EP1814584A2 - Ligands ameliorant des composants endogenes - Google Patents

Ligands ameliorant des composants endogenes

Info

Publication number
EP1814584A2
EP1814584A2 EP05801585A EP05801585A EP1814584A2 EP 1814584 A2 EP1814584 A2 EP 1814584A2 EP 05801585 A EP05801585 A EP 05801585A EP 05801585 A EP05801585 A EP 05801585A EP 1814584 A2 EP1814584 A2 EP 1814584A2
Authority
EP
European Patent Office
Prior art keywords
endogenous target
ligand
target compound
moiety
activity
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.)
Ceased
Application number
EP05801585A
Other languages
German (de)
English (en)
Inventor
Ian M. Tomlinson
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.)
Domantis Ltd
Original Assignee
Domantis Ltd
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 US10/985,847 external-priority patent/US20060002935A1/en
Application filed by Domantis Ltd filed Critical Domantis Ltd
Priority to EP11161978A priority Critical patent/EP2420251A3/fr
Publication of EP1814584A2 publication Critical patent/EP1814584A2/fr
Ceased legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/24Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against cytokines, lymphokines or interferons
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • 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
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/26Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against hormones ; against hormone releasing or inhibiting factors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/21Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/31Immunoglobulins specific features characterized by aspects of specificity or valency multispecific
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/569Single domain, e.g. dAb, sdAb, VHH, VNAR or nanobody®

