WO2006030220A9 - Compositions monovalent for cd40l binding and methods of use - Google Patents

Compositions monovalent for cd40l binding and methods of use

Info

Publication number
WO2006030220A9
WO2006030220A9 PCT/GB2005/003562 GB2005003562W WO2006030220A9 WO 2006030220 A9 WO2006030220 A9 WO 2006030220A9 GB 2005003562 W GB2005003562 W GB 2005003562W WO 2006030220 A9 WO2006030220 A9 WO 2006030220A9
Authority
WO
WIPO (PCT)
Prior art keywords
polypeptide
antibody
cd40l
sequence
amino acid
Prior art date
Application number
PCT/GB2005/003562
Other languages
French (fr)
Other versions
WO2006030220A1 (en
Inventor
Steven Grant
Haiqun Liu
Kevin Moulder
Original Assignee
Domantis Ltd
Steven Grant
Haiqun Liu
Kevin Moulder
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=36074265&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO2006030220(A9) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority to KR1020077008642A priority Critical patent/KR101360140B1/en
Priority to CN200580039317.5A priority patent/CN101061140B/en
Priority to KR1020137005252A priority patent/KR20130039341A/en
Priority to BRPI0515404-9A priority patent/BRPI0515404A/en
Priority to AU2005283932A priority patent/AU2005283932B8/en
Priority to ES05784064.7T priority patent/ES2442455T3/en
Priority to JP2007531827A priority patent/JP5808880B2/en
Application filed by Domantis Ltd, Steven Grant, Haiqun Liu, Kevin Moulder filed Critical Domantis Ltd
Priority to EP05784064.7A priority patent/EP1797126B1/en
Priority to US11/663,247 priority patent/US8524236B2/en
Priority to CA2581017A priority patent/CA2581017C/en
Priority to NZ553847A priority patent/NZ553847A/en
Priority to MX2007003187A priority patent/MX2007003187A/en
Publication of WO2006030220A1 publication Critical patent/WO2006030220A1/en
Publication of WO2006030220A9 publication Critical patent/WO2006030220A9/en
Priority to IL181724A priority patent/IL181724A/en
Priority to NO20071312A priority patent/NO20071312L/en
Priority to HK07108149.6A priority patent/HK1101182A1/en

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
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2875Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the NGF/TNF superfamily, e.g. CD70, CD95L, CD153, CD154
    • 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
    • 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
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • 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
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • 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
    • C07K2317/565Complementarity determining region [CDR]
    • 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®
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding

Definitions

  • CD40 is a 50 IcD cell surface glycoprotein molecule expressed on the surface of mature and immature B cells, macrophages, follicular dendritic cells, thymic epithelium, normal basal epithelium, and some tumor-derived cell lines.
  • the CD40 molecule is a member of the TNF receptor family, and has important signaling functions leading to a variety of downstream effects in various cell types.
  • Early studies showed that cross-linking of CD40 on the B cell surface with an antibody resulted in B cell proliferation and activation.
  • Antibody cross linking of CD40 in the presence of IL-4 induces proliferation and class switching in vitro, B cell aggregation via LFA-I (Gordon et al., 1988, J. Immunol.
  • Anti-CD40 monoclonal antibodies also prime B cells to proliferate in response to agents such as PMA (Gordon et al., 1987, Eur. J. Immunol. 17: 1535) and anti-CD20 antibody (Clark & Ledbetter, 1986, Proc. Natl. Acad. Sci. U.S.A. 83: 4494).
  • a mutant of the Jurkat T cell line was found to constitutively activate human B cells to secrete immunoglobulin (Yellin et al., 1991, J. Immunol. 147: 3389-3395).
  • a monoclonal antibody, termed 5c8, was raised which specifically reacted with the mutant line, but not with the parental Jurkat cell line.
  • the 5c8 antibody immunoprecipitated a 30 kD (more accurately, 29.3 kD, 261 amino acids) cell surface polypeptide and was found to specifically inhibit the B cell helper function of the mutant cell line.
  • T-BAM T-B-cell Activating Molecule
  • CD40L CD40 Ligand
  • gp39 CD154
  • TRAP TRAP
  • CD40L protein shows 82.8% and 77.4% identity at the nucleic acid and amino acid levels, respectively, to a similar protein isolated from murine EL4 thymoma cells. Both of these proteins are ligands for CD40 cell surface antigen expressed on resting B cells. CD40L has also been described as IMD3, a protein involved in hyper-IgM immunodeficiency syndrome.
  • the human gene encoding CD40L maps to chromosome Xq26.3-q27.
  • the gene contains five exons.
  • Deletions, point mutations and frameshift mutations clustering within a limited region of the CD40L extracellular domain have been found to be the basis of a rare X-linked immunodeficiency syndrome (Hyper-IgM immunodeficiency syndrome, HIGMl) characterized by recurrent bacterial infections, very low or absent IgG, IgA and IgE, and normal to increased IgM and IgD serum levels.
  • Hyper-IgM immunodeficiency syndrome characterized by recurrent bacterial infections, very low or absent IgG, IgA and IgE, and normal to increased IgM and IgD serum levels.
  • Causally-related mutations have been found to consist of clustered deletions arising by splice-donor mutations with exon skipping, splice-acceptor mutations with utilization of a cryptic splice site, and deletion/insertion events with the creation of a new splice site.
  • CD40L is expressed on activated, but not resting CD4+ T cells, and was found to play a particularly important role in the humoral immune response, being linked to B cell proliferation, antibody and cytokine production, and cell viability.
  • deletion or mutation of CD40L leads to severe immunodeficiency, both in mice and in humans, characterized by hypogammaglobulinemia and T cell deficits in cell- mediated immunity (Chess, C, 2001, in Therapeutic Immunology, 2 nd edition, Austen, K.F., Burakoff, S., Rosen, F. and Strom, T., eds., Blackwell Sciences, pp. 441-456).
  • CD40L Human CD4+ T cells infected by HIVl, which causes severe dysfunction of cellular immunity, but paradoxically results in intense polyclonal activation of B cells, do not express CD40L. Gene and cell surface expression of the CD40L by activated T cells has been shown to be depressed in a subgroup of patients with common variable immunodeficiency (CVI). Thus, inefficient signaling via CD40 may be responsible, at least in part, for the failure of B cell differentiation in these patients.
  • CVI common variable immunodeficiency
  • CD40L binding to CD40 include, for example, a) rescuing B cells from apoptosis induced by Fas or cross-linking of IgM, b) induction of the co-stimulator molecules CD80 (B7-1) and CD86 (B7-2) which interact with CD28 and CD 152 (CTLA-4) on the surface of activated T cells; c) increased expression of other cell surface activation molecules including CD23, CD54, CD95 and lymphotoxin-a; and d) inducing immunoglobulin class switching (see Chess, supra, and references 25, 44, and 47-60 cited therein).
  • CD40L binding to CD40 also augments the antigen-presenting functions of dendritic cells, inducing maintenance of high levels of MHC class II antigens and upregulation of accessory molecules including CD58 (LF A-3).
  • CD40L induces cytokine production and tumoricidal activity in peripheral blood monocytes.
  • CD40L also co-stimulates the proliferation of activated T cells, and the co-stimulation is accompanied by the production of IFN- ⁇ , TNF- ⁇ and IL2.
  • the expression of CD40L on murine T-helper cells and CD4+ T cells is inhibited by IFN- ⁇ , and is inhibited on T-helper-type 2 cells by TGF- ⁇ .
  • CD40L upregulates the expression of CD54 by cultured Hodgkin and Reed- Sternberg cells.
  • the increased CD54 surface expression is accompanied by increased shedding of surface-bound CD54.
  • CD40L has also been suggested to be important in the induction of tolerance - CD80 and CD86, which are upregulated by CD40L, interact with CD28 to provide essential co-stimulation of T cells, in concert with T cell receptor activation, that results in full activation of T cells.
  • CD80 and CD86-triggered activation of CD28 anergy or tolerance occurs as a consequence of antigen triggering (Linsley & Ledbetter, 1993, Ann. Rev. Immunol. 11: 191-212; Jenkins et al., 1993, Curr Opin. Immunol. 5: 361-367; and Boussiotis et al., 1996, Immunol. Rev. 153: 5- 26).
  • CD40L/CD40 pathway has been implicated in the in vivo priming of CD8+ cytotoxic T lymphocytes (CTSs) by CD4+ T cells.
  • CTSs cytotoxic T lymphocytes
  • CD40L expressed on the surface of activated CD4+ T cells interacts with CD40 expressed on dendritic cells, inducing the dendritic cells to express more MHC, and signaling through CD40 can replace the requirement for CD4+ T-helper cells in priming CD 8+ CTL responses.
  • Blockade of CD40L inhibits CTL priming, emphasizing the vital role of CD40L/CD40 interactions in CTL priming by helper T cells (Ridge et al., 1998, Nature 393: 474-478; Schoenberger et al., 1998, Nature 393: 480-483; Bennett et al., 1998, Nature 393: 478-480).
  • CD40L can also mediate functional interactions of CD4+ T cells with other cells that express CD40, such as fibroblasts, synovial cells and endothelial cells (Yellin et al., 1995, J. Leuko. Biol. 58: 209-216; Yellin et al., 1995, J. Exp. Med. 182: 1857-1864).
  • CD40L induces the expression of CD54 (ICAM-I) and CD106 (VCAM- 1) by fibroblasts, as well as increasing fibroblast IL-6, collagenase and collagen production and inducing fibroblast proliferation.
  • CD40L/CD40 interactions may be involved in the induction of fibrosis associated with autoimmunity and immune responses.
  • CD40L interaction with CD40 induces endothelial cells to express CD62E (E- selectin), ICAM-I and VCAM-I.
  • CD62E E- selectin
  • ICAM-I ICAM-I
  • VCAM-I VCAM-I
  • the upregulation of these adhesion molecules may be involved in the binding of inflammatory cells to vascular endothelium and the subsequent migration of the inflammatory cells to sites of inflammation.
  • CD40L blockade retards the migration of leukocytes through endothelial cell barriers.
  • antibodies to CD40L interfere with the accumulation of inflammatory cells at the site of inflammation.
  • CD40/CD40L interactions have been implicated in diseases having an immune or autoimmune connection.
  • Animal models of immune-related disease in which the CD40L/CD40 pathway has been demonstrated to play a role in the pathology include, for example, murine models of systemic lupus erythematosis (Lupus or SLE; see, e.g., Kalled et al., 1998, J. Immunol. 160: 2158-2165), arthritis (collagen-induced arthritis, see, e.g., Durie et al., 1993, Science 261: 1328-1330), multiple sclerosis (experimental autoimmune encephalomyelitis, EAE; see, e.g., Howard et al., 1999, J. Clin . Invest.
  • autoimmune thyroiditis experimental autoimmune thyroiditis, EAT; see, e.g., Caryanniotis et al., 1997, Immunology 90: 421-426
  • colitis hapten-induced colitis; see, e.g., Stuber et al., 1996, J. Exp. Med. 183: 693-698
  • atherosclerosis and coronary artery disease see, e.g., Mach et al., 1998, Nature 394: 200-203
  • allograft rejection see, e.g., Parker et al., 1995, Proc. Natl. Acad. Sci. U.S.A.
  • CD40L antibody trials for treatment of human immune-related diseases include studies in patients with Lupus (see, e.g., Huang et al., 2002, Arthritis Rheum. 46: 1554-1562).
  • a phase I trial demonstrated that anti-CD40L humanized monoclonal antibody (IDEC-131) is safe and well tolerated by patients with Lupus (Davis et al., 2001, J. Rheumatol. 28: 95-101).
  • a phase II study with the IDEC-131 antibody showed improvement in clinical symptoms, but efficacy of the drug over placebo controls was not demonstrated (Kalunian et al., 2002, Arthritis Rheum. 46: 3251-3258).
  • U.S. Patent Nos. 5,474,771 (Lederman et al.) and 5,876,950 (Siadak et al.) disclose murine monoclonal antibodies specific for different epitopes of human gp39.
  • WO95/06666 Noelle & Foy discloses murine anti-gp39 antibodies.
  • U.S. Patent No. 6,328,964 (Noelle & Claassen) discloses methods for the treatment of multiple sclerosis using gp39-specific antibodies.
  • U.S. Patent No. 5,747,037 (Noelle et al.), and EP0721469B1 (Ledbetter et al.) and its U.S. counterpart U.S. 5,869,049 disclose anti-human monoclonal (mouse) antibodies specific for gp39.
  • U.S. Patent No. 5,876,718 (Noelle et al.) discloses methods of inducing T cell non-responsiveness to transplanted tissues and of treating graft-versus-host disease with anti-gp39 monoclonal (mouse) antibodies.
  • EP0742721B1 discloses methods of inhibiting a 'humoral immune response to a thymus-dependent antigen that use anti-gp39 monoclonal (mouse) antibodies.
  • U.S. Patent No. 6,375,950 describes methods for inducing T cell unresponsiveness to donor tissue or organs in a transplant recipient through use of anti-gp39 monoclonal (murine) antibodies.
  • EP1005372B1 (De Boer et al.) describes methods for the selective killing of autoreactive CD40L+ T cells using anti-CD40L monoclonal (mouse) antibody-toxin fusion proteins.
  • EP0831906B1 (Claassen et al.) describes methods for the treatment of T cell- mediated tissue destruction in autoimmune diseases such as multiple sclerosis using anti-gp39 monoclonal (mouse) antibodies.
  • Antibodies used in therapeutic approaches in the prior art have been divalent antibodies of murine origin.
  • a number of smaller antigen binding fragments of naturally occurring antibodies have been identified following protease digestion. These include, for example, the "Fab fragment” (V L -C L -C H 1-V H ), 'Tab' fragment” (a Fab with the heavy chain hinge region) and "F(ab')2 fragment” (a dimer of Fab' fragments joined by the heavy chain hinge region). Recombinant methods have been used to generate even smaller antigen-binding fragments, referred to as “single chain Fv” (variable fragment) or "scFv,” consisting of V L and V H joined by a synthetic peptide linker.
  • the antigen binding unit of a naturally-occurring antibody (e.g., in humans and most other mammals) is generally known to be comprised of a pair of V regions (V L /VH)
  • camelid species express a large proportion of fully functional, highly specific antibodies that are devoid of light chain sequences.
  • the camelid heavy chain antibodies are found as homodimers of a single heavy chain, dimerized via their constant regions.
  • variable domains of these camelid heavy chain antibodies are referred to as V H H domains and retain the ability, when isolated as fragments of the V H chain, to bind antigen with high specificity ((Hamers-Casterman et al., 1993, Nature 363: 446-448; Gahroudi et al., 1997, FEBS Lett. 414: 521-526).
  • Antigen binding single V H domains have also been identified from, for example, a library of murine V H genes amplified from genomic DNA from the spleens of immunized mice and expressed in E. coli (Ward et al., 1989, Nature 341 : 544-546). Ward et al.
  • dAbs the isolated single V H domains "dAbs," for “domain antibodies.”
  • the term “dAb” will refer herein to an antibody single variable domain (V H or V L ) polypeptide that specifically binds antigen.
  • V H or V L antibody single variable domain
  • a “dAb” binds antigen independently of other V domains; however, as the term is used herein, a “dAb” can be present in a homo- or heteromultimer with other V H or V L domains where the other domains are not required for antigen binding by the dAb, i.e., where the dAb binds antigen independently of the additional V H or V L domains.
  • Antibody single variable domains, for example, V H H are the smallest antigen- binding antibody unit known.
  • human antibodies are preferred, primarily because they are not as likely to provoke an immune response when administered to a patient.
  • isolated non-camelid V H domains tend to be relatively insoluble and are often poorly expressed.
  • Comparisons of camelid V H H with the V H domains of human antibodies reveals several key differences in the framework regions of the camelid VH H domain corresponding to the V H /V L interface of the human V H domains.
  • Trp 103- ⁇ Arg mutation improves the solubility of non-camelid V H domains.
