules
WO2011098449A1 (en) 2010-02-10 2011-08-18 Novartis Ag Methods and compounds for muscle growth
EP3345926A1 (de) 2010-05-06 2018-07-11 Novartis AG Zusammensetzungen und verfahren zur verwendung von therapeutischen antikörpern gegen ldl-assoziiertes protein 6 (lrp6)
EP4234698A2 (de) 2010-05-06 2023-08-30 Novartis AG Zusammensetzungen und verfahren zur verwendung von therapeutischen antikörpern gegen ldl-assoziiertes protein 6 (lrp6)
WO2011138391A1 (en) 2010-05-06 2011-11-10 Novartis Ag Compositions and methods of use for therapeutic low density lipoprotein - related protein 6 (lrp6) multivalent antibodies
WO2011138392A1 (en) 2010-05-06 2011-11-10 Novartis Ag Compositions and methods of use for therapeutic low density lipoprotein -related protein 6 (lrp6) antibodies
WO2012022814A1 (en) 2010-08-20 2012-02-23 Novartis Ag Antibodies for epidermal growth factor receptor 3 (her3)
WO2012045703A1 (en) 2010-10-05 2012-04-12 Novartis Ag Anti-il12rbeta1 antibodies and their use in treating autoimmune and inflammatory disorders
WO2012172495A1 (en) 2011-06-14 2012-12-20 Novartis Ag Compositions and methods for antibodies targeting tem8
WO2013001369A2 (en) 2011-06-28 2013-01-03 Oxford Biotherapeutics Ltd. Therapeutic and diagnostic target
WO2013003625A2 (en) 2011-06-28 2013-01-03 Oxford Biotherapeutics Ltd. Antibodies
WO2013006437A1 (en) 2011-07-01 2013-01-10 Novartis Ag Method for treating metabolic disorders
WO2013010955A1 (en) 2011-07-15 2013-01-24 Morphosys Ag Antibodies that are cross-reactive for macrophage migration inhibitory factor (mif) and d-dopachrome tautomerase (d-dt)
EP3653222A1 (de) 2011-10-14 2020-05-20 Novartis AG Antikörper und verfahren für wnt-signalwegbedingte erkrankungen
WO2013054307A2 (en) 2011-10-14 2013-04-18 Novartis Ag Antibodies and methods for wnt pathway-related diseases
WO2013074840A1 (en) 2011-11-15 2013-05-23 Allergan, Inc. Treatment of dry age related macular degeneration
WO2013084148A2 (en) 2011-12-05 2013-06-13 Novartis Ag Antibodies for epidermal growth factor receptor 3 (her3) directed to domain ii of her3
EP3590538A1 (de) 2011-12-05 2020-01-08 Novartis AG Antikörper für den epidermalen wachstumsfaktorrezeptor 3 (her 3)
WO2013084147A2 (en) 2011-12-05 2013-06-13 Novartis Ag Antibodies for epidermal growth factor receptor 3 (her3)
WO2013093762A1 (en) 2011-12-21 2013-06-27 Novartis Ag Compositions and methods for antibodies targeting factor p
EP3330288A1 (de) 2011-12-21 2018-06-06 Novartis AG Zusammensetzungen die gegen faktor p gerichtete antikörper enthalten
WO2014020331A1 (en) 2012-08-01 2014-02-06 Oxford Biotherapeutics Ltd. Therapeutic and diagnostic target
EP3725805A1 (de) 2012-09-07 2020-10-21 Novartis AG Il-18-bindungsmoleküle
WO2014037899A2 (en) 2012-09-07 2014-03-13 Novartis Ag Il-18 binding molecules
WO2014084859A1 (en) 2012-11-30 2014-06-05 Novartis Ag Molecules and methods for modulating tmem16a activities
WO2014089111A1 (en) 2012-12-05 2014-06-12 Novartis Ag Compositions and methods for antibodies targeting epo