Definitions

  • the invention relates to the use of a ligand comprising a moiety that has a binding site for an endogenous target compound wherein said ligand binds said endogenous target compound and does not substantially inhibit the activity of said endogenous target compound, for the manufacture of a medicament.
  • the ligand does not bind to the active site of the endogenous target compound.
  • the medicament can be for increasing the amount of an endogenous target compound in a subject, for increasing the bioavailability an endogenous target compound in a subject, for increasing the in vivo half-life of an endogenous target compound.
  • the medicament can also be for increasing the activity (e.g., binding activity) of an endogenous target compound.
  • the medicament can be for local or systemic delivery.
  • the amount of endogenous target ligand in the subject about 4 hours after administration of the medicament is increased by at least 1.5 times, relative to the amount before administration of the medicament. In some embodiments, the in vivo half life of the endogenous target compound is increased by at least 1.5 times after administration of the medicament.
  • the ligand inhibits the activity of said endogenous target compound by no more than about 10%, or has an inhibitory concentration 50 (IC50) of at least 1 micromolar.
  • the ligand can comprise two or more copies of said moiety that has a binding site for an endogenous target.
  • the ligand can be a dimer of a moiety that has a binding site for an endogenous target, such as a dAb dimer.
  • the moiety that has a binding site for an endogenous target compound can be an aff ⁇ body, an SpA domain, an LDL receptor class A domain, an EGF domain, an avimer, antibody or an antibody fragment (e.g., a Fv fragment, a single chain Fv fragment, a disulfide bonded Fv fragment, a Fab fragment, a Fab' fragment, a F(ab') 2 fragment, a diabody, an immunoglobulin single variable domain (e.g., a human VH, and a human VL, a VHH)-
  • an immunoglobulin single variable domain e.g., a human VH, and a human VL, a VHH
  • the ligand can further comprise a half-life extending moiety as described herein.
  • the ligand can comprise a half-life extending moiety selected from the group consisting of a polyalkylene glycol moiety, serum albumin or a fragment thereof, transferrin receptor or a transferrin-binding portion thereof, or a moiety comprising a binding site for a polypeptide that enhances half-life in vivo.
  • Suitable a moiety comprising a binding site for a polypeptide that enhances half-life in vivo include an affibody, an SpA domain, an LDL receptor class A domain, an EGF domain, an avimer, an antibody or antibody fragment, such as a Fv fragment, a single chain Fv fragment, a disulfide bonded Fv fragment, a Fab fragment, a Fab' fragment, a F(ab') 2 fragment, a diabody, and an immunoglobulin single variable domain (e.g., human VH, human VL, V HH )-
  • the medicaments and ligands of the invention are substantially non- immunogenic. Li some embodiments, the medicament is a depot formulation.
  • the endogenous target compound is a soluble agonist (e.g., cytokines, growth factors, hormones), soluble receptor (e.g., soluble cytokine receptors, such as soluble TNFRl, soluble TNFR2, soluble IL-I receptor, soluble IL-4 receptor, soluble IL-13 receptor), endogenous receptor antagonist (e.g., IL-I receptor antagonist (IL- Ira)) or an enzyme.
  • the endogenous target compound is a soluble cytokine receptor, such as soluble TNFRl .
  • the ligand binds the endogenous target compound with high affinity, such as a Kd of 1 x 10 '7 M or less.
  • the medicaments or ligands of the invention do not include an antibody, fragment or region thereof disclosed in published US patent application publication no. 2003/0144484. In certain embodiments, the medicaments or ligands of the invention do not inhibit binding of antibody A2 or chimeric antibody A2 (cA2) to human TNF-alpha. In particular embodiments, the medicaments or ligands of the invention do not bind to an epitope included in amino acids 87-108 or an epitope included in both amino acids 59-80 and 87-108 of human TNF (tumour necrosis factor). These amino acids are:
  • the invention also relates to the use of endogenous target compound for the manufacture of a medicament for increasing the activity of said endogenous target, wherein said ligand binds said endogenous target and does not substantially inhibit the activity of said endogenous target compound.
  • the ligand does not bind to the active site of said endogenous target compound, hi some embodiments, the binding activity of the endogenous target ligand is increased, for example the binding activity (e.g., affinity, avidity) can be increased by a factor of at least 10.
  • the invention also relates to a ligand that binds an endogenous target compound having activity suitable for treating a disease in a subject, wherein said ligand does not bind the active site of said endogenous target compound or substantially inhibit the activity of said endogenous target compound, for use in therapy of a disease amenable to treatment with said endogenous target compound.
  • the invention also relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a ligand that binds an endogenous target compound having activity suitable for treating a disease in a subject and a physiologically acceptable carrier, wherein said ligand does not bind the active site of said endogenous target compound or substantially inhibit the activity of said endogenous target compound.
  • the invention also relates to a drug delivery device comprising a pharmaceutical composition of the invention.
  • drug delivery device is a parenteral delivery device, intravenous delivery device, intramuscular delivery device, intraperitoneal delivery device, transdermal delivery device, pulmonary delivery device, intraarterial delivery device, intrathecal delivery device, intraarticular delivery device, subcutaneous delivery device, intranasal delivery device, vaginal delivery device, and rectal delivery device.
  • the drug delivery device can be a syringe, a transdermal delivery device, a capsule, a tablet, a nebulizer, an inhaler, an atomizer, an aerosolizer, a mister, a dry powder inhaler, a metered dose inhaler, a metered dose sprayer, a metered dose mister, a metered dose atomizer, a catheter.
  • the invention relates to a method for increasing the half-life of an endogenous target compound in a subject, comprising administering to a subject in need thereof an effective amount of a ligand comprising a moiety that has a binding site with binding specificity for said endogenous target compound.
  • the invention relates to a method for increasing the amount of an endogenous target compound in a subject, comprising administering to a subject in need thereof an effective amount of a ligand comprising a moiety that has a binding site with binding specificity for said endogenous target compound.
  • the invention relates to a method for increasing the bioavailability of an endogenous target compound in a subject, comprising administering to a subject in need thereof an effective amount of a ligand comprising a moiety that has a binding site with binding specificity for said endogenous target compound.
  • the invention relates to a method for increasing the activity (e.g., binding activity) of an endogenous target compound in a subject, comprising administering to a subject in need thereof an effective amount of a ligand comprising a moiety that has a binding site with binding specificity for said endogenous target compound.
  • the invention relates to a method for treating a subject having a disease that is amenable to treatment with an endogenous target compound in a subject, comprising administering to a subject in need thereof an effective amount of a ligand comprising a moiety that has a binding site with binding specificity for said endogenous target compound.
  • the invention also relates to novel ligands (e.g., dAbs) described herein.
  • novel ligands e.g., dAbs
  • Figure 1 is a graph of the relative concentration of soluble TNFRl detected by ELISA in the serum over time (concentration time curve) following a single intravenous administration of a PEGylated dAb monomer that binds TNFRl (TAR2m-21-23/40K branched PEG; also referred to as PEG anti-TNFRl dAb).
  • the plotted data points are were obtained by ELISA using a 1 :5 dilution of serum.
  • the graph clearly shows that the bioavailability of TNFRl in the systemic circulation was increased by the administration of a PEGylated dAb monomer that binds
  • the level of soluble TNFRl in serum increased after the PEGylated dAb monomer was administerd, reach a peak concentration, and then decreased over time to basal levels as the PEGylated dAb was cleared.
  • the results demonstrate that administering a dAb that binds a soluble receptor can increase the bioavailability of the receptor and increase the level or amount of soluble receptor in the systemic circulation, and that the increased level or amount of soluble receptor in the systemic circulation is cleared and returns to basal levels.
  • the results indicate that the level of soluble receptor in the systemic circulation can be controlled by admininstering a a PEGylated dAb monomer that binds the soluble receptor.
  • the term "ligand” refers to a compound that comprises at least one peptide, polypeptide or protein moiety that has a binding site with binding specificity for a desired endogenous target compound.
  • the moiety that has a binding site for a desired endogenous target compound can be an immunoglobulin single variable domain (e.g., VH, VL, V HH ) that has binding specificity for a desired endogenous target compound (e.g., a soluble receptor protein, endogenous receptor antagonist, cytokine or growth factor).
  • the moiety that has a binding site for an endogenous target compound can also comprises one or more complementarity determining regions (CDRs) of an immunoglobulin single variable domain that has binding specificity for a desired endogenous target compound in a suitable format, such that the moiety has binding specificity for the endogenous target compound.
  • CDRs can be grafted onto a suitable protein scaffold or skeleton, such as an affibody, an SpA scaffold, an LDL receptor class A domain, or an EGF domain.
  • the ligand can be monovalent (e.g., a dAb monomer), bivalent (homobivalent, heterobivalent) or multivalent (homomultivalent, heteromultivalent) as described herein.
  • ligands include polypeptides that consist of a dAb, include polypeptides that consist essentially of such a dAb, polypeptides that comprise a dAb (or the CDRs of a dAb) in a suitable format, such as an antibody format (e.g., IgG-like format, scFv, Fab, Fab', F(ab') 2 ) or a suitable protein scaffold or skeleton, such as an affibody, an SpA scaffold, an LDL receptor class A domain or an EGF domain, dual specific ligands that comprise a dAb that binds a first endogenous target compound and a second dAb that binds another endogenous target compound (e.g., serum albumin), and multispecific ligands as described herein.
  • a suitable format such as an antibody format (e.g., IgG-like format, scFv, Fab, Fab', F(ab') 2 ) or
  • the moiety that has a binding site for an endogenous target compound can also be a protein domain comprising a binding site for a desired target, e.g., a protein domain is selected from an affibody, an SpA domain, an LDL receptor class A domain, an EGF domain, an avimer (see, e.g., U.S. Patent Application Publication Nos. 2005/0053973, 2005/0089932, 2005/0164301).
  • a "ligand" can further comprise one or more additional moieties, that can each independently be a peptide, polypeptide or protein moiety or a non-peptidic moiety (e.g., a polyalkylene glycol, a lipid, a carbohydrate).
  • the ligand can further comprise a half-life extending moiety as described herein (e.g., a polyalkylene glycol moiety, a moiety comprising albumin, an albumin fragment or albumin variant, a moiety comprising transferrin, a transferrin fragment or transferrin variant, a moiety that binds albumin, a moiety that binds neonatal Fc receptor).
  • a half-life extending moiety as described herein (e.g., a polyalkylene glycol moiety, a moiety comprising albumin, an albumin fragment or albumin variant, a moiety comprising transferrin, a transferrin fragment or transferrin variant, a moiety that binds albumin, a moiety that binds neonatal Fc receptor).
  • endogenous target compound refers to soluble compound that is produced by a subject (e.g., a mammal, a human) of interest.
  • Endogenous target compounds include, for example, soluble agonists (e.g., cytokines, growth factors, hormones), soluble receptors (e.g., soluble cytokine receptors, such as soluble TNFRl, soluble, soluble TNFR2, soluble IL-I receptor, soluble IL-4 receptor, soluble IL- 13 receptor), endogenous receptor antagonists (e.g., IL-I receptor antagonist (IL- Ira)), enzymes, and the like.
  • soluble agonists e.g., cytokines, growth factors, hormones
  • soluble receptors e.g., soluble cytokine receptors, such as soluble TNFRl, soluble, soluble TNFR2, soluble IL-I receptor, soluble IL-4 receptor, soluble IL- 13 receptor
  • active site refers to the site or domain of an endogenous target compound (e.g., protein) that interacts with (e.g., binds) an endogenous binding partner of an endogenous target compound (e.g., site or domain of a receptor protein that makes contact with the ligand of the receptor, site or domain of an agonist protein (e.g., a cytokine) that makes contact with the receptor of the agonist, the catalytic domain of an enzyme).
  • an endogenous target compound e.g., protein
  • an endogenous binding partner of an endogenous target compound e.g., site or domain of a receptor protein that makes contact with the ligand of the receptor, site or domain of an agonist protein (e.g., a cytokine) that makes contact with the receptor of the agonist, the catalytic domain of an enzyme.
  • the "active site" of a soluble endogenous compound such as a cytokine, growth factor or hormone is the site on the cytokine, growth factor or hormone that makes contact with the endogenous receptor that binds the cytokine, growth factor or hormone.
  • bioavailability refers to the extent to which an endogenous target compound or ligand-endogenous target compound complex is present or gains access to a desired site of action (e.g., the systemic circulation, the central nervous system, a local site of action (e.g., a particular organ, a particular area of tissue (e.g., muscle, skin))).
  • a desired site of action e.g., the systemic circulation, the central nervous system, a local site of action (e.g., a particular organ, a particular area of tissue (e.g., muscle, skin)
  • Bioavailability of an endogenous target compound can be increased independently of any change is the in vivo half-life of the endogenous target compound, or of any change in the amount of the endogenous target compound in a subject.
  • a ligand may bind an endogenous target compound that has beneficial activity in the systemic circulation but is rapidly distributed into the tissues, and slow the rate of distribution into the tissues. Such a ligand would increase bioavailability of the endogenous target compound in the systemic circulation where the beneficial activity of the endogenous target compound can have therapeutic effect.
  • Binding activity can be increased, for example, by increasing affinity (e.g., increasing the rate of binding (K 0n ) or decreasing the rate of dissociation (K off )) or by increasing avidity.
  • Avidity can be increased, for example, by increasing the number of sites at which a first binding partner and a second binding partner make contact.
  • the phrase "substantially non-immunogenic” means high affinity antibodies that bind ligand, ligand-endogenous target compound or endogenous target compound are not produced with a ligand is administered to a subject.
  • High affinity antibodies bind the ligand, ligand-endogenous target compound complex or endogenous target compound with an affinity of 500 nM or lower, or 300 nM or lower, or 100 nM or lower, or 10 nM or lower, or 1 nM or lower.
  • Antibodies that bind ligand, ligand-endogenous target compound complex or endogenous target compound and the affinity of such antibodies can be identified using any suitable method. Several such methods are well-known in the art.
  • a serum sample can be obtained from a human to whom a ligand has been administered (e.g., about 1 week, 2 weeks, 3 weeks, 4 weeks after administration) and the serum can be tested for presence of antibodies that bind ligand, ligand- endogenous target compound or endogenous target compound using, for example an ELISA or other suitable assay.
  • the affinity of antibodies can be determined using any suitable method, such as by equilibrium dialysis or by surface plasmon resonance.
  • immunoglobulin single variable domain refers to an antibody variable region (V H , VH H , V L ) that specifically binds an antigen or epitope independently of other V regions or domains; however, as the term is used herein, an immunoglobulin single variable domain can be present in a format (e.g., homo- or hetero-multimer) with other variable regions or variable domains where the other regions or domains are not required for antigen binding by the single immunoglobulin variable domain (i.e., where the immunoglobulin single variable domain binds antigen independently of the additional variable domains).
  • Immunoglobulin single variable domain encompasses not only an isolated antibody single variable domain polypeptide, but also larger polypeptides that comprise one or more monomers of an antibody single variable domain polypeptide sequence.
  • a “domain antibody” or “dAb” is the same as an "immunoglobulin single variable domain” polypeptide as the term is used herein.
  • An immunoglobulin single variable domain polypeptide, as used herein refers to a mammalian immunoglobulin single variable domain polypeptide, preferably human, but also includes rodent (for example, as disclosed in WO 00/29004, the contents of which are incorporated herein by reference in their entirety) or camelid VH H dAbs.
  • Camelid dAbs are immunoglobulin single variable domain polypeptides which are derived from species including camel, llama, alpaca, dromedary, and guanaco, and comprise heavy chain antibodies naturally devoid of light chain: V HH - V H H molecules are about ten times smaller than IgG molecules, and as single polypeptides, they are very stable, resisting extreme pH and temperature conditions. "Complementary" Two immunoglobulin domains are "complementary" where they belong to families of structures which form cognate pairs or groups or are derived from such families and retain this feature. For example, a V H domain and a V L domain of an antibody are complementary; two VH domains are not complementary, and two V L domains are not complementary.
  • Complementary domains may be found in other members of the immunoglobulin superfamily, such as the V ⁇ and V ⁇ (or ⁇ and ⁇ ) domains of the T-cell receptor. Domains which are artificial, such as domains based on protein scaffolds which do not bind epitopes unless engineered to do so, are non-complementary. Likewise, two domains based on (for example) an immunoglobulin domain and a fibronectin domain are not complementary.
  • Immunoglobulin family This refers to a family of polypeptides which retain the immunoglobulin fold characteristic of antibody molecules, which contains two ⁇ sheets and, usually, a conserved disulphide bond.
  • Members of the immunoglobulin superfamily are involved in many aspects of cellular and non- cellular interactions in vivo, including widespread roles in the immune system (for example, antibodies, T-cell receptor molecules and the like), involvement in cell adhesion (for example the ICAM molecules) and intracellular signaling (for example, receptor molecules, such as the PDGF receptor).
  • the present invention is applicable to all immunoglobulin superfamily molecules which possess binding domains.
  • the present invention relates to antibodies.
  • Domain A domain is a folded protein structure which retains its tertiary structure independently of the rest of the protein. Generally, domains are responsible for discrete functional properties of proteins, and in many cases may be added, removed or transferred to other proteins without loss of function of the remainder of the protein and/or of the domain.
  • single antibody variable domain is meant a folded polypeptide domain comprising sequences characteristic of antibody variable domains.
  • variable domains and modified variable domains, for example in which one or more loops have been replaced by sequences which are not characteristic of antibody variable domains, or antibody variable domains which have been truncated or comprise N- or C-terminal extensions, as well as folded fragments of variable domains which retain at least in part the binding activity and specificity of the full-length domain.
  • “Repertoire” A collection of diverse variants, for example polypeptide variants which differ in their primary sequence.
  • a library used in the present invention will encompass a repertoire of polypeptides comprising at least 1000 members.
  • “Library” The term library refers to a mixture of heterogeneous polypeptides or nucleic acids. The library is composed of members, each of which have a single polypeptide or nucleic acid sequence. To this extent, library is synonymous with repertoire. Sequence differences between library members are responsible for the diversity present in the library.
  • the library may take the form of a simple mixture of polypeptides or nucleic acids, or may be in the form of organisms or cells, for example bacteria, viruses, animal or plant cells and the like, transformed with a library of nucleic acids.
  • each individual organism or cell contains only one or a limited number of library members.
  • the nucleic acids are incorporated into expression vectors, in order to allow expression of the polypeptides encoded by the nucleic acids.
  • a library may take the form of a population of host organisms, each organism containing one or more copies of an expression vector containing a single member of the library in nucleic acid form which can be expressed to produce its corresponding polypeptide member.
  • the population of host organisms has the potential to encode a large repertoire of genetically diverse polypeptide variants.
  • Antibody An antibody (for example IgG, IgM, IgA, IgD or IgE) or fragment (such as a Fab , F(ab') 2 , Fv, disulphide linked Fv, scFv, closed conformation multispecific antibody, disulphide-linked scFv, diabody) whether derived from any species naturally producing an antibody, or created by recombinant DNA technology; whether isolated from serum, B-cells, hybridomas, transfectomas, yeast or bacteria).
  • Double-specific ligand A ligand comprising a first immunoglobulin single variable domain and a second immunoglobulin single variable domain as herein defined, wherein the variable regions are capable of binding to two different antigens or two epitopes on the same antigen which are not normally bound by a monospecific immunoglobulin.
  • the two epitopes may be on the same hapten, but are not the same epitope or sufficiently adjacent to be bound by a monospecific ligand.
  • the dual specific ligands according to the invention are composed of variable domains which have different specificities, and do not contain mutually complementary variable domain pairs which have the same specificity.
  • Antigen A molecule that is bound by a ligand according to the present invention. Typically, antigens are bound by antibody ligands and are capable of raising an antibody response in vivo. It may be a polypeptide, protein, nucleic acid or other molecule. Generally, the dual specific ligands according to the invention are selected for target specificity against a particular antigen. In the case of conventional antibodies and fragments thereof, the antibody binding site defined by the variable loops (Ll, L2, L3 and Hl, H2, H3) is capable of binding to the antigen.
  • Epitope A unit of structure conventionally bound by an immunoglobulin V J /V L pair. Epitopes define the minimum binding site for an antibody, and thus represent the target of specificity of an antibody. In the case of a single domain antibody, an epitope represents the unit of structure bound by a variable domain in isolation.
  • Universal framework A single antibody framework sequence corresponding to the regions of an antibody conserved in sequence as defined by Kabat ("Sequences of Proteins of Immunological Interest", US Department of Health and Human Services) or corresponding to the human germline immunoglobulin repertoire or structure as defined by Chothia and Lesk, (1987) J. MoI. Biol. 196:910-917.
  • the invention provides for the use of a single framework, or a set of such frameworks, which has been found to permit the derivation of virtually any binding specificity though variation in the hypervariable regions alone.
  • “Half-life” The time taken for the serum concentration of the ligand to reduce by 50%, in vivo, for example due to degradation of the ligand and/or clearance or sequestration of the ligand by natural mechanisms.
  • the ligands of the invention are stabilised in vivo and their half-life increased by binding to molecules which resist degradation and/or clearance or sequestration. Typically, such molecules are naturally occurring proteins which themselves have a long half-life in vivo.
  • the half-life of a ligand is increased if its functional activity persists, in vivo, for a longer period than a similar ligand which is not specific for the half-life increasing molecule.
  • a ligand specific for HSA and a target molecule is compared with the same ligand wherein the specificity for HSA is not present, that it does not bind HSA but binds another molecule. For example, it may bind a second epitope on the target molecule.
  • the half life is increased by 10%, 20%, 30%, 40%, 50% or more. Increases in the range of 2x, 3x, 4x, 5x, 10x, 2Ox, 3Ox, 4Ox, 50x or more of the half life are possible.
  • Substantially identical A first amino acid or nucleotide sequence that contains a sufficient number of identical or equivalent (e.g., with a similar side chain, e.g., conserved amino acid substitutions) amino acid residues or nucleotides to a second amino acid or nucleotide sequence such that the first and second amino acid or nucleotide sequences have similar activities.
  • the second antibody has the same binding specificity and has at least 50% of the affinity of the same.
  • the term "about” is preferably interpreted to mean plus or minus 20%, more preferably plus or minus 10%, even more preferably plus or minus 5%, even more preferably plus or minus 2%, most preferably plus or minus 1%.
  • the terms “low stringency,” “medium stringency,” “high stringency,” or “very high stringency conditions” describe conditions for nucleic acid hybridization and washing. Guidance for performing hybridization reactions can be found in Current Protocols in Molecular Biology, John Wiley & Sons, N.Y. (1989), 6.3.1-6.3.6, which is incorporated herein by reference in its entirety.
  • Aqueous and nonaqueous methods are described in that reference and either can be used.
  • Specific hybridization conditions referred to herein are as follows: (1) low stringency hybridization conditions in 6X sodium chloride/sodium citrate (SSC) at about 45°C, followed by two washes in 0.2X SSC, 0.1% SDS at least at 50°C (the temperature of the washes can be increased to 55 0 C for low stringency conditions); (2) medium stringency hybridization conditions in 6X SSC at about 45 0 C, followed by one or more washes in 0.2X SSC, 0.1% SDS at 6O 0 C; (3) high stringency hybridization conditions in 6X SSC at about 45 0 C, followed by one or more washes in 0.2X SSC, 0.1% SDS at 65 0 C; and preferably (4) very high stringency hybridization conditions are 0.5M sodium phosphate, 7% SDS at 65°C, followed by one or more washes at 0.2X SSC, 1% SDS at 65 0 C. Very
  • the term "competes" means that the binding of a first epitope to its cognate epitope binding domain is inhibited when a second epitope is bound to its cognate epitope binding domain.
  • binding may be inhibited sterically, for example by physical blocking of a binding domain or by alteration of the structure or environment of a binding domain such that its affinity or avidity for an epitope is reduced.
  • sequences similar or homologous are also part of the invention.
  • the sequence identity at the amino acid level can be about 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or higher.
  • the sequence identity can be about 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or higher.
  • substantial identity exists when the nucleic acid segments will hybridize under selective hybridization conditions (e.g., very high stringency hybridization conditions), to the complement of the strand.
  • the nucleic acids may be present in whole cells, in a cell lysate, or in a partially purified or substantially pure form.
  • sequence identity or “sequence identity” or “similarity” between two sequences (the terms are used interchangeably herein) are performed as follows.
  • the sequences are aligned for optimal comparison purposes (e.g., gaps can be introduced in one or both of a first and a second amino acid or nucleic acid sequence for optimal alignment and non-homologous sequences can be disregarded for comparison purposes).
  • the length of a reference sequence aligned for comparison purposes is at least 30%, preferably at least 40%, more preferably at least 50%, even more preferably at least 60%, and even more preferably at least 70%, 80%, 90%, 100% of the length of the reference sequence.
  • amino acid residues or nucleotides at corresponding amino acid positions or nucleotide positions are then compared.
  • a position in the first sequence is occupied by the same amino acid residue or nucleotide as the corresponding position in the second sequence, then the molecules are identical at that position (as used herein amino acid or nucleic acid "homology” is equivalent to amino acid or nucleic acid “identity”).
  • the percent identity between the two sequences is a function of the number of identical positions shared by the sequences, taking into account the number of gaps, and the length of each gap, which need to be introduced for optimal alignment of the two sequences.
  • Amino acid and nucleotide sequence alignments and homology, similarity or identity, as defined herein are preferably prepared and determined using the algorithm BLAST 2 Sequences, using default parameters (Tatusova, T. A. et ah, FEMS Microbiol Lett, i74:187-188 (1999)).
  • the BLAST algorithm version 2.0 is employed for sequence alignment, with parameters set to default values.
  • the BLAST algorithm is described in detail at the world wide web site ("www") of the National Center for Biotechnology Information (".ncbi") of the National Institutes of Health (“nih”) of the U.S. government (“.gov”), in the "/Blast/" directory, in the "blast_help.html” file.
  • the search parameters are defined as follows, and are advantageously set to the defined default parameters.
  • BLAST Basic Local Alignment Search Tool
  • blastp, blastn, blastx, tblastn, and tblastx these programs ascribe significance to their findings using the statistical methods of Karlin and Altschul, 1990, Proc. Natl. Acad. Sci. USA 87(6):2264-8 (see the "blast_help.html” file, as described above) with a few enhancements.
  • the BLAST programs were tailored for sequence similarity searching, for example to identify homologues to a query sequence. The programs are not generally useful for motif- style searching.
  • Altschul et al. (1994) The five BLAST programs available at the National Center for Biotechnology Information web site perform the following tasks:
  • blastp compares an amino acid query sequence against a protein sequence database
  • blastn compares a nucleotide query sequence against a nucleotide sequence database
  • blastx compares the six-frame conceptual translation products of a nucleotide query sequence (both strands) against a protein sequence database; "tblastn” compares a protein query sequence against a nucleotide sequence database dynamically translated in all six reading frames (both strands).
  • tblastx compares the six-frame translations of a nucleotide query sequence against the six-frame translations of a nucleotide sequence database.
  • BLAST uses the following search parameters:
  • HISTOGRAM Display a histogram of scores for each search; default is yes. (See parameter H in the BLAST Manual).
  • DESCRIPTIONS Restricts the number of short descriptions of matching sequences reported to the number specified; default limit is 100 descriptions. (See parameter V in the manual page). See also EXPECT and CUTOFF.
  • ALIGNMENTS Restricts database sequences to the number specified for which high-scoring segment pairs (HSPs) are reported; the default limit is 50. If more database sequences than this happen to satisfy the statistical significance threshold for reporting (see EXPECT and CUTOFF below), only the matches ascribed the greatest statistical significance are reported. (See parameter B in the BLAST Manual).
  • EXPECT The statistical significance threshold for reporting matches against database sequences; the default value is 10, such that 10 matches are expected to be found merely by chance, according to the stochastic model of Karlin and Altschul (1990). If the statistical significance ascribed to a match is greater than the EXPECT threshold, the match will not be reported. Lower EXPECT thresholds are more stringent, leading to fewer chance matches being reported. Fractional values are acceptable. (See parameter E in the BLAST Manual).
  • CUTOFF Cutoff score for reporting high-scoring segment pairs.
  • the default value is calculated from the EXPECT value (see above).
  • HSPs are reported for a database sequence only if the statistical significance ascribed to them is at least as high as would be ascribed to a lone HSP having a score equal to the CUTOFF value. Higher CUTOFF values are more stringent, leading to fewer chance matches being reported. (See parameter S in the BLAST Manual). Typically, significance thresholds can be more intuitively managed using EXPECT.
  • MATRIX Specify an alternate scoring matrix for BLASTP, BLASTX, TBLASTN and TBLASTX.
  • the default matrix is BLOSUM62 (Henikoff & Henikoff, 1992, Proc. Natl. Aacad. Sci. USA 89(22):10915-9).
  • the valid alternative choices include: PAM40, PAM120, PAM250 and IDENTITY.
  • No alternate scoring matrices are available for BLASTN; specifying the MATRIX directive in BLASTN requests returns an error response.
  • STRAND Restrict a TBLASTN search to just the top or bottom strand of the database sequences; or restrict a BLASTN, BLASTX or TBLASTX search to just reading frames on the top or bottom strand of the query sequence.
  • FILTER Mask off segments of the query sequence that have low compositional complexity, as determined by the SEG program of Wootton & Federhen (1993) Computers and Chemistry 17:149-163, or segments consisting of short-periodicity internal repeats, as determined by the XNU program of Claverie & States, 1993, Computers and Chemistry 17:191-201, or, for BLASTN, by the DUST program of Tatusov and Lipman (see the world wide web site of the NCBI). Filtering can eliminate statistically significant but biologically uninteresting reports from the blast output (e.g., hits against common acidic-, basic- or proline-rich regions), leaving the more biologically interesting regions of the query sequence available for specific matching against database sequences.
  • Filtering is only applied to the query sequence (or its translation products), not to database sequences. Default filtering is DUST for BLASTN, SEG for other programs.
  • sequences are masked in their entirety, indicating that the statistical significance of any matches reported against the unfiltered query sequence should be suspect.
  • NCBI-gi causes NCBI gi identifiers to be shown in the output, in addition to the accession and/or locus name.
  • sequence comparisons are conducted using the simple BLAST search algorithm provided at the NCBI world wide web site described above, in the "/BLAST" directory.
  • the invention relates to ligands that comprise a binding moiety that has binding specificity for a desired endogenous target compound, to such ligands for use in therapy and to the use of such ligands for the manufacture of a medicament and to therapeutic methods that comprise administering such ligands.
  • agents that bind soluble endogenous target compounds but do not substantially alter the activity of such compounds such as antibodies and antibody fragments (e.g., Fab fragment, Fab' fragment, F(ab') 2 fragment, Fv fragment (e.g., scFv, disulfide bonded Fv), dAb, diabody), can be used to alter certain properties of the endogenous target compound, and thereby make the endogenous target compound more effective for treating, suppressing or mitigating a disease, or for diagnostic purposes.
  • antibodies and antibody fragments e.g., Fab fragment, Fab' fragment, F(ab') 2 fragment, Fv fragment (e.g., scFv, disulfide bonded Fv), dAb, diabody
  • the invention relates to ligands, as described herein, that bind a soluble endogenous target compound but do not substantially inhibit the activity of the endogenous target compound ⁇ e.g., do not bind the active site of the endogenous target compound), but that are useful for altering certain properties of the endogenous target compounds.
  • a ligand that comprises a binding moiety that has binding specificity for a desired endogenous target compound can bind the endogenous target compound, thereby increasing the hydrodynamic size of the endogenous target compound and increasing the serum half-life of the endogenous target compound.
  • the endogenous target compound retains its biological activity when bound by the ligand (e.g., is active in a complex consisting of ligand and endogenous target compound), and can produce beneficial effects for therapeutic and/or diagnostic purposes.
  • the ligand as described herein, can increase the serum half-life of an endogenous target compound by, for example, reducing the rate of clearance of the endogenous target compound.
  • the amount of endogenous target compound in a subject e.g., in the systemic circulation
  • the increased amount of endogenous target ligand can produce beneficial therapeutic effects that are not produce by lower or normal amount sof endogenous target compound.
  • a ligand that comprise a binding moiety that has binding specificity for a desired endogenous target compound can bind the endogenous target compound and increase the bioavailability of the endogenous target compound independent of any change is the in vivo half-life of the endogenous target compound, or of any change in the amount of the endogenous target compound in a subject.
  • a ligand may bind an endogenous target compound that has beneficial activity in the systemic circulation but is rapidly distributed into the tissues or cleared (e.g., a steroid or peptide hormone), and slow the rate of distribution or clearance.
  • Such a ligand can increase bioavailability of the endogenous target compound in the systemic circulation where the activity of the endogenous target compound can have a beneficial (e.g., therapeutic) effect.
  • a ligand that comprise a binding moiety that has binding specificity for a desired endogenous target compound can bind the endogenous target compound and increase the activity (e.g., binding activity) of the endogenous target compound.
  • a ligand may bind an endogenous target compound such as an endogenous agonist (e.g., a cytokine) or a soluble cytokine receptor and improve the binding activity of the endogenous target compound by increasing the affinity and/or avidity of the endogenous target compound for its endogenous binding partner.
  • a ligand that comprises two or more moieties that have binding sites with binding specificity for a desired cytokine can bind two or more individual cytokine molecules thereby producing a cytokine dimer, trimer, oligomer or multimer.
  • a dimer, trimer, oligomer or multimer will have improved binding activity toward, for example, a cell that expresses the receptor for the cytokine due to higher avidity.
  • the binding activity of soluble receptors e.g., soluble TNFRl
  • a ligand can, for example, bind two soluble TNFRl chains forming a soluble TNFRl dimer that will bind trimeric TNF with higher avidity that a single TNFRl chain.
  • the ligands described herein can be administered systemically, for example, to bind endogenous target ligands in the systemic circulation and produce beneficial effects in the systemic circulation. Additionally, the ligands can be administered locally (e.g., by subcutaneous injection, intramuscular injection, intrarticular injection, intradermal injection, inhalation, and the like) and thereby improve local bioavalability of an endogenous target compound or to increase the amount of endogenous target compound a the site where the ligand is locally administered.
  • the ligands described herein can be administered systemically, for example, to bind endogenous target ligands in the systemic circulation and produce beneficial effects in the systemic circulation. Additionally, the ligands can be administered locally (e.g., by subcutaneous injection, intramuscular injection, intrarticular injection, intradermal injection, inhalation, and the like) and thereby improve local bioavalability of an endogenous target compound or to increase the amount of endogenous target compound a the site where the ligand
  • a depot formulation of a ligand can be administered locally by subcutaneous injection, and the administered ligand can recruit endogenous target ligand to the site of the depot and bind the endogenous target ligand to improve local bioavalability or to produce a locally high amount of endogenous target ligand.