  • Davies & Riechmann (1995, Biotechnology N-. Y. 13: 475-479) also report production of a phage-displayed repertoire of camelized human V H domains and selection of clones that bind hapten with affinities in the range of 100-400 nM, but clones selected for binding to protein antigen had weaker affinities.
  • the present invention provides single domain variable region polypeptides that are linked to polymers which provide increased stability and half- life.
  • polymer molecules e.g., polyethylene glycol; PEG
  • PEG polyethylene glycol
  • PEG modification of proteins has been shown to alter the in vivo circulating half-life, antigenicity, solubility, and resistance to proteolysis of the protein (Abuchowski et al., J Biol. Chem.
  • the invention relates to antibody polypeptides that monovalently bind
  • CD40L Because of the clear importance of CD40L in the production of antibodies, the CD40/CD40L interaction and pathways present important targets for the development of therapeutic approaches for the treatment of diseases and disorders that involve inappropriate or excessive antibody responses, such as autoimmune diseases.
  • Antibody polypeptides that are monovalent for binding of CD40L can inhibit CD40L activity, including binding and activation of CD40 on the B cell surface and downstream effects, while avoiding potential undesirable effects that can occur with antibodies capable of divalent or multivalent binding of CD40L.
  • Monovalent anti-CD40L antibody polypeptides can also be applied to any of a number of uses for which standard divalent antibodies are also used, e.g., in vivo imaging and diagnosis.
  • the antibody polypeptide consists of or comprises a single immunoglobulin variable domain that specifically binds and antagonizes the activity of CD40L, preferably without substantially agonizing CD40 and/or CD40L activity.
  • the antibody polypeptide is a human antibody polypeptide that monovalently binds CD40L, preferably without substantially agonizing CD40 and/or CD40L activity.
  • the invention provides an antibody polypeptide, preferably a human antibody polypeptide, that is monovalent for binding to CD40L (gp39).
  • the human antibody polypeptide dissociates from human
  • the K d for human CD40L can be 25 nM to 20 pM,
  • the antibody polypeptide inhibits the binding of CDAQh to CD40.
  • the binding of the antibody polypeptide to CD40L does not substantially agonize CD40 and/or CD40L activity.
  • the human antibody polypeptide inhibits the binding of CD40 to CD40L, and does not substantially agonize signaling by CD40.
  • the binding of the antibody polypeptide to CD40L does not substantially induce JNK phosphorylation in Jurkat T-cells.
  • the binding of the antibody polypeptide to CD40L does not substantially induce IFN- ⁇ secretion by Jurkat T-cells co-stimulated with anti-CD3 antibody.
  • the presence of the antibody polypeptide in a standard platelet aggregation assay does not result in aggregation of more than 25% over the aggregation observed in a negative control assay performed without the addition of antibody.
  • the human antibody polypeptide comprises a single immunoglobulin variable domain that binds CD40L.
  • the single immunoglobulin variable domain is a VH or a V L domain.
  • the antibody polypeptide is selected from the group consisting of a dAb, a FAb, an scFv, an Fv, or a disulfide-bonded Fv.
  • the human antibody polypeptide is PEG-linked.
  • the PEG is covalently linked to the human antibody polypeptide.
  • the PEG-linked human antibody polypeptide has a hydrodynamic size of at least 24 kD.
  • the PEG is linked to the antibody polypeptide at a cysteine or lysine residue.
  • the total PEG size is from 20 to 60 IcD, inclusive.
  • the PEG-linked human antibody pofypeptide has a hydrodynamic size of at least 200 IcD.
  • the antibody polypeptide has an increased in vivo half-life relative to the same antibody polypeptide composition lacking polyethylene glycol.
  • the t ⁇ -half life of the antibody polypeptide composition is increased by 10% or more. In another embodiment, the t ⁇ -half life of the antibody polypeptide composition is increased by 50% or more. In another embodiment, the t ⁇ -half life of the antibody polypeptide composition is increased by 2X or more. In another embodiment, the t ⁇ -half life of the antibody polypeptide composition is increased by 5X or more, e.g., 1OX, 15X, 2OX, 25X, 3OX, 4OX, or more. In another embodiment, the t ⁇ -half life of the antibody polypeptide composition is increased by 5OX or more.
  • the PEG-linked antibody pol y peptide has a t ⁇ half- life of 0.25 to 6 hours, inclusive. In another embodiment, the t ⁇ half-life is in the range of 30 minutes to 12 hours, inclusive. In another embodiment, the t ⁇ -half life of the antibody polypeptide composition is in the range of 1 to 6 hours.
  • the t ⁇ -half life of the antibody polypeptide composition is increased by 10% or more. In another embodiment, the t ⁇ -half life of the antibody polypeptide composition is increased by 50% or more. In another embodiment, the t ⁇ -half life of the antibody polypeptide composition is increased by 2X or more. In another embodiment, the t ⁇ -half life of the antibody polypeptide composition is increased by 5X or more, e.g., 10X, 15X, 20X, 25X, 30X, 4OX, .or more. In another embodiment, the t ⁇ -half life of the antibody polypeptide composition is increased by 5OX or more.
  • the antibody polypeptide composition has a t ⁇ half- life of 1 to 170 hours, inclusive. In another embodiment, the t ⁇ -half life is in the range of 12 to 48 hours, inclusive. In another embodiment, the t ⁇ -half life is in the range of 12 to 26 hours, inclusive.
  • the present invention provides a dAb containing composition
  • a dAb containing composition comprising a ligand according to the invention having an AUC value (area under the curve) in the range of 1 mg.min/ml or more.
  • the lower end of the range is 5, 10, 15, 20, 30, 100, 200 or
  • a ligand or composition according to the invention has an AUC in the range of up to 600 mg.min/ml. In one embodiment, the upper end of the range is 500, 400, 300, 200, 150, 100, 75 or 50 mg.min/ml.
  • a ligand according to the invention will have an AUC in the range selected from the group consisting of the following: 15 to 150 mg.min/ml, 15 to 100 mg.min/ml, 15 to 75 mg.min/ml, and 15 to 50 mg.min/ml.
  • the antibody polypeptides described herein can be linked to human serum albumin (HSA), which also has the effect of increasing the in vivo half life of the molecule.
  • HSA human serum albumin
  • the human serum albumin coding sequences can be obtained by PCR using primers derived from the cDNA sequence available at GenBank Accession No. NM000477. Such coding sequences can be fused to the coding sequence for a monovalent anti-CD40L antibody polypeptide as described herein, and the fusion can be expressed by one of skill in the art.
  • the t ⁇ -half life of the HSA-linked human antibody polypeptide composition is increased by 10% or more.
  • the t ⁇ -half life of the HSA-linked human antibody polypeptide composition is in the range of 0.25 hours to 6 hours.
  • the t ⁇ -half life of the HSA-linked human antibody polypeptide composition is increased by 10% or more.
  • the t ⁇ -half life of the HSA-linked human antibody polypeptide composition is in the range of 12 to 48 hours.
  • the human antibody polypeptide comprises an amino acid sequence selected from the group consisting of SEQ ID NOs 7-82 and 246-360.
  • the human antibody polypeptide inhibits binding of
  • the IC 50 can preferably be in the range of 20 pM to 1 ⁇ M, 20 pM to 900 nM, 20 pM to 800 nM, 20 pM to 700 nM,
  • nM 200 nM, 20 pM to 100 nM, or 20 pM to 50 nM. Further acceptable or preferred ranges include, for example, 50 pM to 1 ⁇ M, 100 pM to 500 nM, 125 pM to 250 nM,
  • the antibody polypeptide is fused to a second antibody polypeptide which binds a ligand other than CD40L.
  • the antibody polypeptide which binds a ligand other than CD40L binds a ligand selected from the group consisting of HSA, TNF ⁇ , IL-I, IL-2, IL-4, IL-6, IL- 8, IL-12, IL-18, IFN- ⁇ , CD2, CD4, CD8, CTLA4, LFAl, LFA3, VLA4, CD80 (B7- 1), CD28, CD86 (B7-2), and CTLA-4.
  • the human antibody polypeptide is free of an Fc domain.
  • the limits of an Fc domain are set out in Kabat et al. (1991, Sequences of Immunological Interest, 5 th ed. U.S. Dept. Health & Human Services, Washington, D. C; incorporated herein by reference).
  • an Fc domain consists of the CH2-CH3 regions, optionally including a hinge region linked to the CH2.
  • the human antibody polypeptide does not mediate platelet aggregation in a standard platelet aggregation assay.
  • the invention further encompasses a human antibody polypeptide which has an amino acid sequence at least 85% identical to a sequence selected from the group consisting of SEQ ID NOs 7-82 and 246-360, which antibody polypeptide specifically and monovalently binds CD40L.
  • the invention further encompasses an antigen-binding polypeptide, the polypeptide comprising a single immunoglobulin variable domain which specifically and monovalent])' binds CD40L.
  • the invention further encompasses a polypeptide comprising a moiety which specifically binds CD40L, which moiety consists of a single immunoglobulin variable domain.
  • the polypeptide consists of a human single immunoglobulin variable domain.
  • the polypeptide has a Kd for human CD40L in the range of 50 nM to 20 pM, inclusive, as determined by surface plasmon resonance.
  • the Kd for human CD40L can be 25 nM to 20 pM, 10 nM to 20 pM, 5 nm to 20 pM, 1 nM to 20 pM, 0.5 nM to 20 pM, 0.1 nM to 20 pM, 75 pM to 20 pM or even 50 pM to 20 pM.
  • polypeptide inhibits the binding of CD40L to CD40.
  • polypeptide inhibits the binding of CD40 to
  • the CD40L has an IC 50 in the range of 20 pM to 1.5 ⁇ M, inclusive.
  • the IC 50 can be in the range of 20 pM to 1 ⁇ M, 20 pM to 900 nM, 20 pM to 800 nM, 20 pM to 700 nM, 20 pM to 600 nM, 20 pM to 500 nM, 20 pM to 400 nM, 20 pM to 300 nM, 20 pM to 200 nM, 20 pM to 100 nM, or 20 pM to 50 nM.
  • Further acceptable or preferred ranges include, for example, 50 pM to 1 ⁇ M, 100 pM to 500 nM, 125 pM to 250 nM, 150 pM to 200 nM, 150 pM to 100 nM and 200 pM to 50 nM.
  • the binding of the polypeptide to CD40L does not substantially agonize CD40 and/or CD40L activity.
  • the binding of the polypeptide to CD40L does not substantially induce JNK phosphorylation in Jurkat T-cells. In another embodiment, the binding of the polypeptide to CD40L does not substantially induce IFN- ⁇ secretion by Jurkat T-cells co-stimulated with anti-CD3 antibody.
  • the presence of the antibody polypeptide in a standard platelet aggregation assay does not result in aggregation more than 25% over the aggregation observed in a negative control assay lacking antibody polypeptide.
  • the single immunoglobulin variable domain is a human single immunoglobulin variable domain.
  • the single immunoglobulin variable domain is a V H or a V L domain.
  • the polypeptide is PEG-linked. In one embodiment, the PEG is covalently linked. In one preferred embodiment, the PEG-linked antigen- binding polypeptide has a hydrodynamic size of at least 24 kD. In another preferred embodiment, the PEG is linked to the antigen-binding polypeptide at a cysteine or lysine residue. In another preferred embodiment, the total PEG size is from 20 to 60 kD, inclusive. In another preferred embodiment, the PEG-linked antigen-binding polypeptide has a hydrodynamic size of at least 200 kD.
  • the PEG-linked polypeptide has an increased in vivo half-life relative to the same polypeptide composition lacking linked polyethylene glycol.
  • the t ⁇ -half life of the polypeptide composition is increased by 10% or more.
  • the t ⁇ -half life of the polypeptide composition is increased by 50% or more.
  • the t ⁇ -half life of the polypeptide composition is increased by 2X or more.
  • the t ⁇ -half life of the polypeptide composition is increased by 5X or more, e.g., 10X, 15X, 2OX, 25X, 3OX, 4OX, or more.
  • the t ⁇ -half life of the polypeptide composition is increased by 5OX or more.
  • the PEG-linked antibody polypeptide has a toe half- life of 0.25 to 6 hours, inclusive. In another embodiment, the t ⁇ half-life is in the range of 30 minutes to 12 hours, inclusive. In another embodiment, the t ⁇ -half life of the polypeptide composition is in the range of 1 to 6 hours. In another embodiment, the t ⁇ -half life of the polypeptide composition is increased by 10% or more. In another embodiment, the t ⁇ -half life of the polypeptide composition is increased by 50% or more. In another embodiment, the t ⁇ -half life of the polypeptide composition is increased by 2X or more.
  • the t ⁇ -half life of the polypeptide composition is increased by 5X or more, e.g., 10X, 15X, 2OX, 25X, 3OX, 4OX, .or more. In another embodiment, the t ⁇ -half life of the polypeptide composition is increased by 5OX or more.
  • the antibody polypeptide composition has a t ⁇ half- life of 1 to 170 hours, inclusive. In another embodiment, the t ⁇ -half life is in the range of 12 to 48 hours, inclusive. In another embodiment, the t ⁇ -half life is in the range of 12 to 26 hours, inclusive.
  • the composition has an AUC value (area under the curve) in the range of 1 mg.min/ml or more.
  • the lower end of the range is 5, 10, 15, 20, 30, 100, 200 or 300 mg.min/ml.
  • a ligand or composition according to the invention has an AUC in the range of up to 600 mg.min/ml.
  • the upper end of the range is 500, 400, 300, 200, 150, 100, 75 or 50 mg.min/ml.
  • a ligand according to the invention will have an AUC in the range selected from the group consisting of the following: 15 to 150 mg.min/ml, 15 to 100 mg.min/ml, 15 to 75 mg.min/ml, and 15 to 50 mg.min/ml.
  • the antibody polypeptide is linked to human serum albumin (HSA).
  • HSA human serum albumin
  • the antibody polypeptide has an increased in vivo half-life relative to the same polypeptide composition lacking linked HSA.
  • the antibody polypeptide has a t ⁇ -half life that is increased by 10% or more relative to a molecule lacking linked HSA.
  • the t ⁇ -half life of the polypeptide composition is in the range of 0.25 minutes to 6 hours.
  • the t ⁇ -half life of the polypeptide composition is increased by 10% or more.
  • the t ⁇ -half life is in the range of 12 to 48 hours.
  • the antigen-binding polypeptide comprises an amino acid sequence selected from the group consisting of SEQ ID NOs 7-82 and 246-360.
  • the antigen-binding polypeptide is free of an Fc domain.
  • the invention encompasses an immunoglobulin variable domain polypeptide which has an amino acid sequence at least 85% identical to a sequence selected from the group consisting of SEQ ID NOs 7-82 and 246-360, which polypeptide specifically and monovalently binds CD40L.
  • the immunoglobulin variable domain polypeptide antagonizes the binding of CD40L to CD40.
  • the immunoglobulin variable domain polypeptide inhibits the binding of CD40 to CD40L and has an IC 50 in the range of 20 pM to 1.5 ⁇ M, inclusive.
  • the IC50 can be in the range of 20 pM to 1 ⁇ M, 20 pM to 900 nM, 20 pM to 800 nM, 20 pM to 700 nM, 20 pM to 600 nM, 20 pM to 500 nM, 20 pM to 400 nM, 20 pM to 300 nM, 20 pM to 200 nM, 20 pM to 100 nM, or 20 pM to 50 nM.
  • Further acceptable or preferred ranges include, for example, 50 pM to 1 ⁇ M, 100 pM to 500 nM, 125 pM to 250 nM, 150 pM to 200 nM, 150 pM to 100 nM and 200 pM to 50 nM.
  • the immunoglobulin variable domain polypeptide inhibits the interaction of CD40 with CD40L, but does not substantially agonize intracellular signaling by CD40.
  • the binding of the polypeptide to CD40L does not substantially induce JNK phosphorylation in Jurkat
  • the binding of the polypeptide to CD40L does not substantially induce IFN- ⁇ secretion by Jurkat T-cells co-stimulated with anti-CD3 antibody.