EP3851454A1 (de) 2012-12-05 2021-07-21 Novartis AG Zusammensetzungen und verfahren für gegen epo gerichtete antikörper
WO2014122613A1 (en) 2013-02-08 2014-08-14 Novartis Ag Anti-il-17a antibodies and their use in treating autoimmune and inflammatory disorders
WO2015198217A2 (en) 2013-02-08 2015-12-30 Novartis Ag Compositions and methods for long-acting antibodies targeting il-17
EP3656786A1 (de) 2013-02-08 2020-05-27 Novartis AG Anti-il-17a-antikörper und deren verwendung zur behandlung von autoimmun- und entzündungserkrankungen
EP3736293A1 (de) 2013-02-12 2020-11-11 Boehringer Ingelheim International Gmbh Therapeutisches und diagnostisches target für krebs mit dll3-bindenden reagenzien
WO2014133855A1 (en) 2013-02-28 2014-09-04 Caprion Proteomics Inc. Tuberculosis biomarkers and uses thereof
WO2014141064A1 (en) 2013-03-13 2014-09-18 Novartis Ag Notch2 binding molecules for treating respiratory diseases
WO2014159239A2 (en) 2013-03-14 2014-10-02 Novartis Ag Antibodies against notch 3
EP3611189A1 (de) 2013-03-14 2020-02-19 Novartis AG Antikörper gegen notch 3
EP3623380A1 (de) 2013-03-15 2020-03-18 Michael C. Milone Targeting von zytotoxischen zellen mit chimären rezeptoren zur adoptiven immuntherapie
WO2014205300A2 (en) 2013-06-21 2014-12-24 Novartis Ag Lectin-like oxidized ldl receptor1 antibodies and methods of use
WO2014205302A2 (en) 2013-06-21 2014-12-24 Novartis Ag Lectin-like oxidized ldl receptor1 antibodies and methods of use
EP4050033A1 (de) 2013-08-02 2022-08-31 Pfizer Inc. Antikörper gegen cxcr4 und antikörperwirkstoffkonjugate
WO2015015401A2 (en) 2013-08-02 2015-02-05 Pfizer Inc. Anti-cxcr4 antibodies and antibody-drug conjugates
US10144781B2 (en) 2013-08-02 2018-12-04 Pfizer Inc. Anti-CXCR4 antibodies and antibody-drug conjugates
US9708405B2 (en) 2013-08-02 2017-07-18 Pfizer Inc. Anti-CXCR4 antibodies and antibody-drug conjugates
WO2015022658A2 (en) 2013-08-14 2015-02-19 Novartis Ag Methods of treating sporadic inclusion body myositis
WO2015069459A1 (en) 2013-11-05 2015-05-14 Novartis Ag Organic compounds
WO2015090229A1 (en) 2013-12-20 2015-06-25 Novartis Ag Regulatable chimeric antigen receptor
WO2015142661A1 (en) 2014-03-15 2015-09-24 Novartis Ag Regulatable chimeric antigen receptor
EP3811970A1 (de) 2014-03-15 2021-04-28 Novartis AG Regulierbarer chimärer antigenrezeptor
EP3461495A1 (de) 2014-04-24 2019-04-03 Novartis AG Verfahren zur verbesserung oder beschleunigung der körperlichen erholung nach einer hüftbruchoperation
WO2015162590A1 (en) 2014-04-24 2015-10-29 Novartis Ag Methods of improving or accelerating physical recovery after surgery for hip fracture
WO2016014553A1 (en) 2014-07-21 2016-01-28 Novartis Ag Sortase synthesized chimeric antigen receptors
EP4205749A1 (de) 2014-07-31 2023-07-05 Novartis AG Subset-optimierte chimäre antigenrezeptorhaltige zellen
EP3660042A1 (de) 2014-07-31 2020-06-03 Novartis AG T-zellen mit teilmengenoptimierten chimären antigenrezeptoren
EP4122957A1 (de) 2014-08-07 2023-01-25 Novartis AG Angiopoietin-like-4-antikörper und verfahren zur verwendung
WO2016020882A2 (en) 2014-08-07 2016-02-11 Novartis Ag Angiopoetin-like 4 (angptl4) antibodies and methods of use