  • Such an approach is advantageous, for example, to promote wound healing, or to inhibit local inflammatory reactions (e.g., in the skin, in the lung, in a joint).
  • the ligands described herein are generally small polypeptides that can be produced economically and in large quantities using bacterial or yeast expression systems, rather than more expensive mammalian expression systems which produce lower yields.
  • the ligands described herein are administered to harness and exploit the beneficial activities of compounds (e.g., proteins) that are endogenous to the patient, and generally the ligand is composed of protein domains (e.g. a dAb) that are of the same origin as the patient (e.g., human origin) and therefore are substantially non-immunogenic.
  • the invention relates to ligands that comprise a moiety (e.g., dAb) that has a binding site with binding specificity for an endogenous target compound but do not substantially inhibit the activity of said endogenous target compound.
  • a moiety e.g., dAb
  • the ligand does not bind to the active site of an endogenous target compound.
  • the ligands described herein do not substantially inhibit the activity of the endogenous target compounds to which they bind, and the endogenous target compound remains active when bound by the ligand.
  • a soluble receptor is bound by a ligand of the invention, and the soluble receptor in the resulting receptor-lignad complex can bind the endogenous ligand of the soluble receptor,
  • an enzyme is bound by a ligand of the invention, and the enzyme in the resulting enzyme-ligand complex can bind substate and catalyze the enzymatic reaction.
  • the ligand inhibits the activity of the endogenous target compound to which it binds by no more than about 10%, no more than about 9%, no more than about 8%, no more than about 7%, no more than about 6%, no more than about 5%, no more than about 4%, no more than about 3%, no more than about 2%, no more than about 1%, or has substantially no inhibitory effect on the activity of the endogenous target compound.
  • a ligand that does- not substantially inhibit the activity of an endogenous target compound to which it binds inhibits that activity of said endogenous target compound with an high inhibitory concentration 50 (IC50), such as an IC50 of at least 1 nanomolar, at least 10 naomolar, at least 100 nanomolar, at least 1 micromolar, or at least 10 micromolar.
  • IC50 inhibitory concentration 50
  • the ability of a ligand to inhibit of the activity of the endogenous target compound can be assessed using any suitable in vitro or in vivo assay. Many suitable assays are well-known in the art and routinely used to assess inhibitory activity of compounds. For example, the ability of ligands that comprise a moiety that has a binding site for TNFRl to inhibit the activity or TNFRl can be assessed using in vivo models of endotoxin shock or TNF-induced skin necrosis (see, Sheehan et al., J. Exp.
  • ligands per se generally do not have substantial therapeutic activity but are used to harness and exploit the activity of endogenous target compounds for therapeutic or diagnostic purposes. In some embodiments, the ligands have no substantial activity in a suitable in vitro assay for the activity of an endogenous target compound.
  • a ligand that binds an endogenous target compound and increases the half-life of the endogenous target compound will generally not alter the activity of an endogenous target compound in the assay in comparison to the activity of the endogenous target compound in the same assay but without ligand.
  • Ligands e.g., dAbs
  • a desired endogenous target compound such as a receptor protein, a soluble agonist (e.g., a cytokine), or an enzyme, but do not bind the active site of the endogenous target compound
  • a desired endogenous target compound such as a receptor protein, a soluble agonist (e.g., a cytokine), or an enzyme, but do not bind the active site of the endogenous target compound
  • a desired endogenous target compound such as a receptor protein, a soluble agonist (e.g., a cytokine), or an enzyme
  • individual ligands, or moieties that have a binding site for an endogenous target compound or collections of such moieties can be screened in a suitable competition assay in which binding of an endogenous binding partner (e.g., natural ligand, substrate, cofactor) to the endogenous target compound is assessed in the presence of the ligand or moiety (or collection of ligands or moieties) being screened. No substantial reduction (e.g., inhibition) in binding of the endogenous binding partner to the endogenous target compound relative to a suitable control indicates that the ligand or moiety being screened does not bind the active site of the endogenous target compound.
  • an endogenous binding partner e.g., natural ligand, substrate, cofactor
  • Suitable assays for screening ligands and moieties that comprise a binding site for an endogenous target compounds such as enzymes, receptors (e.g., cytokine receptors, growth factor receptors), and soluble agonists (e.g., cytokines, chemokines, growth factors, hormones) are well-known in the art. Additionally, competition assays are routinely used to screen collections of compounds that contain 10 6 to 10 9 or more members. Thus, such screening activity is within the ordinary skill in the art and is not considered burdensome or undue by those in the art.
  • variable domains disclosed in WO 03002609, WO 2004101790, WO 2005035572, WO 2004061026, WO 2004003019, WO 2004058821, WO 9404678, WO 9749805, WO 9923221, WO 9937681, WO 0024884, WO 0043507, WO 0065067, WO 0140310, WO 03035694, WO 03053531, WO 03054015, WO 0305527, WO 2004015425, WO 2004041862, WO 2004041863, WO 2004041865, WO 2004062551, WO 2005044858 and EP1134231 can be screened for the ability to bind a desired endogenous target compound with out binding to the active site in a suitable competition assay.
  • variable domains variable domains, their sequences and methods of production and selection in the foregoing documents are being explicitly incorporated by reference herein to provide the skilled addressee with examples of moieties that have a binding site for an endogenous target compound for use in the invention.
  • the ligands can increase the half-life of the endogenous targets to which they bind, increase the amount of endogenous targets to which they bind in a subject (e.g., the systemic amount, the amount at a desire site or location), and/or increase the bioavailability of the endogenous targets to which they bind.
  • a ligand can increase the half-life of an endogenous target compound to which it binds by, for example, by binding the endogenous target compound and thereby increasing the hydrodynamic size of the endogenous target compound, by binding and stabilizing the endogenous target compound, or by binding and slowing the rate of clearance or metabolism of the endogenous target compound.
  • the ligand will increase the half-life of the endogenous target compound to which it binds by a factor of at least about 1.5, at least about 2, at least about 3, at least about 4, at least about 5, at least about 6, at least about 7, at least about 8, at least about 9, or at least about 10, relative to the half-life of the endogenous target compound in the absence of ligand.
  • an endogenous target compound can have a half-life of about 1 hour, and after administration of a ligand that comprises a moiety with a binding site that binds said endogenous target compound, the half-life of endogenous target compound can be increased to about 1.5 hours, to about 10 hours, or longer.
  • the level or amount of endogenous target compound can increase following administration of a ligand, as described herein, due to the ligands half-life increasing effect.
  • the level or amount of endogenous target ligand e.g., in serum
  • 1 hour, or 2 hours, or 3 hours, or 4 hours, or 5 hours, or 6 hours, or 7 hours, or 8 hours, or 9 hours, or 10 hours, or 11 hours, or 12 hours, or 1 day after administration of a ligand of the invention can be increased by a factor of at least about 1.5, at least about 2, at least about 3, at least about 4, at least about 5, at least about 10, at least about 50, at least about 100, at least about 500, at least about 1,000, at least about 5,000, at least about 10,000, at least about 50,000, or at least about 100,000.
  • Increase levels or amounts of an endogenous target compound can be readily detected using any suitable method.
  • a suitable sample e.g., serum
  • the sample can be, for example, a sample of blood, serum, cerebrospinal fluid, synovial fluid, bronchoalveolar lavage, enema or a biopsy.
  • the amount or level of ligand in such a sample is increased, relative to the amount or level before the ligand is administered.
  • the ligands can increase the bioavailability of an endogenous target compound.
  • the bioavailability can be increase systemically, or at a desired site of action (e.g., at a site where the ligand is locally administered).
  • a ligand may bind an endogenous target compound that has beneficial activity in the systemic circulation but is rapidly distributed into the tissues or cleared (e.g., a steroid or peptide hormone), and slow the rate of distribution or clearance.
  • Such a ligand can increase bioavailability of the endogenous target compound in the systemic circulation where the activity of the endogenous target compound can have a beneficial (e.g., therapeutic) effect.
  • a ligand can increases the bioavailability of an endogenous target compound at a desired site by recruiting the endogenous target compound to that site.
  • a ligand comprising a binding site for a desired endogenous target compound can be locally administered to a patient (e.g., as a depot formulation).
  • the locally administered ligand can bind the desired endogenous target compound, and recruit additional endogenous target compound to the site.
  • an increased level or amount of endogenous target compound can be present at the site to produce beneficial effects.
  • the ligand comprises a moiety with a binding site for TGF beta isoform 3.
  • Such a ligand can be locally administered to bind TGF beta isoform 3 and recruit TGF beta isoform 3 to the site of local administration, and thus promote would healing.
  • Similar ligands that comprise a moiety with a binding site for a desired cell surface target can be prepared using the methods described herein and used, for example, to recruit a desired cell type to a desire site (e.g., site of injury, site of inflammation).
  • a desire site e.g., site of injury, site of inflammation.
  • the ligand will increase bioavailability (e.g., in the systemic circulation, at a desired site of action) of the endogenous target compound to which it binds by a factor of at least about 1.5, at least about 2, at least about 3, at least about 4, at least about 5, at least about 6, at least about 7, at least about 8, at least about 9, or at least about 10, relative to the bioavailability of the endogenous target compound in the absence of ligand.
  • a concentration time curve for the endogenous ligand starting at the time of administration of the ligand can be prepared, and compared to normal systemic levels of the endogenous target ligand to determine the extent of increase in systemic bioavailability.
  • the ligands can increase the activity (e.g., binding activity) of the endogenous targets to which they bind.
  • a ligand may bind an endogenous target compound such as an endogenous agonist (e.g., a cytokine) or a soluble cytokine receptor and improve the binding activity of the endogenous target compound by increasing the affinity and/or avidity of the endogenous target compound for its endogenous binding partner.
  • Ligands that comprises two or more moieties that have binding sites with binding specificity for a desired endogenous target compound can bind two or more individual endogenous target compound molecules thereby producing an endogenous target compound dimer, timer, oligomer or multimer.
  • Such a dimer, trimer, oligomer or multimer will have improved binding activity toward, for example, a cell that expresses the natural binding partner for the endogenous target compound due to higher avidity.
  • a ligand can improve the binding activity of the endogenous target compound.
  • the binding strength (e.g., affinity or avidity) of an endogenous target compound dimer, trimer, oligomer or multimer for its binding partner can be at least about 10, at least about 100, at least about 1000 or at least about 10,000 times stronger than the binding strength of the endogenous target compound as a monomer (i.e., not in a complex with a ligand of the invention).
  • the ligand comprises two or more moieties that have binding sites with binding specificity for a desired endogenous agonist (e.g., a cytokine, a growth factor) and binds two or more endogenous agonist molecules thereby producing an endogenous agonist dimer, trimer, oligomer or multimer.
  • a dimer, trimer, oligomer or multimer will have improved binding activity toward, for example, a cell that expresses the receptor for the endogenous agonist due to higher avidity, and can have improved therapeutic efficacy.
  • the binding activity of a soluble receptor e.g., soluble TNFRl
  • Such a ligand can, for example, bind two soluble receptor chains forming a soluble receptor dimer that will bind the endogenous ligand of the receptor with higher avidity that a single receptor chain.
  • the ligand comprises two or more moieties (dAbs) that have binding specificity for soluble TNFRl but do not bind the active site (the active site of soluble TNFRl is contained within Domains 2 and 3).
  • dAbs moieties
  • Such a ligand can bind two or more soluble TNFRl chains to form a soluble TNFRl dimer, trimer, oligomer or multimer, that has improved binding activity toward its trimeric ligand, TNF, due to increased avidity.
  • such a ligand can improve the TNF-inhibiting activity of soluble TNFRl and improve its therapeutic efficacy.
  • such a ligand can improve the efficacy of soluble TNFRl by a factor of at least about 10, at least about 100, at least about 1,000 or at least about 10,000.
  • a ligand that is a dimer of a dAb that binds Domain 1 of mouse TNFRl is used to demonstrate that ligands that induce dimerization of soluble TNFRl improve efficacy of soluble TNFRl .
  • a ligand can be added to an assay containing human MRC5 cells, human TNF and soluble mouse TNFRl . Soluble mouse TNFRl will compete with human TNFRl on the MRC5 cells for binding to human TNF.
  • the ligand that is a dimer of a dAb that binds Domain 1 of mouse TNFRl binds two soluble mouse TNFRl chains to form a mouse TNFRl dimer that will be much more effective in inhibiting the effects of TNF in the assay.
  • the IC 50 for mouse TNFRl can be reduced from the nanomolar range to the picomolar range (about 10 or about 100 or about 1000 fold reduction) by the addition of the ligand that induces dimerization of soluble TNFRl.
  • ligands that induce dimerization (or trimerization or oligomeriztion) of soluble receptor chains to patients may cause the clustering and activation of transmembrane forms of certain receptors that are expressed on the cell surface.
  • any non-desired effect of such activation e.g, activation of immune cells
  • the interaction of a dimerizing, trimerizing or oligomerizing ligand with the transmembrane form of a receptor can be further reduced by formatting the ligand to increase its hydrodynamic size as described herein.
  • the ligand can be formatted to include a PEG moiety or other half-life extending moiety, such as a moiety that has a binding site for a polypeptide that enhances half-life in vivo (e.g., a dAb that binds serum albumin).
  • a polypeptide that enhances half-life in vivo e.g., a dAb that binds serum albumin.
  • PEGylation of the dAbs dramatically reduced the ability of the dAb to inhibit TNFRl activity in a cell based assay, but had almost no effect on the ability of the dAbs to bind TNFRl in a receptor binding assay that approximates the conditions for binding to soluble TNFRl in vivo.
  • PEGylating dAbs and ligands that comprise two or more dAbs improves selectivity of the dAbs for soluble receptor over the transmembrane form of the receptor.
  • the moiety that has a binding site for an endogenous target compound can be the endogenous target compound itself of a fragment or portion of the endogenous target compound, m some embodiments, such as when the moiety that has a binding site for an endogenous target compound is the endogenous target compound, the ligand can have the activity of the endogenous target compound.
  • administering such a ligand can, as described herein, increase the half-life, increase the bioavailability, increase the amount and/or increase the activity of the endogenous target compound, and will also provide additional endogenous target compound activity due to the presence of a biologically active form of the endogenous target compound in the ligand.
  • the ligand can comprise a moiety that has a binding site for an endogenous target compound that is a portion of the endogenous target compound.
  • the portion of the endogenous target compound can be a portion that binds to the endogenous target compound in vivo, but does not have the activity (e.g., ligand binding activity, receptor binding activity, catalytic activity) of the endogenous target compound.
  • Examples of endogenous target compounds that naturally form dimers, trimers or multiniers include, for example, growth factors (e.g., PDGF which forms homodimers and heterodimers of A and B chains), cytokines (e.g., cytokines of the TNF superfamily (e.g., RANK-L) which form trimers), and receptors (e.g., receptor of the TNF receptor superfamily which form timers).
  • growth factors e.g., PDGF which forms homodimers and heterodimers of A and B chains
  • cytokines e.g., cytokines of the TNF superfamily (e.g., RANK-L) which form trimers
  • receptors e.g., receptor of the TNF receptor superfamily which form timers.
  • Suitable portions of such endogenous target compounds can be easily prepared based on knowledge of the structure of the dimer, trimer or multimer forming domains of the endogenous target compounds, or by preparing peptide or polypeptide fragments of the endogenous target compound and assaying the peptides or polypeptides for the ability to bind the endogenous target compound (e.g., using surface plasmon resonance or any other suitable method).
  • PAD pre-ligand assembly domains
  • PLAD domains from a particular receptor bind to each other in vivo.
  • the PLAD domain of TNFRl will bind another PLAD domain of TNFRl in vivo.
  • the TNF receptor superfamily is an art recognized group of proteins that includes TNFRl (p55, CD120a, p60, TNF receptor superfamily member IA,
  • TNFRSFlA TNFR2 (p75, p80, CD120b, TNF receptor superfamily member IB, TNFRSFlB), CD (TNFRSF3, LT ⁇ R, TNFR2-RP, TNFR-RP, TNFCR, TNF-R-III), OX40 (TNFRSF4, ACT35, TXGPlL), CD40 (TNFRSF5, p50, Bp50), Fas (CD95, TNFRSF6, APO-I, APTI), DcR3 (TNFRSF6B), CD27 (TNFRSF7, Tp55, S 152), CD30 (TNFRSF8, Ki-I, Dl S 166E), CD137 (TNFRSF9, 4-1BB, ILA), TRAILR-I (TNFRSFlOA, DR4, Apo2), TRAIL-R2 (TNFRSFlOB, DR5, KILLER, TRICK2A, TRICKB), TRAILR3 (TNFRSFl OC, DcRl , L
  • PLAD domains are known in the art and other PLAD domains and variants of PLAD domains can be readily isolated and prepared using any suitable methods, such as the methods described in WO 01/58953; U.S. Patent Application Publication No. 2003/0108992 Al; Deng et al, Nature Medicine, doi: 10.1038/nml304 (2005).
  • Many suitable methods for preparing polypeptides, protein fragments, and peptide variants, as well as suitable binding assays, such as the TNFRl recepetor binding assay described herein are well-known and conventional in the art.
  • Exemplary PLAD domains are presented in Table 1.
  • the ligand comprises an moiety that has a binding site with binding specificity for an endogenous target compound which is a PLAD, such as a PLAD of TNFRl, TNFR2, FAS, LT ⁇ R, CD40, CD30, CD27, HVEM, OX40, DR4 or other TNF receptor superfamily member, or a variant of a PLAD domain.
  • a PLAD such as a PLAD of TNFRl, TNFR2, FAS, LT ⁇ R, CD40, CD30, CD27, HVEM, OX40, DR4 or other TNF receptor superfamily member, or a variant of a PLAD domain.
  • the variant of a PLAD domain can, for example, be a PLAD domain of TNFRl, TNFR2, FAS, LT ⁇ R, CD40, CD30, CD27, HVEM, OX40, or DR4, wherein one or more amino acids has been deleted, inserted or substituted, but that retains the ability to bind to the corresponding PLAD of TNFRl, TNFR2, FAS, LT ⁇ R, CD40, CD30, CD27, HVEM, OX40, or DR4.
  • the amino acid sequence of a variant PLAD domain can comprise a region of at least about 10 contiguous amino acids, at least about 15 contiguous amino acids, at least about 20 contiguous amino acids, at least about 25 contiguous amino acids, at least about 30 contiguous amino acids, at least about 35 contiguous amino acids, or at least about 40 contiguous amino acids that are the same as the amino acids in the amino acid sequence of the corresponding PLAD (e.g., PLAD of TNFRl, TNFR2, FAS, LT ⁇ R, CD40, CD30, CD27, HVEM, OX40, DR4).
  • PLAD of TNFRl, TNFR2, FAS, LT ⁇ R, CD40, CD30, CD27, HVEM, OX40, DR4 e.g., PLAD of TNFRl, TNFR2, FAS, LT ⁇ R, CD40, CD30, CD27, HVEM, OX40, DR4
  • the amino acid sequence of a variant PLAD domain can be at least about 80%, at least about 85%, least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% identical to the amino acid sequence of the corresponding PLAD (e.g., PLAD of TNFRl, TNFR2, FAS, LT ⁇ R, CD40, CD30, CD27, HVEM, OX40, or DR4).
  • PLAD of TNFRl, TNFR2, FAS, LT ⁇ R, CD40, CD30, CD27, HVEM, OX40, or DR4 e.g., PLAD of TNFRl, TNFR2, FAS, LT ⁇ R, CD40, CD30, CD27, HVEM, OX40, or DR4
  • Ligands that comprise a binding moiety that has a binding site for an endogenous target compound that is a PLAD or PLAD variant can be formatted as described herein.
  • the ligand can be a dimer, trimer or multimer of a PLAD or PLAD variant, or a fusion protein comprising a PLAD or PLAD variant and a half-life extending moiety (e.g., a moiety that has a binding site for serum albumin or neonatal Fc receptor).
  • Ligands that comprise two or more PLAD or PLAD variants can bind two or more soluble receptor chains to form soluble receptor dimers, trimers or multimers which, as described herein, can have improved binding function in comparison to soluble receptor monomers, hi some circumstances, such a ligand may bind soluble receptor and also bind the transmembrane form of the receptor expressed on the cell surface. If desired, selectivity for binding soluble receptor can be improved by formatting the ligand to have a larger hydrodynamic size, for example using a PEG moiety, or other half-life extending moiety as described herein.
  • the ligand comprises a PLAD (e.g., PLAD of
  • TNFRl TNFR2
  • FAS LT ⁇ R
  • CD40 CD30
  • CD27 CD27
  • HVEM HVEM
  • OX40 OX40
  • DR4 a half-life extending moiety
  • PEG PEG moiety
  • dAb that binds serum albumin or neonatal Fc receptor
  • a ligand that induce dimerization (or trimerization or oligomeriztion) of soluble receptor chains does not substantially agonize cell surface or transmembrane forms of the receptor in a standard cell assay (i.e., when present at a concentration of 1 nM, 10 nM, 100 nM, 1 ⁇ M, 10 ⁇ M, 100 ⁇ M, 1000 ⁇ M or 5,000 ⁇ M, results in no more than about 5% of the receptor-mediated activity induced by natural ligand of the receptor in the assay).
  • the moiety that has a binding site with binding specificity for an endogenous target compound binds the endogenous target compound with high affinity, such as a K d of 300 nM to 5 pM (ie, 3 x 10 "7 to 5 x 10 "12 M), preferably 50 nM to 20 pM, more preferably 5 nM to 200 pM and most preferably 1 nM to 100 pM, for example 1 x 10 "7 M or less, preferably 1 x 10 "8 M or less, more preferably 1 x 10 "9 M or less, advantageously 1 x 10 "10 M or less and most preferably 1 x 10 "11 M or less; and/or a K off rate constant of 5 x 10 "1 s “1 to 1 x 10 "7 s “1 , preferably 1 x 10 "2 s “1 to 1 x 10 "6 s “1 , more preferably 5 x 10 "3 s "1 to 1 x 10 "5 s "1
  • the ligand bind the endogenous target compound with high affinity, such as a K ⁇ of 300 nM to 5 pM (ie, 3 x 10 "7 to 5 x 10 "12 M), preferably 50 nM to 20 pM, more preferably 5 nM to 200 pM and most preferably 1 nM to 100 pM, for example 1 x 10 "7 M or less, preferably 1 x 10 "8 M or less, more preferably 1 x 10 "9 M or less, advantageously 1 x 10 "10 M or less and most preferably 1 x 10 "11 M or less; and/or a K o ff rate constant of 5 x 10 "1 s “1 to 1 x 10 "7 s "1 , preferably 1 x 10 "2 s "1 to 1 x lO "6 s "1 , more preferably 5 x 10 "3 s "1 to 1 x lO
  • the ligand comprises a dAb that binds an endogenous target compound.
  • the endogenous target compound can be, for example, a soluble receptor (e.g., soluble cytokine receptor, soluble growth factor receptor, soluble hormone receptor), an endogenous receptor antagonist (e.g., IL- lra), an enzyme, an endogenous agonist (e.g., a cytokine, a growth factor, a hormone).
  • the ligand is a dAb dimer, trimer, oligomer or multimer.
  • the ligands can comprise a moiety that has a binding site for an endogenous target compound, wherein said moiety that has a binding site for an endogenous target compound is selected from the group consisting of an affibody, an SpA domain, an LDL receptor class A domain, an EGF domain, and an avimer.
  • the ligand can also comprise a moiety that has a binding site for an endogenous target compound wherein the moiety is an antibody or antibody fragment, such as a Fv fragment, a single chain Fv fragment, a disulfide bonded Fv fragment, a Fab fragment, a Fab' fragment, a F(ab') 2 fragment, a diabody, an immunoglobulin single variable domain (e.g., human V H , human VL, VH H >
  • an antibody or antibody fragment such as a Fv fragment, a single chain Fv fragment, a disulfide bonded Fv fragment, a Fab fragment, a Fab' fragment, a F(ab') 2 fragment, a diabody, an immunoglobulin single variable domain (e.g., human V H , human VL, VH H >
  • the ligand can further comprise a half-life extending moiety as described herein.
  • the ligand can comprise a half-life extending moiety selected from the group consisting of a polyalkylene glycol moiety, serum albumin or a fragment thereof, transferrin receptor or a transferrin-binding portion thereof, or a moiety comprising a binding site for a polypeptide that enhances half-life in vivo.
  • Suitable a moieties comprising a binding site for a polypeptide that enhances half- life in vivo include an affibody, an SpA domain, an LDL receptor class A domain, an EGF domain, an avimer, an antibody or antibody fragment, such as a Fv fragment, a single chain Fv fragment, a disulfide bonded Fv fragment, a Fab fragment, a Fab' fragment, a F(ab') 2 fragment, a diabody, and an immunoglobulin single variable domain (e.g., human V H , human V L , V HH )- As described herein, the ligands of the invention do not substantially inhibit the activity of the endogenous target compound to which they bind.
  • the ligand may inhibit the activity that is not associated with the intended therapeutic activity or benefit.
  • a ligand or dAb monomer may bind a soluble receptor and also bind the transmembrane form of the receptor.
  • the transmembrane form of the receptor may have ligand-binding activity and also signaling activity.
  • the ligand of the invention will not inhibit the ligand binding activity (of the soluble and transmembrane forms of the receptor) which acts to inhibit ligand-induced processes, but may inhibit the signaling activity of the transmembrane form of the receptor.
  • exemplary embodiments of ligands that bind soluble TNFRl are described below.
  • Embodiments of ligands that bind soluble TNFRl are illustrative of ligands of the invention that bind other receptors.
  • TNFRl is a transmembrane receptor containing an extracellular region that binds ligand and an intracellular domain that lacks intrinsic signal transduction activity but can associate with signal transduction molecules.
  • the complex of TNFRl with bound TNF contains three TNFRl chains and three TNF chains.
  • TNF ligand is present as a trimer, which is bound by three TNFRl chains.
  • the three TNFRl chains are clustered closely together in the receptor-ligand complex, and this clustering is a prerequisite to TNFRl -mediated signal transduction.
  • multivalent agents that bind TNFRl such as anti-TNFRl antibodies, can induce TNFRl clustering and signal transduction in the absence of TNF and are commonly used as TNFRl agonists.
  • TNFRl agonists See, e.g., Belka et al, EMBO, 14(6):1156-U65 (1995); Mandik-Nayak et al, J. Immunol, 167:1920-1928 (2001).
  • multivalent agents that bind TNFRl are generally not effective antagonists of TNFRl even if they block the binding of TNF ⁇ to TNFRl .
  • the extracellular region of TNFRl comprises a thirteen amino acid amino- terminal segment (amino acids 1-13 of SEQ ID NO:2 (human); amino acids 1-13 of SEQ TD NO:4 (mouse)), Domain 1 (amino acids 14-53 of SEQ ID NO: 2 (human); amino acids 14-53 of SEQ ID NO: 4 (mouse)), Domain 2 (amino acids 54-97 of SEQ ID NO: 2 (human); amino acids 54-97 of SEQ ID NO: 4 (mouse)), Domain 3 (amino acids 98-138 of SEQ ID NO: 2 (human); amino acid 98-138 of SEQ ID NO: 4 (mouse)), and Domain 4 (amino acids 139-167 of SEQ ID NO: 2 (human); amino acids 139-167 of SEQ ID NO: 4 (mouse)) which is followed by a membrane- proximal region (amino acids 168-182 of SEQ ED NO: 2 (human); amino acids 168- 183 SEQ ID
  • TNFRl is shed from the surface of cells in vivo through a process that includes proteolysis of TNFRl in Domain 4 or in the membrane-proximal region (amino acids 168-182 of SEQ ID NO: 2; amino acids 168-183 of SEQ ED NO: 4), to produce a soluble form of TNFRl. Soluble TNFRl retains the capacity to bind TNF ⁇ , and thereby functions as an endogenous inhibitor of the activity of TNF ⁇ .
  • a ligand or dAb monomer that binds soluble TNFRl can also bind transmembrane TNFRl and inhibit TNF ⁇ -induced clustering of TNFRl on the cell surface, which precedes signal transduction through the receptor, but not inhibit binding of TNFa to transmembrane TNFRl and soluble TNFRl.
  • the ligand or dAb monomer does not inhibit the TNF-binding activity of soluble and transmembrane forms of TNFRl but can inhibit the signaling activity of TNFRl.
  • a ligand or dAb monomer of this type can bind Domain 1 or Domain 4 of TNFRl.
  • Such a ligand or dAb monomer can bind soluble TNFRl and transmembrane TNFRl, but does not inhibit the TNF-binding activity of either form of TNFRl .
  • such a ligand or dAb can be administered, for example, to extend the half-life of soluble TNFRl or to enhance the binding activity of soluble TNFRl.
  • the invention provides similar ligands and dAb monomers that bind other soluble receptors but do not inhibit binding of endogenous ligands to soluble receptors.
  • the ligand comprises a dAb that binds Domain 1 of TNFRl and competes with TAR2m-21-23 for binding to mouse TNFRl or competes with TAR2h-205 for binding to human TNFRl .
  • the ligand is a dimer of such a dAb and, if desired, further comprises a PEG moiety.
  • the ligand or dAb monomer is substantially resistant to aggregation. For example, in some embodiments, less than about 10%, less than about 9%, less than about 8%, less than about 7%, less than about 6%, less than about 5%, less than about 4%, less than about 3%, less than about 2% or less than about 1% of the ligand or dAb monomer aggregates when a 1-5 mg/ml, 5-10 mg/ml, 10-20 mg/ml, 20-50 mg/ml, 50-100 mg/ml, 100-200 mg/ml or 200 -500 mg/ml solution of ligand or dAb in a solvent that is routinely used for drug formulation such as saline, buffered saline, citrate buffer saline, water, an emulsion, and, any of these solvents with
  • Aggregation can be assessed using any suitable method, such as, by microscopy, assessing turbidity of a solution by visual inspection or spectroscopy or any other suitable method.
  • aggregation is assessed by dynamic light scattering.
  • Ligands or dAb monomers that are resistant to aggregation provide several advantages. For example, such ligands or dAb monomers can readily be produced in high yield as soluble proteins by expression using a suitable biological production system, such as E. coli, and can be formulated and/or stored at higher concentrations than conventional polypeptides, and with less aggregation and loss of activity.
  • preparation of antigen- or epitope-binding polypeptides intended for in vivo applications includes processes (e.g., gel filtration) that remove aggregated polypeptides. Failure to remove such aggregates can result in a preparation that is not suitable for in vivo applications because, for example, aggregates of an antigen-binding polypeptide that is intended to act as an antagonist can function as an agonist by inducing cross-linking or clustering of the target antigen. Protein aggregates can also reduce the efficacy of therapeutic polypeptide by inducing an immune response in the subject to which they are administered.
  • the aggregation resistant ligands or dAb monomers of the invention can be prepared for in vivo applications without the need to include process steps that remove aggregates, and can be used in in vivo applications without the aforementioned disadvantages caused by polypeptide aggregates.
  • the ligand or dAb monomer unfolds reversibly when heated to a temperature (Ts) and cooled to a temperature (Tc), wherein Ts is greater than the melting temperature (Tm) of the dAb, and Tc is lower than the melting temperature of the dAb.
  • Ts is greater than the melting temperature (Tm) of the dAb
  • Tc is lower than the melting temperature of the dAb.
  • the dAb monomer can unfold reversibly when heated to 8O 0 C and cooled to about room temperature.
  • Such polypeptides are distinguished from polypeptides that aggregate when unfolded or that improperly refold (misfolded polypeptides), i.e., do not regain function.
  • Polypeptide unfolding and refolding can be assessed, for example, by directly or indirectly detecting polypeptide structure using any suitable method.
  • polypeptide structure can be detected by circular dichroism (CD) ⁇ e.g., far- UV CD, near-UV CD), fluorescence ⁇ e.g., fluorescence of tryptophan side chains), susceptibility to proteolysis, nuclear magnetic resonance (NMR), or by detecting or measuring a polypeptide function that is dependent upon proper folding (e.g., binding to target ligand, binding to generic ligand).
  • CD circular dichroism
  • fluorescence ⁇ e.g., fluorescence of tryptophan side chains
  • susceptibility to proteolysis e.g., nuclear magnetic resonance (NMR)
  • NMR nuclear magnetic resonance
  • polypeptide unfolding is assessed using a functional assay in which loss of binding function ⁇ e.g., binding a generic and/or target ligand, binding a substrate) indicates that the polypeptide is unfolded.
  • the extent of unfolding and refolding of a ligand or dAb monomer can be determined using an unfolding or denaturation curve.
  • An unfolding curve can be produced by plotting temperature as the ordinate and the relative concentration of folded polypeptide as the abscissa.
  • the relative concentration of folded ligand or dAb monomer can be determined directly or indirectly using any suitable method (e.g., CD, fluorescence, binding assay).
  • a ligand or dAb monomer solution can be prepared and ellipticity of the solution determined by CD.
  • the ellipticity value obtained represents a relative concentration of folded ligand or dAb monomer of 100%.
  • the ligand or dAb monomer in the solution is then unfolded by incrementally raising the temperature of the solution and ellipticity is determined at suitable increments (e.g., after each increase of one degree in temperature).
  • the ligand or dAb monomer in solution is then refolded by incrementally reducing the temperature of the solution and ellipticity is determined at suitable increments.
  • the data can be plotted to produce an unfolding curve and a refolding curve.
  • the unfolding and refolding curves have a characteristic sigmoidal shape that includes a portion in which the ligand or dAb monomer molecules are folded, an unfolding/refolding transition in which ligand or dAb monomer molecules are unfolded to various degrees, and a portion in which the ligand or dAb monomer molecules are unfolded.
  • the y-axis intercept of the refolding curve is the relative amount of refolded ligand or dAb monomer recovered.
  • a recovery of at least about 50%, or at least about 60%, or at least about 70%, or at least about 75%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95% is indicative that the ligand or dAb monomer unfolds reversibly.
  • reversibility of unfolding of the ligand or dAb monomer is determined by preparing a ligand or dAb monomer solution and plotting heat unfolding and refolding curves.
  • the ligand or dAb monomer solution can be prepared in any suitable solvent, such as an aqueous buffer that has a pH suitable to allow the ligand or dAb monomer to dissolve (e.g., pH that is about 3 units above or below the isoelectric point (pi)).
  • a pH suitable to allow the ligand or dAb monomer to dissolve e.g., pH that is about 3 units above or below the isoelectric point (pi)
  • the ligand or dAb monomer solution is concentrated enough to allow unfolding/folding to be detected.
  • the ligand or dAb monomer solution can be about 0.1 ⁇ M to about 100 ⁇ M, or preferably about 1 ⁇ M to about 10 ⁇ M.
  • the solution can be heated to about ten degrees below the Tm (Tm-10) and folding assessed by ellipticity or fluorescence (e.g., far-UV CD scan from 200 nm to 250 nm, fixed wavelength CD at 235 nm or 225 nm; tryptophan fluorescent emission spectra at 300 to 450 nm with excitation at 298 nm) to provide 100% relative folded ligand or dAb monomer.
  • the solution is then heated to at least ten degrees above Tm (Tm+10) in predetermined increments (e.g., increases of about 0.1 to about 1 degree), and ellipticity or fluorescence is determined at each increment.
  • the ligand or dAb monomer is refolded by cooling to at least Tm-10 in predetermined increments and ellipticity or fluorescence determined at each increment.
  • the solution can be unfolded by incrementally heating from about 25°C to about 100 0 C and then refolded by incrementally cooling to at least about 25 0 C, and ellipticity or fluorescence at each heating and cooling increment is determined.
  • the data obtained can be plotted to produce an unfolding curve and a refolding curve, in which the y-axis intercept of the refolding curve is the relative amount of refolded protein recovered.
  • the ligand or dAb monomer can comprise any suitable immunoglobulin variable domain, and preferably comprises a human variable domain or a variable domain that comprises human framework regions, hi certain embodiments, the dAb monomer comprises a universal framework, as described herein.
  • the universal framework can be a V L framework (V ⁇ or VK) , such as a framework that comprises the framework amino acid sequences encoded by the human germline DPKl, DPK2, DPK3, DPK4, DPK5, DPK6, DPK7, DPK8, DPK9, DPKlO, DPK12, DPK13, DPK15, DPK16, DPK18, DPK19, DPK20, DPK21, DPK22, DPK23, DPK24, DPK25, DPK26 or DPK 28 immunoglobulin gene segment.
  • V ⁇ or VK V L framework
  • the V L framework can further comprises the framework amino acid sequence encoded by the human germline J ⁇ l, J K 2, J K 3, J K 4, or J K 5 immunoglobulin gene segment.
  • the universal framework can be a V H framework, such as a framework that comprises the framework amino acid sequences encoded by the human germline DP4, DP7, DP8, DP9, DPlO, DP31, DP33, DP38, DP45, DP46, DP47, DP49, DP50, DP51, DP53, DP54, DP65, DP66, DP67, DP68 or DP69 immunoglobulin gene segment.
  • the V H framework can further comprises the framework amino acid sequence encoded by the human germline JHI, JH2, JH3, J H 4, J H 4b, J R 5 and J H 6 immunoglobulin gene segment.
  • the dAb monomer ligand comprises the DPK9 VL framework, or a V H framework selected from the group consisting of DP47, DP45 and DP38.
  • the dAb monomer can comprises a binding site for a generic ligand, such as protein A, protein L and protein G.
  • the dAb monomer comprises one or more framework regions comprising an amino acid sequence that is the same as the amino acid sequence of a corresponding framework region encoded by a human germline antibody gene segment, or the amino acid sequences of one or more of said framework regions collectively comprise up to 5 amino acid differences relative to the amino acid sequence of said corresponding framework region encoded by a human germline antibody gene segment.
  • FW4 of the dAb monomer are the same as the amino acid sequences of corresponding framework regions encoded by a human germline antibody gene segment, or the amino acid sequences of FWl, FW2, FW3 and FW4 collectively contain up to 10 amino acid differences relative to the amino acid sequences of corresponding framework regions encoded by said human germline antibody gene segment.
  • the dAb monomer comprises FWl, FW2 and FW3 regions, and the amino acid sequence of said FWl, FW2 and FW3 regions are the same as the amino acid sequences of corresponding framework regions encoded by human germline antibody gene segments.
  • the dAb monomer does not comprise a Camelid immunoglobulin variable domain, or one or more framework amino acids that are unique to immunoglobulin variable domains encoded by Camelid germline antibody gene segments.
  • the ligands (e.g., dAb monomers) of the invention are efficacious in models of diseases when an effective amount is administered.
  • an effective amount is about 1 mg/kg to about 10 mg/kg (e.g., about 1 mg/kg, about 2 mg/kg, about 3 mg/kg, about 4 mg/kg, about 5 mg/kg, about 6 mg/kg, about 7 mg/kg, about 8 mg/kg, about 9 mg/kg, or about 10 mg/kg).