  • the binding of the antibody polypeptide to CD40L does not substantially induce platelet aggregation in a platelet aggregation assay.
  • the antigen-binding polypeptide further comprises a second antibody polypeptide which binds a ligand other than CD40L.
  • the second antibody polypeptide binds a ligand selected from the group consisting of HSA, TNF ⁇ , IL-I, IL-2, IL-4, IL-6, IL- ⁇ , IL-12, IL-18, IFN- ⁇ , CD2, CD4, COS, CTLA4, LFAl, LFA3 and VLA4.
  • the invention relates to an antibody polypeptide comprising an immunoglobulin variable domain which specifically and monovalently binds CD40L (e.g., an anti-CD40L dAb, FAb, an scFv, an Fv, or a disulfide-bonded Fv), and which 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 sequence of one or more of said framework regions collectively comprises 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.
  • CD40L e.g., an anti-CD40L dAb, FAb, an scFv, an Fv, or a disulfide-bonded Fv
  • 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 sequence of one or more
  • the amino acid sequences of FWl, FW2, FW3 and FW4 of the anti-CD40L variable domain or dAb 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 amino acid sequences of FWl, FW2 and FW3 of the anti-CD40L variable domain or dAb are the same as the amino acid sequences of corresponding framework regions encoded by human germline antibody gene segments.
  • the human germline antibody gene segment can be selected from the group consisting of DP47, DP45. DP48 and DPK9.
  • the invention further encompasses a method of antagonizing the binding of CD40 to CD40L in an individual, the method comprising administering a monovalent anti-CD40L antibody polypeptide as described Herein to the individual, wherein the polypeptide antagonizes the binding of CD40 to CD40L in the individual.
  • the invention further encompasses a method of antagonizing an activity of CD40 or CD40L in an individual, the method comprising administering a monovalent anti-CD40L antibody polypeptide as described herein to the individual, wherein the polypeptide antagonizes an activity of CD40 or CD40L or both.
  • the invention further encompasses a composition comprising an extended release formulation comprising a monovalent anti-CD40L antibody polypeptide, preferably, but not limited to, a polypeptide comprising a single immunoglobulin variable domain that binds CD40L.
  • the single immunoglobulin variable domain is a non-human mammalian single immunoglobulin variable domain.
  • the single immunoglobulin variable domain is a human single immunoglobulin variable domain.
  • the invention further encompasses a method of treating or preventing a disease or disorder mediated by CD40L in an individual in need of such treatment, the method comprising administering to the individual a therapeutically effective amount of a composition comprising a monovalent anti-CD40L antibody polypeptide, preferably a composition comprising a single human immunoglobulin variable domain that binds CD40L.
  • the disease or disorder is an autoimmune disease or disorder.
  • the invention further encompasses a method of treating or preventing a symptom of systemic lupus erythematosus (SLE) in an individual, the method comprising administering a monovalent anti-CD40L antibody polypeptide to said individual in an amount effective to treat or prevent a symptom of SLE.
  • SLE systemic lupus erythematosus
  • the invention further encompasses a method of reducing or alleviating a symptom of a disease such as systemic lupus erythematosis, multiple sclerosis, rheumatoid arthritis, allograft rejection, xenograft rejection, and Diabetes, including insulin-dependent Type I Diabetes.
  • the invention further encompasses an antibody polypeptide that is monovalent for binding to CD40L, wherein the antibody polypeptide comprises a universal framework.
  • the universal framework comprises a VH framework selected from the group consisting of DP47, DP45 and DP38, and/or the VL framework is DPK9.
  • the antibody polypeptide comprises a generic ligand binding site.
  • the generic ligand binding site binds a generic ligand selected from the group consisting of protein A, protein L and protein G.
  • the antibody polypeptide comprises a variable domain having 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 the framework regions collectively comprises up to 5 amino acid differences relative to the amino acid sequence of the corresponding framework region encoded by a human germline antibody gene segment.
  • the antibody polypeptide comprises a variable domain, wherein the amino acid sequences of FWl, FW2, FW3 and FW4 are the same as the amino acid sequences of corresponding framework regions encoded by a human germline antibody gene segment, or the antibody 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 the human germline antibody gene segment.
  • the antibody polypeptide comprises an antibody variable domain comprising FWl, FW2 and FW3 regions, and the amino acid sequence of said FWl, FW2 and FW3 are the same as the amino acid sequences of corresponding framework regions encoded by human germline antibody gene segments.
  • the human germline antibody gene segment is selected from the group consisting of DP47, DP45, DP48 and DPK9.
  • the invention includes an antibody single variable domain polypeptide that binds to CD40L, wherein the polypeptide has an amino acid sequence that is identical to the amino acid sequence of DOM8-24, or differs from the amino acid sequence of DOM8-24 at no more than 25 amino acid positions and has a sequence that is at least 80% homologous to the sequence of DOM8-24.
  • the antibody single variable domain polypeptide differs form the amino acid sequence of DOM8- 24 at 25 or fewer amino acid positions, 20 or fewer amino acid positions, 15 or fewer amino acid positions, 10 or fewer amino acid positions, 5 or fewer amino acid positions, 2 or fewer amino acid positions, or as few as one amino acid position.
  • the antibody single variable domain polypeptide is at least 80% homologous to the sequence of DOM8-24, for example, at least 85% homologous, at least 90% homologous, at least 95% homologous, and up to and including 96%, 97%, 98%, or 99% homologous.
  • the invention includes an antibody single variable domain polypeptide that binds to CD40L, wherein the polypeptide has an amino acid sequence that is identical to the amino acid sequence of DOM8-24, or differs from the amino acid sequence of DOM8-24 at no more than 25 amino acid positions and has a CDRl sequence that is at least 50% homologous to the CDRl sequence of DOM8-24, or has a CDR2 sequence that is at least 50% homologous to the CDR2 sequence of DOM8-24, or has a CDR3 sequence that is at least 50% homologous to the CDR3 sequence of DOM8- 24.
  • the invention also includes an antibody single variable domain polypeptide that binds CD40L, wherein the dAb has an amino acid sequence that is identical to the amino acid sequence of DOM8-24, or differs from the amino acid sequence of DOM8-24 at no more than 25 amino acid positions and has a CDRl sequence that is at least 50% homologous to the CDRl sequence of DOM8-24 and has a CDR2 sequence that is at least 50% homologous to the CDR2 sequence of DOM8-24 .
  • the invention also includes an antibody single variable domain polypeptide that binds CD40L, wherein the dAb has an amino acid sequence that is identical to the amino acid sequence of DOM8-24, or differs from the amino acid sequence of DOM8-24 at no more than 25 amino acid positions and has a CDR2 sequence that is at least 50% homologous to the CDR2 sequence of DOM8-24 and has a CDR3 sequence that is at least 50% homologous to the CDR3 sequence of DOM8-24 .
  • the invention also includes an antibody single variable domain polypeptide that binds CD40L, wherein the dAb has an amino acid sequence that is identical to the amino acid sequence of DOM8-24, or differs from the amino acid sequence of DOM8-24 at no more than 25 amino acid positions and has a CDRl sequence that is at least 50% homologous to the CDRl sequence of DOM8-24 and has a CDR3 sequence that is at least 50% homologous to the CDR3 sequence of DOM8-24 .
  • the invention also includes an antibody single variable domain polypeptide that binds CD40L, wherein the dAb has an amino acid sequence that is identical to the amino acid sequence of DOM8-24, or differs from the amino acid sequence of DOM8-24 at no more than 25 amino acid positions and has a CDRl sequence that is at least 50% homologous to the CDRl sequence of DOM8-24 and has a CDR2 sequence that is at least 50% homologous to the CDR2 sequence of DOM8-24 and has a CDR3 sequence that is at least 50% homologous to the CDR3 sequence of DOM8-24.
  • the antibody single variable domain polypeptide that binds to CD40L if not identical in sequence to that of DOM8-24, differs form the amino acid sequence of DOM8-24 at 25 or fewer amino acid positions, 20 or fewer amino acid positions, 15 or fewer amino acid positions, 10 or fewer amino acid positions, 5 or fewer amino acid positions, 2 or fewer amino acid positions, or as few as one amino acid position.
  • the invention also includes a CDAQiL antagonist having a CDRl sequence that is at least 50% homologous to the CDRl sequence of DOM8-24.
  • the invention also includes a CD40L antagonist having a CDR2 sequence that is at least 50% homologous to the CDR2 sequence of DOM8-24.
  • the invention also includes a CD40L antagonist having a CDR3 sequence that is at least 50% homologous to the CDR3 sequence of DOM8-24.
  • the invention also includes a CD40L antagonist having a CDRl sequence that is at least 50% homologous to the CDRl sequence of DOM8-24 and a CDR2 sequence that is at least 50% homologous to the CDR2 sequence of DOM8-24.
  • the invention also includes a CD40L antagonist having a CDR2 sequence that is at least 50% homologous to the CDR2 sequence of DOM8-24 and a CDR3 sequence that is at least 50% homologous to the CDR3 sequence of DOM8-24.
  • the invention also includes a CD40L antagonist having a CDRl sequence that is at least 50% homologous to the CDRl sequence of DOM8-24 and a CDR3 sequence that is at least 50% homologous to the CDR3 sequence of DOM8-24.
  • the invention also includes a CD40L antagonist having a CDRl sequence that is at least 50% homologous to the CDRl sequence of DOM8-24 and a CDR2 sequence that is at least 50% homologous to the CDR2 sequence of DOM8-24 and a
  • the CD40L antagonist inhibits the binding of CD40 to
  • CD40L and/or inhibits an activity of CD40 and/or CD40L, and/or results in no more than 25% platelet aggregation in a platelet aggregation assay.
  • the antagonist results in platelet aggregation of 25% or less, 20% or less, 15% or less,
  • the invention also includes a dual specific ligand comprising a first immunoglobulin single variable domain having a binding specificity to a first antigen and a second single variable domain having a binding activity to a second antigen, wherein the first antigen is CD40L and binding of the second single variable domain to the second antigen acts to increase the half-life of the ligand in vivo.
  • the dual specific ligand is a four chain IgG immunoglobulin.
  • the four chain IgG comprises two dual specific ligands, said dual specific ligands being different in their variable domains.
  • the invention also includes a dual specific ligand comprising an anti-human CD40L dAb and an anti-SA dAb.
  • the dAbs are Camelid V HH domains.
  • first and second immunoglobulin variable domains are heavy chain variable domains; or (ii) the first and the second immunoglobulin variable domains are light chain variable domains.
  • the ligand is provided as an IgG immunoglobulin comprising four heavy chain single variable domains or four light chain single variable domains.
  • the heavy chain can comprise Camelid VHH domains.
  • the first and second domains bind independently, such that the dual specific ligand may simultaneously bind both the first and second antigens.
  • the first single variable domain has a dissociation constant (Kd) of 1x10 '8 M or less for human CD40L, and a K Off rate constant of IxIO "3 s "1 or less, as determined by surface plasmon resonance.
  • the second single variable domain is specific for serum albumin (SA) and has a dissociation constant (K d ) of InM to 500 ⁇ m for SA, as determined by surface plasmon resonance.
  • the second domain binds SA in a standard ligand binding assay with an IC50 of InM to 500 ⁇ M.
  • the second single variable domain may be specific for SA, and comprise the amino acid sequence of MSA-] 6 or a sequence that is at least 80% homologous thereto.
  • the second single variable domain may be specific for SA, and comprise the amino acid sequence of MSA-26 or a sequence that is at last 80% homologous thereto.
  • the anti-CD40L variable domain or dAb comprises a universal framework.
  • the anti-CD40L variable domain or dAb may also comprise a V H framework selected from the group consisting of DP47, DP45 and DP38; or a V L framework which is DPK9.
  • the dual specific ligand or dAb can comprise a binding site for a generic ligand.
  • the generic ligand binding site is selected from the group consisting of protein A, protein L and protein G binding site.
  • the anti-CD40L variable domain or dAb 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 sequence of one or more of said framework regions collectively comprises 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.
  • the amino acid sequences of FWl, FW2, FW3 and FW4 of the anti-CD40L variable domain or dAb 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 amino acid sequences of said FWl, FW2 and FW3 of the anti-CD40L variable domain or 'dAb are the same as the amino acid sequences of corresponding framework regions encoded by human germline antibody gene segments.
  • the human germline antibody gene segments are preferably selected from the group consisting of DP47, DP45, DP48 and DPK9.
  • the invention also includes a method for producing a dual specific ligand as described herein, comprising a first immunoglobulin single variable domain having a binding specificity for CD40L and a second single immunoglobulin single variable domain having a binding specificity for a protein which increases the half-life of the ligand in vivo, the method comprising the steps of:selecting a first variable domain by its ability to bind CD40L; selecting a second variable domain by its ability to bind to said protein; combining the variable domains; and selecting the ligand by its ability to bind to CD40L and said protein.
  • the first variable domain is selected for binding to CD40L in absence of a complementary variable domain.
  • the invention also includes nucleic acid encoding a dual specific ligand described herein.
  • the nucleic acid may comprise the nucleic acid sequence of MS A- 16 or a sequence that is at least 80% homologous thereto, or alternatively may comprise, the nucleic acid sequence of MSA-26 or a sequence that is at least 70% homologous thereto.
  • the nucleic acid may be incorporated into a vector, which may be incorporated into a host cell.
  • the invention also includes a pharmaceutical composition
  • a pharmaceutical composition comprising a dual specific ligand as described herein and a pharmaceutically acceptable excipient, carrier or diluent.
  • the invention also includes a dAb monomer specific for CD40L, which monomer has a dissociation constant (Ka) of 1x10 "8 M or less for human CD40L, and a K off rate constant of 1x10 "3 s "1 or less, as determined by surface plasmon resonance.
  • the dAb monomer specific for CD40L has a dissociation constant (Kd) of 1x10 "7 M or less, as determined by surface plasmon resonance.
  • the dAb monomer has binding specificity to CD40L with a dissociation constant (K d ) of IxIO "8 M or less, as determined by surface plasmon resonance.
  • the dAb monomer has binding specificity to CD40L with a dissociation constant (K d ) of 5OnM to 2OpM, as determined by surface plasmon resonance.
  • the monomer inhibits binding of CD40 to CD40L with an IC 50 of 5OnM or less.
  • the dAb monomer has binding specificity to CD40L with a K O ff rate constant of 1x10 "3 s "1 or less, 1x10 "4 s “1 or less, 1x10 "5 s “1 or less, or 1x10 "6 s “1 or less,as determined by surface plasmon resonance.
  • the dAb monomer neutralizes CD40L in a standard assay with an ND50 of 5OnM or less.
  • In invention also includes a dual specific ligand comprising first and second heavy chain single variable domains, or first and second light chain single variable domains, wherein the first variable domain is an anti-CD40L dAb monomer.
  • the second variable domain has binding specificity for an antigen other than CD40L.
  • the second variable domain has binding specificity for an antigen selected from the group consisting of EPO receptor, ApoE, Apo-SAA, BDNF 5 Cardiotrophin-1, EGF, EGF receptor, ENa-78, Eotaxin, Eotaxin-2, Exodus-2, EpoR, FGF-acidic, FGF-basic, fibroblast growth factor- 10, FLT3 ligand, Fractalkine (CX3C), GDNF, G-CSF, GM-CSF, GF-bl, insulin, IFN-g, IGF-I, IGF-II, IL-Ia, U-Ib, IL-2, 11-3, IL-4, IL-5, IL-6, IL-7, IL-8 (72 a.a.), IL-8 (77 a.a.), IL-9, IL-10, IL-11, IL- 12, IL-13, IL-15, EL-16, IL-17, IL-18 (IGIF), Inhibin a
  • an antigen
  • Mullerian inhibitory substance monocyte colony inhibitor' factor, monocyte attractant protein, M-CSF, MDC (67 a.a.), MDC (69 a.a.), MCP-I (MCAF), MCP-2, MCP-3, MCP-4, MDC (67 a.a.), MIG, MIP-Ia, MIP-Ib 5 MIP-Sa, MIP-3b, MIP-4, myeloid progenitor inhibitor factor-1 (MPIF-I), NAP -2, Neurturin, Nerve growth factor, b-NGF, NT-3, NT-4, Oncostatin M, PDGF-AA, PDGF-AB, PDGF-BB, PF-4, RANTES, SDFIa, DFGIb SCF, SCGF, stem cell factor, (SCF), TARC, TGF-a, TGF-b, TGF-b2, ⁇ TGF-b3, tumour necrosis factor (TNF), TNF-a s TNF-b, TNF receptor I
  • the invention also includes an antibody polypeptide that antagonizes or inhibits the binding of DOM8-24 to CD40L, or an antibody polypeptide that binds to the same epitope of CD40L bound by DOM8-24.