WO2016020880A2 (en) 2014-08-07 2016-02-11 Novartis Ag Angiopoietin-like 4 antibodies and methods of use
EP3967709A1 (de) 2014-09-17 2022-03-16 Novartis AG Targeting von zytotoxischen zellen mit chimären rezeptoren zur adoptiven immuntherapie
WO2016044605A1 (en) 2014-09-17 2016-03-24 Beatty, Gregory Targeting cytotoxic cells with chimeric receptors for adoptive immunotherapy
WO2016057841A1 (en) 2014-10-08 2016-04-14 Novartis Ag Compositions and methods of use for augmented immune response and cancer therapy
WO2016057846A1 (en) 2014-10-08 2016-04-14 Novartis Ag Compositions and methods of use for augmented immune response and cancer therapy
WO2016098079A2 (en) 2014-12-19 2016-06-23 Novartis Ag Compositions and methods for antibodies targeting bmp6
EP4056588A1 (de) 2015-04-08 2022-09-14 Novartis AG Cd20-therapien, cd22-therapien und kombinationstherapien mit einer zelle zur expression des chimären antigen-rezeptors (car) cd19
WO2016164731A2 (en) 2015-04-08 2016-10-13 Novartis Ag Cd20 therapies, cd22 therapies, and combination therapies with a cd19 chimeric antigen receptor (car) - expressing cell
WO2016193872A2 (en) 2015-06-05 2016-12-08 Novartis Ag Antibodies targeting bone morphogenetic protein 9 (bmp9) and methods therefor
WO2016207858A1 (en) 2015-06-26 2016-12-29 Novartis Ag Factor xi antibodies and methods of use
WO2017021893A1 (en) 2015-08-03 2017-02-09 Novartis Ag Methods of treating fgf21-associated disorders
WO2017042701A1 (en) 2015-09-09 2017-03-16 Novartis Ag Thymic stromal lymphopoietin (tslp)-binding antibodies and methods of using the antibodies
EP3842457A1 (de) 2015-09-09 2021-06-30 Novartis AG Moleküle zur bindung von thymus-stroma-lymphopoietin (tslp) und verfahren zur verwendung der moleküle
WO2017103895A1 (en) 2015-12-18 2017-06-22 Novartis Ag Antibodies targeting cd32b and methods of use thereof
WO2017189724A1 (en) 2016-04-27 2017-11-02 Novartis Ag Antibodies against growth differentiation factor 15 and uses thereof
WO2017203450A1 (en) 2016-05-25 2017-11-30 Novartis Ag Reversal binding agents for anti-factor xi/xia antibodies and uses thereof
WO2017216724A1 (en) 2016-06-15 2017-12-21 Novartis Ag Methods for treating disease using inhibitors of bone morphogenetic protein 6 (bmp6)
WO2018116255A1 (en) 2016-12-23 2018-06-28 Novartis Ag Factor xi antibodies and methods of use
WO2018146594A1 (en) 2017-02-08 2018-08-16 Novartis Ag Fgf21 mimetic antibodies and uses thereof
WO2018158398A1 (en) 2017-03-02 2018-09-07 INSERM (Institut National de la Santé et de la Recherche Médicale) Antibodies having specificity to nectin-4 and uses thereof
WO2018175460A1 (en) 2017-03-24 2018-09-27 Novartis Ag Methods for preventing and treating heart disease
WO2018229715A1 (en) 2017-06-16 2018-12-20 Novartis Ag Compositions comprising anti-cd32b antibodies and methods of use thereof
WO2019003104A1 (en) 2017-06-28 2019-01-03 Novartis Ag METHOD FOR PREVENTING AND TREATING URINARY INCONTINENCE
WO2019081983A1 (en) 2017-10-25 2019-05-02 Novartis Ag CD32B TARGETING ANTIBODIES AND METHODS OF USE