  • the models of chronic inflammatory disease described herein are recognized by those skilled in the art as being predictive of therapeutic efficacy in humans.
  • the prior art does not suggest using ligands that comprise a moiety that binds an endogenous target compound, such as a soluble cytokine receptor (e.g., soluble TNFRl) in these models, or that they would be efficacious.
  • a soluble cytokine receptor e.g., soluble TNFRl
  • the ligand or dAb monomer is efficacious in the standard mouse collagen-induced arthritis model.
  • administering an effective amount of the ligand can reduce the average arthritic score of the summation of the four limbs in the standard mouse collagen-induced arthritis model, for example, by about 1 to about 16, about 3 to about 16, about 6 to about 16, about 9 to about 16, or about 12 to about 16, as compared to a suitable control.
  • administering an effective amount of the ligand can delay the onset of symptoms of arthritis in the standard mouse collagen-induced arthritis model, for example, by about 1 day, about 2 days, about 3 days, about 4 days, about 5 days, about 6 days, about 7 days, about 10 days, about 14 days, about 21 days or about 28 days, as compared to a suitable control.
  • administering an effective amount of the ligand can result in an average arthritic score of the summation of the four limbs in the standard mouse collagen-induced arthritis model of 0 to about 3, about 3 to about 5, about 5 to about 7, about 7 to about 15, about 9 to about 15, about 10 to about 15, about 12 to about 15, or about 14 to about 15.
  • the ligand or dAb monomer is efficacious in the mouse ⁇ ARE model of arthritis.
  • administering an effective amount of the ligand can reduce the average arthritic score in the mouse ⁇ ARE model of arthritis, for example, by about 0.1 to about 2.5, about 0.5 to about 2.5, about 1 to about 2.5, about 1.5 to about 2.5, or about 2 to about 2.5, as compared to a suitable control.
  • administering an effective amount of the ligand can delay the onset of symptoms of arthritis in the mouse ⁇ ARE model of arthritis by, for example, about 1 day, about 2 days, about 3 days, about 4 days, about 5 days, about 6 days, about 7 days, about 10 days, about 14 days, about 21 days or about 28 days, as compared to a suitable control.
  • administering an effective amount of the ligand can result in an average arthritic score in the mouse ⁇ ARE model of arthritis of 0 to about 0.5, about 0.5 to about 1, about 1 to about 1.5, about 1.5 to about 2, or about 2 to about 2.5.
  • the ligand or dAb monomer is efficacious in the mouse ⁇ ARE model of inflammatory bowel disease (IBD).
  • IBD inflammatory bowel disease
  • administering an effective amount of the ligand can reduce the average acute and/or chronic inflammation score in the mouse ⁇ ARE model of IBD, for example, by about 0.1 to about 2.5, about 0.5 to about 2.5, about 1 to about 2.5, about 1.5 to about 2.5, or about 2 to about 2.5, as compared to a suitable control.
  • administering an effective amount of the ligand can delay the onset of symptoms of IBD in the mouse ⁇ ARE model of IBD by, for example, about 1 day, about 2 days, about 3 days, about 4 days, about 5 days, about 6 days, about 7 days, about 10 days, about 14 days, about 21 days or about 28 days, as compared to a suitable control, hi another example, administering an effective amount of the ligand can result in an average acute and/or chronic inflammation score in the mouse ⁇ ARE model of IBD of 0 to about 0.5, about 0.5 to about 1, about 1 to about 1.5, about 1.5 to about 2, or about 2 to about 2.5.
  • the ligand or dAb monomer is efficacious in the mouse dextran sulfate sodium (DSS) induced model of IBD.
  • DSS dextran sulfate sodium
  • administering an effective amount of the ligand can reduce the average severity score in the mouse DSS model of IBD, for example, by about 0.1 to about 2.5, about 0.5 to about 2.5, about 1 to about 2.5, about 1.5 to about 2.5, or about 2 to about 2.5, as compared to a suitable control.
  • administering an effective amount of the ligand can delay the onset of symptoms of IBD in the mouse DSS model of IBD by, for example, about 1 day, about 2 days, about 3 days, about 4 days, about 5 days, about 6 days, about 7 days, about 10 days, about 14 days, about 21 days or about 28 days, as compared to a suitable control.
  • administering an effective amount of the ligand can result in an average severity score in the mouse DSS model of IBD of 0 to about 0.5, about 0.5 to about 1, about 1 to about 1.5, about 1.5 to about 2, or about 2 to about 2.5.
  • the ligand or dAb monomer is efficacious in the mouse tobacco smoke model of chronic obstructive pulmonary disease (COPD).
  • COPD chronic obstructive pulmonary disease
  • COPD chronic obstructive pulmonary disease
  • the ligand of dAb monomer are efficacious in an animal model of asthma (see, Coffman RL et al, J. Exp. Med.
  • the ligand or dAb monomer is secreted in a quantity of at least about 0.5 mg/L when expressed in E. coli or in Pichia species (e.g., P. pastoris).
  • the dAb monomer is secreted in a quantity of at least about 0.75 mg/L, at least about 1 mg/L, at least about 4 mg/L, at least about 5 mg/L, at least about 10 mg/L, at least about 15 mg/L, at least about 20 mg/L, at least about 25 mg/L, at least about 30 mg/L, at least about 35 mg/L, at least about 40 mg/L, at least about 45 mg/L, or at least about 50 mg/L, or at least about 100 mg/L, or at least about 200 mg/L, or at least about 300 mg/L, or at least about 400 mg/L, or at least about 500 mg/L, or at least about 600 mg/L, or at least about 700 mg/L, or at least about 800 mg/L, at
  • the dAb monomer is secreted in a quantity of at least about 1 mg/L to at least about lg/L, at least about 1 mg/L to at least about 750 mg/L, at least about 100 mg/L to at least about 1 g/L, at least about 200 mg/L to at least about 1 g/L, at least about 300 mg/L to at least about 1 g/L, at least about 400 mg/L to at least about 1 g/L, at least about 500 mg/L to at least about lg/L, at least about 600 mg/L to at least about 1 g/L, at least about 700 mg/L to at least about 1 g/L, at least about 800 mg/L to at least about lg/L, or at least about 900 mg/L to at least about lg/L when expressed in E.
  • the ligands and dAb monomers described herein can be secretable when expressed in E. coli or in Pichia species (e.g., P. pastoris), they can be produced using any suitable method, such as synthetic chemical methods or biological production methods that do not employ E. coli or Pichia species.
  • the ligand is an antibody that has binding specificity for an endogenous target compound or an antigen-binding fragment thereof, such as an Fab fragment, Fab' fragment, F(ab') 2 fragment or Fv fragment (e.g., scFV).
  • the antagonist or ligand is monovalent, such as a dAb or a monovalent antigen-binding fragment of an antibody, such as an Fab fragment, Fab' fragment, or Fv fragment.
  • a domain that comprises the CDRs of a dAb that binds an endogenous target compound e.g., CDRs grafted onto a suitable protein scaffold or skeleton, eg an affibody, an SpA scaffold, an LDL receptor class A domain or an ⁇ GF domain
  • an endogenous target compound e.g., CDRs grafted onto a suitable protein scaffold or skeleton, eg an affibody, an SpA scaffold, an LDL receptor class A domain or an ⁇ GF domain
  • a suitable protein scaffold or skeleton e.g an affibody, an SpA scaffold, an LDL receptor class A domain or an ⁇ GF domain
  • the domain is selected from an affibody, an SpA domain, an LDL receptor class A domain or an ⁇ GF domain.
  • the disclosure as a whole is to be construed accordingly to provide disclosure of antagonists, ligands and methods using such domains in place of a dAb.
  • the ligands of the invention can be formatted (e.g., extended half-life formats) as described herein.
  • the invention relates to ligands that comprise a moiety (e.g., dAb) that has a binding site with binding specificity for an endogenous target compound but do not substantially inhibit the activity of said endogenous target compound.
  • a moiety e.g., dAb
  • the ligand does not bind to the active site of an endogenous target compound.
  • the invention relates to compositions comprising a ligand that comprise a moiety (e.g., dAb) that has a binding site with binding specificity for an endogenous target compound, but does not substantially inhibit the activity of said endogenous target compound, and a pharmaceutically acceptable carrier, diluent or excipient.
  • a pharmaceutically acceptable carrier e.g., diluent or excipient.
  • the invention also relates to therapeutic and diagnostic methods that employ the ligands or compositions of the invention.
  • the ligands described herein may be used for in vivo therapeutic and prophylactic applications, in vivo diagnostic applications and the like.
  • the ligands e.g., dAb monomers
  • the antagonists and/or ligands can be administered to treat, suppress or prevent a inflammatory diseases (e.g., acute and/or chronic inflammation), cancers and neoplasms (e.g., lymphomas (e.g., B cell lymphomas, T cell lymphomas, acute myeloid lymphoma), multiple myeloma, lung cancer, carcinomas), metabolic diseases (e.g., diabetes, obesity), allergic hypersensitivity, bacterial or viral infection, autoimmune disorders (which include, but are not limited to, Type I diabetes, asthma, multiple sclerosis, arthritis (e.g., rheumatoid arthritis, juvenile rheumatoid arthritis, psoriatic arthritis, spondylarthropathy (e.g., ankylosing spondylitis
  • a inflammatory diseases e.g., acute and/or chronic inflammation
  • cancers and neoplasms e.g.,
  • prevention involves administration of the protective composition prior to the induction of the disease.
  • suppression refers to administration of the composition after an inductive event, but prior to the clinical appearance of the disease.
  • Treatment involves administration of the protective composition after disease symptoms become manifest.
  • the invention relates to the use of a ligand, as described herein, for the manufacture of a medicament that can be administered to a patient to harness and exploit the beneficial activities of endogenous compounds to produce beneficial effects (e.g., therapeutically beneficial effects, diagnostically beneficial effects).
  • beneficial effects e.g., therapeutically beneficial effects, diagnostically beneficial effects.
  • the invention relates to use of a ligand for the manufacture of a medicament for increasing the amount of said endogenous target compound in a subject (e.g., the amount of the endogenous target compound in the systemic circulation, the amount at a particular site, such as a particular organ, tissue or region (e.g., in a joint, in the lung, in a muscle, in site of the skin, in the CNS)), for increasing the bioavailability said endogenous target compound in a subject (e.g., systemically or at a desired site of action (e.g., in a joint, in the lung, in a muscle, in site of the skin, in the CNS)), for increasing the in vivo half-life of said endogenous target compound, or for treating a disease that is amenable to treatment with an endogenous target compound (e.g., such as a disease as described herein), for example.
  • the medicament can be for systemic or local administration.
  • the invention relates to the use of a ligand comprising a moiety that has a binding site for an endogenous target compound for the manufacture of a medicament for increasing the half-life of said endogenous target compound in a subject.
  • the medicament can be administered to a patient to increase the half-life of an endogenous target compound to which the ligand binds by, for example, by binding the endogenous target compound and thereby increasing the hydrodynamic size of the endogenous target compound, by binding and stabilizing the endogenous target compound, or by binding and slowing the rate of clearance or metabolism of the endogenous target compound.
  • the ligand will increase the half-life of the endogenous target compound to which it binds by a factor of at least about 1.5, at least about 2, at least about 3, at least about 4, at least about 5, at least about 6, at least about 7, at least about 8, at least about 9, or at least about 10, relative to the half-life of the endogenous target compound in the absence of ligand.
  • an endogenous target compound can have a half-life of about 1 hour, and after administration of a ligand that comprises a moiety with a binding site that binds said endogenous target compound, the half-life of endogenous target compound can be increased to about 1.5 hours, to about 10 hours, or longer.
  • the invention relates to use of a ligand comprising a moiety that has a binding site for an endogenous target compound for the manufacture of a medicament for increasing the amount of said endogenous target compound in a subject.
  • the level or amount of endogenous target compound can increase following administration of a ligand, as described herein, for example, due to the ligands half- life increasing effect.
  • the level or amount of endogenous target ligand e.g., in serum
  • 1 hour, or 2 hours, or 3 hours, or 4 hours, or 5 hours, or 6 hours, or 7 hours, or 8 hours, or 9 hours, or 10 hours, or 11 hours, or 12 hours, or 1 day after administration of a ligand of the invention can be increased by a factor of at least about 1.5, at least about 2, at least about 3, at least about 4, at least about 5, at least about 10, at least about 50, at least about 100, at least about 500, at least about 1,000, at least about 5,000, at least about 10,000, at least about 50,000, or at least about 100,000.
  • Increase levels or amounts of an endogenous target compound can be readily detected using any suitable method.
  • a suitable sample e.g., serum
  • the sample can be, for example, a sample of blood, serum, cerebrospinal fluid, synovial fluid, bronchoalveolar lavage, enema or a biopsy.
  • the amount or level of ligand in such a sample is increased, relative to the amount or level before the ligand is administered.
  • the invention relates to use of a ligand comprising a moiety that has a binding site for an endogenous target compound for the manufacture of a medicament for increasing the bioavailability said endogenous target compound in a subject.
  • the medicament can be administered to a subject to increase bioavailability of an endogenous target compound systemically, or at a desired site of action (e.g., at a site where the ligand is locally administered).
  • a ligand may bind an endogenous target compound that has beneficial activity in the systemic circulation but is rapidly distributed into the tissues or cleared (e.g., a steroid or peptide hormone), and slow the rate of distribution or clearance.
  • Such a ligand can increase bioavailability of the endogenous target compound in the systemic circulation where the beneficial activity of the endogenous target compound can have a beneficial (e.g., therapeutic) effect.
  • a ligand can increases the bioavailability of an endogenous target compound at a desired site by recruiting the endogenous target compound to that site.
  • a ligand comprising a binding site for a desired endogenous target compound can be locally administered to a patient (e.g., as a depot formulation). The locally administered ligand can bind the desired endogenous target compound, and recruit additional endogenous target compound to the site.
  • an increased level or amount of endogenous target compound can be present at the site to produce beneficial effects.
  • the ligand comprises a moiety with a binding site for TGF beta isoform 3.
  • TGF beta isoform 3 can be locally administered to bind TGF beta isoform 3 and recruit TGF beta isoform 3 to the site of local administration, and thus promote would healing.
  • the ligand will increase bioavailability (e.g., in the systemic circulation, at a desired site of action) of the endogenous target compound to which it binds by a factor of at least about 1.5, at least about 2, at least about 3, at least about 4, at least about 5, at least about 6, at least about 7, at least about 8, at least about 9, or at least about 10, relative to the bioavailability of the endogenous target compound in the absence of ligand.
  • a concentration time curve for the endogenous ligand starting at the time of administration of the ligand can be prepared, and the curve and area under the curve (AUC) can be compared to normal systemic levels of the endogenous target ligand to determine the extent of increase in systemic bioavailability.
  • the invention relates to use of a ligand comprising a moiety that has a binding site for an endogenous target compound for the manufacture of a medicament for increasing the activity of said endogenous target.
  • the ligands can increase the activity (e.g., binding activity) of the endogenous targets to which they bind.
  • a ligand may bind an endogenous target compound such as an endogenous agonist (e.g., a cytokine) or a soluble cytokine receptor and improve the binding activity of the endogenous target compound by increasing the affinity and/or avidity of the endogenous target compound for its endogenous binding partner.
  • Ligands that comprises two or more moieties that have binding sites with binding specificity for a desired endogenous target compound can bind two or more individual endogenous target compound molecules thereby producing an endogenous target compound dimer, trimer, oligomer or multimer. Such a dimer, trimer, oligomer or multimer will have improved binding activity toward, for example, a cell that expresses the natural binding partner for the endogenous target compound due to higher avidity.
  • such a ligand can improve the binding activity of the endogenous target compound.
  • the binding strength (e.g., affinity or avidity) of an endogenous target compound dimer for its binding partner can be at least about 10, at least about 100, at least about 1,000 or at least about 10,000 times stronger than the binding strength of the endogenous target compound as a monomer (i.e., not in a complex with a ligand of the invention).
  • the ligand comprises two or more moieties (dAbs) that have binding specificity for soluble TNFRl but do not bind the active site (the active sire of soluble TNFRl is contained within Domains 2 and 3).
  • dAbs moieties
  • Such a ligand can bind two or more soluble TNFRl chains to form a soluble TNFRl dimer, timer, oligomer or multimer, that has improved binding activity toward its trimeric ligand, TNF, due to increased avidity.
  • such a ligand can improve the TNF-inhibiting activity of soluble TNFRl and improve its therapeutic efficacy.
  • such a ligand can improve the efficacy of soluble TNFRl by a factor of at least about 10, at least about 100, at least about 1,000 or at least about 10,000.
  • a ligand that is a dimer of a dAb that binds Domain 1 of mouse TNFRl is used to demonstrate that ligands that induce dimerization of soluble TNFRl improve efficacy of soluble TNFRl .
  • a ligand can be added to an assay containing human MRC5 cells, human TNF and soluble mouse TNFRl . Soluble mouse TNFRl will compete with human TNFRl on the MRC5 cells for binding to human TNF.
  • the ligand that is a dimer of a dAb that binds Domain 1 of mouse TNFRl binds two soluble mouse TNFRl chains to form a mouse TNFRl dimer that will be much more effective in inhibiting the effects of TNF in the assay.
  • the IC 50 for mouse TNFRl can be reduced from the nanomolar range to the picomolar range (about 10 or about 100 or about 1000 fold reduction) by the addition of the ligand that induces dimerization of soluble TNFRl.
  • ligand that induce dimerization (or trimerization or oligomeriztion) of soluble receptor chains does not substantially agonize cell surface or transmembrane forms of the receptor in a standard cell assay (i.e., when present at a concentration of 1 nM, 10 nM, 100 nM, 1 ⁇ M, 10 ⁇ M, 100 ⁇ M, 1000 ⁇ M or 5,000 ⁇ M, results in no more than about 5% of the receptor-mediated activity induced by natural ligand of the receptor in the assay).
  • the invention relates to the use of a ligand comprising two or more moieties that have a binding site for an endogenous target compound for the manufacture of a medicament for increasing the binding activity of said endogenous target compound, wherein said ligand binds said endogenous target, does not bind the active site of said endogenous target, and does not substantially inhibit the binding activity of said endogenous target compound.
  • the invention also relates to a ligand that binds an endogenous target compound having activity suitable for treating a disease in a subject, wherein said ligand does not bind the active site of said endogenous target compound or substantially inhibit the activity of said endogenous target compound, for use in therapy of a disease amenable to treatment with said endogenous target compound.
  • the ligand can increase the in vivo half-life of said endogenous target compound, increase the amount of said endogenous target compound in a subject, increase the bioavailability of said endogenous target compound, and/or increase the binding activity of said endogenous compound.
  • the invention relates to use of a ligand comprising a binding moiety that has a binding site for an endogenous target compound for increasing the half-life, bioavailability or activity of said endogenous compound, wherein said binding moiety that has a bindng site for an endogenous compound is said endogenous compound or a portion or variant thereof, and wherein said ligand binds said endogenous target compound and does not substantially inhibit the activity of said endogenous target compound.
  • the invention relates to use of a ligand comprising a binding moiety that has a binding site for a member of the TNF receptor superfamily for increasing the half- life, bioavailability or activity of said said member of the TNF receptor superfamily, wherein said ligand binds said member of the TNF receptor superfamily and does not substantially inhibit the activity of said member of the TNF receptor superfamily, and wherein said binding moiety that has a binding site for a member of the TNF receptor superfamily is a pre-ligand assembly domain (PLAD) or a variant thereof.
  • PLAD pre-ligand assembly domain
  • the invention relates to a method for increasing the half-life of an endogenous target compound in a subject, comprising administering to a subject in need thereof an effective amount of a ligand comprising a moiety that has a binding site with binding specificity for said endogenous target compound.
  • the invention relates to a method for increasing the amount of an endogenous target compound in a subject, comprising administering to a subject in need thereof an effective amount of a ligand comprising a moiety that has a binding site with binding specificity for said endogenous target compound.
  • the invention relates to a method for increasing the bioavailability of an endogenous target compound in a subject, comprising administering to a subject in need thereof an effective amount of a ligand comprising a moiety that has a binding site with binding specificity for said endogenous target compound.
  • the invention relates to a method for increasing the activity (e.g., binding activity) of an endogenous target compound in a subject, comprising administering to a subject in need thereof an effective amount of a ligand comprising a moiety that has a binding site with binding specificity for said endogenous target compound.
  • the invention relates to a method for treating a subject having a disease that is amenable to treatment with an endogenous target compound in a subject, comprising administering to a subject in need thereof an effective amount of a ligand comprising a moiety that has a binding site with binding specificity for said endogenous target compound.
  • the invention relates to a method for increasing the half-life, bioavailability or activity of an endogenous target compound, comprising administering to a subject in need thereof an effective amount of a ligand comprising a moiety that has a binding site with binding specificity for said endogenous target compound, wherein said binding moiety that has a bindng site for an endogenous compound is said endogenous compound or a portion or variant thereof, and wherein said ligand binds said endogenous target compound and does not substantially inhibit the activity of said endogenous target compound.
  • the invention relates to a method for increasing the half-life, bioavailability or activity of for a member of the TNF receptor superfamily, comprising administering to a subject in need thereof an effective amount of a ligand comprising a binding moiety that has a binding site for a member of the TNF receptor superfamily, wherein said ligand binds said member of the TNF receptor superfamily and does not substantially inhibit the activity of said member of the TNF receptor superfamily, and wherein said binding moiety that has a binding site for a member of the TNF receptor superfamily is a pre-ligand assembly domain (PLAD) or a variant thereof.
  • PLAD pre-ligand assembly domain
  • ligands of the invention involve the administration of ligands according to the invention to a recipient mammal, such as a human.
  • Dual-specific and Multi-specific ligands e.g., dual-specific antibody formats
  • Dual- or Multispecific ligands can allow the cross-linking of two antigens, for example to produce high avidity dimers, trimers or multimers of soluble receptors.
  • Substantially pure ligands of at least 90 to 95% homogeneity are preferred for administration to a mammal, and 98 to 99% or more homogeneity is most preferred for pharmaceutical uses, especially when the mammal is a human.
  • the ligands may be used diagnostically or therapeutically (including extracorporeally) or in developing and performing assay procedures, immunofluorescent stainings and the like (Lefkovite and Pernis, (1979 and 1981) Immunological Methods, Volumes I and II, Academic Press, NY).
  • the ligands will be utilised in purified form together with pharmacologically appropriate carriers.
  • these carriers include aqueous or alcoholic/aqueous solutions, emulsions or suspensions, any including saline and/or buffered media.
  • Parenteral vehicles include sodium chloride solution, Ringer's dextrose, dextrose and sodium chloride and lactated Ringer's.
  • Suitable physiologically-acceptable adjuvants, if necessary to keep a polypeptide complex in suspension may be chosen from thickeners such as carboxymethylcellulose, polyvinylpyrrolidone, gelatin and alginates.
  • Intravenous vehicles include fluid and nutrient replenishers and electrolyte replenishers, such as those based on Ringer's dextrose. Preservatives and other additives, such as antimicrobials, antioxidants, chelating agents and inert gases, may also be present (Mack (1982) Remington's Pharmaceutical Sciences, 16th Edition). A variety of suitable formulations can be used, including extended release formulations.
  • the route of administration of pharmaceutical compositions according to the invention may be any of those commonly known to those of ordinary skill in the art.
  • the selected ligands thereof of the invention can be administered to any patient in accordance with standard techniques.
  • the administration can be by any appropriate mode, including parenterally, intravenously, intramuscularly, intraperitoneally, transdermally, via the pulmonary route, or also, appropriately, by direct infusion with a catheter.
  • Parenteral administration can also be by intraarterial, intrathecal, intraarticular, subcutaneous, or other suitable injection or infusion. Additional suitable modes of administration include pulmonary administration, intranasal administration, intravaginal, intrarectal administration (e.g., by suppository or enema).
  • the dosage and frequency of administration will depend on the age, sex and condition of the patient, concurrent administration of other drugs, counterindications and other parameters to be taken into account by the clinician.
  • Administration can be local (e.g., local delivery to the lung by pulmonary administration, e.g., intranasal administration) or systemic as indicated.
  • the ligands of the present invention may be used as separately administered compositions or in conjunction with other agents. These can include various immunotherapeutic drugs, such as cylcosporine, methotrexate, adriamycm or cisplatinum, and immunotoxins.
  • compositions can include "cocktails" of various cytotoxic or other agents in conjunction with the antagonists (e.g., ligands) of the present invention, or even combinations of ligands according to the present invention having different specificities, such as ligands selected using different target antigens or epitopes, whether or not they are pooled prior to administration.
  • the ligand and any additional agent are administered in a manner that provides an overlap of therapeutic effect.
  • the ligand comprising a moiety that has a binding site for an endogenous target compound is administered together with a low dose of the endogenous target compound (e.g., a recombinant form of the endogenous target compound).
  • the ligand can, for example, increase the half-life or bioavailability of the endogenous target compound, or increase the activity of the endogenous target compound, rendering the low does therapeutically effective.
  • This therapeutic approach is advantageous because side effects associated with higher doses of agents for treating disease (e.g., recombinant forms of endogenous target proteins) can be avoided.
  • a ligand that has been preloaded with an endogenous target compound is administered.
  • Preloaded ligands are well suited for local administration to provide high bioavailablity of an endogenous target ligand that is normally present at sub-therapeutic levels or that is difficult to recruit to a site where the activity of the endogenous target ligand is desired.
  • Ligands according to the invention which able to bind to extracellular targets involved in endocytosis (e.g. Clathrin) can be endocytosed, enabling access to intracellular targets.
  • dual or multispecific ligands provide a means by which a binding domain (e.g., a dAb monomer) that is specificity able to bind to an intracellular target can be delivered to an intracellular environment.
  • a binding domain e.g., a dAb monomer
  • This strategy requires, for example, a dual-specific ligand with physical properties that enable it to remain functional inside the cell.
  • a well folding ligand may not need to be disulphide free.
  • dual- or multi-specific ligands may be used to target cytokines and other molecules which cooperate synergistically in therapeutic situations in the body of an organism.
  • the invention therefore provides a method for synergising the activity of two or more binding domains (e.g., dAbs) that bind cytokines or other molecules, comprising administering a dual- or multi-specific ligand capable of binding to said two or more molecules (e.g., cytokines).
  • the dual- or multi-specific ligand may be any dual- or multi- specific ligand, including a ligand composed of complementary and/or non- complementary domains, a ligand in an open conformation, and a ligand in a closed conformation.
  • this aspect of the invention relates to combinations of V H domains and V L domains, V H domains only and V L domains only.
  • Synergy in a therapeutic context may be achieved in a number of ways. For example, target combinations may be therapeutically active only if both targets are targeted by the ligand, whereas targeting one target alone is not therapeutically effective. In another embodiment, one target alone may provide some low or minimal therapeutic effect, but together with a second target the combination provides a synergistic increase in therapeutic effect.
  • Animal model systems which can be used to screen the effectiveness of the ligands in protecting against or treating the disease are available. Suitable animal model can be used, such as the models disclosed herein.
  • the ligands of this invention can be lyophilised for storage and reconstituted in a suitable carrier prior to use. This technique has been shown to be effective with conventional immunoglobulins and art-known lyophilisation and reconstitution techniques can be employed. It will be appreciated by those skilled in the art that lyophilisation and reconstitution can lead to varying degrees of antibody activity loss (e.g. with conventional immunoglobulins, IgM antibodies tend to have greater activity loss than IgG antibodies) and that use levels may have to be adjusted upward to compensate.
  • compositions containing the present ligands or a cocktail thereof can be administered for prophylactic and/or therapeutic treatments.
  • an adequate amount to accomplish at least partial inhibition, suppression, modulation, killing, or some other measurable parameter, of a population of selected cells is defined as a "therapeutically-effective dose”. Amounts needed to achieve this dosage will depend upon the severity of the disease and the general state of the patient's own immune system, but generally range from 0.005 to 5.0 mg of ligand per kilogram of body weight, with doses of 0.05 to 2.0 mg/kg/dose being more commonly used.
  • compositions containing the present ligands or cocktails thereof may also be administered in similar or slightly lower dosages, to prevent, inhibit or delay onset of disease (e.g., to sustain remission or quiescence, or to prevent acute phase).
  • onset of disease e.g., to sustain remission or quiescence, or to prevent acute phase.
  • the skilled clinician will be able to determine the appropriate dosing interval to treat, suppress or prevent disease.
  • an antagonist of TNFRl e.g., ligand
  • it can be administered up to four times per day, twice weekly, once weekly, once every two weeks, once a month, or once every two months, at a dose off, for example, about 10 ⁇ g/kg to about 80 mg/kg, about 100 ⁇ g/kg to about 80 mg/kg, about 1 mg/kg to about 80 mg/kg, about 1 mg/kg to about 70 mg/kg, about 1 mg/kg to about 60 mg/kg, about 1 mg/kg to about 50 mg/kg, about 1 mg/kg to about 40 mg/kg, about 1 mg/kg to about 30 mg/kg, about 1 mg/kg to about 20 mg/kg , about 1 mg/kg to about 10 mg/kg, about 10 ⁇ g/kg to about 10 mg/kg, about 10 ⁇ g/kg to about 5 mg/kg, about 10 ⁇ g/kg to about 2.5 mg/kg, about 1 mg/kg, about 2 mg/kg, about
  • the antagonist of TNFRl (e.g., ligand) is administered to treat, suppress or prevent a chronic inflammatory disease once every two weeks or once a month at a dose of about 10 ⁇ g/kg to about 10 mg/kg (e.g., about 10 ⁇ g/kg, about 100 ⁇ g/kg, about 1 mg/kg, about 2 mg/kg, about 3 mg/kg, about 4 mg/kg, about 5 mg/kg, about 6 mg/kg, about 7 mg/kg, about 8 mg/kg, about 9 mg/kg or about 10 mg/kg.).
  • a dose of about 10 ⁇ g/kg to about 10 mg/kg e.g., about 10 ⁇ g/kg, about 100 ⁇ g/kg, about 1 mg/kg, about 2 mg/kg, about 3 mg/kg, about 4 mg/kg, about 5 mg/kg, about 6 mg/kg, about 7 mg/kg, about 8 mg/kg, about 9 mg/kg or about 10 mg/kg.
  • the ligands can also be administered (e.g., systemically or locally), for example, at a dose of about 1 mg/day to about 10 mg/day (e.g., 10 mg/day, 9 mg/day, 8 mg/day, 7 mg/day, 6 mg/day, 5 mg/day, 4 mg/day, 3 mg/day, 2 mg/day, or 1 mg/day).
  • a dose of about 1 mg/day to about 10 mg/day e.g., 10 mg/day, 9 mg/day, 8 mg/day, 7 mg/day, 6 mg/day, 5 mg/day, 4 mg/day, 3 mg/day, 2 mg/day, or 1 mg/day.
  • the agent can be locally administered to pulmonary tissue at a dose of about 1 ⁇ g/kg/day to about 200 ⁇ g/kg/day (e.g., about 10 ⁇ g/kg/day, about 20 ⁇ g/kg/day, about 30 ⁇ g/kg/day, about 40 ⁇ g/kg/day, about 50 ⁇ g/kg/day, about 60 ⁇ g/kg/day, about 70 ⁇ g/kg/day, about 80 ⁇ g/kg/day, about 90 ⁇ g/kg/day, about 100 ⁇ g/kg/day, about 110 ⁇ g/kg/day, about 120 ⁇ g/kg/day, about 130 ⁇ g/kg/day, about 140 ⁇ g/kg/day, about 150 ⁇ g/kg/day, about 160 ⁇ g/kg/day, about 170 ⁇ g/kg/day, about 180 ⁇ g/kg/day, or about 190 ⁇ g/kg/day).
  • Treatment or therapy performed using the compositions described herein is considered “effective” if one or more symptoms are reduced (e.g., by at least 10% or at least one point on a clinical assessment scale), relative to such symptoms present before treatment, or relative to such symptoms in an individual (human or model animal) not treated with such composition or other suitable control. Symptoms will obviously vary depending upon the disease or disorder targeted, but can be measured by an ordinarily skilled clinician or technician.
  • Such symptoms can be measured, for example, by monitoring the level of one or more biochemical indicators of the disease or disorder (e.g., levels of an enzyme or metabolite correlated with the disease, affected cell numbers, etc.), by monitoring physical manifestations (e.g., inflammation, tumor size, etc.), or by an accepted clinical assessment scale, for example, the Expanded Disability Status Scale (for multiple sclerosis), the Irvine Inflammatory Bowel Disease Questionnaire (32 point assessment evaluates quality of life with respect to bowel function, systemic symptoms, social function and emotional status - score ranges from 32 to 224, with higher scores indicating a better quality of life), the Quality of Life Rheumatoid Arthritis Scale, the St.
  • biochemical indicators of the disease or disorder e.g., levels of an enzyme or metabolite correlated with the disease, affected cell numbers, etc.
  • physical manifestations e.g., inflammation, tumor size, etc.
  • an accepted clinical assessment scale for example, the Expanded Disability Status Scale (for multiple
  • a sustained (e.g., one day or more, preferably longer) reduction in disease or disorder symptoms by at least 10% or by one or more points on a given clinical scale is indicative of "effective" treatment.
  • prophylaxis performed using a composition as described herein is "effective” if the onset or severity of one or tnore symptoms is delayed, reduced or abolished relative to such symptoms in a similar individual (human or animal model) not treated with the composition.
  • a composition containing a ligand or cocktail thereof according to the present invention may be utilised in prophylactic and therapeutic settings to aid in the alteration, inactivation, killing or removal of a select target cell population in a mammal, hi addition, the selected repertoires of polypeptides described herein may be used extracorporeally or in vitro selectively to kill, deplete or otherwise effectively remove a target cell population from a heterogeneous collection of cells.
  • Blood from a mammal may be combined extracorporeally with the ligands, e.g. antibodies, cell-surface receptors or binding proteins thereof whereby the undesired cells are killed or otherwise removed from the blood for return to the mammal in accordance with standard techniques.
  • a composition containing an ligand according to the present invention may be utilised in prophylactic and therapeutic settings to aid in the alteration, inactivation, killing or removal of a select target cell population in a mammal.
  • the selected repertoires of polypeptides described herein maybe used extracorporeally or in vitro selectively to kill, deplete or otherwise effectively remove a target cell population from a heterogeneous collection of cells.
  • Blood from a mammal may be combined extracorporeally with the ligands, e.g. antibodies, cell- surface receptors or binding proteins thereof whereby the undesired cells are killed or otherwise removed from the blood for return to the mammal in accordance with standard techniques.
  • Endogenous Target Compounds Suitable endogenous target compounds include, for example, soluble cytokine receptors (e.g., soluble TNFRl, soluble TNFR2, soluble IL-I receptor, soluble IL-4 receptor, soluble IL- 13 receptor), endogenous receptor antagonists (IL- lra, IL-6ra), enzymes (e.g., beta- glucocerebrosidase (E.C.
  • soluble cytokine receptors e.g., soluble TNFRl, soluble TNFR2, soluble IL-I receptor, soluble IL-4 receptor, soluble IL- 13 receptor
  • endogenous receptor antagonists IL- lra, IL-6ra
  • enzymes e.g., beta- glucocerebrosidase (E.C.
  • blood factors e.g., Factor II, Factor VIII
  • erythropoietin human growth hormone, TPO, interferon-alpha, interferon-beta, interferon-gamma, GLP-I, OXM
  • sex hormones e.g., testosterone, estrogen
  • bone morphogenic proteins e.g., BMP-I, BMP-2, BMP-3, BMP-4, BMP-5, BMP-6, BMP-7
  • TGF- beta transforming growth factor beta
  • TGF-beta isoform 1
  • TGF-beta isoform 2
  • IL-IO IL-2
  • granulocyte macrophage colony stimulating factor granulocyte colony stimulating factor
  • insulin glucagon, GIP.
  • Suitable endogenous target compounds include cytokines, such as interleukins, interferons, colony stimulating factores and chemokines.
  • cytokines include, interleukins, ILl, IL2, IL3, IL4, IL5, IL6, IL7, IL8, IL9, ILlO, ILI l, IL12, IL13, IL14, IL15 (IL-T), IL16, IL17 5 IL17B, IL17C, IL17E, IL17F, ILl 8, IL19, IL20, IL21, IL22, IL23, IL24, IL25, IL26, IL27, IL28A, IL28B, IL29, IL30, IL-31 and IL-32, and interferons IFN-alpha, IFN-beta, IFN-delta, IFN- gamma, IFN-kappa, IFN-lambda-1
  • Suitable endogenous target compounds include chemokines such as, CCFl 8, CCLl, CCL2, CCL3, CCL4, CCL5, CCL6, CCL7, CCL8, CCL9, CCLlO, CCLI l, CCL12, CCL13, CCL14, CCL15, CCL16, CCL17, CCL18, CCL19, CCL20, CCL21, CCL22, CCL23, CCL24, CCL25, CCL26, CCL27, CCL28, CXCLl,
  • chemokines such as, CCFl 8, CCLl, CCL2, CCL3, CCL4, CCL5, CCL6, CCL7, CCL8, CCL9, CCLlO, CCLI l, CCL12, CCL13, CCL14, CCL15, CCL16, CCL17, CCL18, CCL19, CCL20, CCL21, CCL22, CCL23, CCL24, CCL25, CCL26, CCL27, CCL28
  • Suitable endogenous target compounds include colony stimulating factors (CSFs).
  • CSFs are cytokines that stimulate the proliferation of specific pluripotent stem cells of the bone marrow in adults.
  • Granulocyte-CSF G-CSF
  • M-CSF Macrophage-CSF
  • Granulocyte-macrophage-CSF GM- CSF
  • Epo is also considered a CSF as well as a growth factor, since it stimulates the proliferation of erythrocyte colony-forming units.