  • the invention also includes a dual specific ligand comprising a . first immunoglobulin single variable domain having a binding specificity to a first antigen and a second single variable domain having a binding activity to a second antigen, wherein the first antigen is CD40L and the second single variable domain is an Antigen Presenting Cell surface antigen or a T cell surface antigen.
  • the Antigen Presenting Cell surface antigen can be selected from one of the group consisting of dendritic cell surface antigens, activated macrophage surface antigens, activated B cell surface antigens, co-stimulatory signal pathway surface antigens, and MHC.
  • the MHC is class II, and the class II can be alpha or beta.
  • the Antigen Presenting Cell surface antigen may be selected from the group consisting of CD28, Inducible costimulatory molecule (ICOS), CD27, CD30, OX40, CD45, CD69, CD3, CD70, Inducible costimulatory molecule ligand (ICOSL) 5 OX40L, CD80, CD86, HVEM (Herpes Virus Entry Mediator), and LIGHT, but is preferably one of CD28, Inducible costimulatory molecule (ICOS), CD27, CD30, OX40, CD45, CD69, or CD3.
  • the surface antigen is preferably a B7 gene surface antigen such as B7-2 or B7-1.
  • the term "human” when applied to an antibody polypeptide or to an immunoglobulin variable domain means that the polypeptide has a sequence derived from a human immunoglobulin.
  • a sequence is "derived from” a human immunoglobulin coding sequence when the sequence is either: a) isolated from a human individual or from cells or a cell line from a human individual; b) isolated from a library of cloned human antibody gene sequences (or a library of human antibody V domain sequences); or c) when a cloned human antibody gene sequence (or a cloned human V region sequence (including, e.g., a germline V gene segment)) was used to generate one or more diversified sequences that were then selected for binding to a desired target antigen.
  • human as applied herein to an antibody polypeptide or to an immunoglobulin variable domain does not encompass an immunoglobulin from another species, e.g., mouse, camel, etc., that has been "humanized” through grafting of human constant region sequences onto an antibody polypeptide (i.e., replacing non-human constant regions with human constant regions) or through grafting of human V region framework sequences onto an immunoglobulin variable domain from a non-human mammal (i.e., replacing non-human framework regions of a V domain with human framework regions).
  • a human variable domain has at least 85% amino acid similarity (including, for example, 87%, 90%, 93%, 95%, 97%, 99% or higher similarity) to a naturally-occurring human immunoglobulin variable domain sequence.
  • domain refers to 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 immunoglobulin variable domain is meant a folded polypeptide domain which comprises a sequence characteristic of immunoglobulin variable domains and which specifically binds an antigen (e.g., dissociation constant of 500 nM or less).
  • a "single immunoglobulin variable domain” therefore includes complete antibody variable domains as well as 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 a dissociation constant of 500 nM or less (e.g., 450 nM or less, 400 nM or less, 350 nM or less, 300 nM or less, 250 nM or less, 200 nM or less, 150 nM or less, 100 nM or less) and the target antigen specificity of the full- length domain.
  • 500 nM or less e.g., 450 nM or less, 400 nM or less, 350 nM or less, 300 nM or less, 250 nM or less, 200 nM or less, 150 nM or less, 100 nM or less
  • An antibody single variable domain polypeptide refers to a mammalian single immunoglobulin variable domain polypeptide, preferably human, but also includes rodent (for example, as disclosed in WO00/29004, the contents of which are incorporated herein in their entirety) or camelid V HH dAbs.
  • Camelid dAbs are antibody 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: VHH- VHH molecules are about 10x smaller than IgG molecules, and as single polypeptides, they are very stable, resisting extreme pH and temperature conditions. Moreover, camelid antibody single variable domain polypeptides are resistant to the action of proteases. Camelid antibodies are described in, for example, U.S. Pat. Nos. 5,759,808; 5,800,988; 5,840,526; 5,874,541; 6,005,079; and 6,015,695, the contents of each of which are incorporated herein in their entirety.
  • Camelid V HH antibody single variable domain polypeptides useful according to the invention include a class of camelid antibody single variable domain polypeptides having human-like sequences, wherein the class is characterized in that the V HH domains carry an amino acid from the group consisting of glycine, alanine, valine, leucine, isoleucine, proline, phenylalanine, tyrosine, tryptophan, methionine, serine, threonine, asparagine, or glutamine at position 45, such as for example L45, and further comprise a tryptophan at position 103 according to the Kabat numbering.
  • Humanized camelid V HH polypeptides are taught for example in WO04/041862, the teachings of which are incorporated herein in their entirety. It will be understood by one of skill in the art that naturally occurring camelid antibody single variable domain polypeptides may be modified according to the teachings of WO04/041862 (e.g., amino acid substitutions at positions 45 and 103) to generate humanized camelid V HH polypeptides. Also included in the present invention are antibody single variable domain polypeptides which are nurse shark V HH - Nurse shark dAbs are antibody single variable domain polypeptides derived from the nurse shark, that comprise heavy chain antibodies naturally devoid of light chain: V HH - Nurse Shark V HH dAbs are described, for example, in Greenberg et al. (Nature 374 pp 168- 173 1995) and U.S. 20050043519.
  • single immunoglobulin variable domain polypeptide encompasses not only an isolated single immunoglobulin variable domain polypeptide, but also larger polypeptides that comprise a monomer of a single immunoglobulin variable domain polypeptide sequence.
  • dAb is equivalent to a "single immunoglobulin variable domain polypeptide" as the term is used herein.
  • the binding to antigen e.g., CD40L
  • the single immunoglobulin V domain without a requirement for a complementary V domain.
  • the terms “antibody single variable domain polypeptide”, “antibody single variable domain”, “single antibody variable domain”, and “single immunoglobulin variable domain” are understood to be equivalent.
  • sequence characteristic of immunoglobulin variable domains refers to an amino acid sequence that is homologous, over 20 or more, 25 or more, 30 or more, 35 or more, 40 or more, 45 or more, or even 50 or more contiguous amino acids, to a sequence comprised by an immunoglobulin variable domain sequence. Sequences similar or homologous (e.g., at least about 70% sequence identity) to the sequences disclosed herein are also part of the invention. In some embodiments, the sequence identity at the amino acid level can be about 80%, 85%,
  • sequence identity can be about 70%, 75%, 80%, 85%, 90%, 91%, 92%,
  • 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 “similarity” refers to the degree with which two nucleotide or amino acid sequences structurally resemble each other.
  • sequence “similarity” is a measure of the degree to which amino acid sequences share similar amino acid residues at corresponding positions in an alignment of the sequences. Amino acids are similar to each other where their side chains are similar. Specifically, “similarity” encompasses amino acids that are conservative substitutes for each other. A “conservative” substitution is any substitution that has a positive score in the blosum62 substitution matrix (Hentikoff and Hentikoff, 1992, Proc. Natl. Acad. Sci. USA 89: 10915-10919).
  • sequence A is n% similar to sequence B
  • sequence B n% of the positions of an optimal global alignment between sequences A and B consists of identical amino acids or conservative substitutions.
  • Optimal global alignments can be performed using the following parameters in the Needleman-Wunsch alignment algorithm:
  • LG is the length of the gap.
  • Typical conservative substitutions are among Met, VaI, Leu and lie; among Ser and Thr; among the residues Asp, GIu and Asn; among the residues GIn, Lys and Arg; or aromatic residues Phe and Tyr.
  • two sequences are “homologous” or “similar” to each other where they have at least 70%, 80%, or 85% sequence similarity to each other, including, e.g., 90%, 95%, 97%, 99% or even 100% sequence similarity, when aligned using either the Needleman-Wunsch algorithm or the "BLAST 2 sequences" algorithm described by Tatusova & Madden, 1999, FEMS Microbiol Lett. 174:247-
  • Blosum 62 matrix is the default matrix.
  • the te ⁇ ns "inhibit,” “inhibits” and “inhibited” refer to a decrease in a given measurable activity (e.g., binding activity) by at least 10% relative to a reference. Where inhibition is desired, such inhibition is preferably at least 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or more, up to and including 100%, i.e., complete inhibition or absence of the given activity.
  • a given measurable activity e.g., binding activity
  • the term “substantially inhibits” refers to a decrease in a given measurable activity (e.g., the binding of CD40L to CD40) by at least 50% relative to a reference.
  • substantially inhibits refers to a decrease in a given measurable activity of at least 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% and up to and including 100% relative to a reference.
  • inhibits the binding with reference to the binding of an antibody polypeptide binding to CD40L, or CD40 binding to CD40L, refers to a decrease in binding by at least 10% relative to a reference.
  • Inhibits the binding preferably refers to a decrease in binding of at least 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or more, up to and including 100%.
  • the terms “activate,” “activates” and “activated” refer to an increase in a given measurable activity by at least 5% relative to a reference, for example, at least 10%, 25%, 50%, 75% or even 100%.
  • the term “antagonist” refers to an agent that inhibits at least one activity mediated by CD40L.
  • CD40 inhibits the binding of CD40 to CD40L, and/or results in no more than 25% platelet activation and/or aggregation in a platelet aggregation assay or platelet activation assay as described herein, and preferably results in 25% or less platelet activation and/or aggregation, 20% or less, 15% or less, 10% or less, 5% or less, and as little as zero platelet activation and/or aggregation.
  • An activity is "antagonized” if the activity (i.e., CD40L mediated activity, binding of CD40 or CD40L, or platelet activation and/or aggregation) is reduced by at least 10%, and preferably at least 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 97% or even 100% (i.e., no activity) in the presence, relative to the absence of an antagonist.
  • An antagonist as the term is used herein preferably comprises a single immunoglobulin variable domain that binds monovalently to CD40L.
  • agonist refers to an agent that activates at least one activity mediated by CD40L, either alone or when combined with another co- stimulus, relative to a reference.
  • An activity is “agonized” if the activity is increased by at least 10%, e.g., 50%, in the presence, relative to the absence of an agonist.
  • epitope refers to a unit of structure conventionally bound by an immunoglobulin VVV 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 immunoglobulin variable domain, an epitope represents the unit of structure bound by a single variable domain in isolation. That is, the binding site is provided by one, single immunoglobulin variable domain.
  • extended release or the equivalent terms “controlled release” or “slow release” refer to drug formulations that release active drug, such as a polypeptide drug, over a period of time following administration to an individual. Extended release of polypeptide drugs, which can occur over a range of desired times, e.g., minutes, hours, days, weeks or longer, depending upon the drug formulation, is in contrast to standard formulations in which substantially the entire dosage unit is available for immediate absorption or immediate distribution via the bloodstream.
  • Preferred extended release formulations result in a level of circulating drug from a single administration that is sustained, for example, for 8 hours or more, 12 hours or more, 24 hours or more, 36 hours or more, 48 hours or more, 60 hours or more, 72 hours or more 84 hours or more, 96 hours or more, or even, for example, for 1 week or 2 weeks or more, for example, 1 month or more,
  • a "CD40L activity” is an activity involving or resulting from the binding of CD40L to CD40, and includes, but is not limited to binding to CD40 (assayed, for example, according to the method described in Example 6), activation of Jun-N-terminal Kinase (JNK), the induction of T cells to produce and secrete cytokines including, for example, IL-IO, IFN- ⁇ and TNF- ⁇ , and the mediation of platelet activation and/or aggregation. Assays for these activities are provided herein below.
  • the term "does not substantially agonize” means that a given agent, e.g., an anti-CD40L antibody polypeptide, does not activate one or more of the CD40L activities including Jun-N-terminal kinase activation (phosphorylation) in Jurkat T cells and induction of IFN- ⁇ production or secretion in anti-CD3-stimulated Jurkat T cells, as the term “activate” is defined herein.
  • does not substantially agonize means that the agent does not activate more than 20% of the activity which is activated by CD40 binding to CD40L, preferably, the agent does not activate more than 10%, 8%, 5%, 3%, or more than 2% or less, including zero activation, of the activity which is activated by CD40 binding to CD40L.
  • antibody polypeptide refers to a polypeptide which either is an antibody or is a part of an antibody, modified or unmodified, which retains the ability to specifically bind antigen.
  • antibody polypeptide includes an antigen-binding heavy chain, light chain, heavy chain-light chain dimer, Fab fragment, F(ab')2 fragment, dAb, or an Fv fragment, including a single chain Fv (scFv).
  • scFv single chain Fv
  • the term "monovalent” means that a given antibody polypeptide or single immunoglobulin variable domain polypeptide can bind only a single molecule of its target.
  • Naturally-occurring antibodies are generally divalent, in that they have two functional antigen-binding arms, each comprising a VH and a VL domain. Where steric hindrance is not an issue, a divalent antibody can bind two separate molecules of the same antigen. In contrast, a "monovalent” antibody has the capacity to bind only one such antigen molecule.
  • a "monovalent" antibody can also comprise more than one antigen binding site, e.g., two antigen bindi ⁇ g sites, but the binding sites must be for different antigens, such that the antibody can only bind one molecule of CD40L at a time.
  • the antigen- binding domain of a monovalent antibody can comprise a V H and a V L domain, but preferably comprises only a single immunoglobulin variable domain, i.e., a V H or a V L domain, that has the capacity to bind CD40L without the need for a corresponding V L or V H domain, respectively.
  • a monovalent antibody lacks the capacity to cross link molecules of a single antigen.
  • standard platelet aggregation assay means the assay described in the section herein below, entitled “Platelet Aggregation Assaj'.”
  • linked refers to the attachment of a polymer moiety, such as PEG to an amino acid residue of an antibody polypeptide. Attachment of a PEG polymer to an amino acid residue of an antibody polypeptide, e.g., an anti-CD40L dAb, is referred to as "PEGylation” and may be achieved using several PEG attachment moieties including, but not limited to N-hydroxylsuccinimide (NHS) active ester, succinimidyl propionate (SPA), maleimide (MAL), vinyl sulfone (VS), or thiol.
  • NHS N-hydroxylsuccinimide
  • SPA succinimidyl propionate
  • MAL maleimide
  • VS vinyl sulfone
  • a PEG polymer, or other polymer can be linked to an antibody polypeptide at either a predetermined position, or may be randomly linked to the an antibody polypeptide molecule. It is preferred, however, that the PEG polymer be linked to an antibody polypeptide at a predetermined position.
  • a PEG polymer may be linked to any residue in the an antibody polypeptide, however, it is preferable that the polymer is linked to either a lysine or cysteine, which is either naturally occurring in the antibody polypeptide, or which has been engineered into the antibody polypeptide, for example, by mutagenesis of a naturally occurring residue in the antibody polypeptide to either a cysteine or lysine.
  • PEG-linkage can also be mediated through a peptide linker attached to an antibody polypeptide. That is, the PEG moiety can be attached to a peptide linker fused to an antibody polypeptide, where the linker provides the site, e.g., a free cysteine or lysine, for PEG attachment.
  • linker provides the site, e.g., a free cysteine or lysine, for PEG attachment.
  • "linked” can also refer to the association of two or more antibody polypeptides, e.g., dAb monomers, to form a dimer, trimer, tetramer, or other multimer.