WO2019099440A1 (en) 2017-11-14 2019-05-23 Arcellx, Inc. Multifunctional immune cell therapies
WO2019102353A1 (en) 2017-11-22 2019-05-31 Novartis Ag Reversal binding agents for anti-factor xi/xia antibodies and uses thereof
WO2019229658A1 (en) 2018-05-30 2019-12-05 Novartis Ag Entpd2 antibodies, combination therapies, and methods of using the antibodies and combination therapies
DE202019005887U1 (de) 2018-07-03 2023-06-14 Marengo Therapeutics, Inc. Anti-TCR-Antikörpermoleküle und Verwendungen davon
US11965025B2 (en) 2018-07-03 2024-04-23 Marengo Therapeutics, Inc. Method of treating solid cancers with bispecific interleukin-anti-TCRß molecules
WO2020010250A2 (en) 2018-07-03 2020-01-09 Elstar Therapeutics, Inc. Anti-tcr antibody molecules and uses thereof
US11845797B2 (en) 2018-07-03 2023-12-19 Marengo Therapeutics, Inc. Anti-TCR antibody molecules and uses thereof
WO2020069409A1 (en) 2018-09-28 2020-04-02 Novartis Ag Cd19 chimeric antigen receptor (car) and cd22 car combination therapies
WO2020069405A1 (en) 2018-09-28 2020-04-02 Novartis Ag Cd22 chimeric antigen receptor (car) therapies
WO2020131935A1 (en) 2018-12-18 2020-06-25 Novartis Ag Reversal binding agents for anti-factor xi/xia antibodies and uses thereof
WO2020183133A1 (en) 2019-03-08 2020-09-17 Oxford Genetics Limited Method of selecting for antibodies
GB201903233D0 (en) 2019-03-08 2019-04-24 Oxford Genetics Ltd Method of selecting for antibodies
WO2021026310A1 (en) 2019-08-06 2021-02-11 L.E.A.F. Holdings Group Llc Processes of preparing polyglutamated antifolates and uses of their compositions
WO2021053559A1 (en) 2019-09-18 2021-03-25 Novartis Ag Entpd2 antibodies, combination therapies, and methods of using the antibodies and combination therapies
WO2021053560A1 (en) 2019-09-18 2021-03-25 Novartis Ag Combination therapy with entpd2 and cd73 antibodies
WO2021116119A1 (en) 2019-12-09 2021-06-17 INSERM (Institut National de la Santé et de la Recherche Médicale) Antibodies having specificity to her4 and uses thereof
WO2021228956A1 (en) 2020-05-12 2021-11-18 INSERM (Institut National de la Santé et de la Recherche Médicale) New method to treat cutaneous t-cell lymphomas and tfh derived lymphomas
WO2022190010A1 (en) 2021-03-09 2022-09-15 Molecular Partners Ag Novel darpin based cd33 engagers
WO2022190018A1 (en) 2021-03-09 2022-09-15 Molecular Partners Ag Novel darpin based cd123 engagers
WO2022216993A2 (en) 2021-04-08 2022-10-13 Marengo Therapeutics, Inc. Multifuntional molecules binding to tcr and uses thereof
WO2023170296A1 (en) 2022-03-11 2023-09-14 Inserm (Institut National De La Sante Et De La Recherche Medicale) Nucleic acid system to specifically reprogram b and t cells and uses thereof
WO2023217904A1 (en) 2022-05-10 2023-11-16 Institut National de la Santé et de la Recherche Médicale Syncitin-1 fusion proteins and uses thereof for cargo delivery into target cells
WO2024052503A1 (en) 2022-09-08 2024-03-14 Institut National de la Santé et de la Recherche Médicale Antibodies having specificity to ltbp2 and uses thereof