  • IL-3 secreted primarily from T-cells
  • multi-CSF since it stimulates stem cells to produce all forms of hematopoietic cells.
  • Suitable endogenous target compounds include Erythropoietin (Epo).
  • Epo is synthesized by the kidney and is the primary regulator of erythropoiesis. Epo stimulates the proliferation and differentiation of immature erythrocytes; it also stimulates the growth of erythoid progenitor cells (e.g. erythrocyte burst- forming and colony-forming units) and induces the differentiation of erythrocyte colony- forming units into proerythroblasts.
  • erythoid progenitor cells e.g. erythrocyte burst- forming and colony-forming units
  • IGF-I insulin growth factor
  • IGF-I is a growth factor structurally related to insulin.
  • IGF-I is the primary protein involved in responses of cells to growth hormone (GH): that is, IGF-I is produced in response to GH and then induces subsequent cellular activities, particularly on bone growth. It is the activity of IGF-I in response to GH that gave rise to the term somatomedin.
  • GH growth hormone
  • IGF-I has autocrine and paracrine activities in addition to the initially observed endocrine activities on bone.
  • the IGF-I receptor like the insulin receptor, has intrinsic tyrosine kinase activity. Owing to their structural similarities IGF-I can bind to the insulin receptor but does so at a much lower affinity than does insulin itself.
  • IGF-II Insulin-Like Growth Factor-I
  • IGF-II Insulin-Like Growth Factor-I
  • the IGF-II receptor is identical to the mannose-6-phosphate receptor that is responsible for the integration of lysosomal enzymes (which contain mannose-6-phosphate residues) to the lysosomes.
  • Suitable endogenous target compounds include tumor necrosis factor beta.
  • TNF-beta also called lymphotoxin
  • TNF-beta is characterized by its ability to kill a number of different cell types, as well as the ability to induce terminal differentiation in others.
  • One significant non-proliferative response to TNF-beta is an inhibition of lipoprotein lipase present on the surface of vascular endothelial cells.
  • the predominant site of TNF-beta synthesis is T-lymphocytes, in particular the special class of T-cells called cytotoxic T-lymphocytes (CTL cells).
  • CTL cells cytotoxic T-lymphocytes
  • the induction of TNF- beta expression results from elevations in IL-2 as well as the interaction of antigen with T-cell receptors.
  • Suitable endogenous target compounds include tumor necrosis factor alpha
  • TNF-alpha TNF-alpha
  • TNF-alpha also called lymphotoxin B, or cachectin
  • TNF- alpha shares only 36% amino acid sequence homology with TNF-beta. Yet, the tertiary structures of the two proteins are remarkably similar and both bind to TNF receptors TNFR-I and TNFR-2.
  • TNF-alpha is a trimeric protein encoded within the major histocompatibility complex. It was first identified in its 17 kd secreted form, but further research then showed that a noncleaved 27kd precursor form also existed in transmembrane form.
  • Stimulated macrophages produce 27kd TNF-alpha, which can either bind directly to TNFR-I and TNFR-2 receptors through cell-to-cell contact or undergo cleavage and bind in its soluble form.
  • the cytokine is produced by several types of cells, but especially by macrophages.
  • Low levels of TNF-alpha promote the remodeling or replacement of injured and senescent tissue by stimulating fibroblast growth.
  • Additional beneficial functions of TNF-alpha include its role in the immune response to bacterial, and certain fungal, viral, and parasitic invasions as well as its role in the necrosis of specific tumors. Lastly it acts as a key mediary in the local inflammatory immune response.
  • TNF-alpha is an acute phase protein which initiates a cascade of cytokines and increases vascular permeability, thereby recruiting macrophage and neutrophils to a site of infection. TNF-alpha secreted by the macrophage causes blood clotting which serves to contain the infection. TNF-alpha also exhibits chronic effects (eg in inflammation) and prolonged overproduction of TNF-alpha also results in a condition known as cachexia, which is characterized by anorexia, net catabolism, weight loss and anemia and which occurs in illnesses such as cancer and AIDS.
  • Suitable endogenous target compounds include cytokines and growth factors in hematopoiesis, such as, CD34, CIL (colony-inhibiting lymphokine), Daniplestim, GADS (GRB2 related adapter downstream of she), Hematopoietic cell growth factor, Hematopoietic cell kinase, Hematopoietic cell phosphatase, Hematopoietic consensus tyrosine-lacking kinase, Hematopoietic colony stimulating factor-309, Hematopoietin-1, Hematopoietin-2, Hemoregulatory peptide, HIM, Hiwi, HnudC, progenipoietin, progenipoietin-1, progenipoietin-2, progenipoietin-4, Promegapoietin, Promegapoietin-1, Promegapoietin-la,
  • CSF colony stimulating factor
  • GM-CSF GM-CSF
  • M-CSF M-CSF
  • Epo SCF(stem cell factor)
  • SCPF SCPF(stem cell proliferation factor)
  • various Interleukins ILl, IL3, IL4, IL5, IL6, ILIl, IL12
  • LIF TGF-beta
  • MIP-I -alpha TNF-alpha
  • TNF-alpha also many other low molecular weight factors
  • cytokines also many other low molecular weight factors.
  • Many of these proteins are multifunctional. They act on very early differentiation stages or at later stages; they can also act in a lineage-specific manner or may influence more than one lineage.
  • Proliferation and maturation of committed progenitors is controlled by late-acting lineage-specific factors such as Epo, M-CSF, G-CSF, and IL5.
  • Multipotential progenitors beginning active cell proliferation are regulated by several overlapping cytokines, including IL3, GM-CSF, and IL4.
  • Triggering of cycling by dormant primitive progenitors and maintenance of B-cell potential of the primitive progenitors appears to require interactions of early acting cytokines including IL6, G-CSF, ILl 1, ILl 2, LIF, and SCF.
  • hematopoietins have been classified into several subgroups.
  • type-1 hematopoietins is occasionally used to describe those factors involved in the regulation of hematopoiesis that act directly on some cell types.
  • This group includes IL3 (multi-CSF) and GM-CSF.
  • Factors that synergise with colony stimulating factors but that by themselves do not possess intrinsic colony stimulating activity have been referred to occasionally as type-2 hematopoietins. They include ILl, IL4, IL5 and IL6.
  • Type-3 hematopoietins are those modulatng the hematopoietic growth by stimulating the release of colony stimulating factors by their respective producer cells. They include ILl , IL2, TNF-beta and IFN-garnma.
  • TGF-beta transforming growth factor-beta
  • MIP- 1 -alpha macrophage inflammatory protein- 1 -alpha
  • Suitable endogenous target compounds include endogenous compounds involved in angiogenesis.
  • endogenous compounds involved in angiogenesis include, 16K Prolactin, ADAMTS-I, ADAMTS-8, Adrenomedullin, Angio-associated migratory cell protein, Angiogenin, angiogenin-related protein, Angiomodulin, Angiomotin, Angiopoietin-1, Angiopoietin-2, Angiopoietin-3, Angiopoietin-4, Angiopoietin-5, Angiopoietin-like 1, angiopoietin-like 2, angiopoietin-like 3, angiopoietin-like 4, Angiopoietin-related protein-2, Angiostatin, Angiotensin-2, Angiotropin, ARP-2, B 16-Fl melanoma autocrine motility factor, brain-specific angiogenesis inhibitor- 1, brain-specific angiogenesis inhibitor-2
  • Factors modulating growth, chemotactic behavior and/or functional activities of smooth muscle cells include Activin A , Adrenomedullin, aFGF, ANF, Angiogenin , Angiotensin-2 , Betacellulin , bFGF, CLAF , ECDGF (endothelial cell-derived growth factor ), ET(Endothelins), Factor X , Factor Xa, HB-EGF, Heart derived inhibitor of vascular cell proliferation, IFN- gamma, ILl, LDGF(Leiomyoma-derived growth factor), MCP-I, MDGF(macrophage-derived growth factor, monocyte-derived growth factor), NPY, Oncostatin M, PD-ECGF, PDGF, Prolactin, Protein S, SDGF(smooth muscle cell- derived growth factor), SDMF(Smooth muscle cell-derived migration factor), Tachykinins, TGF-beta, Thrombospond
  • Factors modulating growth, chemotactic behavior and/or functional activities of vascular endothelial cells include AcSDKP, aFGF, ANF, Angiogenin, angiomodulin, Angiotropin, AtT20-ECGF, B61 , bFGF, bFGF inducing activity, CAM-RF, ChDI, CLAF, ECGF, ECI, EDMF, EGF, EMAP-2, Neurothelin(see: EMMPRIN), Endostatin, Endothelial cell growth inhibitor, Endothelial cell-viability maintaining factor, Epo, FGF-5, IGF-2(see: Growth-promoting activity for vascular endothelial cells), HBNF, HGF, HUAF, IFN-gamma, ILl, K-FGF, LIF, MD-ECI, MECIF, NPY, Oncostatin M, PD-ECGF, PDGF, PF4, PlGF, Prolactin, TNF-alpha, TNF
  • protein factors detected initially due to some other biological activities and later shown to promote angio genesis are protein factors detected initially due to some other biological activities and later shown to promote angio genesis.
  • the list of protein factors angiogenically active in vivo includes fibroblast growth factors(see: FGF), Angiogenin, Angiopoietin-1, EGF, HGF, NPY, VEGF, TNF-alpha, TGF-beta, PD-ECGF, PDGF, IGF, IL8, Growth hormone.
  • Fibrin fragment E has been shown also to have angiogenic activity.
  • factors such as Angiopoietin-1 which do not behave as classical growth factors for endothelial cells but play a prominent role in vasculogenic and angiogenic processes.
  • PF4 and a 16 kDa fragment of Prolactin are inhibitory in vivo.
  • Suitable endogenous target compounds include endogenous compounds involved in formation and maintenance of the central and/or peripheral nervus systems, such as neurotrophic factors which enhance neuronal differentiation, induce proliferation, influence synaptic functions, and promote the survival of neurons that are normally destined to die during different phases of the development of the central and peripheral nervous system.
  • neurotrophins such as BDNF(brain-derived neurotrophic factor), NGF, NT-3 (neurotrophin-3), NT-4, NT-5, NT-6, NT-7, CNTF (ciliary neuronotrophic factor), GDNF (Glial cell line-derived neurotrophic factor), and Purpurin.
  • Growth factors such as bFGF or LIF are frequently also counted among the neurotrophins due to their trophic activities on a number of neurons.
  • BDNF, NGF and NT-3 are sometimes referred to also collectively as the NGF protein family as NGFis the founder member of this family of proteins. It has been possible, by combination of structural elements from NGF, BDNFand NT-3, to engineer the multifunctional Pan-Neurotrophin-1 (PNT-I) that efficiently activates all trk receptors and displays multiple neurotrophic specificities.
  • PNT-I Pan-Neurotrophin-1
  • NSE neuronal survival factor
  • EGF EGF
  • HBNF hep arm-binding neurite-promoting factor
  • IGF-2 IGF-2
  • aFGF and bFGF PDGF
  • NSE neuron-specific enolase
  • Activin A Activin A
  • Neural antiproliferative protein affecting neural cells see also: Neural antiproliferative protein, Astrostatine, GGIF (glial growth inhibitory factor).
  • Neurite promoting factors do not promote neuronal survival or general growth by themselves but are required in addition to one or more neurotrophic factors to induce outgrowth of axonal or dendritic processes.
  • Factors with NPF activity include NGF, S 100,
  • GMF-beta glia maturation factor
  • proteoglycans merosin
  • collagens cell adhesion molecules
  • laminin laminin
  • Suitable endogenous target compounds include endogenous compounds involved in wound healing.
  • Platelets are a rich source of locally active growth factors and cytokines .
  • platelet-derived factors include adenosine dinucleotide (which induces platelet aggregation and also stimulates cell proliferation and migration), Beta-Thromboglobulin, bFGF, CTAP-3 (Connective tissue activating protein-3), EGF, Eosinophil chemotactic polypeptide-1 (i. e.
  • RANTES f-ECGF
  • Fibroblast derived endothelial cell growth factor fibronectin (which serves as an early matrix ligand for platelet aggregation)
  • HCI human collagenase inhibitor
  • HGF hepatocyte growth factor
  • HRF histamine-releasing factors
  • IGF-BP-3 HCI
  • NAP-2 neurotrophil-activating protein-2
  • NAP-4 neutraltrophil- activating protein-4
  • PBP platelet basic protein
  • PD-ECGF platelet-derived endothelial cell growth factor
  • PDGF PF4, platelet activating factor (PAF, involved also in platelet aggregation), serotonin (which induces vascular permeability and is a chemoattractant for neutrophils), Somatostatin, TGF-alpha, TGF-beta, thromboxane A2 (which is involved in vasoconstriction, platelet aggregation, and chemotaxis ), Vitronectin.
  • the first cells to appear in the wound area are neutrophils.
  • Neutrophil numbers reach peak levels approximately 24 hours after injury. Their migration is stimulated by various chemotactic factors and cytokines, including complement factors, ILl, TNF- alpha, TGF-beta, and chemokinessuch as IL8, GRO-alpha, PF4, MCP-I, IP-IO, mig, and also by bacterial polysaccharides.
  • Neutrophils adhere to the endothelium by means of selectins, which function as neutrophil receptors on the endothelial cell surface. Integrin receptors on the neutrophil cell surfaces facilitate binding of neutrophils to the extracellular matrix.
  • Neutrophils do not appear to play a critical role in wound healing in the absence of infections as wounds can heal in animals in which neutrophils are depleted. Neutrophils are removed by tissue macrophages when they are no longer needed.
  • Monocytes appear approximately 24 hours after injury and peak at 48 hours post-injury. Since monocytes mature into macrophages they can be considered an essential source of cytokines driving repair processes. Macrophages and monocytes are also attracted by a variety of chemokines. These chemokinescontribute to the spatially and temporally different infiltration of leukocyte subsets and thus integrate inflammatory and reparative processes during wound repair. Tissue macrophages are the cells that essentially control and regulate the wound healing process and wounds cannot heal without the participation of these cells as shown by experiments involving depletion of wound macrophages. The differentiation of macrophages is initiated by several specific cytokines.
  • Macrophages also control the degradation of the extracellular matrix and regulate remodeling of the wound matrix. Macrophages secrete cytokines and growth factors including TGF-beta, FGF, VEGF, and chemokines such as JE.
  • TGF-beta appears to be the major factor responsible for the formation of granulation tissue and the synthesis of proteins of the extracellular matrix and thus has been referred to as a "wound hormone".
  • TGF-beta is a member of one of the most complex groups of cytokine superfamilies, consisting of various TGF-beta isoforms and other family members, for example, Activin A and BMP.
  • the complexity of the wound healing process is illustrated by the observation that manipulation of the ratios of TGF-beta superfamily members, particularly the ratio of TGF-beta-lrelative to TGF-beta-3, reduces scarring and fibrosis.
  • Re-epithelialization is mediated by chemotactic and mitogenic growth factors of the EGF family of growth factors.
  • Leptin has been shown to be a potent growth factor for keratinocytes during wound healing.
  • the final phase of wound healing is characterized by the gradual replacement of granulation tissues by connective tissue. This process also requires locally acting cytokines. However, little is known about the factors and mechanisms that eventually restrain tissue growth once the repair process has been completed.
  • the synthesis of collagen and proteinase inhibitors is stimulated, among other things, by TGF-beta and related factors. Closing of the wound and the evolution of a scar is associated with a striking decrease in cellularity, including disappearance of typical myofibroblasts. It has been suggested that cell death by apoptosis is the mechanism responsible for the evolution of granulation tissue into a scar.
  • Fetal wound healing is conspicuous for its absence of scarring. There is some evidence that fetal and adult fibroblasts display phenotypic differences in terms of migratory activity, moto genie response to cytokines and the synthesis of motogenic cytokines, growth factors , and matrix macromolecules.
  • cytokines By manipulating the actions of growth factord and cytokines, it is possible to accelerate or modify wound healing. Animal experiments and also clinical experience have demonstrated that the topical administration of various cytokines, including bFGF, EGF, KGF, PDGF, TGF-beta, either alone or in combination, considerably accelerates wound healing by stimulating granulation tissue formation and enhancing epithelialization
  • cytokines including bFGF, EGF, KGF, PDGF, TGF-beta, either alone or in combination, considerably accelerates wound healing by stimulating granulation tissue formation and enhancing epithelialization
  • Suitable endogenous target compounds include acute phase proteins of inflammation. Suitable examples of such proteins and their functions are provided in table 2.
  • Type-1 acute phase proteins are those that require the synergistic action of IL6 and ILl for maximum synthesis.
  • Type-1 proteins are C-reactive protein, serum amyloid A and alpha- 1 acid glycoprotein.
  • Type-2 acute phase proteins are those that require IL6 only for maximal induction.
  • Type-2 proteins are fibrinogen chains, haptoglobin, and alpha-2-Macroglobulin. Expression of genes encoding Type-2 acute phase proteins is suppressed rather than being enhanced frequently by ILl (Ramadori et al; Fey et al).
  • cytokines additive, synergistic, co ⁇ operative, and antagonistic effects between cytokines and other mediator substances influencing the expression of acute phase proteins do occur and have been observed in almost all combinations. Many cytokines also show differential effects, inducing the synthesis of one or two acute phase proteins but not others. For example, Activin A induces a subset of acute phase proteins in HepG2 cells. Bacterial lipopolysaccharides and several cytokines (mainly ILl, IL6 and TNF but also LIF, CNTF, oncostatin M, ILl 1, and cardiotrophin-1) are involved in the induction of SAA synthesis and some of these cytokines act synergistically (Benigni et al).
  • ILl and also IFN-gamma reduce some of the effects of IL6. Some of the effects of IL2 and IL6 are antagonized by TGF-beta. The combined action of two or even more cytokines may produce effects that no factor on its own would be able to achieve.
  • ILl, IL6 TNF-alpha and TGF-beta induce the synthesis of antichymotrypsin and at the same time repress the synthesis of albumin and AFP (alpha-Fetoprotein).
  • the synthesis of fibrinogen is induced by IL6 and this effect is, in turn, suppressed by ILl-alpha, TNF-alpha or TGF-beta-1.
  • Suitable endogenous target compounds include endogenous compounds involved in haemostasis and coagulation cascades. Apart from being involved in the regulation of capillary permeability and vessel tonus the endothelium of blood vessels plays a decisive role in maintaining a non-thrombogenic surface by providing activators and inhibitors of coagulation and fibrinolysis. The endothelium is also involved in modulation of various immunological processes.
  • ILl induces, among other things, the synthesis of various colony-stimulating factors, IL6, TNF-alpha, prostaglandins, platelet activating factor (PAF), plasminogen activator inhibitor (PAI).
  • PGE2 platelet activating factor
  • PAI plasminogen activator inhibitor
  • An important endogenous regulator of ILl is PGE2 which inhibits secretion of ILl and TNF-alpha.
  • TNF-alpha induces the synthesis of tissue thromboplastin (TPL) which participates in the formation of the factor X activator complex that catalyses generation of thrombin. The complex also activates factor IX.
  • TNF-alpha also induces the synthesis of ILl by endothelial cells and this ILl can also promote production of TPL by endothelial cells.
  • the synthesis of TNF-alpha can also be induced by ILl .
  • TNF-alpha and ILl also induce the synthesis of cell surface antigens.
  • the expression of adhesion molecules increases adhesion of lymphocytes and leukocytes at the vessel wall and thus facilitates transendothelial migration of these cells.
  • TNF-alpha and ILl down-regulate the Protein C inactivation system by inhibiting the synthesis of thrombomodulin.
  • Thrombomodulin complexed with thrombin activates protein C which can then form complexes with membrane-bound protein S. These complexes inhibit factor Va.
  • Activated protein C also neutralizes plasminogen activator inhibitor (PAI) by complexation.
  • TNF-alpha and also ILl thus reduce inactivation of factor Va.
  • Suitable endogenous target compounds involved in coagulation include, Kallikrein, Factor XII, Factor XIb, Factor XI, Factor XIa, Factor IX, Factor IXa, Factor VIII, Factor Villa, Factor X, Factor Xa, Factor Va, Factor XIIIa, Prothrombin, Thrombin, Fibrinogen, Fibrin, Plasminigen, Plasmin.
  • Suitable endogenous target compounds also include hormones, such as stress hormones (e.g., Adrenaline, Adrenocorticotropic hormone, Corticosterone, Epinephrine, Growth Hormone, Hydrocortisone), fluid/electrolyte and vascular hormones (e.g., Aldosterone, Androstenedione, Vasopressin, Bradykinin, Calcitonin, Neurotensin), liver and digestive hormones (e.g., Cholecystokinin, Cholesterol, VIP), reproductive hormones (e.g., Chorionic Gonadotrophin, Epogen, Estradiol, Estriol, Estrone, Etiocholanolone, FSH, Lactogenic Hormone, Lutenizing Hormone, Testosterone, Oxytocin, Progesterone, Prolactin, Gonadotrophin), sugar handling hormones (e.g., Glucagon, Insulin), thyroid hormones (e.g., T2, T3,
  • Table 4 provides a non-exhaustive list of suitable endogenous target compounds and preferred therapeutics for targeting the endogenous compound with a ligand as described herein.
  • the "Exemplary Identifier” column provides Chemical Abstracts Services (CAS) Registry Numbers (published by the American Chemical Society) and/or Genbank Accession Numbers ((e.g., Locus ID, NP_XXXXX (Reference Sequence Protein), and XP_XXXXX (Model Protein) identifiers available through the national Center for Biotechnology Information (NCBI) webpage at www.ncbi.nlm.nih.gov) that correspond to entries in the CAS Registry or Genbank database which contain an amino acid sequence of the endogenous target compount or of a fragment or variant of the endogenous target compound.
  • NCBI National Center for Biotechnology Information
  • the Summary pages associated with each of these CAS and Genbank and GenSeq Accession Numbers as well as the cited journal publications (e.g., PubMed ID number (PMID)) are each incorporated by reference in their entireties, particularly with respect to the amino acid sequences described therein.
  • the "Biological activity” column describes biological activities associated with the endogenous target molecule.
  • the "Preferred Indication” column describes disease, disorders, and/or conditions that may be treated, prevented, diagnosed, or ameliorated by a ligand that comprises a moiety that has a binding site with binding specificity for the indicated endogenous target compound.
  • the invention also relates to a ligand that comprises a moiety having a binding site with binding specificity for any of the endogenous target compounds listed in Table 4 for use in the manufacture of a medicament for treating any of the corresponding preferred indications disclosed in Table 4.
  • the invention also relates to a method for treating any of the preferred indication listed in Table 4, comprising admininstering to a subject in need thereof an effective amount of a ligand that comprises a moiety having a binding site with
  • the presented lists of endogenous target compounds is by no means exhaustive.
  • the antigen(s) may be selected from this list.
  • the ligand comprises a dAb that binds TNFRl and a second dAb or epitope binding domain that binds any one of the these antigens, hi such embodiments, the multispecific ligand can comprise any combination of immunoglobulin variable domains (e.g., V H V H , V H V L , V L V L ).
  • the invention provides a ligand or dAb monomer (e.g., dual specific ligand comprising such a dAb) that binds to serum albumin (SA) with a K ⁇ j of InM to 500 ⁇ M (ie, x 10 '9 to 5 x 10 '4 ), preferably 100 nM to 10 ⁇ M.
  • SA serum albumin
  • K ⁇ j of InM to 500 ⁇ M ie, x 10 '9 to 5 x 10 '4
  • a dual specific ligand comprising a first anti-SA dAb and a second dAb to another target,
  • the affinity (eg Kd and/or Koff as measured by surface plasmon resonance, eg using BiaCore) of the second dAb for its target is from 1 to 100000 times (preferably 100 to 100000, more preferably 1000 to 100000, or 10000 to 100000 times) the affinity of the first dAb for SA.
  • the first dAb binds SA with an affinity of approximately 10 ⁇ M
  • the second dAb binds its target with an affinity of 100 pM.
  • the serum albumin is human serum albumin (HSA).
  • the first dAb (or a dAb monomer) binds SA (eg, HSA) with a K ⁇ j of approximately 50, preferably 70, and more preferably 100, 150 or 200 nM.
  • the dAb monomer that binds SA resists aggregation, unfolds reversibly and/or comprises a framework region as described above for dAb monomers that bind TNFRl.
  • Ligands and dAb monomers can be formatted as mono or multispecific antibodies or antibody fragments or into mono or multispecific non-antibody structures.
  • Suitable formats include, any suitable polypeptide structure in which an antibody variable domain or one or more of the CDRs thereof can be incorporated so as to confer binding specificity for antigen on the structure.
  • a variety of suitable antibody formats are known in the art, such as, IgG-like formats, chimeric antibodies, humanized antibodies, human antibodies, single chain antibodies, bispecific antibodies, antibody heavy chains, antibody light chains, homodimers and heterodimers of antibody heavy chains and/or light chains, antigen-binding fragments of any of the foregoing (e.g., a Fv fragment (e.g., single chain Fv (scFv), a disulfide bonded Fv), a Fab fragment, a Fab' fragment, a F(ab') 2 fragment), a single variable domain (e.g., V H , V L , V HH ), a dAb, and modified versions of any of the foregoing (e.g., modified by the covalent attachment of polyalkylene glycol (e.g., polyethylene glycol, polypropylene glycol, polybutylene glycol) or other suitable polymer). See, PCT/GB03/002804, filed June 30, 2003, which designated the
  • ligands, including dAb monomers, dimers and trimers can be linked to an antibody Fc region, comprising one or both of C H 2 and C H 3 domains, and optionally a hinge region.
  • vectors encoding ligands linked as a single nucleotide sequence to an Fc region may be used to prepare such polypeptides.
  • Ligands and dAb monomers can also be combined and/or formatted into non-antibody multi-ligand structures to form multivalent complexes, which bind target molecules with the same antigen, thereby providing superior avidity.
  • natural bacterial receptors such as SpA can been used as scaffolds for the grafting of CDRs to generate ligands which bind specifically to one or more epitopes. Details of this procedure are described in US 5,831,012.
  • Other suitable scaffolds include those based on fibronectin and affibodies. Details of suitable procedures are described in WO 98/58965.
  • Other suitable scaffolds include lipocallin and CTLA4, as described in van den Beuken et ah, J. MoI. Biol.
  • Protein scaffolds may be combined; for example, CDRs may be grafted on to a CTLA4 scaffold and used together with immunoglobulin V H or V L domains to form a ligand. Likewise, fibronectin, lipocallin and other scaffolds may be combined
  • antibody chains and formats e.g., IgG-like formats, chimeric antibodies, humanized antibodies, human antibodies, single chain antibodies, bispecific antibodies, antibody heavy chains, antibody light chains, homodimers and heterodimers of antibody heavy chains and/or light chains
  • suitable expression constructs and/or culture of suitable cells e.g., hybridomas, heterohybridomas, recombinant host cells containing recombinant constructs encoding the format.
  • formats such as antigen-binding fragments of antibodies or antibody chains ⁇ e.g., a Fv fragment ⁇ e.g., single chain Fv (scFv), a disulfide bonded Fv), a Fab fragment, a Fab' fragment, a F(ab') 2 fragment
  • a Fv fragment ⁇ e.g., single chain Fv (scFv), a disulfide bonded Fv), a Fab fragment, a Fab' fragment, a F(ab') 2 fragment
  • the ligand can be formatted as a dual specific ligand or a multispecific ligand, for example as described in WO 03/002609, the entire teachings of which are incorporated herein by reference.
  • the dual specific ligands comprise immunoglobulin single variable domains that have different binding specificities.
  • Such dual specific ligands can comprise combinations of heavy and light chain domains.
  • the dual specific ligand may comprise a VH domain and a V L domain, which may be linked together in the form of an scFv (e.g., using a suitable linker such as Gly 4 Ser), or formatted into a bispecific antibody or antigen-binding fragment theref (e.g. F(ab') 2 fragment).
  • the dual specific ligands do not comprise complementary V H /V L pairs which form a conventional two chain antibody antigen- binding site that binds antigen or epitope co-operatively.
  • the dual format ligands comprise a V J /V L complementary pair, wherein the V domains have different bindng specificities.
  • the dual specific ligands may comprise one or more C H or C L domains if desired.
  • a hinge region domain may also be included if desired.
  • Such combinations of domains may, for example, mimic natural antibodies, such as IgG or IgM, or fragments thereof, such as Fv, scFv, Fab or F(ab') 2 molecules.
  • the dual specific ligand of the invention comprises only two variable domains although several such ligands may be incorporated together into the same protein, for example two such ligands can be incorporated into an IgG or a multimeric immunoglobulin, such as IgM.
  • a plurality of dual specific ligands are combined to form a multimer. For example, two different dual specific ligands are combined to create a terra-specific molecule.
  • variable regions of a dual-specific ligand produced according to the method of the present invention may be on the same polypeptide chain, or alternatively, on different polypeptide chains.
  • variable regions are on different polypeptide chains, then they may be linked via a linker, generally a flexible linker (such as a polypeptide chain), a chemical linking group, or any other method known in the art.
  • the multispecific ligand possesses more than one epitope binding specificity.
  • the multi-specific ligand comprises two or more epitope binding domains, such dAbs or non-antibody protein domain comprising a binding site for an epitope, e.g., an affibody, an SpA domain, an LDL receptor class A domain, an EGF domain, an avimer.
  • Multispecific ligands can be formatted further as described herein.
  • the ligand is an IgG-like format.
  • Such formats have the conventional four chain structure of an IgG molecule (2 heavy chains and two light chains), in which one or more of the variable regions (V H and or V L ) have been replaced with a dAb or single variable domain of a desired specificity.
  • each of the variable regions (2 V H regions and 2 V L regions) is replaced with a dAb or single variable domain.
  • the dAb(s) or single variable domain(s) that are included in an IgG-like format can have the same specificity or different specificities, hi some embodiments, the IgG-like format is tetravalent and can have one, two, three or four specificities.
  • the IgG-like format can be monospecific and comprises 4 dAbs that have the same specificity; bispecific and comprises 3 dAbs that have the same specificity and another dAb that has a different specificity; bispecific and comprise two dAbs that have the same specificity and two dAbs that have a common but different specificity; trispecific and comprises first and second dAbs that have the same specificity, a third dAbs with a different specificity and a fourth dAb with a different specificity from the first, second and third dAbs; or tetraspecific and comprise four dAbs that each have a different specificity.
  • Antigen- binding fragments of IgG-like formats e.g., Fab, F(ab') 2 , Fab', Fv, scFy
  • the IgG-like formats or antigen-binding fragments thereof do not crosslink TNFRl.
  • Ligands can be formatted to extend in vivo serum half life. Increased in vivo half-life is useful in in vivo applications of immunoglobulins, especially antibodies and most especially antibody fragments of small size such as dAbs. Such fragments (Fvs, disulphide bonded Fvs, Fabs, scFvs, dAbs) are rapidly cleared from the body, which can severely limit clinical applications.
  • An ligand e.g., a dAb monomer
  • a polyalkyleneglycol group e.g. polyethyleneglycol (PEG) group, polypropyleneglycol group
  • serum albumin e.g. polyethyleneglycol (PEG) group, polypropyleneglycol group
  • transferrin transferrin receptor
  • the ligand is PEGylated.
  • the PEGylated ligand binds an endogenous target compound with substantially the same affinity as the same ligand that is not PEGylated.
  • the ligand can be a PEGylated dAb monomer that binds an endogenous target compound, wherein the PEGylated dAb monomer binds said endogenous target compound with an affinity that differs from the affinity of dAb in unPEGylated form by no more than a factor of about 1000, preferably no more than a factor of about 100, more preferably no more than a factor of about 10, or with affinity substantially unchanged affinity relative to the unPEGylated form.
  • Small ligands such as a dAb monomer
  • a dAb monomer can be formatted as a larger antigen- binding fragment of an antibody or as and antibody (e.g., formatted as a Fab, Fab', F(ab) 2 , F(ab') 2 , IgG, scFv).
  • the hydrodynaminc size of an antagonist (e.g., ligand, dAb monomer) and its serum half-life can also be increased by conjugating or linking the antagonist to a binding domain (e.g., antibody or antibody fragment) that binds an antigen or epitope that increases half-live in vivo, as described herein.
  • the ligand e.g., dAb monomer
  • an anti ⁇ serum albumin or anti-neonatal Fc receptor antibody or antibody fragment e.g an anti-SA or anti-neonatal Fc receptor dAb, Fab, Fab' or scFv, or to an anti-SA affibody or anti-neonatal Fc receptor affibody.
  • Hydrodynamic size of the ligands (e.g., dAb monomers and multimers) of the invention may be determined using methods which are well known in the art. For example, gel filtration chromatography may be used to determine the hydrodynamic size of a ligand. Suitable gel filtration matrices for determining the hydrodynamic sizes of ligands, such as cross-linked agarose matrices, are well known and readily available.
  • the size of a ligand format e.g., the size of a PEG moiety attached to a bining moiety that has a binding site for an endogenous target compound, can be varied depending on the desired application.
  • ligand is intended to leave the circulation and enter into peripheral tissues, it is desirable to keep the hydrodynamic size of the ligand low to facilitate extravazation from the blood stream.
  • size of the ligand can be increased, for example by formatting an and Ig like protein or by addition of a 30 to 60 kDa PEG moiety.
  • suitable albumin, albumin fragments or albumin variants for use in ligand according to the invention are described in WO 2005/077042A2, which is incorporated herein by reference in its entirety.
  • albumin, albumin fragments or albumin variants can be used in the present invention:
  • Albumin fragment or variant comprising or consisting of amino acids 1-387 of SEQ ID NO:1 in WO 2005/077042A2;
  • Albumin or fragment or variant thereof, comprising an amino acid sequence selected from the group consisting of: (a) amino acids 54 to 61 of SEQ ID NO: 1 in WO 2005/077042A2; (b) amino acids 76 to 89 of SEQ ID NO: 1 in
  • WO 2005/077042A2 amino acids 92 to 100 of SEQ ID NO:1 in WO 2005/077042A2; (d) amino acids 170 to 176 of SEQ ID NO:1 in WO 2005/077042A2; (e) amino acids 247 to 252 of SEQ ID NO:1 in WO 2005/077042A2; (f) amino acids 266 to 277 of SEQ ID NO:1 in WO 2005/077042A2; (g) amino acids 280 to 288 of SEQ ID NO: 1 in WO
  • albumin, fragments and analogs for use in a ligand according to the invention are described in WO 03/076567 A2, which is incorporated herein by reference in its entirety.
  • albumin, fragments or variants can be used in the present invention:
  • HA Human serum albumin
  • a (one or more) half-life extending moiety eg, albumin, transferrin and fragments and analogues thereof
  • it can be conjugated using any suitable method, such as, by direct fusion to the binding moiety that has a binding site for an endogenous target compound, for example by using a single nucleotide construct that encodes a fusion protein, wherein the fusion protein is encoded as a single polypeptide chain with the half-life extending moiety located N- or C-terminally to the TNFRl binding moiety.
  • conjugation can be achieved by using a peptide linker between moieties, eg, a peptide linker as described in WO 03/076567 A2 or WO 2004/003019 (these linker disclosures being incorporated by reference in the present disclosure to provide examples for use in the present invention).
  • a peptide linker between moieties eg, a peptide linker as described in WO 03/076567 A2 or WO 2004/003019 (these linker disclosures being incorporated by reference in the present disclosure to provide examples for use in the present invention).
  • a polypeptide that enhances serum half-life in vivo is a polypeptide which occurs naturally in vivo and which resists degradation or removal by endogenous mechanisms which remove unwanted material from the organism ⁇ e.g., human).
  • a polypeptide that enhances serum half-life in vivo can be selected from proteins from the extracellular matrix, proteins found in blood, proteins found at the blood brain barrier or in neural tissue, proteins localized to the kidney, liver, lung, heart, skin or bone, stress proteins, disease-specific proteins, or proteins involved in Fc transport.
  • Suitable polypeptides that enhance serum half-life in vivo include, for example, transferrin receptor specific ligand-neuropharmaceutical agent fusion proteins (see U.S. Patent No. 5,977,307, the teachings of which are incorporated herein by reference), brain capillary endothelial cell receptor, transferrin, transferrin receptor (e.g., soluble transferrin receptor), insulin, insulin-like growth factor 1 (IGF 1) receptor, insulin-like growth factor 2 (IGF 2) receptor, insulin receptor, blood coagulation factor X, ⁇ l-antitrypsin and HNF l ⁇ .
  • transferrin receptor specific ligand-neuropharmaceutical agent fusion proteins see U.S. Patent No. 5,977,307, the teachings of which are incorporated herein by reference
  • brain capillary endothelial cell receptor transferrin, transferrin receptor (e.g., soluble transferrin receptor)
  • insulin insulin-like growth factor 1
  • IGF 2 insulin
  • Suitable polypeptides that enhance serum half-life also include alpha- 1 glycoprotein (orosomucoid; AAG), alpha- 1 antichymotrypsin (ACT), alpha- 1 microglobulin (protein HC; AIM), antithrombin III (AT III), apolipoprotein A-I (Apo A-I), apolipoprotein B (Apo B), ceruloplasmin (Cp), complement component C3 (C3), complement component C4 (C4), Cl esterase inhibitor (Cl INH), C-reactive protein (CRP), ferritin (FER), hemopexin (HPX), lipoprotein(a) (Lp(a)), mannose-binding protein (MBP), myoglobin (Myo), prealbumin (transthyretin; PAL), retinol-binding protein (RBP), and rheumatoid factor (RF).
  • alpha- 1 glycoprotein orosomucoid
  • AAG alpha- 1 antichy
  • Suitable proteins from the extracellular matrix include, for example, collagens, laminins, integrins and fibronectin.
  • Collagens are the major proteins of the extracellular matrix.
  • about 15 types of collagen molecules are currently known, found in different parts of the body, e.g. type I collagen (accounting for 90% of body collagen) found in bone, skin, tendon, ligaments, cornea, internal organs or type II collagen found in cartilage, vertebral disc, notochord, and vitreous humor of the eye.
  • Suitable proteins from the blood include, for example, plasma proteins (e.g., fibrin, ⁇ -2 microglobulin, serum albumin, fibrinogen (e.g., fibrinogen A, fibrinogen B), serum amyloid protein A, haptoglobin, profilin, ubiquitin, uteroglobulin and ⁇ -2- microglobulin), enzymes and enzyme inhibitors (e.g., plasminogen, lysozyme, cystatin C, alpha-1-antitrypsin and pancreatic trypsin inhibitor), proteins of the immune system, such as immunoglobulin proteins (e.g., IgA, IgD, IgE, IgG, IgM, immunoglobulin light chains (kappa/lambda)), transport proteins (e.g., retinol binding protein, ⁇ -1 microglobulin), defensins (e.g., beta-defensin 1, neutrophil defensin 1, neutrophil defensin
  • Suitable proteins found at the blood brain barrier or in neural tissue include, for example, melanocortin receptor, myelin, ascorbate transporter and the like.
  • Suitable polypeptides that enhances serum half-life in vivo also include proteins localized to the kidney (e.g., polycystin, type IV collagen, organic anion transporter Kl, Heymann's antigen), proteins localized to the liver (e.g., alcohol dehydrogenase, G250), proteins localized to the lung (e.g., secretory component, which binds IgA), proteins localized to the heart (e.g., HSP 27, which is associated with dilated cardiomyopathy), proteins localized to the skin (e.g., keratin), bone specific proteins such as morphogenic proteins (BMPs), which are a subset of the transforming growth factor ⁇ superfamily of proteins that demonstrate osteogenic activity (e.g., BMP-2, BMP-4, BMP-5, BMP-6, BMP-7