  • Antibody polypeptide monomers can be linked to form a multimer by several methods known in the art, including, but not limited to, expression of the antibody polypeptide monomers as a fusion protein, linkage of two or more monomers via a peptide linker between monomers, or by chemically joining monomers after translation, either to each other directly, or through a linker by disulfide bonds, or by linkage to a di-, tri- or multivalent linking moiety (e.g., a multi-arm PEG).
  • dAb multimers are specifically contemplated herein, e.g., in the context of dual- or multi-specific antibody polypeptide constructs, it is emphasized that for any given antibody polypeptide construct, the construct should only be able to bind one molecule of CD40L, i.e., the constructs can have only one CD40L-binding element, and cannot cross link CD40L.
  • polymer refers to a macromolecule made up of repeating monomeric units, and can refer to a synthetic or naturally occurring polymer such as an optionally substituted straight or branched chain polyalkylene, polyalkenylene, or polyoxyalkylene polymer or a branched or unbranched polysaccharide.
  • PEG polyethylene glycol
  • PEG polymer refers to polyethylene glycol, and more specifically can refer to a derivitized form of PEG, including, but not limited to N-hydroxylsuccinimide (NHS) active esters of PEG such as succinimidyl propionate, benzotriazole active esters, PEG derivatized with maleimide, vinyl sulfones, or thiol groups.
  • NHS N-hydroxylsuccinimide
  • Particular PEG formulations can include PEG-O-CH 2 CH 2 CH 2 -CO 2 -NHS; PEG-O-CH 2 -NHS; PEG-O-CH 2 CH 2 -CO 2 -NHS; PEG-S-CH 2 CH 2 -CO-NHS; PEG- O 2 CNH-CH(R)-CO 2 -NHS; PEG-NHCO-CH 2 CH 2 -CO-NHS; and PEG-O-CH 2 -CO 2 - NHS; where R is (CH?) 4 )NHCO 2 (mPEG).
  • PEG polymers useful in the invention may be linear molecules, or may be branched wherein multiple PEG moieties are present in a single polymer. Some particularly preferred PEG conformations that are useful in the invention include, but are not limited to the following:
  • a "sulfhydryl-selective reagent” is a reagent which is useful for the attachment of a PEG polymer to a thiol-containing amino acid. Thiol groups on the amino acid residue cysteine are particularly useful for interaction with a sulfhydryl-selective reagent. Sulfhydryl-selective reagents which are useful for such attachment include, but are not limited to maleimide, vinyl sulfone, and thiol.
  • sulfhydryl-selective reagents for coupling to cysteine residues is known in the art and may be adapted as needed according to the present invention (See Eg., Zalipsky, 1995, Bioconjug. Chem. 6:150; Greenwald et al., 2000, Crit. Rev. Ther. Drug Carrier Syst. 17:101; Herman et al., 1994, Macromol. Chem. Phys. 195:203).
  • PEG or another agent e.g., HSA
  • PEG or another agent e.g., HSA
  • "retains activity” refers to a level of activity of a PEG-linked antibody polypeptide which is at least 10% of the level of activity of a non-PEG-linked antibody polypeptide, preferably at least 20%, 30%, 40%, 50%, 60%, 70%, 80% and up to 90%, preferably up to 95%, 98%, and up to 100% of the activity of a non-PEG-linked antibody polypeptide comprising the same antigen- binding domain or domains.
  • the activity of a PEG-linked antibody polypeptide compared to a non-PEG linked antibody variable domain should be determined on an antibody polypeptide molar basis; that is equivalent numbers of moles of each of the PEG-linked and non-PEG-linked antibody polypeptides should be used in each trial.
  • determining whether a particular PEG-linked antibody polypeptide "retains activity" it is preferred that the activity of a PEG-linked antibody polypeptide be compared with the activity of the same antibody polypeptide in the absence of PEG.
  • the term "in vivo half-life” refers to the time taken for the serum concentration of a ligand (e.g., a single immunoglobulin variable domain) 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.
  • a ligand e.g., a single immunoglobulin variable domain
  • the anti CD40L antibody polypeptides or single immunoglobulin variable domain polypeptides described herein can be stabilized in vivo and their half-life increased by binding to molecules, such as PEG, which resist degradation and/or clearance or sequestration.
  • the half-life of an antibody polypeptide is increased if its functional activity persists, in vivo, for a longer period than a similar antibody polypeptide which is not linked to a PEG polymer.
  • the half life of a PEGylated antibody polypeptide is increased by 10%, 20%, 30%, 40%, 50% or more relative to a non-PEGylated antibody polypeptide. Increases in the range of 2x, 3x, 4x, 5x, 10x, 2Ox, 3Ox, 40x, 5Ox or more of the half life are possible.
  • a PEG-linked antibody single variable domain has a half- life of between 0.25 and 170 hours, preferably between 1 and 100 hours, more preferably between 30 and 100 hours, and still more preferably between 50 and 100 hours, and up to 170, 180, 190, and 200 hours or more.
  • resistant to degradation or “resists degradation” with respect to a PEG or other polymer-linked antibody polypeptide monomer or multimer means that the PEG- or other polymer-linked antibody polypeptide monomer or multimer is degraded by no more than 10% when exposed to pepsin at pH 2.0 for 30 minutes and preferably not degraded at all.
  • hydrodynamic size refers to the apparent size of a molecule (e.g., a protein molecule) based on the diffusion of the molecule through an aqueous solution.
  • the diffusion, or motion of a protein through solution can be processed to derive an apparent size of the protein, where the size is given by the "Stokes radius” " or “hydrodynamic radius” of the protein particle.
  • the “hydrodynamic size” of a protein depends on both mass and shape (conformation), such that two proteins having the same molecular mass may have differing hydrodynamic sizes based on the overall conformation of the protein. Hydrodynamic size is measured, for example, by size exclusion chromatography.
  • the hydrodynamic size of a PEG-linked antibody polypeptide can be in the range of 24 kDa to 500 kDa; 30 to 500 kDa; 40 to 500 kDa; 50 to 500 IcDa; 100 to 500 kDa; 150 to 500 IcDa; 200 to 500 IcDa; 250 to 500 kDa; 300 to 500 IcDa; 350 to 500 IcDa; 400 to 500 IcDa and 450 to 500 IcDa.
  • the hydrodynamic size of a PEGylated antibody polypeptide of the invention is 30 to 40 kDa; 70 to 80 IcDa or 200 to 300 IcDa.
  • the polypeptide should have a hydrodynamic size of between 50 and 100 IcDa.
  • the polypeptide preparation should have a hydrodynamic size of greater than 200 kDa.
  • IC5 0 refers to the concentration of an inhibitor necessary to inhibit a given activity by 50%.
  • IC 50 is determined by assaying a given activity, e.g., binding of CD40L to CD40, in the presence of varying amounts of the inhibitor (e.g., monovalent anti-CD40L antibody polypeptide), and plotting the inhibitor concentration versus the activity being targeted. Binding of CD40L to CD40 is measured herein by the method described in Example 6. Alternatively, SPR can be used.
  • fused to an antibody polypeptide means that a polypeptide is fused to a given antibody through use of recombinant DNA techniques.
  • an antibody “fused to” another polypeptide e.g., to another antibody of different binding specificity, does not exist in nature and is generated through recombinant means.
  • the term “fused to an antibody polypeptide” also encompasses the linkage of a polypeptide to a given antibody polypeptide through, for example, disulfide or other chemical linkages, where the fused polypeptide is not naturally found fused to the antibody polypeptide. Recombinant and chemical methods of fusing a polypeptide to another polypeptide, e.g., to an antibody, are well known in the art.
  • Fc domain refers to the constant region antibody sequences comprising CH2 and CH3 constant domains as delimited according to Kabat et al., supra.
  • the Fc portion of the heavy chain polypeptide has the ability to self-associate, a function which facilitates the formation of divalent antibodies.
  • the term "lacks an Fc domain” means that a given antibody polypeptide lacks at least the portion of an immunoglobulin Fc domain (as such domains are defined according to Kabat et al., supra) sufficient to mediate the dimerization of Fc-containing antibody polypeptides. Dimerization of Fc-containing antibody polypeptides is measured, for example, by chromatographic methods or by surface plasmon resonance. An antibody polypeptide lacking an Fc domain avoids Fc-platelet interactions and therefore avoids induction of platelet aggregation.
  • symptom of disease refers to a decrease of the a symptom by at least 10% based on a a clinically measurable parameter, or by at least one point on a clinically-accepted scale of disease or symptom severity.
  • symptom of systemic lupus erythematosus refers to any of the clinically relevant symptoms of SLE known to those of skill in the art.
  • Non-limiting examples include the accumulation of IgG autoantibodies (e.g., against nuclear antigens such as chromatin, snRHPs (especially Ul, Sm, Ro/SSA and La/SSB), phospholipids and cell surface molecules), hemolytic anemia, thrombocytopenia, leukopenia, glomerulonephritis, vasculitis, arthritis, and serositis).
  • a reduction in such a symptom is a reduction by at least 10% in a clinically measurable parameter, or by at least one point on a clinically-accepted scale of disease severity.
  • the phrase "specifically binds" refers to the binding of an antigen by an immunoglobulin variable domain with a dissociation constant (K d ) of 1 ⁇ M or lower as measured by surface plasmon resonance analysis using, for example, a BIAcoreTM surface plasmon resonance system and BIAcore T kinetic evaluation software (e.g., version 2.1).
  • K d dissociation constant
  • the affinity or Kd for a specific binding interaction is preferably about 500 nM or lower, more preferably about 300 nM or lower.
  • a "generic ligand” is a ligand that binds a substantial proportion of functional members in a given repertoire, e.g., in a phage display library.
  • the same generic ligand can bind many members of the repertoire regardless of their target ligand specificities.
  • the presence of a functional generic ligand binding site indicates that the repertoire member is expressed and folded correctly.
  • binding of the generic ligand to its binding site provides a method for preselecting functional polypeptides from a repertoire of polypeptides.
  • Generic ligands include, for example, Protein A, Protein G and Protein L.
  • universal framework refers to a single antibody framework sequence corresponding to the regions of an antibody conserved in sequence as defined by Kabat (supra) 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.
  • Figure 1 shows gel analysis of the quality of biotin-labeled CD40L used in the screening procedures described herein, (a) 1 ⁇ g of non-bio tinylated-CD40L (Lane 1) and 0.3 ⁇ g of biotin-CD40L (Lane 2) were analysed on SDS-PAGE and detected by Simply Blue Safe-Stain, (b) 0.1 ⁇ g of biotin-CD40L (Lane 1) and 0.02 ⁇ g of biotin- CD40L (Lane 2) were detected by Western-blot probing with 1 :5000 Streptavidin- HRP.
  • Figure 2 shows a graphical representation of a dose response receptor binding assay (RBA) readout, analysing the inhibition of CD40L binding to CD40-Fc by dAbs DOM-IO, -20, -27, -30, -31, -62, -77, titrated from 1 ⁇ M down to 10 pM.
  • dAbs DOM-20, -30, and -31 are the most potent with IC 5 0 values of approximately 8 nM.
  • Figure 3 shows a graphical representation of a dose response receptor binding assay readout, analysing the inhibition of CD40L binding to CD40-Fc by dAbs DOM- 4. and DOM-5, titrated from 1 ⁇ M down to 500 pM.
  • the IC 50 values for dAbs DOM- 5 and DOM-4 are approximately 3 nM and 100 nM respectively.
  • Figure 4 shows a graphical representation of a dose response receptor binding assay readout, analysing the inhibition of CD40L binding to CD40-Fc by dAb DOM- 24, titrated from 100 nM down to 0.5 pM. The data were curve-fitted using GraphPad Prism software.
  • Figure 5 shows the sequence of the VH framework based on germline sequence DP47 - JH4b (SEQ E) NO: 1, amino acid sequence; SEQ E) NO: 2, nucleotide sequence - both sense and antisense strands are shown - SEQ E) NO: 2 is the top strand (sense) and SEQ E ) NO: 476 is the lower strand (anti-sense)).
  • Figure 6 shows the sequence of the V ⁇ framework based on germline sequence DPK9 - J ⁇ l (SEQ E ) NO: 3, amino acid sequence; SEQ E) NO: 4, nucleotide sequence — both sense and antisense strands are shown - SEQ E) NO: 4 is the top strand (sense) and SEQ E) NO: 477 is the lower strand (anti-sense)).
  • LCDRs LCDRs
  • Figure 7 shows a schematic representation of the CD40L binding assay used herein, e.g., in Example 6.
  • Figure 8 shows various GASl secretion signal peptide coding sequences.
  • GAS wt The natural occurring sequence in yeast.
  • GAS E.Coli The nucleotide sequence according to optimal E. CoIi codon usage (Wada et al. 1992 NAR 20 p 2111).
  • GAS leader AT AT rich nucleotide sequence.
  • AU nucleotide sequences encode the same amino acid sequence. Yellow (light grey in greyscale) indicates nucleotides that are similar for all sequences. Blue (dark grey in greyscale) indicates nucleotides that are similar to the wt sequence. White (white in greyscale) indicates nucleotides that are different from the wt sequence.
  • Figure 9 shows the results of -a receptor binding assay which demonstrates the affinity of PEGylated DOM8-24cys with either 3OK PEG MAL or 40K PEG2-MAL.
  • Figure 10 shows the results of an assay to assess the simultaneous binding of a dual specific dimer to HSA and CD40L (shaded bars). Binding to control BSA antigen is also shown (solid bars).
  • Figure 11 shows the results of an assay to assess the simultaneous binding of a dual specific Fab to HSA and CD40L (shaded bars). Binding to control skimmed mild powder antigen is also shown (solid bars).
  • Figure 12 shows the results of FACS analysis of the inhibitory effect of monomeric DOM-24 (grey dotted line). Control stimulated cells ae shown as the solid black line and a control dAb is shown as the grey solid line.
  • Figure 13 shows the results of FACS analysis of the inhibitory effect of the Vk dAb DOM-116 (dotted line). Control stimulated cells are shown as the solid black line and a control dAb is shown as the grey solid line.
  • the invention provides antibody polypeptides that are monovalent for binding to CD40L.
  • Monovalency for CD40L binding removes the possibility for cross- linking that occurs with prior art antibodies, and which plays a role in undesirable side effects observed with anti-CD40L monoclonal antibodies.
  • cross-linked CD40L may in turn cross-link cell surface CD40 and result in agonism of CD40 signaling activity.
  • the anti- CD40L antibodies disclosed herein not only inhibit or antagonize the binding of CD40L to CD40, they do not substantially agonize CD40 and/or CD40L activity.
  • the antibodies monovalent for CD40L binding are human antibody polypeptides.
  • Human antibody polypeptides can be administered to human patients while largely avoiding the anti-antibody immune response often provoked by the administration of antibodies from other species, e.g., mouse.
  • murine antibodies can be "humanized” by grafting human constant domains onto the murine antigen-binding domains, human antibodies as disclosed herein are produced without the need for laborious and time-consuming genetic manipulation of a murine antibody sequence.
  • the heavy and light polypeptide chains of antibodies comprise variable (V) regions that directly participate in antigen interactions, and constant (C) regions that provide structural support and function in non-antigen-specific interactions with immune effectors.
  • the antigen binding domain of a conventional antibody is comprised of two separate domains: a heavy chain variable domain (V H ) and a light chain variable domain (VL: which can be either V ⁇ or Vx).
  • the antigen binding site itself is formed by six polypeptide loops: three from the V H domain (Hl, H2 and H3) and three from the V L domain (Ll, L2 and L3).
  • C regions include the light chain C regions (referred to as C L regions) and the heavy chain C regions (referred to as C H I, C H 2 and C H 3 regions).
  • a naturally-occurring antibody generally comprises two antigen binding domains and is therefore divalent.