Also Published As

Publication number Publication date
WO2006009888A2 (en) 2006-01-26
US7803907B2 (en) 2010-09-28
US20100292167A1 (en) 2010-11-18
KR20070041721A (ko) 2007-04-19
AU2005265150A1 (en) 2006-01-26
AU2005265150B2 (en) 2012-04-12
CA2569191A1 (en) 2006-01-26
EP2151453A1 (de) 2010-02-10
MXPA06014796A (es) 2007-04-23
WO2006009888A3 (en) 2006-07-06
KR101238074B1 (ko) 2013-07-15
US20060177831A1 (en) 2006-08-10
US7968681B2 (en) 2011-06-28
EP1776134A4 (de) 2009-08-26
EP1776134A2 (de) 2007-04-25
CA2569191C (en) 2013-11-19
JP2008503216A (ja) 2008-02-07

Similar Documents

Publication Publication Date Title
US20060008844A1 (en) c-Met kinase binding proteins
EP2198890B1 (de) Kombinatorische Bibliotheken von Monomer-Domänen
US20040175756A1 (en) Methods for using combinatorial libraries of monomer domains
US20050048512A1 (en) Combinatorial libraries of monomer domains
JP4369662B2 (ja) 単量体ドメインのコンビナトリアルライブラリー
US20050053973A1 (en) Novel proteins with targeted binding
US20050089932A1 (en) Novel proteins with targeted binding
US20050164301A1 (en) LDL receptor class A and EGF domain monomers and multimers
US7786262B2 (en) IL-6 binding proteins
JP2008503216A5 (de)
KR20120110171A (ko) 리간드 결합능력을 가진 헤테로-다량체 변형 유비퀴틴 단백질을 확인하는 방법
US7820790B2 (en) IL-6 binding proteins
CA2587463A1 (en) Protein scaffolds and uses therof

Legal Events

Date Code Title Description
AS Assignment

Owner name: STEMMER, DR WILLLEM P.C., CALIFORNIA

Free format text: SECURITY AGREEMENT;ASSIGNOR:AVIDIA RESEARCH INSTITUTE;REEL/FRAME:015490/0075

Effective date: 20041222

Owner name: ALLOY VENTURES 2002, L.P., CALIFORNIA

Free format text: SECURITY AGREEMENT;ASSIGNOR:AVIDIA RESEARCH INSTITUTE;REEL/FRAME:015490/0075

Effective date: 20041222

Owner name: ALLOY PARTNERS 2002, L.P., CALIFORNIA

Free format text: SECURITY AGREEMENT;ASSIGNOR:AVIDIA RESEARCH INSTITUTE;REEL/FRAME:015490/0075

Effective date: 20041222

Owner name: MAXYGEN, INC., CALIFORNIA

Free format text: SECURITY AGREEMENT;ASSIGNOR:AVIDIA RESEARCH INSTITUTE;REEL/FRAME:015490/0075

Effective date: 20041222

Owner name: WOODY, DR JAMES N, CALIFORNIA

Free format text: SECURITY AGREEMENT;ASSIGNOR:AVIDIA RESEARCH INSTITUTE;REEL/FRAME:015490/0075

Effective date: 20041222

Owner name: VAN VLASSELAER, DR PETER, CALIFORNIA

Free format text: SECURITY AGREEMENT;ASSIGNOR:AVIDIA RESEARCH INSTITUTE;REEL/FRAME:015490/0075

Effective date: 20041222

AS Assignment

Owner name: AVIDIA RESEARCH INSTITUTE, CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:STEMMER, WILLEM P.C.;PERLROTH, D. VICTOR;SATYAL, SANJEEV;REEL/FRAME:015796/0979;SIGNING DATES FROM 20050215 TO 20050228

AS Assignment

Owner name: AVIDIA RESEARCH INSTITUTE, CALIFORNIA

Free format text: TERMINATION OF PATENT SECURITY INTEREST;ASSIGNORS:ALLOY VENTURES 2002, L.P.;ALLOY PARTNERS 2002, L.P.;WOODY, DR. JAMES N.;AND OTHERS;REEL/FRAME:015963/0826

Effective date: 20050422

AS Assignment

Owner name: AVIDIA, INC., CALIFORNIA

Free format text: CHANGE OF NAME;ASSIGNOR:AVIDIA RESEARCH INSTITUTE;REEL/FRAME:017371/0432

Effective date: 20050422

AS Assignment

Owner name: AMGEN MOUNTAIN VIEW, INC., CALIFORNIA

Free format text: CHANGE OF NAME;ASSIGNOR:AVIDIA, INC.;REEL/FRAME:019052/0766

Effective date: 20061024

STCB Information on status: application discontinuation

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