  • Suitable disease-specific proteins include, for example, antigens expressed only on activated T-cells, including LAG-3 (lymphocyte activation gene), osteoprotegerin ligand (OPGL; see Nature 402, 304-309 (1999)), OX40 (a member of the TNF receptor family, expressed on activated T cells and specifically up- regulated in human T cell leukemia virus type-I (HTLV-I)-producing cells; see Immunol. 165 (l):263-70 (2000)).
  • LAG-3 lymphocyte activation gene
  • osteoprotegerin ligand OPGL
  • OX40 a member of the TNF receptor family, expressed on activated T cells and specifically up- regulated in human T cell leukemia virus type-I (HTLV-I)-producing cells; see Immunol. 165 (l):263-70 (2000)).
  • Suitable disease-specific proteins also include, for example, metalloproteases (associated with arthritis/cancers) including CG6512 Drosophila, human paraplegin, human FtsH, human AFG3L2, murine ftsH; and angiogenic growth factors, including acidic fibroblast growth factor (FGF-I), basic fibroblast growth factor (FGF-2), vascular endothelial growth factor/vascular permeability factor (VEGF/VPF), transforming growth factor- ⁇ (TGF ⁇ ), tumor necrosis factor-alpha (TNF- ⁇ ), angiogenin, interleukin-3 (IL-3), interleukin-8 (IL- 8), platelet-derived endothelial growth factor (PD-ECGF), placental growth factor (PlGF), midkine platelet-derived growth factor-BB (PDGF), and fractalkine.
  • metalloproteases associated with arthritis/cancers
  • FGF-I acidic fibroblast growth factor
  • FGF-2 basic fibroblast growth factor
  • Suitable polypeptides that enhance serum half-life in vivo also include stress proteins such as heat shock proteins (HSPs).
  • HSPs are normally found intracellularly. When they are found extracellularly, it is an indicator that a cell has died and spilled out its contents. This unprogrammed cell death (necrosis) occurs when as a result of trauma, disease or injury, extracellular HSPs trigger a response from the immune system. Binding to extracellular HSP can result in localizing the compositions of the invention to a disease site.
  • Suitable proteins involved in Fc transport include, for example, Brambell receptor (also known as FcRB). This Fc receptor has two functions, both of which are potentially useful for delivery.
  • the functions are (1) transport of IgG from mother to child across the placenta (2) protection of IgG from degradation thereby prolonging its serum half-life. It is thought that the receptor recycles IgG from endosomes. (See, Holliger et al, Nat Biotechnol 15(7):632-6 (1997).)
  • Assays for Assessing Endogenous Target Compound Function The following assays, suitable variations thereof and other suitable assays can be used to assess activity of endogenous target compounds, and to assess the whether a ligand substantially inhibits the activity of an endogenous target compound to which it binds.
  • ABC-I function may be assayed by measuringapolipoprotein-mediatedlipid efflux from cultured cells (J Clin Invest 1999Oct; 104(8): R25-31). aFGFl- pseudomonas exotoxin chimera function may be assayed in vitro using a cytotoxicity assay (Proc Natl Acad Sci USA 1989 Jun; 86(11): 4215-4219).
  • rnhibin A activity may be assayed in vitro by measuring its differentiation-inducing activity toward mouse Friend erythroleukemia (MEL) cells and human K-562 cells (Proc Natl Acad Sci USA 1988 Apr; 85(8): 2434-2438); or its suppression of the secretion of pituitary follicle-stimulating hormone (Biol Reprod 2000 Sep; 63(3): 865-871).
  • MEL mouse Friend erythroleukemia
  • K-562 Proc Natl Acad Sci USA 1988 Apr; 85(8): 2434-2438
  • Biol Reprod 2000 Sep; 63(3): 865-871 Biol Reprod 2000 Sep; 63(3): 865-871.
  • Inhibin beta C activity may be assayed in vitro by measuring its differentiation- inducing activity toward mouse Friend erythroleukemia (MEL) cells and human K- 562 cells (Proc Natl Acad Sci USA 1988 Apr; 85(8): 2434-2438); or its suppression of the secretion of pituitary follicle-stimulating hormone (Biol Reprod 2000 Sep; 63(3): 865-871).
  • Adenosine deaminase activity may be assayed in vitro by measuring purine catabolism (MoI Cell Biol 1985 Apr; 5(4): 762-767).
  • Adiposin function may be assayed in vitro by measuring cAMP accumulation in adiposin stimulated mouse melanoma cells.
  • ASP function may be assayed in vitro by measuring inhibition of alpha- MSH-stimulated cAMP accumulation in mouse melanoma cells (Hum MoI Genet 1995 Feb; 4(2): 223-230).
  • Myelin may be assayed in vitro by measurement of its phosphorylation by MAP kinase (J Neurochem 1999 Sep; 73(3): 1090-1097); or its effect on synaptic transmission between neurons (Eur Neurol. 1988; 28(2): 57-63).
  • Myelin Basic Protein may be assayed in vitro by measurement of its phosphorylation by MAP kinase (J Neurochem 1999 Sep; 73(3): 1090-1097); or its effect on synaptic transmission between neurons (Eur Neurol. 1988; 28(2): 57-63).
  • Collagen function may be measured using an in vitro collagen fibril stability assay (Cell MoI Life Sci 2000 May; 57(5): 859-863); or an in vitro cell adhesion assay (J Cell Biochem Oct. 1, 1997; 67(1): 75-83).
  • Alpha glucosidase can be assayed by hydrolysis of chromogenic artificial substrate p-Nitrophenyl a-D-glucoside. Bergmeyer, H. U.
  • Alpha-galactosidase can be assayed by hydrolysis of chromogenic artificial substrate p-Nitrophenyl a- D-galactosidase to p-Nitrophenol and D- galactose- Rietra et al., (1975) "Properties of the residual alpha-galactosidase activity in the tissues of a Fabry hemizygote" . Clin Chim Acta; 62(3) : 401 - 13.
  • Alpha-L iduronidase can be assayed by hydrolysis of the substrate A- methylumbelliferyl alpha-L-iduronide is followed in a fluorometric assay (Hopwood et al. (1979), Clin Chim Acta.; 92: 257-65); other assays are found in Thompson (1978) "Substrates for the assay of alpha-L-iduronidase". Clin Chim Acta; 89(3): 435-46
  • Angiopoeitin 1 activity may be assayed in vitro using a capillary sprouting assay. (Curr Biol Apr. 23, 1998; 8(9): 529-532).
  • Angiopoeitin 2 activity may be assayed in vitro using a capillary sprouting assay (Curr Biol Apr. 23, 1998; 8(9): 529-532).
  • Angiostatin can be assayed in an endothelial proliferation assay (Kringle domains of Human Angiostatin. Cao et al (1996) J. Biol. Chem. 271 29461-29467.
  • ADMP activity may beassayed in vitro by measuring its ability to downregulate the dorsalizing factors noggin, goosecoid or follistatin. (Development 1995 Dec;121(12): 4293-4301).
  • TRAIL can be assayed in an apoptosis assay (TRAIL- R2: a novel apoptosis-mediating receptor for TRAIL, Walczak et al. (1996) EMBOJ 16: 5386-5397).
  • Arresten function may be assayed in vitro by measuring its ability to inhibit endothelial cell proliferation, migration, tube formation, and Matrigel neovascularization (Cancer Res May 1, 2000;60(9): 2520-6).
  • Arylsulfatase B activity may be assayed by in vitro measurement of hydrolysis of sulfates of N- Acetyl-D-galactosamine (J Biol Chem Feb. 25, 1990;265(6): 3374-3381). Asparaginase activity may be assayed in vitro using an asparaginase enzymatic assay (Anal Biochem Apr. 10, 2000;280(l): 42-45). rBPI activity may be assayed in vitro using an antibacterial assay (J Biol Chem Nov. 5, 1987;262(31): 14891- 14894).
  • BDNF can be assayed in a neuronal growth and synaptic activity assay (BDNF enhances neuronal growth and synaptic activity in hippocampal cell cultures. Bartrupet al (1997) Neuroreport 1; 8(17): 3791-4 Daniels L B, Glew R H, Radin N S, Vunnam R R).
  • B-glucocerebrosidase can be assayed using a fluorometric assay for Gaucher's disease using conduritol-beta-epoxide with liver as the source of Beta- glucosidase (Clin Chim Acta. Sep. 25, 1980; 106(2): 155-63; Johnson W G, Gal A E, Miranda A F, Pentchev P G.
  • BMP-2 can be assayed as described by Wang, E. A et al. Recombinant humanbone morphogenetic protein induces boneformation. Proc. NatAcad. Sci. 87: 2220-2224,1990.
  • BT-SD function may beassayed in vitro by using a superoxide dismutaseassay (Nucleic Acids Res Mar. 25, 1985; 13(6): 2017- 34).
  • BRCAl activity may be assayed by measuring alterations in expression of p21WAFl/CIPl (Oncogene Jun. 11, 1998;16(23): 3069-82).
  • BRCA2 activity may be assayed by measuring alterations in expression of p21 WAF1/CIP1 (Oncogene Jun. 11, 1998;16(23): 3069-82).
  • the vasodilatory activity of CGRP can be assayed using the aortic ring vasodilation assay described in Pharmacol Res. 1999 Mar; 39(3): 217-20; endothelial and osteoblast cell proliferation activities can be measured in vitro (Eur J Pharmacol. Dec. 15, 2000; 409(3): 273-8; Proc Natl Acad Sci U.S.A. 1990 May; 87(9): 3299-303).
  • Calreticulin activity can be measured in vitro using calcium imaging assays
  • CD4 function maybe assayed in vitro by measuring g ⁇ l20 binding (Viral Immunol 2000; 13(4): 547-554); or altered monocyte responses tomcytokines following gpl20 binding (J Immunol Oct. 15, 1998; 161(8): 4309-4317).
  • CD40 ligand can ve assayed as described by Hollenbaugh D, et al., The human T cell antigen gp39, a member of the TNF gene family, is a ligand for the CD40 receptor: expression of a soluble form of gp39 with B cell co-stimulatory activity. EMBO J. 1992 Dec; 11(12): 4313-21.
  • Chemokine binding proteins can be assayed using receptor binding assays (J Biol Chem May 9, 1997; 272(19): 12495-12504). CNTF may be assayed in vitro using neuronal proliferation and survival assays (EMBO J. Apr. 2, 2001 ; 20(7) 1692-1703).
  • Contortrostatin activity may be measured in vitro by measuring binding to integrins alphavbeta3 andnalphavbeta5, inhibition ofnplatelet aggregation, and inhibition of cancer cell adhesion to fibronectin and vitronectin. (Arch Biochem Biophys Mar. 15, 2000;375(2): 278-288).
  • the activity of CRF binding protein can be measured using a CRF binding assay (Peptides Jan. 22, 2001; 22(1): 47-56).
  • CTLA4 activity can be measured using a T cell activation assay (J Immunol Mar. 1, 2001; 166(5): 3143-3150).
  • Decorin function may be measured using an in vitro collagen fibril stability assay (Cell MoI Life Sci 2000 May;57(5): 859-863); or an in vitro cell adhesion assay (J Cell Biochem Oct. 1, 1997;67(1): 75-83).
  • DeI-I function may be assayed in vitro using an alphavbeta3 integrin adhesion assay.
  • Dnase activity can be measured using the DNA degradation assay described in J Biochem (Tokyo). 1982 Oct; 92(4): 1297-303.
  • Ectoapyrase activity may be assayed in vitro using an ectoapyrase assay (J Biol Chem Sep. 18, 1998; 273(38): 24814-24821).
  • EGF can be assayed using a cell growth assay (J Biol Chem Mar. 31, 1995; 270(13): 7495-500)
  • EMAP II activity may be assayed in vitro using a capillary sprouting assay (Curr Biol Apr. 23, 1998; 8(9): 529-532).
  • FGF-I can be assayed using a cell proliferation assay: Cell, vol.50, no.5, pp.729-737 (Aug. 1987). Proc Natl Acad Sci U.S.A, vol.86, no.3, pp.802-806 (1989).
  • FGF-2 can be assayed using a roliferation assay using NR6R-3T3 cells (Rizzino 1988 Cancer Res. 48: 4266).
  • Fibrolase activity may be assayed in vitro using a fibrinolytic assay. (Thromb Res May 15, 1994; 74(4): 355-367).
  • FLT3 can be assayed in a proliferation assay using a Flt-3 transformed pro B-cell line (Hannum 1994 Nature 368: 643).
  • FoUitropin activity may be assayed in vitro by measuring cAMP production in cells expressing the FSH receptor (J Reprod Immunol 2001 Jan; 49(1): 1-19).
  • GDNF activity may be assayed in vitro by measuring increases in Ret tyrosine phosphorylation in response to GDNF treatment (MoI Cell Biol Mar. 15, 1995; (3): 1613-1619).
  • Gelsolin activity may be assayed in vitro by measuring proteolysis of actin (Nature 1987 Jan 22-28; 325(6102): 362-364).
  • GGF2 activity may be assayed in vitro by measuring activation of ERBB receptor tyrosine kinases in human rhabdomyosarcoma cells.(Int J Cancer JuI. 1, 2000;87(l): 29-36).
  • Glucagon's glucogenic activity is mediated by a high affinity glucagons receptor.
  • Biological acitivity of recombinant glucagon can be assessed by direct binding assays (Science Mar. 12, 1993; 259(5101): 1614-6).
  • HBNF activity may be assayed in vitro using a neurite outgrowth assay. (J Biol Chem Oct. 27, 1995; 270(43): 25992-25999).
  • hCG receptor binding and activation can be measured to assess biological activity of recombinant hCG (J Biol Chem Oct. 5, 1993; 268(28): 20851-4).
  • Heat shock proteins can be assayed by administration of HSP- peptide complexes in two UV-induced carcinoma models in mice (US 5,837,251).
  • Interleukin 1 can be assayed by 1) binding to IL-I receptors in YT-NCI or C3H/HeJ cells (Carter et al., Nature 344: 633-638, 1990); 2) induction of endothelial cell- leukocyte adhesion (Carter et al., Nature 344: 633-638, 1990); 3) proliferation assays on A375-C6 cells (Murai T et al., J. Biol. Chem.
  • IL-lra can be assayed by 1) competition for IL-I binding to IL-I receptors in YT-NCI or C3H/HeJ cells (Carter et al., Nature 344: 633-638, 1990); X) inhbition of IL-I -induced endothelial cell-leukocyte adhesion (Carter et al., Nature 344: 633-638, 1990); 3) proliferation assays on A375-C6 cells, a human melanoma cell line highly susceptible to the antiproliferative action of IL-I (Murai T et al., J.
  • IL-10 can be assayed by 1) binding of IL-10 to NK cells (carson WE et al., Blood 85: 3577-3585, 1995); 2) inhibition of TNF-alpha production by macrophages (Riley J K et al., J. Biol. Chem. 274: 16513-16521, 1999); 3) inhibition of macrophagem proliferation (O'Farrell A- M et al., EMBO 17: 1006- 1018, 1998); MC-9 proliferation: Thompson- Snipes et al (1991) J. Exp. Med.
  • IL-11 can be assayed in hematopoietic cell proliferation assay.
  • Interleukin- 11 enhances human megakaryocytopoiesis in vitro. Blood. Jan. 15, 1992; 79(2): 327-31.
  • IL-12 can be assayed in a natural killer (NK) cell cytotoxicity assay and interferon-gamma (IFN- gamma) release assay.
  • NK natural killer
  • IFN- gamma interferon-gamma
  • ILl 8 binding protein activity may be assayed in vitro by measuring its ability to inhibit the early ThI response (Immunity 1999 Jan; 10(1): 127-136).
  • IL2- diphtheria toxin chimera function may be assayed in vitro using a cytotoxicity assay.
  • IL-4 can be assayed in a cytotoxicity assay in normal T lymphocytes. (PMID: 8144944); RAMOS Augmentation of CD23 expression: Siegel & Mostowski (1990) J Immunol Methods 132, 287-295.
  • IL-4 recetpor activity can be measured by ability to inhibit IL-4 dependent proliferation of TF-I cells (Kitamura, T et al., 1989, J. Cell. PPhysiol. 140:323).
  • IL-8 can be assayed in an assay that monitors calcium reflux in IL8R bearing cells (Holmes et al (1991)Science 253, 1278-80).
  • Interferon gama can measure activity in anti-viral assay using HeIa cells infected withEMC virus (Meager, A. 1987, Lymphokines and Interferons, A Practical Approach, Clemens, MJ et al., eds., IRL Press, p.
  • IP-10 activity may beassayed in vitro using a rat artery smooth musclecell chemotaxis assay.
  • IL-3 activity may be measured in vitro using a haematopoietic cell differentiation assay(Science 1985; 228(4701): 810-815).
  • KGF- 1 activity may be assayed in vitro using an epithelial cell proliferation assay.
  • Kistrin activity may be assayed in vitro by assaying thrombolysis, reocclusion, and bleeding associated with administration of recombinant tissue-type plasminogen activator (rt-PA) in a canine model of coronary artery thrombosis (Circulation 1991 Mar; 83(3): 1038-1047).
  • Kunitz protease inhibitor 1 activity may be assayed in vitro by measuring inhibitory activity toward HGF activator (J Biol Chem Mar. 7, 1997; 272(10): 6370-6376). Lactotransferrin activity can be measured using an in vitro viral inhibition assay (J Med Microbiol 1998 Nov; 47(11): 1015-8), and antimicrobial assays (J Clin Invest Sep.
  • Leptin can be assayed by in vivo modulation of food intake, reduction in body weight, and lowering of insulin and glucose levels in ob/ob mice, radioimmunoassay (RIA) and activation of the leptin receptor in a cell-based assay.
  • RIA radioimmunoassay
  • LD? activity may be measured in vitro using a haematopoietic cell differentiation assay. (Science 1985; 228(4701): 810-815).
  • LFA-3 activity may be assayed in vitro by measuring its ability to inhibit T-cell function. (JExp Med JuI. 1, 1993;178(1): 211-222).
  • Lys plasminogen activity can be measured using an in vitro fibrinolysis assay (J Biol Chem Dec. 25, 1992; 267(36): 26150-6). Maspin activity can be measured using in vitro angiogenesis assays (Nat Med 2000 Feb; 6(2): 196-9). Methioninase activity maybe assayed in vitro as described by Ito et al. (J Biochem (Tokyo), 1975 Nov; 78(5): 1105-1107). MTP activity may be assayed in vitro by using an apoB-containing lipoprotein secretion assay. (J Biol Chem Sep. 2, 1994; 269(35): 21951-21954).
  • NGF activity may be assayed by measurement of CREB transcription factor activation in sympathetic neurons in culture (Science Dec. 17, 1999;286(5448): 2358-2361).
  • Neutral endopeptidase activity may be assayed in vitro by measuring proteolysis of bombesin-,like peptides (Proc NatlmAcad Sci U.S.A. Dec. 1, 1991; 88(23): 10662- 10666).
  • NIF activity may be assayed in vitro using a polymorphonuclear leukocyte adhesion assay (MoI Pharm 1999 Nov; 56(5): 926-932).
  • Noggin activity may be assayed by measuring TNF receptor responses to TNF stimulation of HeLa cells. (J Biol Chem Feb.
  • NT-3 activity may be assayed in vitro by measuring its ability to proliferate cultured NC progenitor cells grown in a serum-free defined medium (Proc Natl Acad Sci U.S.A Mar. 1, 1992; 89(5): 1661- 1665).
  • OP-I activity may be assayed in vitro by measuring VEGF expression in fetal rat calvaria cells (MoI Cell Endocrinol JuI. 20, 1999;153(l-2): 113-124).
  • Osteoprotegerin activity may be assayed in vitro using a coculture assay for osteoclastogenesis, a bone resorption assay using fetal long-bone organ culture system, a dentine resorption assay, or a fibroblast proliferation assay (FASEB J.
  • Plasma PAF acetylhydrolase activity may be determined by the method of Stafforini et al. (Stroke 1997 Dec; 28(12): 2417-20).
  • Patched function may be assayed in vitro by measuring binding of its ligand, Sonic hedgehog (Nature Nov. 14, 1996; 384(6605): 129-134).
  • PDGF activity may be assayed in vitro by measuring its ability toinduce tyrosine phosphorylation on the PDGF receptor (J Biol Chem May 25, 1989;264(15): 8905-8912).
  • PEDF function may be assayed in vitro using a neurite outgrowth assay (J Biol Chem Oct. 27, 1995; 270(43): 25992-25999).
  • Plasminogen activator inhibitor can be assayed as described by Wallen, P., Biochemistry of plasminogen, hi: Kline D. L., Reddy, K. N. N., eds. Fibrinolysis. Boca Raton, FL: CRC Press, 1980: 1-25; Saksela, O.,
  • PGP activity may be assayed in vitro by measuring agonism of fetal liver tyrosine kinase- 3 and granulocyte colony- stimulating factor (Exp Hematol 2001 Jan; 29(1): 41-50).
  • PMP activity may be assayed in vitro by measuing the ability to induce megakaryocytopoiesis in CD43+ cells purified from bone marrow, mobilized peripheral blood progenitor cells, or umbilical cord (J Hematother 1999 Apr; 8(2): 199-208).
  • Prosaptide efficacy can be measured through multiple dose regimens using diabetic rats (Anesthesiology 2000 Nov; 93(5): 1271-8).
  • Ranpirnase activity in vitro may be assayed by using ribonuclease and cytotoxic activity assays (J MoI Biol Apr. 19, 1996; 257(5): 992-1007).
  • Relaxin activity may be assayed in vitro using an inhibition of KCl-induced rat uterine contractions in vitro assay (Can J Physiol Pharmacol 1981 May; 59(5): 507-12).
  • Retinal S-Antigen activity may be assayed in vitro by measuring activity of cyclic GMP- gated channels in rod photoreceptor cells (J Biol Chem Nov. 25, 1994;
  • RhLH activity may be assayed in vitro by measuring Fura-2 fluorescence in isolated porcine thecal cells in response to RhLH stimulation (Endocrinology 2000 Jun; 141(6): 2220-2228).
  • Saruplase activity may be measured in vitro using a plasminogen cleavage assay (J Biol Chem Jan. 18, 2001; epub ahead of print).
  • Complement receptor type 1 may be assayed in vitro by measuring C3b and C4b binding. (J Exp Med Nov. 1, 1988; 168(5): 1699-1717).
  • Sonic hedgehog function may be assayed in vitro by measuring its binding to patched (Nature 1996 Nov. 14; 384(6605): 129-134).
  • Staphylokinase activity may be measured in vitro using a plasminogen cleavage assay (J Biol Chem Jan. 18, 2001; epub ahead of print).
  • SCF activity may be assayed by measuring mast cell survival, proliferation, or production of pro- inflammatory cytokines (Immunol Rev 2001 Feb; 179: 57-60).
  • Streptokinase activity may be measured in vitro using a plasminogen activation assay (J Biol Chem Jan. 18, 2001; epub ahead of print).
  • Superoxide dismutase activity may be assayed in vitro using a superoxide dismutase assay (Nucleic Acids Res Mar. 25, 1985; 13(6): 2017-34).
  • T1/ST2 function may be assayed in vitro using a Th2 cell activation assay (J Immunol Mar. 1, 2001 ; 166(5): 3143-3150).
  • TGF beta 1 activity may be assayed in vitro by measuring induction of renal fibroblast proliferation via induction of FGF-2 (Kidney Int 2001 Feb; 59(2): 579-592).
  • TGF Beta 2 function may be assayed in vitro by measuring inhibition of iNOS transcription (Inflamm Res 1997 Sep; 46(9): 327-331).
  • TGF-beta 3 can be assayed for stimulation of production of prostaglandin E2 (PGE2) and bone resorption in neonatal mouse calvaria in organ culture (PNAS USA 1985 JuI; 82(13): 4535-4538), or stimulation of the synthesis of collagen, osteopontin, osteonectin, and alkaline phosphatase, and the ability to stimulate replication in osteoblast-like cells (J Biol Chem 1987;262: 2869, J Biol Chem 1988; 263: 13916, J Cell Biol 1988; 106: 915, J Cell Physiol 1987; 133: 426, Endocrinology 1987; 121: 212, Endocrinology 1986;19: 2306, and J Cell Biol 1987; 105: 457).
  • Thrombopoietin can be assayed to determine regulation of growth and differentiation of megakaryocytes (MoI Cell Biol 2001 Apr; 21(8): 2659-2670; Exp Hematol 2001 Jan; 29(1): 51-58; Leukemia 2000 Oct; 14(10): 1751-1756).
  • Tie- 2/Tek function may be assayed by measuringits phosphorylation in reponse to stimulation by angiopoietin (Jut Immunol 1998 Aug; 10(8): 1217-1227).
  • Tissue factor pathway inhibitor may be assayed for inactivation of factor Xa and inhibition of the Vila-tissue factor complex of the extrinsic coagulation pathway (J Biol Chem May 5, 1998; 263(13): 6001-6004; Thromb Haemost 1998; 79(2): 306-309).
  • TNF binding protein activity may be assayed by measuring inhibition of PIP5K activation in response to TNF stimulation of HeLa cells. (J Biol Chem Feb. 28, 1997; 272(9): 5861-5870).
  • TNF binding protein activity may be assayed by measuring inhibition of PJP5K activation in response to TNF stimulation of HeLa cells. (J Biol Chem Feb. 28, 1997;272(9): 5861-5870).
  • TNF receptor activity may be assayed by measuring increases of PIP5K activation in response to TNF stimulation of HeLa cells (J Biol Chem Feb. 28, 1997; 272(9): 5861-5870).
  • t-PA can be assayed as described in Wallen, P., Biochemistry of plasminogen. In: Kline D. L., Reddy, K. N. N., eds. Fibrinolysis. Boca Raton, FL: CRC Press, 1980: 1-25; Saksela, O., Rifkin, D. B., Cell- associated plasminogen activation: Regulation and physiological functions.
  • IFN-tau activity may be assayed in vitro by measuring IFN-tau induced production of an acidic 70 kD protein in cultured explants of endometrium prepared from ewes on day 13 of the estrous cycle. (MoI Endocrinol 1990 Oct; 4(10): 1506-1514).
  • Troponin I activity may be assayed in vitro using a myofibril binding assay (J Muscle Res Cell Motil 1999 Nov; 20(8): 755-760).
  • Urate oxidase activity may be assayed in vitro using an assay for uricase-catalyzed oxidation of uric acid (Anal Chem May 15, 1999; 71(10): 1928-1934); or by immunoassay of recombinant urate oxidase (J Pharm Sci 1996 Sep; 85(9): 955-959).
  • Urokinase activity may be measured in vitro using a plasminogen cleavage assay. Sazonova et al. (J Biol Chem Jan. 18, 2001, electronic publication prior to print).
  • VEGF-I activity may beassayed in vitro using an endothelial cell proliferation assay. (Proc Natl Acad Sci U.S.A.
  • Binding ligands as described herein according to the invention can be prepared according to previously established techniques, used in the field of antibody engineering, for the preparation of scFv, "phage” antibodies and other engineered antibody molecules. Techniques for the preparation of antibodies are for example described in the following reviews and the references cited therein: Winter & Milstein, (1991) Nature 349:293-299; Pluckthun (1992) Immunological Reviews 130:151-188; Wright et al, (1992) Crti. Rev. Immunol.l2:125-168; Holliger, P. & Winter, G. (1993) Curr. Op. Biotechn. 4, 446-449; Carter, et al. (1995) J. Hematother.
  • Suitable techniques employed for selection of antibody variable domains with a desired specificity employ libraries and selection procedures which are known in the art.
  • Natural libraries Marks et al (1991) J. MoI. Biol, 222: 581; Vaughan et al (1996) Nature Biotech., 14: 309) which use rearranged V genes harvested from human B cells are well known to those skilled in the art.
  • Synthetic libraries Hoogenboom & Winter (1992) J. MoI. Biol, 227: 381; Barbas et al. (1992) Proc. Natl. Acad. ScL USA, 89: 4457; Nissim et al. (1994) EMBO J., 13: 692; Griffiths et al.
  • V H and/or VL libraries may be selected against target antigens or epitopes separately, in which case single domain binding is directly selected for, or together.
  • Bacteriophage lambda expression systems may be screened directly as bacteriophage plaques or as colonies of lysogens, both as previously described (Huse et al. (1989) Science, 246: 1275; Caton and Koprowski (1990) Proc. Natl.
  • a selection display system is a system that permits the selection, by suitable display means, of the individual members of the library by binding the generic and/or target ligands. Selection protocols for isolating desired members of large libraries are known in the art, as typified by phage display techniques. Such systems, in which diverse peptide sequences are displayed on the surface of filamentous bacteriophage (Scott and Smith (1990) Science, 249: 386), have proven useful for creating libraries of antibody fragments (and the nucleotide sequences that encoding them) for the in vitro selection and amplification of specific antibody fragments that bind a target antigen (McCafferty et al, WO 92/01047).
  • the nucleotide sequences encoding the variable regions are linked to gene fragments which encode leader signals that direct them to the periplasmic space of E. coli and as a result the resultant antibody fragments are displayed on the surface of the bacteriophage, typically as fusions to bacteriophage coat proteins (e.g., pill or pVIII). Alternatively, antibody fragments are displayed externally on lambda phage capsids (phagebodies).
  • phagebodies lambda phage capsids
  • An advantage of phage-based display systems is that, because they are biological systems, selected library members can be amplified simply by growing the phage containing the selected library member in bacterial cells. Furthermore, since the nucleotide sequence that encode the polypeptide library member is contained on a phage or phagemid vector, sequencing, expression and subsequent genetic manipulation is relatively straightforward.
  • RNA molecules are selected by alternate rounds of selection against a target ligand and PCR amplification (Tuerk and Gold (1990) Science, 249: 505; Ellington and Szostak (1990) Nature, 346: 818).
  • a similar technique may be used to identify DNA sequences which bind a predetermined human transcription factor (Thiesen and Bach (1990) Nucleic Acids Res., 18: 3203; Beaudry and Joyce (1992) Science, 257: 635; WO92/05258 and WO92/14843).
  • in vitro translation can be used to synthesise polypeptides as a method for generating large libraries.
  • These methods which generally comprise stabilised polysome complexes, are described further in WO88/08453, WO90/05785, WO90/07003, WO91/02076, WO91/05058, and WO92/02536.
  • Alternative display systems which are not phage-based, such as those disclosed in WO95/22625 and WO95/11922 (Affymax) use the polysomes to display polypeptides for selection.
  • a still further category of techniques involves the selection of repertoires in artificial compartments, which allow the linkage of a gene with its gene product.
  • a selection system in which nucleic acids encoding desirable gene products may be selected in microcapsules formed by water-in-oil emulsions is described in WO99/02671, WO00/40712 and Tawfik & Griffiths (1998) Nature Biotechnol 16(7), 652-6.
  • Genetic elements encoding a gene product having a desired activity are compartmentalised into microcapsules and then transcribed and/or translated to produce their respective gene products (RNA or protein) within the microcapsules.
  • Genetic elements which produce gene product having desired activity are subsequently sorted. This approach selects gene products of interest by detecting the desired activity by a variety of means.
  • Libraries intended for selection may be constructed using techniques known in the art, for example as set forth above, or may be purchased from commercial sources. Libraries which are useful in the present invention are described, for example, in WO99/20749.
  • PCR polymerase chain reaction
  • PCR is performed using template DNA (at least lfg; more usefully, 1-1000 ng) and at least 25 pmol of oligonucleotide primers; it maybe advantageous to use a larger amount of primer when the primer pool is heavily heterogeneous, as each sequence is represented by only a small fraction of the molecules of the pool, and amounts become limiting in the later amplification cycles.
  • a typical reaction mixture includes: 2 ⁇ l of DNA, 25 pmol of oligonucleotide primer, 2.5 ⁇ l of 1OX PCR buffer 1 (Perkin-Elmer, Foster City, CA), 0.4 ⁇ l of 1.25 ⁇ M dNTP, 0.15 ⁇ l (or 2.5 units) of Taq DNA polymerase (Perkin Elmer, Foster City, CA) and deionized water to a total volume of 25 ⁇ l.
  • Mineral oil is overlaid and the PCR is performed using a programmable thermal cycler. The length and temperature of each step of a PCR cycle, as well as the number of cycles, is adjusted in accordance to the stringency requirements in effect.
  • Annealing temperature and timing are determined both by the efficiency with which a primer is expected to anneal to a template and the degree of mismatch that is to be tolerated; obviously, when nucleic acid molecules are simultaneously amplified and mutagenised, mismatch is required, at least in the first round of synthesis.
  • the ability to optimise the stringency of primer annealing conditions is well within the knowledge of one of moderate skill in the art.
  • An annealing temperature of between 30 °C and 72 0 C is used.
  • Initial denaturation of the template molecules normally occurs at between 92 0 C and 99 0 C for 4 minutes, followed by 20-40 cycles consisting of denaturation (94-99 0 C for 15 seconds to 1 minute), annealing (temperature determined as discussed above; 1-2 minutes), and extension (72 0 C for 1-5 minutes, depending on the length of the amplified product).
  • Final extension is generally for 4 minutes at 72 0 C, and may be followed by an indefinite (0-24 hour) step at 4 0 C.
  • Combining Single Variable Domains Domains useful in the invention may be combined by a variety of methods known in the art, including covalent and non-covalent methods.
  • Preferred methods include the use of polypeptide linkers, as described, for example, in connection with scFv molecules (Bird et al, (1988) Science 242:423-426). Discussion of suitable linkers is provided in Bird et al. Science 242, 423-426; Hudson et al , Journal Immunol Methods 231 (1999) 177-189; Hudson et al, Proc Nat Acad Sci USA 85, 5879-5883. Linkers are preferably flexible, allowing the two single domains to interact.
  • the linkers used in diabodies, which are less flexible, may also be employed (Holliger et al, (1993) PNAS (USA) 90:6444-6448).
  • the linker employed is not an immunoglobulin hinge region.
  • Variable domains may be combined using methods other than linkers. For example, the use of disulphide bridges, provided through naturally-occurring or engineered cysteine residues, may be exploited to stabilise V H 'V H 'V L 'V L or VR- V L dimers (Reiter et al, (1994) Protein Eng. 7:697-704) or by remodelling the interface between the variable domains to improve the "fit" and thus the stability of interaction (Ridgeway et al, (1996) Protein Eng. 7:617-621; Zhu et al, (1997) Protein Science 6:781-788).
  • Other techniques for joining or stabilising variable domains of immunoglobulins, and in particular antibody VH domains may be employed as appropriate.
  • a ligand e.g., dAb monomer, dual-specific ligand
  • binding is tested using monoclonal phage ELISA.
  • Phage ELISA may be performed according to any suitable procedure: an exemplary protocol is set forth below. Populations of phage produced at each round of selection can be screened for binding by ELISA to the selected antigen or epitope, to identify "polyclonal" phage antibodies. Phage from single infected bacterial colonies from these populations can then be screened by ELISA to identify "monoclonal" phage antibodies.
  • the diversity of the selected phage monoclonal antibodies may also be assessed by gel electrophoresis of PCR products (Marks et al 1991, supra; Nissim et al 1994 supra), probing (Tomlinson et al, 1992) J. MoI. Biol. 227, 776) or by sequencing of the vector DNA.
  • variable domains are selected from V-gene repertoires selected for instance using phage display technology as herein described, then these variable domains comprise a universal framework region, such that is they may be recognised by a specific generic ligand as herein defined.
  • the use of universal frameworks, generic ligands and the like is described in WO99/20749.
  • V-gene repertoires are used variation in polypeptide sequence is preferably located within the structural loops of the variable domains.
  • the polypeptide sequences of either variable domain may be altered by DNA shuffling or by mutation in order to enhance the interaction of each variable domain with its complementary pair.
  • DNA shuffling is known in the art and taught, for example, by Stemmer, 1994, Nature 370: 389-391 and U.S. Patent No. 6,297,053, both of which are incorporated herein by reference. Other methods of mutagenesis are well known to those of skill in the art.
  • nucleic acid molecules and vector constructs required for selection, preparation and formatting ligands may be constructed and manipulated as set forth in standard laboratory manuals, such as Sambrook et al. (1989,) Molecular Cloning: A Laboratory Manual, Cold Spring Harbor, USA.
  • the manipulation of nucleic acids useful in the present invention is typically carried out in recombinant vectors.
  • vector refers to a discrete element that is used to introduce heterologous DNA into cells for the expression and/or replication thereof.
  • Methods by which to select or construct and, subsequently, use such vectors are well known to one of ordinary skill in the art.
  • Numerous vectors are publicly available, including bacterial plasmids, bacteriophage, artificial chromosomes and episomal vectors. Such vectors may be used for simple cloning and mutagenesis; alternatively gene expression vector is employed.
  • a vector of use according to the invention may be selected to accommodate a polypeptide coding sequence of a desired size, typically from 0.25 kilobase (kb) to 40 kb or more in length
  • a suitable host cell is transformed with the vector after in vitro cloning manipulations.
  • Each vector contains various functional components, which generally include a cloning (or "polylmker") site, an origin of replication and at least one selectable marker gene. If given vector is an expression vector, it additionally possesses one or more of the following: enhancer element, promoter, transcription termination and signal sequences, each positioned in the vicinity of the cloning site, such that they are operatively linked to the gene encoding a ligand according to the invention.
  • Both cloning and expression vectors generally contain nucleic acid sequences that enable the vector to replicate in one or more selected host cells.
  • this sequence is one that enables the vector to replicate independently of the host chromosomal DNA and includes origins of replication or autonomously replicating sequences.
  • origins of replication or autonomously replicating sequences are well known for a variety of bacteria, yeast and viruses.
  • the origin of replication from the plasmid pBR322 is suitable for most Gram-negative bacteria, the 2 micron plasmid origin is suitable for yeast, and various viral origins (e.g. SV 40, adenovirus) are useful for cloning vectors in mammalian cells.
  • the origin of replication is not needed for mammalian expression vectors unless these are used in mammalian cells able to replicate high levels of DNA, such as COS cells.
  • a cloning or expression vector may contain a selection gene also referred to as selectable marker.
  • This gene encodes a protein necessary for the survival or growth of transformed host cells grown in a selective culture medium. Host cells not transformed with the vector containing the selection gene will therefore not survive in the culture medium.
  • Typical selection genes encode proteins that confer resistance to antibiotics and other toxins, e.g. ampicillin, neomycin, methotrexate or tetracycline, complement auxotrophic deficiencies, or supply critical nutrients not available in the growth media.
  • an E. co/z ' -selectable marker for example, the ⁇ -lactamase gene that confers resistance to the antibiotic ampicillin.
  • E. coli plasmids such as pBR322 or a pUC plasmid such as pUC 18 or pUC 19.
  • Expression vectors usually contain a promoter that is recognised by the host organism and is operably linked to the coding sequence of interest. Such a promoter may be inducible or constitutive.
  • operably linked refers to a juxtaposition wherein the components described are in a relationship permitting them to function in their intended manner.
  • a control sequence "operably linked" to a coding sequence is ligated in such a way that expression of the coding sequence is achieved under conditions compatible with the control sequences.
  • Promoters suitable for use with prokaryotic hosts include, for example, the ⁇ - lactamase and lactose promoter systems, alkaline phosphatase, the tryptophan (trp) promoter system and hybrid promoters such as the tac promoter. Promoters for use in bacterial systems will also generally contain a Shine-Delgarno sequence operably linked to the coding sequence.
  • the preferred vectors are expression vectors that enables the expression of a nucleotide sequence corresponding to a polypeptide library member.
  • selection with the first and/or second antigen or epitope can be performed by separate propagation and expression of a single clone expressing the polypeptide library member or by use of any selection display system.
  • the preferred selection display system is bacteriophage display.
  • phage or phagemid vectors may be used, eg pITl or pIT2.
  • Leader sequences useful in the invention include pelB, stll, ompA, phoA, bla and pelA.
  • phagemid vectors which have an E. coli.
  • the vector contains a ⁇ -lactamase gene to confer selectivity on the phagemid and a lac promoter upstream of a expression cassette that consists (N to C terminal) of a pelB leader sequence (which directs the expressed polypeptide to the periplasmic space), a multiple cloning site (for cloning the nucleotide version of the library member), optionally, one or more peptide tag (for detection), optionally, one or more TAG stop codon and the phage protein pill.
  • a pelB leader sequence which directs the expressed polypeptide to the periplasmic space
  • a multiple cloning site for cloning the nucleotide version of the library member
  • one or more peptide tag for detection
  • TAG stop codon optionally, one or more TAG stop codon and the phage protein pill.
  • the vector is able to replicate as a plasmid with no expression, produce large quantities of the polypeptide library member only or produce phage, some of which contain at least one copy of the polypeptide-pIII fusion on their surface.
  • Construction of vectors encoding ligands according to the invention employs conventional ligation techniques. Isolated vectors or DNA fragments are cleaved, tailored, and religated in the form desired to generate the required vector. If desired, analysis to confirm that the correct sequences are present in the constructed vector can be performed in a known fashion. Suitable methods for constructing expression vectors, preparing in vitro transcripts, introducing DNA into host cells, and performing analyses for assessing expression and function are known to those skilled in the art.
  • telomere sequence The presence of a gene sequence in a sample is detected, or its amplification and/or expression quantified by conventional methods, such as Southern or Northern analysis, Western blotting, dot blotting of DNA, RNA or protein, in situ hybridisation, immunocytochemistry or sequence analysis of nucleic acid or protein molecules. Those skilled in the art will readily envisage how these methods maybe modified, if desired.
  • Skeletons may be based on immunoglobulin molecules or may be non- immunoglobulin in origin as set forth above.
  • Preferred immunoglobulin skeletons as herein defined includes any one or more of those selected from the following: an immunoglobulin molecule comprising at least (i) the CL (kappa or lambda subclass) domain of an antibody; or (ii) the CHl domain of an antibody heavy chain; an immunoglobulin molecule comprising the CHl and CH2 domains of an antibody heavy chain; an immunoglobulin molecule comprising the CHl, CH2 and CH3 domains of an antibody heavy chain; or any of the subset (ii) in conjunction with the CL (kappa or lambda subclass) domain of an antibody.
  • a hinge region domain may also be included.