  • a number of smaller antigen binding fragments of naturally occurring antibodies have been identified following protease digestion. These include, for example, the "Fab fragment” (V L -C L -C H 1-VH), "Fab' fragment” (a Fab with the heavy chain hinge region), and "F(ab') 2 fragment” (a dimer of Fab' fragments joined by the heavy chain hinge region). Recombinant methods have been used to generate such fragments and to generate even smaller antigen-binding fragments, e.g., those referred to as “single chain Fv” (variable fragment) or "scFv,” consisting of V L and V H joined by a synthetic peptide linker (VL-linker-Vn).
  • Fab fragment V L -C L -C H 1-VH
  • Fab' fragment a Fab with the heavy chain hinge region
  • F(ab') 2 fragment a dimer of Fab' fragments joined by the heavy chain hinge region
  • Fab fragments, Fab' fragments and scFv fragments are monovalent for antigen binding, as they each comprise only one antigen binding domain comprising one V R /V L dimer. Even smaller monovalent antibody fragments are the "domain antibodies,” or "dAbs,” which comprise only a single immunoglobulin variable domain, e.g., V H or VL, that alone specifically binds antigen, i.e., without the need for a complementary V L or VH domain, respectively.
  • dAb will refer herein to a single immunoglobulin variable domain
  • V H or V L polypeptide that specifically binds antigen.
  • A- "dAb” binds antigen independently of other V domains; however, a “dAb” can be present in a homo- or heteromultimer with other V H or V L domains where the other domains are not required for antigen binding by the dAb, i.e., where the dAb binds antigen independently of the additional V H or V L domains.
  • the preparation of single immunoglobulin variable domains is described and exemplified herein below.
  • Monovalent antibody polypeptides can be generated in several different ways.
  • the nucleic acid sequence encoding heavy and light chains of an antibody known to bind CD40L can be manipulated to generate a number of different antibody polypeptides that are monovalent for CD40L binding.
  • monovalent antigen- binding polypeptide constructs such as Fab fragments, scFv, dAbs, or even bispecific antibodies (i.e., antibodies that comprise two different antigen-binding moieties and can therefore bind two separate antigens, preferably simultaneously) that are monovalent for CD40L.
  • one means of generating monovalent antibody polypeptides specific for CD40L is to amplify and express the V H and V L regions of the heavy chain and light chain gene sequences isolated, for example, from a hybridoma (e.g., a mouse hybridoma) that expresses anti-CD40L monoclonal antibody.
  • a hybridoma e.g., a mouse hybridoma
  • the boundaries of V H and V L domains are set out by Kabat et al. (1991, supra).
  • the information regarding the boundaries of the V H and V L domains of heavy and light chain genes is used to design PCR primers that amplify the V domain from a heavy or light chain coding sequence encoding an antibody known to bind CD40L.
  • the amplified V domains are inserted into a suitable expression vector, e.g., pHEN-1 (Hoogenboom et al., 1991, Nucleic Acids Res. 19: 4133-4137) and expressed, e.g., as a fusion of the V H and V L in an scFv or other suitable monovalent format.
  • pHEN-1 Hoogenboom et al., 1991, Nucleic Acids Res. 19: 4133-4137
  • the resulting polypeptide is then screened for high affinity monovalent binding to CD40L.
  • screening for binding is performed as known in the art or as described herein below.
  • library screening methods can be used to identify monovalent CD40L-specific binding proteins.
  • Phage display technology provides an approach for the selection of antibody polypeptides which bind a desired target from among large, diverse repertoires of antibody polypeptides.
  • These phage-antibody libraries can be grouped into two categories: natural libraries which use rearranged V genes harvested from human B cells (Marks et al., 1991, J. MoI.
  • Methods involving genetic display packages are well-suited for the selection of monovalent CD40L-specif ⁇ c antibody constructs because they generally express only monovalent fragments, rather than whole, divalent antibodies, on the display packages.
  • Methods for the preparation of phage display libraries displaying various antibody fragments are described in the preceding references. Such methods are also described, for example, in U.S. Patent No. 6,696,245, which is incorporated herein by reference.
  • the methods described in the '245 patent generally involve the randomization of selected regions of immunoglobulin gene coding regions, in particular V H and V L coding regions, while leaving other regions non-randomized (see below).
  • the '245 patent also describes the generation of scFv constructs comprising individually randomized V H and V L domains.
  • the V H gene is produced by the recombination of three gene segments, V H , D and JH- In humans, there are approximately 51 functional V H segments (Cook and Tomlinson (1995) Immunol Today 16: 237), 25 functional D segments (Corbett et al. (1997) J. MoI. Biol. 268: 69) and 6 functional JH segments (Ravetch et al. (1981) Cell 27: 583), depending on the haplotype.
  • the V H segment encodes the region of the polypeptide chain which forms the first and second antigen binding loops of the V H domain (Hl and H2), while the V H , D and JH segments combine to form the third antigen binding loop of the VH domain (H3).
  • V L ⁇ ene ⁇ s produced by the recombination of only two gene segments, V L and JL.
  • V L and JL there are approximately 40 functional V ⁇ segments (Schable and Zachau (1993) Biol. Chem. Hoppe-Seyler 374: 1001), 31 functional V ⁇ segments (Williams et al. (1996) J. MoI. Biol. 264: 220; Kawasaki et al. (1997) Genome Res. 7: 250), 5 functional J ⁇ segments (Hieter et al. (1982) J. Biol. Chem. 257: 1516) and 4 functional J ⁇ segments (Vasicek and Leder (1990) J. Exp. Med. 172: 609), depending on the haplotype.
  • V L segment encodes the region of the polypeptide chain which forms the first and second antigen binding loops of the V L domain (Ll and L2), while the V L and JL segments combine to form the third antigen binding loop of the V L domain (L3).
  • Antibodies selected from this primary repertoire are believed to -be sufficiently diverse to bind almost all antigens with at least moderate affinity.
  • High affinity antibodies are produced in vivo by "affinity maturation" of the rearranged genes, in which point mutations are generated and selected by the immune system on the basis of improved binding.
  • the 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. While, in one approach, diversity can be added to S3'nthetic repertoires at any site in the CDRs of the various antigen-binding loops, this 'approach results in a greater proportion of V domains that do not properly fold and therefore contribute to a lower proportion of molecules with the potential to bind antigen.
  • an antibody polypeptide comprises the amino acid sequence of a given human germline V region gene segment, e.g., V H germline gene segment DP-47, or V ⁇ germline gene segment DPK9.
  • variable region polypeptides can be used for the production of scFvs or Fabs, e.g., an scFv or Fab comprising (i) an antibody heavy chain variable domain (V H ), or antigen binding fragment thereof, which comprises the amino acid sequence of germline VH segment DP-47 and (ii) an antibody light chain variable domain (V L ), or antigen binding fragment thereof, which comprises the amino acid sequence of germline V ⁇ segment DPK9.
  • V H antibody heavy chain variable domain
  • V L antibody light chain variable domain
  • variable region polypeptides can also be expressed as dAbs and screened for high affinity binding to CD40L.
  • the repertoire can be cloned into or generated in a vector suitable for phage display, e.g., a lambda or filamentous bacteriophage display vector and is then screened for binding to a given target antigen, e.g., CD40.L.
  • a single immunoglobulin variable domain is a folded polypeptide domain which comprises sequences characteristic of immunoglobulin variable domains and which specifically binds an antigen (e.g., dissociation constant of 500 nM or less), and which binds antigen as a single variable domain; that is, there is one binding site provided by a single immunoglobulin variable domain without any complementary variable domain.
  • an antigen e.g., dissociation constant of 500 nM or less
  • a single immunoglobulin variable domain therefore includes complete antibody variable domains as well as 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 a dissociation constant of 500 nM or less (e.g., 450 nM or less, 400 nM or less, 350 nM or less, 300 nM or less, 250 nM or less, 200 nM or less, 150 nM or less, 100 nM or less) and the target antigen specificity of the full- length domain.
  • 500 nM or less e.g., 450 nM or less, 400 nM or less, 350 nM or less, 300 nM or less, 250 nM or less, 200 nM or less, 150 nM or less, 100 nM or less
  • an antibody single variable domain useful in the invention is selected from the group of VH and VL, including Vk apP a and Vi am b da -
  • the single immunoglobulin variable domains of use herein are preferably "human" as that term is defined herein.
  • Single immunoglobulin variable domains are prepared in a number of ways. For each of these approaches, well-known methods of preparing (e.g., amplifying, mutating, etc.) and manipulating nucleic acid sequences are applicable.
  • One means of preparing single immunoglobulin variable domains is to amplify and express the V H or V L region of a heavy chain or light chain gene for a
  • V H or VL domain of a known anti-CD40L antibody coding region can be amplified and expressed as a single domain (or as a fusion of a single domain) and evaluated for binding to CD40L.
  • the boundaries of V H and V L domains are set out by Kabat et al. (1991, supra).
  • the information regarding the boundaries of the V H and V L domains of heavy and light chain genes is used to design PCR primers that amplify the V domain from a cloned heavy or light chain coding sequence encoding an antibody known to bind CD40L.
  • the amplified V domain is inserted into a suitable expression vector, e.g., pHEN-1 (Hoogenboom et al., 1991, Nucleic Acids Res. 19: 4133-4137) and expressed, either alone or as a fusion with another polypeptide sequence.
  • pHEN-1 Hoogenboom et al., 1991, Nucleic Acids Res. 19: 4133-4137
  • V H or V L domains preferably human V H or V L domains
  • phage display panning against the desired antigen.
  • Methods for the construction of bacteriophage display libraries and lambda phage expression libraries are well known in the art, and taught, for example, by: McCafferty et al., 1990, Nature 348: 552; Kang et al., 1991, Proc. Natl. Acad. Sci. U.S.A., 88: 4363; Clackson et al., 1991, Nature 352: 624; Lowman et al., 1991, Biochemistry 30: 10832; Burton et al., 1991, Proc.
  • Fab phage display libraries are taught, for example, by U.S. 5,922,545.
  • scFv phage libraries are taught, for example, by Huston et al., 1988, Proc. Natl. Acad. Sci U.S.A. 85: 5879-5883; Chaudhary et al., 1990, Proc. Natl. Acad. Sci U.S.A.
  • the repertoire of V H or V L domains can be a naturally-occurring repertoire of immunoglobulin sequences or a synthetic repertoire.
  • a naturally-occurring repertoire is one prepared, for example, from immunoglobulin-expressing cells harvested from one or more individuals. Such repertoires can be "naive,” i.e., prepared, for example, from human fetal or newborn immunoglobulin-expressing cells, or rearranged, i.e., prepared from, for example, adult human B cells.
  • Natural repertoires are described, for example, by Marks et al., 1991, J. MoI. Biol. 222: 581 and Vaughan et al., 1996, Nature Biotech. 14: 309. If desired, clones identified from a natural repertoire, or any repertoire, for that matter, that bind the target antigen are then subjected to mutagenesis and further screening in order to produce and select variants with improved binding characteristics.
  • Synthetic repertoires of single immunoglobulin variable domains are prepared by artificially introducing diversity into a cloned V domain. Synthetic repertoires are described, for example, by Hoogenboom & Winter, 1992, J. MoI. Biol. 227: 381; Barbas et al., 1992, Proc. Natl. Acad. Sci. U.S.A. 89: 4457; Nissim et al., 1994, EMBO J. 13: 692; Griffiths et al, 1994, EMBO J. 13: 3245; DeKriuf et al., 1995, J. MoI. Biol. 248: 97; and WO 99/20749.
  • V H domain repertoires are prepared in V H or VK backgrounds, based on artificially diversified germline V H or VK sequences.
  • the V H domain repertoire can be based on cloned germline V H gene segments V3-23/DP47 (Tomlinson et al., 1992, J. MoI. Biol. 227: 7768) and JH4b.
  • the V ⁇ domain repertoire can be based, for example, on germline V ⁇ gene segments O2/O12/DPK9 (Cox et al., 1994, Eur. J. Immunol. 24: 827) and J ⁇ l. Diversity is introduced into these or other gene segments by, for example, PCR mutagenesis.
  • DVT codon (A/G/T) (A/G/QT )
  • DVC codon (A/G/T)(A/G/C)C
  • DVY codon (A/G/T)(A/G/C)(C/T).
  • the DVT codon encodes 22% serine and 11% tyrosine, asgpargine, glycine, alanine, aspartate, threonine and cysteine, which most closely mimics the distribution of amino acid residues for the antigen binding sites of natural human antibodies.
  • Repertoires are made using PCR primers having the selected degenerate codon or codons at each site to be diversified. PCR mutagenesis is well known in the art.
  • diversity is introduced into the sequence of human germline V H gene segments V3-23/DP47 (Tomlinson et al., 1992, J. MoI. Biol. 227: 7768) and JH4b using the NNK codon at sites H30, H31, H33, H35, H50, H52, H52a, H53, H55, H56, H58, H95, H97 and H98, corresponding to diversity in CDRs 1, 2 and 3, with the numbering as used in U.S. 6,696,245.
  • diversity is also introduced into the sequence of human germline V H gene segments V3-23/DP47 and JH4b, for example, using the NNK codon at sites H30, H31, H33, H35, H50, H52, H52a, H53, H55, H56, H58, H95, H97, H98, H99, HlOO, HlOOa and HlOOb, corresponding to diversity in CDRs 1, 2 and 3,with the numbering as used in U.S. 6,696,245.
  • V ⁇ gene segments O2/O12/DPK9 and J ⁇ l for example, using the NNK codon at sites L30, L31, L32, L34, L50, L53, L91, L92, L93, L94 and L96, corresponding to diversity in CDRs 1, 2 and 3,with the numbering as used in U.S. 6,696,245.
  • nucleic acid molecules and vector constructs required for the performance of the present invention are available in the art and are 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 in the present invention is typicalfy 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.
  • vectors are publicly available, including bacterial plasmids, bacteriophage, artificial chromosomes and episomal vectors. Such vectors may be used for simple cloning and mutagenesis; alternatively, as is typical of vectors in which repertoire (or pre-repertoire) members of the invention are carried, a gene expression vector is employed.
  • a vector of use according to the invention is selected to accommodate a polypeptide coding sequence of a desired size, typically from 0.25 kilobase (kb) to 40 kb 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 "polylinker”) site, an origin of replication and at least one selectable marker gene. If a 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 polypeptide repertoire member 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.
  • a cloning or expression vector also contains 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. coli-selectable marker for example, the ⁇ - lactamase gene that confers resistance to the antibiotic ampicillin, is of use.
  • E. coli plasmids such as pBR322 or a pUC plasmid such as pUC18 or pUC19.
  • Expression vectors usually contain a promoter that is recognized 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-Dalgarno sequence operably linked to the coding sequence.
  • the preferred vectors are expression vectors that enable the expression of a nucleotide sequence corresponding to a polypeptide library member. Thus, selection is performed by separate propagation and expression of a single clone expressing the polypeptide library member or by use of any selection display system. As described above, a preferred selection display system uses bacteriophage display. Thus, phage or phagemid vectors can be used. Preferred vectors are phagemid vectors, which have an E. coli origin of replication (for double stranded replication) and also a phage origin of replication (for production of single-stranded DNA).
  • the vector contains a ⁇ -lactamase or other selectable marker 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 tags (for detection), optionally, one or more TAG stop codons and the phage protein pill.
  • the vector encodes, rather than the pelB leader sequence, a eukaryotic GASl leader sequence which serves to direct the secretion of the fusion polypeptide to the periplasmic space in E. coli or to the medium in eukaryotic cell systems.
  • a eukaryotic GASl leader sequence which serves to direct the secretion of the fusion polypeptide to the periplasmic space in E. coli or to the medium in eukaryotic cell systems.
  • 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.
  • a preferred vector is the pHENl phagemid vector (Hoogenboom et al., 1991, Nucl. Acids Res. 19: 4133-4137; sequence is available, e.g., as SEQ ID NO: 7 in WO 03/031611), in which the production of pill fusion protein is under the control of the LacZ promoter, which is inhibited in the presence of glucose and induced with IPTG. When grown in suppressor strains of E.
  • the gene III fusion protein is produced and packaged into phage, while growth in non-suppressor strains, e.g., HB2151, permits the secretion of soluble fusion protein into the bacterial periplasm and into the culture medium. Because the expression of gene III prevents later infection with helper phage, the bacteria harboring the phagemid vectors are propagated in the presence of glucose before infection with VCSM 13 helper phage for phage rescue.
  • vectors employs conventional ligation techniques. Isolated vectors or DNA fragments are cleaved, tailored, and re- ligated in the form desired to generate the required vector. If desired, sequence analysis to confirm that the correct sequences are present in the constructed vector is performed using standard methods. 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. 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 hybridization, immunocytochemistry or sequence analysis of nucleic acid or protein molecules. Those skilled in the art will readily envisage how these methods may be modified, if desired.
  • a repertoire of single immunoglobulin variable domains on the surface of phage selection is performed by contacting the phage repertoire with immobilized target antigen (e.g., CD40L and/or an epitope bound by DOM8-24), washing to remove unbound phage, and propagation of the bound phage, the whole process frequently referred to as "panning.”
  • immobilized target antigen e.g., CD40L and/or an epitope bound by DOM8-24
  • This process is applicable to the screening of single immunoglobulin variable domains as well as other antibody fragments that can be expressed on a display library, e.g., scFv, Fab, etc.
  • phage are pre-selected for the expression of properly folded member variants by panning against an immobilized generic ligand (e.g., protein A or protein L) that is only bound by folded members.
  • an immobilized generic ligand e.g., protein A or protein L
  • This has the advantage of reducing the proportion of non-functional members, thereby increasing the proportion of members likely to bind a target antigen.
  • Pre-selection with generic ligands is taught in WO 99/20749. The screening of phage antibody libraries is generally described, for example, by Harrison et al., 1996, Meth. Enzymol. 267: 83-109.
  • Screening is commonly performed using purified antigen immobilized on a solid support, for example, plastic tubes or wells, or on a chromatography matrix, for example Sepharose ⁇ (Pharmacia). Screening or selection can also be performed on complex antigens, such as the surface of cells (Marks et al., 1993, BioTechnology 11: 1145; de Kruif et al., 1995, Proc. Natl. Acad. Sci. U.S.A. 92: 3938). Another alternative involves selection by binding biotinylated antigen in solution, followed by capture on streptavidin-coated beads.
  • panning is performed by immobilizing antigen (generic or specific) on tubes or wells in a plate, e.g., Nunc MAXISORPTM immunotube 8 well strips.
  • Wells are coated with 150 ⁇ l of antigen (100 ⁇ g/ml in PBS) and incubated overnight.
  • the wells are then washed 3 times with PBS and blocked with 400 ⁇ l PBS-2% skim milk (2%MPBS) at 37 0 C for 2 hr.
  • the wells are rinsed 3 times with PBS and phage are added in 2%MPBS.
  • the mixture is incubated at room temperature for 90 minutes and the liquid, containing unbound phage, is removed.
  • Bound phage are eluted by adding 200 ⁇ l of freshly prepared 100 mM triethylamine, mixing well and incubating for 10 min at room temperature. Eluted phage are transferred to a tube containing 100 ⁇ l of IM Tris-HCl, pH 7.4 and vortexed to neutralize the triethylamine. Exponentially-growing E. coli host cells (e.g., TGl) are infected with, for example, 150 ml of the eluted phage by incubating for 30 min at 37 0 C.
  • E. coli host cells e.g., TGl
  • Infected cells are spun down, resuspended in fresh medium and plated in top agarose. Phage plaques are eluted or picked into fresh cultures of host cells to propagate for analysis or for further rounds of selection. One or more rounds of plaque purification are performed ' if necessary to ensure pure populations of selected phage. Other screening approaches are described by Harrison et al., 1996, supra.
  • variable domain fusion proteins are easily produced in soluble form by infecting non-suppressor strains of bacteria, e.g., HB2151 that permit the secretion of soluble gene III fusion protein.
  • non-suppressor strains of bacteria e.g., HB2151 that permit the secretion of soluble gene III fusion protein.
  • the fusion polypeptide can be secreted by eukaryotic (e.g., yeast or mammalian) or prokaryotic (e.g., E. coli) ceils.
  • the V domain sequence can be sub-cloned into an appropriate expression vector to produce soluble protein according to methods known in the art.
  • variable domain polypeptides or other monovalent antibody polypeptides secreted into the periplasmic space or into the medium of bacteria are harvested and purified according to known methods (Harrison et al., 1996, supra). Skerra & Pluckthun (1988, Science 240: 1038) and Breitling et al. (1991, Gene 104: 147) describe the harvest of antibody polypeptides from the periplasm,, and Better et al. (1988, Science 240: 1041) describes harvest from the culture supernatant.
  • purification can also be achieved by binding to generic ligands, such as protein A or Protein L.
  • the variable domains can be expressed with a peptide tag, e.g., the Myc, HA or 6X- His tags, which facilitates purification by affinity chromatography.
  • monovalent anti-CD40L antibody polypeptides are concentrated by any of several methods well known in the art, including, for example, ultrafiltration, diafiltration and tangential flow filtration.
  • the process of ultrafiltration uses semi-permeable membranes and pressure to separate molecular species on the basis of size and shape. The pressure is provided by gas pressure . or by centrifugation.
  • Commercial ultrafiltration products are widely available, e.g., from Millipore (Bedford, MA; examples include the CentriconTM and MicroconTM concentrators) and Vivascience (Hannover, Germany; examples include the VivaspinTM concentrators).
  • a molecular weight cutoff smaller than the target polypeptide usually 1/3 to 1/6 the molecular weight of the target polypeptide, although differences of as little as 10 kD can be used successfully
  • the polypeptide is retained when solvent and smaller solutes pass through the membrane.
  • a molecular weight cutoff of about 5 kD is useful for concentration of anti- CD40L single immunoglobulin variable domain polypeptides described herein.
  • Diafiltration which uses ultrafiltration membranes with a "washing" process, is used where it is desired to remove or exchange the salt or buffer in a polypeptide preparation.
  • the polypeptide is concentrated by the passage of solvent and small solutes through the membrane, and remaining salts or buffer are removed by dilution of the retained polypeptide with a new buffer or salt solution or water, as desired, accompanied by continued ultrafiltration.
  • new buffer is added at the same rate that filtrate passes through the membrane.
  • a diafiltration volume is the volume of polypeptide solution prior to the start of diafiltration - using continuous diafiltration, greater than 99.5% of a fully permeable solute can be removed by washing through six diafiltration volumes with the new buffer.
  • the process can be performed in a discontinuous manner, wherein the sample is repeatedly diluted and then filtered back to its original volume to remove or exchange salt or buffer and ultimately concentrate the polypeptide.
  • Equipment for diafiltration and detailed methodologies for its use are available, for example, from Pall Life Sciences (Ann Arbor, MI) and Sartorius AG/Vivascience (Hannover, Germany).
  • Tangential flow filtration also known as “cross-flow filtration,” also uses ultrafiltration membrane. Fluid containing the target polypeptide is pumped tangentially along the surface of the membrane. The pressure causes a portion of the fluid to pass through the membrane while the target polypeptide is retained above the filter. In contrast to standard .ultrafiltration, however, the retained molecules do not accumulate on the surface of the membrane, but are carried along by the tangential flow. The solution that does not pass through the filter (containing the target polypeptide) can be repeatedly circulated across the membrane to achieve the desired degree of concentration.
  • Protein concentration is measured in a number of ways that are well known in the art. These include, for example, amino acid analysis, absorbance at 280 inn, the "Bradford” and “Lowry” methods, and SDS-PAGE. The most accurate method is total hydrolysis followed by amino acid analysis by HPLC, concentration is then determined then comparison with the known sequence of the single immunoglobulin variable domain polypeptide. While this method is the most accurate, it is expensive and time-consuming. Protein determination by measurement of UV absorbance at 280 nm faster and much less expensive, yet relatively accurate and is preferred as a compromise over amino acid analysis. Absorbance at 280 nm was used to determine protein concentrations reported in the Examples described herein.
  • the SDS-PAGE method uses gel electrophoresis and Coomassie Blue staining in comparison to known concentration standards, e.g., known amounts of a single immunoglobulin variable domain polypeptide. Quantitation can be done by eye or by densitometry.
  • Single human immunoglobulin variable domain antigen-binding polypeptides described herein retain solubility at high concentration (e.g., at least 4.8 mg ( ⁇ 400 ⁇ M) in aqueous solution (e.g., PBS), and preferably at least 5 mg/ml (-417 ⁇ M), 10 mg/ml (-833 ⁇ M), 20 mg/ml (-1.7 mM), 25 mg/ml (-2.3 mM), 30 mg/ml (-2.5 mM), 35 mg/ml (-2.9 mM), 40 mg/ml (-3.3 mM), 45 mg/ml (-3.75 mM), 50 mg/ml (-4.2 mM), 55 mg/ml (-4.6 mM), 60 mg/ml (-5.0 mM), 65 mg/ml (-5.4 mM), 70 mg/ml (-5.8 mM), 75 mg/ml (-6.3 mM), 100 mg/ml (-8.33 mM), 150 mg/ml (
  • single immunoglobulin variable domain polypeptides A full length conventional four chain antibody, e.g., IgG is about 150 kD in size.
  • single immunoglobulin variable domains which all have a general structure comprising 4 framework (FW) regions and 3 CDRs, have a size of approximately 12 IcD, or less than 1/10 the size of a conventional antibody.
  • single immunoglobulin variable domains are approximately 1 A the size of an scFv molecule (-26 IcD), and approximately 1/5 the size of a Fab molecule (-60 kD).
  • the size of a single immunoglobulin variable domain-containing structure disclosed herein is 100 kD or less, including structures of, for example, about 90 kD or less, 80 kD or less, 70 kD or less, 60 IcD or less, 50 IcD or less, 40 kD or less, 30 kD or less, 20 kD or less, down to and including about 12 kD, or a single immunoglobulin variable domain in isolation.
  • solubility of a polypeptide is primarily determined by the interactions of the amino acid side chains with the surrounding solvent. Hydrophobic side chains tend to be localized internally as a polypeptide folds, away from the solvent- interacting surfaces of the polypeptide. Conversely, hydrophilic residues tend to be localized at the solvent-interacting surfaces of a polypeptide. Generally, polypeptides having a primary sequence that permits the molecule to fold to expose more hydrophilic residues to the aqueous environment are more soluble than one that folds to expose fewer hydrophilic residues to the surface. Thus, the arrangement and number of hydrophobic and hydrophilic residues is an important determinant of solubility.
  • polypeptide solubility can be maintained or enhanced by the addition of glycerol (e.g., -10% v/v) to the solution.
  • glycerol e.g., -10% v/v
  • specific amino acid residues have been identified in conserved residues of human V H domains that vary in the VH domains of camelid species, which are generally more soluble than human V H domains. These include, for example, GIy 44 (GIu in camelids), Leu 45 (Arg in camelids) and Trp 47 (GIy in camelids). Amino acid residue 103 of V H is also implicated in solubility, with mutation from Trp to Arg tending to confer increased VH solubility.
  • single immunoglobulin variable domain polypeptides are based on the DP47 germline VH gene segment or the DPK9 germline V ⁇ gene segment.
  • these germline gene segments are capable, particularly when diversified at selected structural locations described herein, of producing specific binding single immunoglobulin variable domain polypeptides that are highly soluble.
  • the four framework regions which are preferably not diversified, can contribute to the high solubility of the resulting proteins.
  • the substitution or addition of a hydrophobic residue at the solvent-exposed surface, relative to a molecule of known solubility that has a less hydrophobic or even hydrophilic residue exposed in that position is expected to decrease the relative solubility of the polypeptide.
  • the substitution or addition of a more hydrophilic residue at such a location is expected to increase the relative solubility. That is, a change in the net number of hydrophilic or hydrophobic residues located at the surface of the molecule (or the overall hydrophobic or hydrophilic nature of the surface-exposed residues) relative to a single immunoglobulin variable domain polypeptide structure with known solubility can predict the relative solubility of a single immunoglobulin variable domain polypeptide.
  • the antigen-binding affinity of an antibody polypeptide can be conveniently measured by SPR using the BIAcore system (Pharmacia Biosensor, Piscataway, NJ.).
  • BIAcore Pharmaacia Biosensor, Piscataway, NJ.
  • antigen is coupled to the BIAcore chip at known concentrations, and variable domain polypeptides are introduced.
  • Specific binding between the variable domain polypeptide and the immobilized antigen results in increased protein concentration on the chip matrix and a change in the SPR signal. Changes in SPR signal are recorded as resonance units (RU) and displayed with respect to time along the Y axis of a sensorgram. Baseline signal is taken with solvent alone (e.g., PBS) passing over the chip.
  • solvent alone e.g., PBS
  • the net difference between baseline signal and signal after completion of antibody polypeptide injection represents the binding value of a given sample.
  • K O ff off rate
  • K 0n on rate
  • Kd dissociation rate
  • SPR can be used to monitor antagonism of CD40L binding to CD40 by a monovalent anti-CD40L antibody preparation by measuring the displacement or inhibition of binding of CD40L to CD40 caused the monovalent antibody preparation.
  • SPR can also be used to monitor the dimerization, or preferably, the lack of dimerization, occurring via Fc region in antibody preparations as described herein.
  • High affinity is dependent upon the complementarity between a surface of the antigen and the CDRs of the antibody or antibody fragment.
  • Complementarity is determined by the type and strength of the molecular interactions possible between portions of the target and the CDR, for example, the potential ionic interactions, van der Waals attractions, hydrogen bonding or other interactions that can occur.
  • CDR3 tends to contribute more to antigen binding interactions than CDRs 1 and 2, probably due to its generally larger size, which provides more opportunity for favorable surface interactions. (See, e.g., Padlan et al., 1994, MoL Immunol. 31: 169-217; Chothia & Lesk, 1987, J. MoL Biol. 196: 904-917; and Chothia et al., 1985, J.
  • a monovalent anti-CD40L antibody polypeptide e.g., a single immunoglobulin variable domain polypeptide
  • another antibody polypeptide is linked to another antibody polypeptide to form a heterodimer in which each individual antibody polypeptide is capable of binding a different cognate antigen.
  • Fusing antibody polypeptides, such as single immunoglobulin variable domains, as heterodimers, wherein each monomer binds a different target antigen can produce a dual-specific ligand capable, for example, of bridging the respective target antigens.
  • Such dual specific ligands may be used to target cytokines and other molecules which cooperate synergistically in therapeutic situations in the body of an organism.
  • a method for synergising the activity of two or more cytokines comprising administering a dual specific antibody heterodimer capable of binding to the two or more cytokines.
  • Non-limiting examples of second targets for anti-CD40L dual specific antibody polypeptides include the following: TNF- ⁇ ; IL-I; IL-2; IL-4; IL-6; IL-8; IL- 12; IL-18; IFN- ⁇ ; CD2; CD4; CD8; CTLA4; LFAl; LFA3, VLA4, CD80, B7-1, CD28, CD86, B7-2, and CTLA-4.
  • second targets useful according to the invention include CD80, B7-1, CD28, CD86, B7-2, and CTLA-4. These targets are thought to be involved in a co-stimulatory pathway critical for T-cell activation (termed, co-stimulatory signal pathway antigens).
  • This pathway includes activation of the molecule CD28 on the surface of T cells.
  • This molecule can receive a costimulatory signal delivered by a ligand on B cells or other APCs.
  • Ligands for CD28 include members of the B7 family of B lymphocyte activation antigens, such as B7-1 and/or B7-2 (Freedman, A. S. et al. (1987) J. Immunol. 137, 3260-3267; Freeman, G. J. et al. (1989) J. Immunol. 143, 2714-2722; Freeman, G. J. et al. (1991) J. Exp. Med. 174, 625-631; Freeman, G. J. et al.