  • Such combinations of domains may, for example, mimic natural antibodies, such as IgG or IgM, or fragments thereof, such as Fv, scFv, Fab or F(ab') 2 molecules. Those skilled in the art will be aware that this list is not intended to be exhaustive.
  • Each epitope binding domain comprises a protein scaffold and one or more CDRs which are involved in the specific interaction of the domain with one or more epitopes.
  • an epitope binding domain according to the present invention comprises three CDRs.
  • Suitable protein scaffolds include any of those selected from the group consisting of the following: those based on immunoglobulin domains, those based on fibronectin, those based on affibodies, those based on CTLA4, those based on chaperones such as GroEL, those based on lipocallin and those based on the bacterial Fc receptors SpA and SpD. Those skilled in the art will appreciate that this list is not intended to be exhaustive.
  • the members of the immunoglobulin superfamily all share a similar fold for their polypeptide chain.
  • antibodies are highly diverse in terms of their primary sequence
  • comparison of sequences and crystallo graphic structures has revealed that, contrary to expectation, five of the six antigen binding loops of antibodies (Hl, H2, Ll, L2, L3) adopt a limited number of main-chain conformations, or canonical structures (Chothia and Lesk (1987) J. MoI. Biol, 196: 901; Chothia et al. (1989) Nature, 342: 877).
  • Analysis of loop lengths and key residues has therefore enabled prediction of the main-chain conformations of Hl, H2, Ll, L2 and L3 found in the majority of human antibodies (Chothia et al.
  • H3 region is much more diverse in terms of sequence, length and structure (due to the use of D segments), it also forms a limited number of main-chain conformations for short loop lengths which depend on the length and the presence of particular residues, or types of residue, at key positions in the loop and the antibody framework (Martin et al. (1996) J. MoI Biol, 263: 800; Shirai et al (1996) FEBS Letters, 399: 1).
  • Libraries of ligands and/or domains can be designed in which certain loop lengths and key residues have been chosen to ensure that the main-chain conformation of the members is known.
  • these are real conformations of immunoglobulin superfamily molecules found in nature, to minimise the chances that they are non-functional, as discussed above.
  • Germline V gene segments serve as one suitable basic framework for constructing antibody or T- cell receptor libraries; other sequences are also of use. Variations may occur at a low frequency, such that a small number of functional members may possess an altered main-chain conformation, which does not affect its function.
  • Canonical structure theory is also of use to assess the number of different main-chain conformations encoded by ligands, to predict the main-chain conformation based on ligand sequences and to choose residues for diversification which do not affect the canonical structure. It is known that, in the human V ⁇ domain, the Ll loop can adopt one of four canonical structures, the L2 loop has a single canonical structure and that 90% of human V ⁇ domains adopt one of four or five canonical structures for the L3 loop (Tomlinson et al. (1995) supra); thus, in the V ⁇ domain alone, different canonical structures can combine to create a range of different main-chain conformations.
  • V ⁇ domain encodes a different range of canonical structures for the Ll, L2 and L3 loops and that V ⁇ and V ⁇ domains can pair with any V H domain which can encode several canonical structures for the Hl and H2 loops
  • the number of canonical structure combinations observed for these five loops is very large. This implies that the generation of diversity in the main-chain conformation may be essential for the production of a wide range of binding specificities.
  • by constructing an antibody library based on a single known main-chain conformation it has been found, contrary to expectation, that diversity in the main-chain conformation is not required to generate sufficient diversity to target substantially all antigens.
  • the single main-chain conformation need not be a consensus structure - a single naturally occurring conformation can be used as the basis for an entire library.
  • the dual-specific ligands of the invention possess a single known main-chain conformation.
  • the single main-chain conformation that is chosen is preferably commonplace among molecules of the immunoglobulin superfamily type in question.
  • a conformation is commonplace when a significant number of naturally occurring molecules are observed to adopt it.
  • the natural occurrence of the different main-chain conformations for each binding loop of an immunoglobulin domain are considered separately and then a naturally occurring variable domain is chosen which possesses the desired combination of main-chain conformations for the different loops. If none is available, the nearest equivalent may be chosen. It is preferable that the desired combination of main-chain conformations for the different loops is created by selecting germline gene segments which encode the desired main-chain conformations.
  • the selected germline gene segments are frequently expressed in nature, and most preferable that they are the most frequently expressed of all natural germline gene segments.
  • hi designing ligands (e.g., dAbs) or libraries thereof the incidence of the different main-chain conformations for each of the six antigen binding loops may be considered separately.
  • ligands e.g., dAbs
  • Hl, H2, Ll, L2 and L3 a given conformation that is adopted by between 20% and 100% of the antigen binding loops of naturally occurring molecules is chosen.
  • its observed incidence is above 35% (i.e. between 35% and 100%) and, ideally, above 50% or even above 65%.
  • H3 loops Since the vast majority of H3 loops do not have canonical structures, it is preferable to select a main-chain conformation which is commonplace among those loops which do display canonical structures. For each of the loops, the conformation which is observed most often in the natural repertoire is therefore selected.
  • the most popular canonical structures (CS) for each loop are as follows: Hl - CS 1 (79% of the expressed repertoire), H2 - CS 3 (46%), Ll - CS 2 of V ⁇ (39%), L2 - CS 1 (100%), L3 - CS 1 of V ⁇ (36%) (calculation assumes a ⁇ : ⁇ ratio of 70:30, Hood et al. (1967) Cold Spring Harbor Symp. Quant.
  • V H segment 3-23 DP-47
  • J H segment JH4b the V ⁇ segment 02/012
  • J ⁇ segment J ⁇ l the V H segments DP45 and DP38 are also suitable. These segments can therefore be used in combination as a basis to construct a library with the desired single main-chain conformation.
  • the natural occurrence of combinations of main-chain conformations is used as the basis for choosing the single main-chain conformation.
  • the natural occurrence of canonical structure combinations for any two, three, four, five or for all six of the antigen binding loops can be determined.
  • the chosen conformation is commonplace in naturally occurring antibodies and most preferable that it observed most frequently in the natural repertoire.
  • the most frequent combination of canonical structures is determined and then combined with the most popular conformation for the H3 loop, as a basis for choosing the single main-chain conformation.
  • ligands e.g., dAbs
  • libraries for use in the invention can be constructed by varying the binding site of the molecule in order to generate a repertoire with structural and/or functional diversity. This means that variants are generated such that they possess sufficient diversity in their structure and/or in their function so that they are capable of providing a range of activities.
  • the desired diversity is typically generated by varying the selected molecule at one or more positions.
  • the positions to be changed can be chosen at random or are preferably selected.
  • the variation can then be achieved either by randomisation, during which the resident amino acid is replaced by any amino acid or analogue thereof, natural or synthetic, producing a very large number of variants or by replacing the resident amino acid with one or more of a defined subset of amino acids, producing a more limited number of variants.
  • H3 region of a human tetanus toxoid-binding Fab has been randomised to create a range of new binding specificities (Barbas et al. (1992) Proc. Natl. Acad. ScL USA, 89: 4457). Random or semi-random H3 and L3 regions have been appended to germline V gene segments to produce large libraries with unmutated framework regions (Hoogenboom & Winter (1992) J. MoI. Biol, 227: 381; Barbas et al (1992) Proc. Natl. Acad.
  • the binding site for the target is most often the antigen binding site.
  • residues in the antigen binding site are varied.
  • These residues are extremely diverse in the human antibody repertoire and are known to make contacts in high-resolution antibody/antigen complexes.
  • positions 50 and 53 are diverse in naturally occurring antibodies and are observed to make contact with the antigen.
  • the conventional approach would have been to diversify all the residues in the corresponding Complementarity Determining Region (CDRl) as defined by Kabat et al. (1991, supra), some seven residues compared to the two diversified in the library for use according to the invention.
  • CDRl Complementarity Determining Region
  • antibody diversity is the result of two processes: somatic recombination of germline V, D and J gene segments to create a naive primary repertoire (so called germline and junctional diversity) and somatic hypermutation of the resulting rearranged V genes.
  • somatic recombination of germline V, D and J gene segments to create a naive primary repertoire (so called germline and junctional diversity)
  • somatic hypermutation of the resulting rearranged V genes The analysis of human antibody sequences has shown that diversity in the primary repertoire is focused at the centre of the antigen binding site whereas somatic hypermutation spreads diversity to regions at the periphery of the antigen binding site that are highly conserved in the primary repertoire (see Tomlinson et al. (1996) J. MoI. Biol, 256: 813).
  • an initial 'naive' repertoire can be created where some, but not all, of the residues in the antigen binding site are diversified.
  • the term "naive” refers to antibody molecules that have no pre-dete ⁇ nined target. These molecules resemble those which are encoded by the immunoglobulin genes of an individual who has not undergone immune diversification, as is the case with fetal and newborn individuals, whose immune systems have not yet been challenged by a wide variety of antigenic stimuli.
  • This repertoire is then selected against a range of antigens or epitopes. If required, further diversity can then be introduced outside the region diversified in the initial repertoire. This matured repertoire can be selected for modified function, specificity or affinity.
  • Naive repertoires of binding domains for the construction of ligands in which some or all of the residues in the antigen binding site are varied are known in the art. (See, WO 2004/058821, WO 2004/003019, and WO 03/002609).
  • the "primary" library mimics the natural primary repertoire, with diversity restricted to residues at the centre of the antigen binding site that are diverse in the germline V gene segments (germline diversity) or diversified during the recombination process (junctional diversity).
  • residues which are diversified include, but are not limited to, H50, H52, H52a, H53, H55, H56, H58, H95, H96, H97, H98, L50, L53, L91, L92, L93, L94 and L96.
  • diversity is restricted to residues that are diversified during the recombination process (junctional diversity) or are highly somatically mutated).
  • residues which are diversified include, but are not limited to: H31, H33, H35, H95, H96, H97, H98, L30, L31, L32, L34 and L96.
  • diversification of chosen positions is typically achieved at the nucleic acid level, by altering the coding sequence which specifies the sequence of the polypeptide such that a number of possible amino acids (all 20 or a subset thereof) can be incorporated at that position.
  • the most versatile codon is NNK, which encodes all amino acids as well as the TAG stop codon.
  • the NNK codon is preferably used in order to introduce the required diversity.
  • Other codons which achieve the same ends are also of use, including the NNN codon, which leads to the production of the additional stop codons TGA and TAA.
  • a feature of side-chain diversity in the antigen binding site of human antibodies is a pronounced bias which favours certain amino acid residues. If the amino acid composition of the ten most diverse positions in each of the V H , V K and V ⁇ regions are summed, more than 76% of the side-chain diversity comes from only seven different residues, these being, serine (24%), tyrosine (14%), asparagine (11%), glycine (9%), alanine (7%), aspartate (6%) and threonine (6%).
  • This bias towards hydrophilic residues and small residues which can provide main-chain flexibility probably reflects the evolution of surfaces which are predisposed to binding a wide range of antigens or epitopes and may help to explain the required promiscuity of antibodies in the primary repertoire.
  • the distribution of amino acids at the positions to be varied preferably mimics that seen in the antigen binding site of antibodies.
  • Such bias in the substitution of amino acids that permits selection of certain polypeptides (not just antibody polypeptides) against a range of target antigens is easily applied to any polypeptide repertoire.
  • There are various methods for biasing the amino acid distribution at the position to be varied including the use of tri-nucleotide mutagenesis, see WO97/08320), of which the preferred method, due to ease of synthesis, is the use of conventional degenerate codons.
  • codons (AGT)(AGC)T, (AGT)(AGC)C and (AGT)(AGC)(CT) - that is, DVT, DVC and DVY, respectively using IUPAC nomenclature - are those closest to the desired amino acid profile: they encode 22% serine and 11% tyrosine, asparagine, glycine, alanine, aspartate, threonine and cysteine.
  • libraries are constructed using either the DVT, DVC or DVY codon at each of the diversified positions.
  • Antagonists of TNFRl can be identified by the ability to inhibit TNF-induced cytotoxicity in mouse L929 fibroblasts (Evans, T. (2000) Molecular Biotechnology 15, 243-248). Briefly, L929 cells plated in microtitre plates are incubated overnight with antagonist of TNFRl, 100 pg/ml TNF and lmg/ml actinomycin D (Sigma, Poole, UK).
  • Antagonists of TNFRl can be identified by the ability to inhibit TNF-induced secretion of IL-8 by human HeLa cells (method adapted from that of Akeson, L. et al (1996) Journal of Biological Chemistry 271, 30517-30523, describing the induction of IL-8 by IL-I in HUVEC; here we look at induction by human TNF alpha and we use HeLa cells instead of the HUVEC cell line). Briefly, HeLa cells can be plated in microtitre plates were incubated overnight with antagonist of TNFRl and 300pg/ml TNF.
  • IL-8 concentration is measured using a sandwich ELISA (R&D Systems), or other suitable method.
  • Antagonists of TNFRl inhibit IL-8 secreation, and less IL-8 is detected in the supernatant compared with the TNF only control.
  • mice transgenic for human TNF (TgI 97) were administered in groups of 10 animals with weekly intraperitoneal doses of anti-TNF dAbs which had been reformatted either as dimers with a 40k branched PEG (termed PEG TAR1-5-19) or as a fusion with an anti-serum albumin dAb (termed TAR1-5-19/MSA trimer). Saline administration was used as a control. After 7 weeks of drug administration the serum of the animals was analysed for circulating TNF levels using an anti-TNF sandwich ELISA kit.
  • CDl mice were administered a single intravenous lmg/kg dose of an anti- mouse TNFRl dAb which had been reformatted (extende half-life formats) with either a 40k branched PEG (termed PEG anti-TNFRl dAb) or as a fusion with an anti-serum albumin dAb (termed anti-TNFRl /anti-S A dimer). Serum was examined at various time points for levels of soluble TNFRl using a anti-TNFRl sandwich ELISA as detailed below.
  • TNFRl F96 Maxisorp 96 well flat bottom immunoplates (Nunc, Cat No: 439454) were coated with anti-mouse sTNF Rl / TNFRSFlA antibody (50ul per well at lug/ml in PBS). Plates were incubated for lhr at room temperature. Plates were washed three times with 200ul PBS. Non-specific binding was blocked with 3% BSA/PBS for lhr at room temperature. Plates were washed three times with 200ul PBS/0.05% Tween-20.
  • Dilutions of recombinant mouse sTNF Rl were prepared in PBS/1% BSA at 5000ng ml '1 , 500ng ml "1 , 50ng ml "1 and 5ng ml "1 and 50ul added to the well.
  • serum from mice immunised with TAR2m-21-23/40K branched PEG was prepared at 1/5, 1/50, 1/500 and 1/5000 dilutions in PBS/1%BSA and 50ul added to the well. Plates were incubated for 1 hour, room temperature. Plates were washed three times with 200ul PBS/0.05% Tween-20.
  • Biotinylated anti-mouse sTNF Rl antibody (R & D Systems, cat # AHFOl) was prepared at lug/ml and 50ul added to the well. Plates were incubated for 1 hour, room temperature. Plates were washed three times with 200ul PBS/0.05% Tween-20. Peroxidase-conjugated IgG fraction monoclonal mouse anti-biotin (Stratech,
  • Cat No: 200-032-096 was prepared at 160ng ml-1 and 50ul added to the well. Plates were incubated for 1 hour, room temperature. Plates were washed three times with 200ul PBS/0.05% Tween-20 then three times with PBS.
  • PEG anti-TNFRl dAb were plotted against time ( Figure 1). The results clearly show the increasing levels of TNFRl in the serum before falling back to basal levels following clearance of the dAb.
  • Example 3 dAbs That Bind Receptor Molecules But Do Not Bind The Active Site.
  • This example illustrates a method for identify dAbs that bind a desired receptor molecule but does not bind the active site of the receptor.
  • the approach is illustrated using tumor necrosis factor 1 receptor (TNFRl), and is generally applicable to receptor molecules.
  • TNFRl tumor necrosis factor 1 receptor
  • Chimeric receptor molecules made up of different murine TNFRl domains and human TNFRl domains (Banner DW, et al. Cell, 75(3):431-45 (1993).) such that the molecule contains the four defined extracellular domains of TNFRl but that these vary in origin between mouse and human TNFRl proteins were produced.
  • the produced chimeric receptors shared properties of both human TNFRl and mouse TNFRl according to the differing domain roles and functionality.
  • the molecules provided a means for the assessment of the domain specificity of dAbs, antibodies and antigen-binding fragments thereof and other molecules (eg protein domains such as affibodies, LDL receptor domains, or EGF domains) that bind human or mouse TNFRl .
  • Human and mouse TNFRl sequences were previously cloned into the Pichia expression vector pPicZalpha (Invitrogen) via EcoRl and Notl restriction endonuclease sites.
  • the template mouse TNFRl DNA (and consequently chimeric receptor constructs ending with a murine domain 4) contained a 3 ' 6x Histidine tag.
  • Human TNFRl ( and consequently chimeric receptor constructs ending with a human domain 4) contained both Myc and 6x Histidine tags in sequence at the 3' end.
  • Typical PCR reacts were set up was as follows: 25 ⁇ l of 1OX RubyTaq PCR buffer containing polymerase; 2 ⁇ l of first primer (from 10 ⁇ M stock); 2 ⁇ l of second primer (from 10 ⁇ M stock); 1 ⁇ l (100 ng) full length TNFRl template DNA; 20 ⁇ l of dH 2 O (to a final volume of 50 ⁇ l).
  • the reactions were set up in thin walled tubes and placed into a therniocycler where the reaction was performed according to the following parameters.
  • H human domain
  • M TnOUSe domain
  • MHHH mouse Domain 1
  • PCR products generated these initial PCRs were cut out from a 1% agarose gel and purified using a gel purification kit (Qiagen) before elution into 50 ⁇ l dH 2 0.
  • Assembly PCR also known as 'pull-through' or Splicing by Overlap Extension (SOE) see Gene, 75:77(l):61-8 (1989)
  • SOE Overlap Extension
  • Assembly PCR allows the primary PCR products to be brought together without digest or ligation, making use of the complementary ends of the Primary PCR products.
  • the primary products are brought together and denatured before their complementary ends are allowed to anneal together in the presence of Taq DNA polymerase and dNTPs.
  • Several cycles of reannealing and extension result in fill-in of the complementary strands and the production of a full-length template.
  • Primers that flank the now full-length construct cassette are added and a conventional PCR was run to amplify the assembled product.
  • SOE PCRs were performed in order to anneal together and amplify the various TNFRl domains derived from the initial PCRs described above. Assembly SOE PCRs were set up as follows: 40 ⁇ l 10 x PCR buffer containing MgCl 2 ; ⁇ 2 ⁇ l (100 ng) cleaned product of initial PCR 1; ⁇ 2 ⁇ l (100 ng) cleaned product of initial PCR 2; 36 ⁇ l dH 2 O (to final volume of 80 ⁇ l).
  • SOE primer mix was added after the assembly step as follows: 2 ⁇ l 5' flanking primer (Primer 1); 2 ⁇ l 3' flanking primer (Primer 2); 10 ⁇ l 1Ox PCR Buffer; 6 ⁇ l dH2O (to final volume 20 ⁇ l).
  • the PCR reactions were performed using the program described below. The initial assembly cycles required approximately 45 minutes after which the thermocycler was set to pause at 94 0 C. 20 ⁇ l of primer mix was added to each reaction and mixed.
  • PCR products were checked by running 3-5 ⁇ l of each reaction on a 1% agarose gel.
  • pPicZalpha vector (Invitrogen) was sequentially digested with EcoRI and Notl enzymes prior to Chromaspin TE- 1000 gel filtration column (Clontech, Mountain View, CA) purification.
  • the ligated chimeric constructs were transformed into HB2151 electrocompetent E. coli cells and recovered for an hour in low salt LB media prior to plating on low salt LB agar with 0.25 ⁇ g/ml ZEOCIN, antibiotic formulation containing Phleomycin D (Cayla, Toulouse, France), for 24 hrs at 37 0 C. Individual colonies were then sequence verified to ensure the correct sequence of the chimeric construct within the expression vector and large scale Maxiprep plasmid preparations made of each chimeric construct vector.
  • the nucleotide sequences of prepared chimeric constructs are presented below.
  • the chimeric constructs were named according to origin of their domains (running from Domain 1 on the left to Domain 4 on the right). For example,
  • HMMM contains human Domain 1 and mouse Domains 2-4. Chimeric proteins that contain mouse domain 4 have only His tags and lack the spacer region between the transmembrane region and Domain 4.
  • TNFRl chimeric construct Preparation of TNFRl chimeric construct and transformation into Pichiapastoris.
  • the plasmid DNA generated by each maxiprep was digested with the infrequent cutting restriction endonuclease Pmel in order to linearise the DNA prior to pichia transformation.
  • the linearised DNA was subsequently cleaned by phenol/chloroform extraction and ethanol precipitation, before resuspension in 30 ⁇ l of dH 2 O. 10 ⁇ l of the linearised DNA solution was mixed with 80 ⁇ l of electro- competent KM71H Pichia cells for 5 minutes prior to electroporation at 1.5kV, 200 ⁇ , 25 ⁇ F.
  • An individual transformant colony for each construct was picked into 5 ml of BMGY as a starter culture and grown for 24 hrs at 3O 0 C. This culture was used to inoculate 500 ml of BMGY media which was grown for 24 hrs at 3O 0 C before cells were harvested by centrifugation at 1500-3000g for 5 minutes at room temp. Cells were then resuspended in 100 ml of BMMY and grown for 4 days with staggered increases in methanol concentration (0.5% day 1, 1% day 2, 1.5% day 3 and 2% day 4). After expression supernatant was recovered after centrifugation of the cultures at 330Og for 15 minutes.
  • Culture supernatants were initially buffered through addition of 10 mM final concentration imidazole and 2x PBS. His-tagged protein was batch absorbed for 4 hours (shaking) at room temperature through addition of Nickel-NT A resin. The supernatant/resin mix was then flowed into a poly-prep column (Biorad). Resin was then washed with 10 column volumes of 2xPBS before elution using 250 mM imidazole Ix PBS. After buffer exchange the chimeric construct expression was deglycosylated using the EndoH deglycosylase before verification by SDS-PAGE.
  • TNFRl Extracellular region Genbank accession 339914178 CTGGTCCCTCACCTAGGGGACAGGGAGAAGAGAGATAGTGTGTGTCCCCAAGG AAAATATATCCACCCTCAAAATAATTCGATTTGCTGTACCAAGTGCCACAAAGG AACCTACTTGTACAATGACTGTCCAGGCCCGGGGCAGGATACGGACTGCAGGG AGTGTGAGAGCGGCTCCTTCACCGCTTCAGAAAACCACCTCAGACACTGCCTCA GCTGCTCCAAATGCCGAAAGGAAATGGGTCAGGTGGAGATCTCTTCTTGCACAG TGGACCGGGACACCGTGTGTGGCTGCAGGAAGAACCAGTACCGGCATTATTGG AGTGAAAACCTTTTCCAGTGCTTCAATTGCAGCCTCTGCCTCAATGGGACCGTG CACCTCTCCTGCCAGGAGAAACAGAACACCGTGTGCACCTGCCATGCAGGTTTC TTTCTAAGAAAACGAGTGTGTCTCCTGTAGTAACTGTAAGAA
  • the encoded extracellular region of human TNFRl has the following amino acid sequence.
  • LVPHLGDREKRDSVCPQGKYIHPQNNSICCTKCHKGTYLYNDCPGPGQDTDCRECE SGSFTASENHLRHCLSCSKCRKEMGQVEISSCTVDRDTVCGCRKNQYRHYWSENLF QCFNCSLCLNGTVHLSCQEKQNTVCTCHAGFFLRENECVSCSNCKKSLECTKLCLP QIENVKGTEDSGTT (SEQ ID NO:2)
  • Murine (Mus musculus) TNFRl extracellular region Genbank accession 31560798) CTAGTCCCTTCTCTTGGTGACCGGGAGAAGAGGGATAGCTTGTGTCCCCAAGGA AAGTATGTCCATTCTAAGAACAATTCCATCTGCTGCACCAAGTGCCACAAAGGA ACCTACTTGGTGAGTGACTGTCCGAGCCCAGGGCGGGATACAGTCTGCAGGGA GTGTGAAAAGGGCACCTTTACGGCTTCCCAGAATTACCTCAGGCAGTGTCTCAG TTGCAAGACATGTCGGAAAGAAATGTCCCAGGTGGAGATCTCTCCTTGCCAAGC TGACAAGGACACGGTGTGTGGCTGTAAGGAGAACCAGTTCCAACGCTACCTGA GTGAGACACACTTCCAGTGCGTGGACTGCAGCCCCTGCTTCAACGGCACCGTGA CAATCCCCTGTAAGGAGACTCAGAACACCGTGTGTAACTGCCATGCAGGGTTCT TTCTGAGAAAGTGAGTGCGTCCCTTGCAGGG
  • the encoded extracellular region of murine (Mus musculus) TNFRl has the following amino acid sequence.
  • LVPSLGDREKRDSLCPQGKYVHSKNNSICCTKCHKGTYLVSDCPSPGRDTVCRJECE KGTFTASQNYLRQCLSCKTCRKEMSQVEISPCQADKDTVCGCKENQFQRYLSETHF QCVDCSPCFNGTVTIPCKETQNTVCNCHAGFFLRESECVPCSHCKKNEECMKLCLP PPLANVTNPQDSGTA (SEQ ID NO:4)
  • the domain specificity of anti-TNFRl dAbs was determined using surface plasmon resonance (SPR) ('Detection of immuno-complex formation via surface plasmon resonance on gold-coated diffraction gratings.' Biosensors. 1987- 88;3(4):211-25.) to determine the ability of antibodies to bind fully human or mouse biotinylated TNFRl that was immobilized on a SPR chip surface, after the antibodies had been incubated and equilibrated with an excess of the chimeric receptors described above.
  • SPR surface plasmon resonance
  • flow of an anti-TNFRl dAb over the TNFRl surface generates an SPR signal indicating that amount of dAb that binds TNFRl immoblized on the SPR chip. If the dAb is pre-incubated and equilibrated with a chimeric molecule that comprises the domain(s) of TNFRl that the particular dAb binds, then flow of this mixture over the TNFRl surface will produce a smaller SPR signal relative to the dAb alone.
  • the dAb is pre-incubated and equilibrated with a chimeric molecule that does not comprises the domain(s) of TNFRl that the particular dAb binds, then flow of this mixture over the TNFRl surface will produce a SPR signal that is about the same as the signal obtained using dAb alone.
  • TNFRl surface is determined by the species specificity of the anti-TNFRl dAb to be tested. Therefore anti-human TNFRl dAbs were evaluated using a surface coated with human TNFRl and anti-mouse TNFRl dAbs were evaluated using a chip coated with mouse TNFRl .
  • Biotinylated TNFRl was diluted in the appropriate SPR buffer and run across a streptavidin (SA) sensor chip in a BIACORE 3000 SPR instrument (Biacore International AB, Uppsala, Sweden). A low flow-rate (5-10 ⁇ l/minute) was used in order to maximise the contact time between the biotinylated TNFRl and the streptavidin surface. Flow continued until the streptavidin surface was saturated with biotinylated material, in order to generate a chip with maximal TNFRl surface. The chip typically bound several hundred to several thousand SPR response units of the biotinylated material.
  • a successful competition experiment requires initial optimisation of the concentration of anti-TNFRl dAb such that the minimum amount of dAb is flowed over the surface that gives a significant SPR signal. Within a certain concentration range, the dAb will bind the surface in a dose dependent manner so that the number of RUs of dAb bound reflect the concentration of dAb flowed across the chip surface.
  • the anti-TNFRl dAbs were titrated in a 10-fold dilution series of Biacore buffer ranging from a 1 in 10 dilution to a 1 in 1,000,000 dilution. The dilutions were then individually and sequentially injected across the TNFRl chip surface, starting with the most dilute sample. The maximal number of RUs achieved at each dilution were measured. After each injection the TNFRl surface was regenerated to remove bound anti-TNFRl dAb where necessary using a suitable SPR regeneration buffer. Using this method the minimal concentration of anti-TNFRl dAb required to generate a signal representing approximately IOORU was determined.
  • anti- TNFRl dAb/chimeric TNFRl mixes were set up. Mixes were set up such that the final concentration of anti-TNFRl dAb was identical to the optimal concentration determined previously. Reactions were typically set up in 100 ⁇ l volumes containing 50 microliters of a 2 x concentrate of anti-TNFRl dAb, 40 microliters of Biacore buffer and 10 microliters of neat, purified chimeric protein. Typical concentrations for the final mix were about 10-100 ⁇ M of chimeric protein and about 10-100 nM anti-TNFRl dAb. Mixtures were allowed to equilibrate for 30 minutes at room temperature. Competition Biacore experiment
  • each anti-TNFRl dAb/chimeric TNFRl mixture was sequentially run over the TNFRl SPR surface and the number of response units measured. After each mixture was injected, the surface was regenerated to remove bound anti-TNFRl dAb on before the next mixture was injected.
  • the different responses generated using the different chimerics enabled determination of the TNFRl domains bound by particular dAbs.
  • TNFRl TNFRl
  • TAR2h-205 binds Domain 1 of human TNFRl.
  • CDRl is flanked by ⁇
  • CDR2 is flanked by ⁇
  • CDR3 is flanked by ⁇ — .
  • the presented amino acid sequences are continuous with not gaps.
  • Chimeric receptor proteins such as the chimeric TNFRl proteins described herein can be used in assays or screens to isolate agents (e.g., antibodies, dAbs) that bind to particular domains within a desired receptor receptor.
  • agents e.g., antibodies, dAbs
  • TNFRl as a model for a desired receptor protein, these methods describe the addition of chimeric proteins to crude antibody preparations prior to their screening for receptor binding either by ELISA or surface plasmon resonance. Additionally they describe the use of chimeric proteins coated on a surface (e.g., ELISA plate or SPR chip) and the screening of antibodies through testing of their binding to chimeric proteins on this surface. Similar methods can be used to identify agents (e.g., antibodies, dAbs) that bind to desired domains (e.g., out side of the active site) of other receptors.
  • agents e.g., antibodies, dAbs
  • This method can be used to rapidly isolate antibodies or antibody fragments (e.g., dAbs) that bind specific domains of TNFRl from a large repertoire of antibodies or antibody fragments of unknown specificity.
  • a 96 well assay plate will be coated overnight at 4 0 C with lOO ⁇ l per well of chimeric TNFRl.
  • Wells will be washed 3 times with 0.1% TPBS (Phosphate buffered saline containing Tween-20 at a concentration of 0.1%). 200 ⁇ l per well of 1% TPBS will be added to block the plate, and the plate incubated for 1-2 hours at room temperature.
  • TPBS Phosphate buffered saline containing Tween-20 at a concentration of 0.1%)
  • Wells will then be washed 3 times with PBS before addition of 50 ⁇ l of bacterial supernatant or periprep, containing the soluble antibody or antibody fragment (that contain the c-Myc epitope tag), in 50 ⁇ l 0.2% TPBS.
  • the plate will then be incubated for 1 hour at room temperature. After this the plate will be washed 5 times with 0.1% TPBS (0.1% Tween-20 in PBS).
  • lOO ⁇ l of a primary anti-c-Myc mouse monoclonal will then be added in 0.1% TPBS to each well and the plate will be incubated for 1 hour at room temperature. This primary antibody solution will be discarded and the plate will then be washed 5 times with 0.1% TPBS.
  • This method can be used to rapidly screen a diverse sets of crude antibody or antibody fragment preparations that bind TNFRl in order to determine their domain binding specificity.
  • a 96 well assay plate will be coated overnight at 4 0 C with lOO ⁇ l per well of murine or human TNFRl (either human or mouse). Wells will be washed 3 times with 0.1% TPBS (Phosphate buffered saline containing Tween-20 at a concentration of 0.1%). 200 ⁇ l per well of 1% TPBS (1% Tween-20 in PBS) will be added to block the plate, and the plate incubated for 1-2 hours at room temperature. Wells will then be washed 3 times with PBS .
  • bacterial supernatants or peripreps will be pre-equilibriated with a pre-optimised concentration of chimeric TNFRl protein in solution.
  • 50 ⁇ l of this crude bacterial preparatiori/chimeric protein mix, containing the soluble antibody or antibody fragment will then be added to the ELISA plate.
  • the plate will be incubated for 1 hour at room temperature.
  • the plate will be washed 5 times with 0.1% TPBS (0.1% Tween-20 in PBS), and lOO ⁇ l of a primary detecting antibody (or Protein A-HRP or Protein L HRP) will be added in 0.1% TPBS, to each well and the plate will be incubated for 1 hour at room temperature.
  • a primary detecting antibody or Protein A-HRP or Protein L HRP
  • This primary antibody solution will be discarded and the plate will be washed 5 times with 0.1% TPBS. If required lOO ⁇ l of a prediluted secondary antibody/HRP conjugate from goat will then be added, and the plate will be incubated for 1 hour at room temperature. The secondary antibody will then be discarded and the plate will be washed 6 times with 0.1% TPBS followed by 2 washes with PBS. 50 ⁇ l of TMB peroxidase solution will be added to each well and the plate will be left at room temperature for 2-60 minutes. The reaction will be stopped by the addition of 50 ⁇ l of IM hydrochloric acid. The OD at 450 nni of the plate will be read in a 96-well plate reader within 30 minutes of acid addition. A reduction in ELISA signal will be indicative of the antibody binding the chimeric TNFRl domains rather than the full TNFRl coated on the plate, and therefore, that the antibody binds one of the domains within the chimeric protein.
  • This method can be used to rapidly screen diverse sets of crude antibody or antibody fragment preparations that bind TNFRl for those antibodies or antibody fragments that compete with a reference antibody or antibody fragment (e.g., TAR2m-21-23) for binding to TNFRl or bind a desired domain of TNFRl (e.g., domain 1).
  • the method uses a reference antibody or antibody fragment and test antibody or antibody fragment (e.g., a population of antibodies to be screened) that contain different detectable tags (epitope tags).
  • a 96 well assay plate will be coated overnight at 4°C with lOO ⁇ l per well of murine or human TNFRl. Wells will be washed 3 times with 0.1% TPBS (Phosphate buffered saline containing Tween-20 at a concentration of 0.1%). 200 ⁇ l per well of 1% TPBS (1% Tween-20 in PBS) will be added to block the plate, and the plate will be incubated for 1-2 hours at room temperature. Wells will then be washed 3 times with PBS. At the same time the crude antibody preparations to be tested will be mixed with a pre-optimised concentration of reference antibody or antibody fragment (e.g., domain 1-binding antibody; TAR2m-21-23) in solution.
  • TPBS Phosphate buffered saline containing Tween-20 at a concentration of 0.1%). 200 ⁇ l per well of 1% TPBS (1% Tween-20 in PBS) will be added to block the plate, and the plate will be in
  • this antibody does not include the same detection tags as present in the antibodies being screened for domain binding specificity.
  • 50 ⁇ l of this crude antibody/reference antibody mix will be added to the ELISA plate. The plate will then be incubated for 1 hour at room temperature. After, the plate will be washed 5 times with 0.1% TPBS, lOO ⁇ l of a primary detecting antibody (that binds the tag present only on the antibody population being screened) will be added in 0.1% TPBS to each well, and the plate will be incubated for 1 hour at room temperature. This primary antibody solution will be discarded and the plate will then be washed 5 times with 0.1% TPBS.
  • a reduction in ELISA signal in the presence of the reference antibody or antibody fragment, in comparison to the ELISA signal for the same antibody preparation without competing reference antibody or antibody fragment, will be indicative that the particular antibody or antibody fragment competes with the reference antibody or antibody fragment for binding to TNFRl, and binds the same domain of TNFRl as the reference antibody or antibody fragment.
  • ELISA Screening The ELISA methods described above can be readily adapted to a format that uses surface plasmon resonance, for example using a BIACORE 3000 SPR instrument (Biacore International AB, Uppsala, Sweden).
  • a BIACORE 3000 SPR instrument Biacore International AB, Uppsala, Sweden.
  • the chimeric protein will be either immobilized on the SPR chip, or the chimeric protein will be equilibrated with crude bacterial supernatant containing anti-TNFRl antibodies or antibody fragments, and the resultant mixture flowed over a SPR chip coated with full length human TNFRl or murine TNFRl .
  • MRC-5 cell assay The activities of certain dAbs that bind human TNFRl were assessed in the following MRC-5 cell assay.
  • the assay is based on the induction of IL-8 secretion by TNF in MRC-5 cells and is adapted from the method described in Alceson, L. et al. Journal of Biological Chemistry 27.7:30517-30523 (1996), describing the induction of IL-8 by IL-I in HUVEC.
  • the activity of the dAbs was assayed by assessing IL-8 induction by human TNFa using MRC-5 cells instead of the HUVEC cell line. Briefly, MRC-5 cells were plated in microtitre plates and the plates were incubated overnight with dAb and human TNFa (300 pg/ml).
  • the culture supernatant was aspirated and the IL-8 concentration in the supernatant was measured via a sandwich ELISA (R&D Systems).
  • Anti-TNFRl dAb activity resulted in a decrease in IL-8 secretion into the supernatant compared with control wells that were incubated with TNFa only.
  • Anti-TNF dAbs were tested for the ability to inhibit the binding of TNF to recombinant TNF receptor 1 (p55). Briefly, Maxisorp plates were incubated overnight with 30mg/ml anti-human Fc mouse monoclonal antibody (Zymed, San Francisco, USA). The wells were washed with phosphate buffered saline (PBS) containing 0.05% Tween-20 and then blocked with 1% BSA in PBS before being incubated with lOOng/ml TNF receptor 1 Fc fusion protein (R&D Systems, Minneapolis, USA). Anti-TNF dAb was mixed with TNF which was added to the washed wells at a final concentration of 10ng/ml. TNF binding was detected with 0.2mg/ml biotinylated anti-TNF antibody (HyCuIt biotechnology, Uben,
  • the receptor binding assay which utilises a recombinant form of receptor, containing only the extracellular portion of the receptor in a non-cell based assay format, is shown on the left, where the effects on PEGylation are minimal, and in the case of 5K PEG, no difference in binding was observed.
  • PEGylation of dAbs substantially reduced the effectiveness of the dAbs in the assay. In some cases, effectiveness with reduced up to a factor of 33-fold.
  • the data collected in cell based assay where based on the ability of the dAbs to block receptor activity at the cell surface. The results of the study show that PEGylation of dAbs had a much greater effect on the ability of the dAbs to inhibit function of TNFRl on the cell surface than on the ability to bind TNFRl in a cell free assay.
  • This example illustrates a method for identify dAbs that bind a desired endogenous agonist molecule but do not bind the active site of the endogenous agonist.
  • the approach is illustrated using erythropoietin (epo), and is generally applicable to endogenous agonists.
  • Domain antibodies that bind EPO can be identified and isolated using any suitable screening method.
  • a cell line that expresses recombinant EPO receptor and that is dependent upon hematopoietic factors for growth can be used in an assay to identify dAbs that bind EPO but do not bind in the active site, and therefore do not prevent binding of EPO to the EPO receptor.
  • Ba/F3 cells which is a murine IL-3 dependent cell line that has been transfected with human EPO receptor and undergoes epo-dependent growth can be used. (D'Andrea et al., Blood 82:46-52 (1993).
  • Ba/F3 cells can be cultured in a suitable culture dish, the culture media can be removed and the cells can be washed using a suitable buffer such as PBS. The cells can then be cultured for about 48 hours in culture media that contains EPO.
  • Domain antibodies to be assayed can be added to individual cultures (e.g., individual wells of a suitable cell culture assay plate). After about 48 hours, cell proliferation can be assessed using any suitable method, such as the dimethylthiazol diphenyl- tetrazolium brimide (MTT) reduction assay. A lack of inhibition of cell proliferation is wells that contain a dAb relative to control wells that contain culture media but not dAb will indicate that the dAb does not bind the active site of epo.
  • MTT dimethylthiazol diphenyl- tetrazolium brimide

Abstract

L'invention se rapporte à des ligands qui comprennent une fraction (par ex. : dAb) comportant un site de liaison présentant une spécificité de liaison pour un composé endogène cible, mais qui n'inhibent sensiblement pas l'activité de ce composé endogène cible. De préférence, le ligand ne se lie pas au site actif d'un composé endogène cible. Cette invention concerne en outre l'utilisation d'un tel ligand pour produire un médicament servant à augmenter la demi-vie, la biodisponibilité, l'activité, ou la quantité d'un composé endogène cible auquel le ligand se lie.
EP05801585A 2004-11-10 2005-11-10 Ligands ameliorant des composants endogenes Ceased EP1814584A2 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
EP11161978A EP2420251A3 (fr) 2004-11-10 2005-11-10 Ligands ameliorant des composants endogenes

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US10/985,847 US20060002935A1 (en) 2002-06-28 2004-11-10 Tumor Necrosis Factor Receptor 1 antagonists and methods of use therefor
PCT/GB2005/004319 WO2006051288A2 (fr) 2004-11-10 2005-11-10 Ligands ameliorant des composants endogenes

Publications (1)

Publication Number Publication Date
EP1814584A2 true EP1814584A2 (fr) 2007-08-08

Family

ID=36282598

Family Applications (2)

Application Number Title Priority Date Filing Date
EP11161978A Withdrawn EP2420251A3 (fr) 2004-11-10 2005-11-10 Ligands ameliorant des composants endogenes
EP05801585A Ceased EP1814584A2 (fr) 2004-11-10 2005-11-10 Ligands ameliorant des composants endogenes

Family Applications Before (1)

Application Number Title Priority Date Filing Date
EP11161978A Withdrawn EP2420251A3 (fr) 2004-11-10 2005-11-10 Ligands ameliorant des composants endogenes

Country Status (11)

Country Link
EP (2) EP2420251A3 (fr)
JP (1) JP2008519813A (fr)
KR (2) KR20070084069A (fr)
CN (2) CN101724071A (fr)
AU (1) AU2005303584A1 (fr)
BR (1) BRPI0517569A (fr)
CA (1) CA2587206A1 (fr)
MA (2) MA29427B1 (fr)
NO (2) NO20071788L (fr)
RU (2) RU2401842C2 (fr)
WO (1) WO2006051288A2 (fr)

Families Citing this family (31)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9321832B2 (en) 2002-06-28 2016-04-26 Domantis Limited Ligand
KR20120133403A (ko) 2004-06-01 2012-12-10 도만티스 리미티드 증강된 혈청 반감기를 가지는 이특이성 융합 항체
WO2006036568A2 (fr) * 2004-09-13 2006-04-06 Arrowsmith Technologies Llp Absorption in vivo d'un ligand par des anticorps
CA2589802A1 (fr) * 2004-12-02 2006-06-08 Domantis Limited Compositions, fusions et conjugues de domaine plad
US20140113860A1 (en) 2006-02-03 2014-04-24 Prolor Biotech Ltd. Long-acting polypeptides and methods of producing and administering same
US9249407B2 (en) 2006-02-03 2016-02-02 Opko Biologics Ltd. Long-acting coagulation factors and methods of producing same
US8048849B2 (en) 2006-02-03 2011-11-01 Modigene, Inc. Long-acting polypeptides and methods of producing same
US10221228B2 (en) 2006-02-03 2019-03-05 Opko Biologics Ltd. Long-acting polypeptides and methods of producing and administering same
US10351615B2 (en) 2006-02-03 2019-07-16 Opko Biologics Ltd. Methods of treatment with long-acting growth hormone
US8946155B2 (en) 2006-02-03 2015-02-03 Opko Biologics Ltd. Long-acting polypeptides and methods of producing and administering same
US20150038413A1 (en) 2006-02-03 2015-02-05 Opko Biologics Ltd. Long-acting polypeptides and methods of producing and administering same
US9458444B2 (en) 2006-02-03 2016-10-04 Opko Biologics Ltd. Long-acting coagulation factors and methods of producing same
US9663778B2 (en) 2009-07-09 2017-05-30 OPKO Biologies Ltd. Long-acting coagulation factors and methods of producing same
CN102010473A (zh) * 2010-11-10 2011-04-13 曹鹏 重组胃泌酸调节素融合蛋白及其制备和应用
SG10201806648TA (en) 2011-07-01 2018-09-27 Ngm Biopharmaceuticals Inc Compositions, uses and methods for treatment of metabolic disorders and diseases
RS59076B1 (sr) * 2012-03-16 2019-09-30 Regeneron Pharma Glodari koji eksprimiraju imunoglobulinske sekvence senzitivne na ph
CN104487082A (zh) 2012-04-19 2015-04-01 奥普科生物制品有限公司 长效胃泌酸调节素变体及其生产方法
BR122020018510B1 (pt) 2012-11-20 2023-03-14 Opko Biologics Ltd Método para aumentar incrementalmente o tamanho hidrodinâmico de um fator de coagulação ativado viia
US9963494B2 (en) 2012-11-28 2018-05-08 Ngm Biopharmaceuticals, Inc. Methods of using compositions comprising variants and fusions of FGF19 polypeptides for reducing glucose levels in a subject
CN105008548B (zh) 2012-12-27 2020-11-27 恩格姆生物制药公司 用于调节胆汁酸体内稳态以及治疗胆汁酸紊乱和疾病的方法
RU2560699C2 (ru) * 2013-08-02 2015-08-20 федеральное государственное автономное образовательное учреждение высшего профессионального образования "Национальный исследовательский ядерный университет МИФИ" (НИЯУ МИФИ) Способ создания наноразмерной диагностической метки на основе конъюгатов наночастиц и однодоменных антител
US20150158926A1 (en) 2013-10-21 2015-06-11 Opko Biologics, Ltd. Long-acting polypeptides and methods of producing and administering same
WO2016018931A1 (fr) 2014-07-30 2016-02-04 Ngm Biopharmaceuticals, Inc. Compositions et méthodes d'utilisation pour le traitement de troubles métaboliques
IL251834B2 (en) 2014-10-23 2023-09-01 Ngm Biopharmaceuticals Inc Pharmaceutical compositions containing peptide variants and methods of using them
AU2015339130C1 (en) 2014-10-31 2021-03-18 Ngm Biopharmaceuticals, Inc. Compositions and methods of use for treating metabolic disorders
WO2016203482A2 (fr) 2015-06-19 2016-12-22 Opko Biologics Ltd. Facteurs de coagulation à action prolongée et leurs procédés de production
CA3003616C (fr) 2015-11-09 2020-07-28 Ngm Biopharmaceuticals, Inc. Methodes de traitement de troubles associes aux acides biliaires
HUE064463T2 (hu) 2016-07-11 2024-03-28 Opko Biologics Ltd Hosszantartó hatású VII. koagulációs faktor és az elõállítására vonatkozó eljárások
CN110128536A (zh) * 2018-02-02 2019-08-16 暨南大学 抗肿瘤干细胞标志蛋白cd133的单域抗体及其应用
CN109468294A (zh) * 2018-11-06 2019-03-15 青岛古高生物科技有限公司 一种血红素-GroEL复合体及其制备方法和应用
WO2021214501A1 (fr) 2020-04-22 2021-10-28 Genima Innovations Marketing Gmbh Décontamination antivirale dans le tuyau d'aspiration de canal ou dans l'antichambre d'un masque respiratoire

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997006251A1 (fr) * 1995-08-04 1997-02-20 Lek, Tovarna Farmacevtskih In Kemic^¿Nih Izdelkov, D.D. ANTICORPS MONOCLONAUX DIRIGES CONTRE LES RECEPTEURS SOLUBLES p55 ET p75 DU FACTEUR α DE NECROSE TUMORALE TNF-α DE MEME QUE CONTRE TNF-α ET SES ANALOGUES
WO2001058953A2 (fr) * 2000-02-11 2001-08-16 The Government Of The United States Of America As Represented By The Secretary, Department Of Health And Human Services Identification d"un nouveau domaine dans la famille des recepteurs du facteur de necrose tumorale mediant l"assemblage et la fonction du recepteur de pre-ligand
WO2006038027A2 (fr) * 2004-10-08 2006-04-13 Domantis Limited Antagonistes et leurs methodes d'utilisation

Family Cites Families (58)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0467416A1 (fr) * 1983-09-01 1992-01-22 Hybritech Incorporated Compositions d'anticorps d'agents therapeutiques à période de demi-vie sérique prolongée
SU1618761A1 (ru) 1987-04-29 1991-01-07 Институт Белка Ан Ссср Способ получени пептидов и белков в бесклеточной системе трансл ции
DE3889783T2 (de) * 1987-07-07 1994-09-08 Hybrisens Ltd Verwendung von Antikörper-Antigen-Wechselwirkung zum Schutz oder Modulation der biologischen Aktivität.
GB8725529D0 (en) 1987-10-30 1987-12-02 Delta Biotechnology Ltd Polypeptides
US5225540A (en) * 1988-04-26 1993-07-06 Du Pont Merck Pharmaceutical Company Monoclonal antibodies to tissue plasminogen activator (t-pa) which prolong its functional half-life
AU649217B2 (en) 1988-11-18 1994-05-19 Regents Of The University Of California, The Method for site-specifically incorporating unnatural amino acids into proteins
SU1705302A1 (ru) 1988-12-22 1992-01-15 Институт Белка Ан Ссср Способ препаративной экспрессии генов в бесклеточной системе сопр женной транскрипции/трансл ции
ATE92107T1 (de) 1989-04-29 1993-08-15 Delta Biotechnology Ltd N-terminale fragmente von menschliches serumalbumin enthaltenden fusionsproteinen.
CA2064754C (fr) 1989-07-31 1996-06-18 Vladimir Ivanovich Baranov Methode de preparation de polypeptides dans un systeme de translation sans cellules
US5977307A (en) 1989-09-07 1999-11-02 Alkermes, Inc. Transferrin receptor specific ligand-neuropharmaceutical agent fusion proteins
CA2067194C (fr) 1989-10-05 2003-03-18 Glenn Kawasaki Synthese sans cellules et isolement de nouveaux genes et polypeptides
GB9015198D0 (en) 1990-07-10 1990-08-29 Brien Caroline J O Binding substance
WO1992002536A1 (fr) 1990-08-02 1992-02-20 The Regents Of The University Of Colorado Evolution polypeptidique systematique par traduction inverse
WO1992005258A1 (fr) 1990-09-20 1992-04-02 La Trobe University Gene encodant une enzyme de l'orge
CA2104698A1 (fr) 1991-02-21 1992-08-22 John J. Toole Aptameres specifiques de biomolecules et methodes de production
US6277969B1 (en) 1991-03-18 2001-08-21 New York University Anti-TNF antibodies and peptides of human tumor necrosis factor
DK1087013T3 (da) 1992-08-21 2009-05-11 Univ Bruxelles Immunoglobuliner uden lette kæder
AU8124694A (en) 1993-10-29 1995-05-22 Affymax Technologies N.V. In vitro peptide and antibody display libraries
SE9400088D0 (sv) 1994-01-14 1994-01-14 Kabi Pharmacia Ab Bacterial receptor structures
US5605793A (en) 1994-02-17 1997-02-25 Affymax Technologies N.V. Methods for in vitro recombination
GB9416721D0 (en) 1994-08-18 1994-10-12 Short Brothers Plc A bias yarn assembly forming device
WO1997008320A1 (fr) 1995-08-18 1997-03-06 Morphosys Gesellschaft Für Proteinoptimierung Mbh Banques de proteines/(poly)peptides
US5837251A (en) 1995-09-13 1998-11-17 Fordham University Compositions and methods using complexes of heat shock proteins and antigenic molecules for the treatment and prevention of neoplastic diseases
ATE374248T1 (de) 1996-06-27 2007-10-15 Vlaams Interuniv Inst Biotech Antikörpermoleküle, die spezifisch mit dem aktiven zentrum oder dem aktiven spalt eines zielmoleküls interagieren
AU8800598A (en) 1997-06-20 1999-01-04 Innogenetics N.V. B7-binding molecules for treating immune diseases
ATE487790T1 (de) 1997-07-07 2010-11-15 Medical Res Council In-vitro-sortierverfahren
GB9722131D0 (en) 1997-10-20 1997-12-17 Medical Res Council Method
CN1203178C (zh) 1997-10-27 2005-05-25 尤尼利弗公司 多价抗原结合蛋白
EP1051493A2 (fr) 1998-01-26 2000-11-15 Unilever Plc Procede servant a preparer des fragments d'anticorps
EP1002861A1 (fr) 1998-10-26 2000-05-24 Unilever Plc Protéines se liant aux antigènes comprenant un liant conférant une flexibilité conformationnelle limitée
IL127127A0 (en) 1998-11-18 1999-09-22 Peptor Ltd Small functional units of antibody heavy chain variable regions
GB9900298D0 (en) 1999-01-07 1999-02-24 Medical Res Council Optical sorting method
AU2291700A (en) 1999-01-19 2000-08-07 Unilever Plc Method for producing antibody fragments
AU4484400A (en) 1999-04-23 2000-11-10 University Of Washington Prostate-specific polynucleotides, polypeptides and their methods of use
JP2003502024A (ja) 1999-05-14 2003-01-21 メディカル リサーチ カウンシル タンパク質骨格および単量体ポリペプチドを多量体化するためのその使用
US7358096B1 (en) 1999-11-29 2008-04-15 Conopco, Inc. Immobilisation of proteins
DE60138333D1 (de) 2000-03-14 2009-05-28 Unilever Nv Variabele Domänen der schweren Kette eines Antikörpers gegen menschliche Ernährungslipasen und deren Verwendungen
US20050053973A1 (en) 2001-04-26 2005-03-10 Avidia Research Institute Novel proteins with targeted binding
US20050089932A1 (en) 2001-04-26 2005-04-28 Avidia Research Institute Novel proteins with targeted binding
EP1399484B1 (fr) 2001-06-28 2010-08-11 Domantis Limited Ligand a double specificite et son utilisation
WO2004003019A2 (fr) * 2002-06-28 2004-01-08 Domantis Limited Ligand
US6608742B2 (en) 2001-07-06 2003-08-19 Schweitzer Engineering Laboratories, Inc. Voltage and current source selection system for use in the protection, automation and control of an electric power system
JP2005289809A (ja) 2001-10-24 2005-10-20 Vlaams Interuniversitair Inst Voor Biotechnologie Vzw (Vib Vzw) 突然変異重鎖抗体
DE10160133A1 (de) 2001-12-07 2003-06-26 Transmit Technologietransfer Verwendung von koagulationsaktivem Antithrombin III zur Therapie von Angiogenese-abhängigen Erkrankungen
WO2003053531A1 (fr) 2001-12-11 2003-07-03 Majorem Ltd. Moteur de jeu en reseau persistant en temps reel multijoueur massif
AU2003210806A1 (en) 2002-03-05 2003-09-22 Eli Lilly And Company Heterologous g-csf fusion proteins
US20040071705A1 (en) * 2002-06-21 2004-04-15 Dyax Corporation Serum protein-associated target-specific ligands and identification method therefor
AU2003251238A1 (en) 2002-08-07 2004-02-25 Umc Utrecht Holding B.V. Modulation of platelet adhesion based on the surface exposed beta-switch loop of platelet glycoprotein ib-alpha
WO2004041865A2 (fr) 2002-11-08 2004-05-21 Ablynx N.V. Anticorps a domaine unique stabilises
US6683132B1 (en) 2002-12-19 2004-01-27 Eastman Chemical Company Self-crosslinking aqueous acetoacetate-functionalized sulfonated alkyd systems
JP2006523090A (ja) * 2002-12-27 2006-10-12 ドマンティス リミテッド リガンドに、そしてリガンド受容体に特異的な二重特異性単一ドメイン抗体
CN101412759A (zh) 2003-01-10 2009-04-22 埃博灵克斯股份有限公司 治疗性多肽,其同源物、其片段及其在调节血小板介导的聚集方面的应用
EP1627062A1 (fr) 2003-05-14 2006-02-22 Domantis Limited Procede de recuperation de polypeptides qui se deplient de facon reversible a partir d'un repertoire de polypeptides
PL1639011T3 (pl) * 2003-06-30 2009-05-29 Domantis Ltd Pegilowane przeciwciała jednodomenowe (dAb)
CA2543360A1 (fr) 2003-10-24 2005-05-06 Joost A. Kolkman Multimeres et monomeres comprenant des domaines de recepteur de lipoproteines de basse densite de classe a et egf
DK1687338T3 (da) 2003-11-07 2011-02-07 Ablynx Nv Camelidae enkeltdomæne-antistoffer VHH, der er rettet mod epidermal vækstfaktor-receptor og anvendelser deraf
MXPA06009072A (es) 2004-02-09 2007-03-29 Human Genome Sciences Inc Proteinas de fusion de albumina.
CA2561732A1 (fr) * 2004-03-30 2005-10-13 Takeda Pharmaceutical Company Limited Medicament a base d'anticorps

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997006251A1 (fr) * 1995-08-04 1997-02-20 Lek, Tovarna Farmacevtskih In Kemic^¿Nih Izdelkov, D.D. ANTICORPS MONOCLONAUX DIRIGES CONTRE LES RECEPTEURS SOLUBLES p55 ET p75 DU FACTEUR α DE NECROSE TUMORALE TNF-α DE MEME QUE CONTRE TNF-α ET SES ANALOGUES
WO2001058953A2 (fr) * 2000-02-11 2001-08-16 The Government Of The United States Of America As Represented By The Secretary, Department Of Health And Human Services Identification d"un nouveau domaine dans la famille des recepteurs du facteur de necrose tumorale mediant l"assemblage et la fonction du recepteur de pre-ligand
WO2006038027A2 (fr) * 2004-10-08 2006-04-13 Domantis Limited Antagonistes et leurs methodes d'utilisation

Also Published As

Publication number Publication date
AU2005303584A1 (en) 2006-05-18
RU2401842C2 (ru) 2010-10-20
MA29221B1 (fr) 2008-02-01
EP2420251A3 (fr) 2013-03-13
MA29427B1 (fr) 2008-05-02
JP2008519813A (ja) 2008-06-12
WO2006051288A2 (fr) 2006-05-18
CN101724071A (zh) 2010-06-09
RU2007112509A (ru) 2008-11-27
KR20070084069A (ko) 2007-08-24
NO20072475L (no) 2007-08-06
EP2420251A2 (fr) 2012-02-22
BRPI0517569A (pt) 2008-10-14
CA2587206A1 (fr) 2006-05-18
CN101098712A (zh) 2008-01-02
KR20070089930A (ko) 2007-09-04
RU2007117196A (ru) 2008-12-20
NO20071788L (no) 2007-07-06
WO2006051288A3 (fr) 2006-08-17

Similar Documents

Publication Publication Date Title
EP1814584A2 (fr) Ligands ameliorant des composants endogenes
US20070298041A1 (en) Ligands That Enhance Endogenous Compounds
AU2005291017B2 (en) Single domain antibodies against TNFRl and methods of use therefor
AU2003244817B2 (en) Antigen-binding immunoglobulin single variable domains and dual-specific constructs
EP2559702B1 (fr) Domaines variables d'anticorps isolés contre la sérum albumine
US20060257406A1 (en) Ligand
EP1777235A1 (fr) Compositions et méthodes pour le traitement des maladies inflammatoires
KR20080077238A (ko) 인터루킨 1 수용체 타입 1에 결합하는 비경쟁적 도메인항체 포맷
KR20080077237A (ko) 인터루킨 1 수용체 타입 1에 결합하는 경쟁적 도메인 항체포맷
US20080008713A1 (en) Single domain antibodies against tnfr1 and methods of use therefor
US20100168393A1 (en) Antibody Polypeptide Libray Screening and Selected Antibody Polypeptides
US20080233129A1 (en) Ligand that has binding specificity for IL-4 and/or lL-13
US9028822B2 (en) Antagonists against TNFR1 and methods of use therefor
MX2007005650A (es) Ligandos que mejoran compuestos endogenos.
AU2013273775A1 (en) Antibody single variable domains against serum albumin
US20110223168A1 (en) Ligand that has binding specificity for il-4 and/or il-13

Legal Events

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

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20070607

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LI LT LU LV MC NL PL PT RO SE SI SK TR

AX Request for extension of the european patent

Extension state: HR

RAX Requested extension states of the european patent have changed

Extension state: HR

Payment date: 20070607

REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 1106145

Country of ref document: HK

17Q First examination report despatched

Effective date: 20090414

REG Reference to a national code

Ref country code: HK

Ref legal event code: WD

Ref document number: 1106145

Country of ref document: HK

REG Reference to a national code

Ref country code: DE

Ref legal event code: R003

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

Free format text: STATUS: THE APPLICATION HAS BEEN REFUSED

18R Application refused

Effective date: 20150315