  • B7-1 and B7-2 are also ligands for another molecule, CTLA4, present on the surface of activated T cells. Accordingly, the present invention contemplates that members of the CD28 signalling pathway may be useful second targets for the dual-specific format anti-CD40L antibody polypeptides.
  • the invention encompasses anti-CD40L antibody polypeptides, e.g., CD40L- binding single immunoglobulin variable domain clones, and clones with substantial sequence similarity or homology to them that also bind target antigen with high affinity.
  • substantial sequence similarity or homology is at least 85% similarity or homology.
  • sequence identity is calculated 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.
  • sequence “similarity” is a measure of the degree to which amino acid sequences share similar amino acid residues at corresponding positions in an alignment of the sequences. Amino acids are similar to each other where their side chains are similar. Specifically, “similarity” encompasses amino acids that are conservative substitutes for each other. A “conservative” substitution is any substitution that has a positive score in the blosum62 substitution matrix (Hentikoff and Hentikoff, 1992, Proc. Natl. Acad. Sci. USA 89: 10915-10919). By the statement “sequence A is n% similar to sequence B" is meant that n% of the positions of an optimal global alignment between sequences A and B consists of identical amino acids or conservative substitutions. Optimal global alignments can be performed using the following parameters in the Needleman-Wunsch alignment algorithm: For polypeptides:
  • Substitution matrix blosum62.
  • substitution matrix 10 for matches, 0 for mismatches.
  • Typical conservative substitutions are among Met, VaI, Leu and lie; among Ser and Thr; among the residues Asp, GIu and Asn; among the residues GIn, Lys and Arg; or aromatic residues Phe and Tyr.
  • degree most often as a percentage of similarity between two polypeptide sequences, one considers the number of positions at which identity or similarity is observed between corresponding amino acid residues in the two polypeptide sequences in relation to the entire lengths of the two molecules being compared.
  • BLAST Basic Local Alignment Search Tool
  • the BLAST algorithm "BLAST 2 Sequences” is available at the world wide web site ("www") of the National Center for Biotechnology Information (“.ncbi”), of the National Library of Medicine (“.nlm”) of the National Institutes of Health (“nih”) of the U.S. government (“.gov”), in the "/blast/” directory, sub-directories "bl2seq/bl2.html.” This algorithm aligns two sequences for comparison and is described by Tatusova & Madden, 1999, FEMS Microbiol Lett. 174:247-250.
  • a first sequence encoding a single immunoglobulin variable domain polypeptide is substantially similar to a second coding sequence if the first sequence hybridizes to the second sequence (or its complement) under highly stringent hybridization conditions (such as those described by Sambrook et al., Molecular Cloning, Laboratory Manuel, Cold Spring, Harbor Laboratory press, New York).
  • Highly stringent hybridization conditions refer to hybridization in 6X SSC at about 45°C, followed by one or more washes in 0.2X SSC, 0.1% SDS at 65°C.
  • Very highly stringent hybridization conditions refer to hybridization in 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.
  • a monovalent anti-CD40L antibody polypeptides as described herein bind to CD40L yet do not substantially agonize CD40 signaling.
  • Activation of the CD40L/CD40 pathway manifests a number of different outcomes that can be measured in order to assess the effect of a given monovalent anti-CD40L antibody polypeptide on the activity of the pathway.
  • at least one of the following CD40L assays can be used:
  • JNK Jun-N-Terminal Kinase
  • a monovalent anti-CD40L antibody polypeptide to activate this signaling pathway is measured as follows. Human leukemic Jurkat cells are stimulated with a positive control agonistic anti-CD40L antibody (2 ⁇ g/ml monoclonal anti-human or anti-mouse gp39/CD40L antibody (Pharmingen, San Diego, CA, USA) or isotype matched hamster or mouse immunoglobulins (Dianova, Hamburg, Germany)), monovalent anti-CD40L antibody polypeptide, or a negative control irrelevant antibody as described by Brenner et al., 1997, FEBS Lett.
  • a positive control agonistic anti-CD40L antibody 2 ⁇ g/ml monoclonal anti-human or anti-mouse gp39/CD40L antibody (Pharmingen, San Diego, CA, USA) or isotype matched hamster or mouse immunoglobulins (Dianova, Hamburg, Germany)
  • monovalent anti-CD40L antibody polypeptide or a negative control irrelevant
  • the cells are lysed and the extract assayed for phosphorylated JNK via colorimetric assay (e.g., TiterzymeTM colorimetric (EIA) phospho-JNKl/2 immunoassay kit, by Assay Designs Inc., Catalog # 900-106).
  • colorimetric assay e.g., TiterzymeTM colorimetric (EIA) phospho-JNKl/2 immunoassay kit, by Assay Designs Inc., Catalog # 900-106.
  • Co-stimulation of T cells with anti-CD3 Ab and CD40L has been shown to upregulate the production of IL-IO, IFN- ⁇ and TNF- ⁇ by those cells.
  • the ability of a monovalent anti-CD40L antibody polypeptide to activate this signaling pathway is measured as follows. Human leukemic Jurkat T cells (or freshly isolated CD4+ T cells) are plated into 96 well plates containing immobilized anti-CD3 antibody. The cells are then cultured for 72 hours in the presence of a positive control agonistic anti- CD40L antibody, CD40L, monovalent anti-CD40L antibody polypeptide, or a negative control irrelevant antibody, as described by Blair et al., 2000, J. Exp. Med. 191 : 651-660.
  • IFN- ⁇ (or IL-10 of TNF- ⁇ ) is then quantitated in the supernatant by sandwich ELISA using an IFN-g standard to generate a standard curve from which all other unknowns can be calculated.
  • An increase in IFN-g (e.g., by 5% or more) for anti-CD40L-stimulated cells over non-stimulated cells indicates agonism by the antibody polypeptide.
  • Divalent anti-CD40L antibodies tend to cause platelet aggregation, which is likely associated with the thromboembolic events observed in clinical trials of divalent anti-CD40L antibodies in the prior art.
  • Monovalent anti-CD40L antibody polypeptides as described herein preferably do not substantially mediate or agonize CD40L-mediated platelet aggregation.
  • the "standard platelet aggregation assay" is as follows:
  • Platelets are prepared at 2.5x10 5 /ml and left stirring in a 500-Ca lumi- aggregometer (or its equivalent, e.g., a Platelet Aggregation Profiler (BioData, Horsham, PA)). Platelets are partially activated by the addition of a dilution series of 0.1-10 ⁇ M ADP (the lO ⁇ M ADP induces aggregation, and is used as a positive control - lower concentrations activate platelets but do not induce aggregation).
  • CD40L mediated platelet aggregation is stimulated by addition of either anti-CD40L monoclonal antibodies (i.e., divalent monoclonal antibodies, available from, e.g., Pharmingen, San Diego, CA, USA) or soluble CD40/Fc fusion protein (available from R&D Systems). The reaction is allowed to proceed for between 3 and 5 minutes. Stimulation of platelet aggregation above the mininimal aggregation/activation achieved with ADP alone is plotted against stimulating anti- CD40L or CD40/Fc concentration. The percentage of platelet aggregation is measured by the change in light transmittance following addition of antibody polypeptide being tested or positive control peptide. A value greater than that observed for negative control lacking antibody and amounting to 25% or more of the positive control value (divalent anti-CD40L or CD40/Fc fusion) is considered to be indicative of induction of platelet aggregation.
  • anti-CD40L monoclonal antibodies i.e., divalent mono
  • platelet activation can be determined by assaying for CD62P expression in platelets (e.g., using anti-CD26P antibodies and flow cytometry), assaying for monocyte-platelet- conjugate formation, assaying for platelet closure time under high shear conditions
  • the present invention provides PEGylated monovalent anti-CD40L antibody polypeptides which have increased half-life and preferably also resistance to degradation without a loss in activity (e.g., binding affinity) relative to non- PEGylated antibody polypeptides.
  • Monovalent anti-CD40L antibody polypeptides according to this aspect can be coupled, using methods known in the art to polymer molecules (preferably PEG) useful for achieving the increased half-life and degradation resistance properties encompassed by the present invention.
  • Polymer moieties which can be utilized in the invention can be synthetic or naturally occurring and include, but are not limited to straight or branched chain polyalkylene, polyalkenylene or polyoxyalkylene polymers, or a branched or unbranched polysaccharide such as a homo- or heteropolysaccharide.
  • Preferred examples of synthetic polymers which may be used in the invention include straight or branched chain poly(ethylene glycol) (PEG), poly(propylene glycol), or poly(vinyl alcohol) and derivatives or substituted forms thereof.
  • Particularly preferred substituted polymers useful in the invention include substituted PEG, including methoxy(polyethylene glycol).
  • Naturally occurring polymer moieties which may be used according to the invention in addition to or in place of PEG include lactose, amylose, dextran, or glycogen, as well as derivatives thereof which would be recognized by one of skill in the art.
  • Derivatized forms of polymer molecules of the invention include, for example, derivatives which have additional moieties or reactive groups present therein to permit interaction with amino acid residues of the dAb polypeptides described herein.
  • Such derivatives include N- hydroxylsuccinimide (NHS) active esters, succinimidyl propionate polymers, and sulfhydryl-selective reactive agents such as maleimide, vinyl sulfone, and thiol.
  • NHS N- hydroxylsuccinimide
  • Particularly preferred derivatized polymers include, but are not limited to PEG polymers having the formulae: PEG-O-CH 2 CH 2 CH 2 -CO 2 -NHS; PEG-O-CH 2 -NHS; PEG-O-CH 2 CH 2 -CO 2 -NHS; PEG-S-CH 2 CH 2 -CO-NHS; PEG-O 2 CNH-CH(R)-CO 2 - NHS; PEG-NHCO-CH 2 CH 2 -CO-NHS; and PEG-O-CH 2 -CO 2 -NHS; where R is (CH 2 ) 4 )NHCO 2 (mPEG).
  • PEG polymers useful in the invention may be linear molecules, or may be branched wherein multiple PEG moieties are present in a single polymer.
  • Some particularly preferred PEG derivatives which are useful in the invention include, but are not limited to the following: mPEG-MAL mPEG2-MAL
  • the reactive group (e.g., MAL, NHS, SPA, VS, or Thiol) may be attached directly to the PEG polymer or may be attached to PEG via a linker molecule.
  • the size of polymers useful in the invention can be in the range of between 500 Da to 60 kDa, for example, between 1000 Da and 60 kDa, 10 kDa and 60 kDa, 20 kDa and 60 kDa, 30 kDa and 60 kDa, 40 kDa and 60 kDa, and up to between 50 kDa and 60 kDa.
  • the polymers used in the invention, particularly PEG can be straight chain polymers or can possess a branched conformation.
  • the polymer molecules useful in the invention when attached to a monovalent anti-CD40L antibody polypeptide, will yield a molecule having an average hydrodynamic size of between 24 and 500 kDa.
  • the hydrodynamic size of a polymer molecule used herein refers to the apparent size of a molecule (e.g., a protein molecule) based on the diffusion of the molecule through an aqueous solution. The diffusion, or motion of a protein through solution can be processed to derive an apparent size of the protein, where the size is given by the Stokes radius or hydrodynamic radius of the protein particle.
  • the hydrodynamic size of a protein depends on both mass and shape (conformation), such that two proteins having the same molecular mass may have differing hydrodynamic sizes based on the overall conformation of the protein.
  • the hydrodynamic size of a PEG- linked monovalent anti-CD40L antibody polypeptide e.g., an anti-CD40L single immunoglobulin variable domain as described herein, can be in the range of 24 IdDa to 500 IdDa; 30 to 500 IcDa; 40 to 500 kDa; 50 to 500 kDa; 100 to 500 IvDa; 150 to 500 IdDa; 200 to 500 kDa; 250 to 500 IdDa; 300 to 500 IdDa; 350 to 500 IdDa; 400 to 500 IdDa and 450 to 500 kDa.
  • the hydrodynamic size of a PEGylated antibody polypeptide as described herein is 30 to 40 IdDa; 70 to 80 IdDa or 200 to 300 IdDa.
  • the size of a polymer molecule attached to a monovalent anti-CD40L antibody polypeptide may thus be varied depending upon the desired application. For example, where the PEGylated antibody polypeptide is intended to leave the circulation and enter into peripheral tissues, it is desirable to keep the size of the attached polymer low to facilitate extravazation from the blood stream. Alternatively, where it is desired to have the PEGylated antibody polypeptide remain in the circulation for a longer period of time, a higher molecular weight polymer can be used (e.g., a 30 to 60 kDa polymer).
  • the polymer (PEG) molecules useful in the invention can be attached to antibody polypeptides using methods that are well known in the art.
  • the first step in the attachment of PEG or other polymer moieties to an antibody polypeptide is the substitution of the hydroxyl end-groups of the PEG polymer by electrophile- containing functional groups.
  • PEG polymers are attached to either cysteine or lysine residues present in the antibody polypeptide.
  • the cysteine and lysine residues can be naturally occurring, or can be engineered into the antibody polypeptide molecule.
  • cysteine residues can be recombinantly engineered at the C-terminus of antibody polypeptides, or residues at specific solvent accessible locations in the antibody polypeptide can be substituted with cysteine or lysine.
  • a PEG moiety is attached to a cysteine residue which is present in the hinge region at the C-terminus of an antibody polypeptide.
  • a PEG moiety or other polymer is attached to a cysteine or lysine residue which is either naturally occurring at or engineered into the N-terminus of antibody single variable domain polypeptide of the invention.
  • a PEG moiety or other polymer is attached to an antibody single variable domain according to the invention at a cysteine or lysine residue (either naturally occurring or engineered) which is at least 2 residues away from (e.g., internal to) the C- and/or N-terminus of the antibody single variable domain polypeptide.
  • the PEG polymer(s) is attached to one or more cysteine or lysine residues present in a framework region (FWs) and one or more heterologous CDRs of a single immunoglobulin variable domain.
  • CDRs and framework regions are those regions of an immunoglobulin variable domain as defined in the Kabat database of Sequences of Proteins of Immunological Interest (Kabat et al., 1991, supra).
  • a PEG polymer is linked to a cystine or lysine residue in the VH framework segment DP47, or the V k framework segment DPK9.
  • Cysteine and/or lysine residues of DP47 which may be linked to PEG according to the invention include the cysteine at positions 22, or 96 and the lysine at positions 43, 65, 76, or 98 of SEQ ID NO: 1 ( Figure 5).
  • Cysteine and/or lysine residues of DPK9 which may be linked to PEG according to the invention include the cysteine residues at positions 23, or 88 and the lysine residues at positions 39, 42, 45, 103, or 107 of SEQ ID NO: 3 ( Figure 6).
  • specific cysteine or lysine residues may be linked to PEG in the V H canonical framework region DP38, or DP45.
  • Solvent accessible residues in any given antibody can be determined using methods known in the art such as analysis of the crystal structure of the antibody polypeptide. For example, using the solved crystal structure of the V H dAb HEL4 (SEQ ID NO: 3; a dAb that binds hen egg lysozyme), the residues Gln-13, Pro- 14, GIy- 15, Pro-41, Gly-42, Lys-43, Asp-62,
  • Lys-65, Arg-87, Ala-88, Glu-89, GIn-112, Leu-115, Thr-117, Ser-119, and Ser-120 have been identified as being solvent accessible, and according to the present invention would be attractive candidates for mutation to cysteine or lysine residues for the attachment of a PEG polymer.
  • a PEG polymer is linked to multiple solvent accessible cysteine or lysine residues, or to solvent accessible residues which have been mutated to a cysteine or lysine residue.
  • only one solvent accessible residue is linked to PEG, either where the particular antibody polypeptide only possesses one solvent accessible cysteine or lysine (or residue modified to a cysteine or lysine) or where a particular solvent accessible residue is selected from among several such residues for PEGylation.
  • PEG attachment schemes which are useful in the invention are provided by the company Nektar (SanCarlos, CA).
  • Nektar SanCarlos, CA
  • active esters of PEG polymers which have been derivatized with N-hydroxylsuccinimide, such as succinimidyl propionate may be used.
  • PEG polymers which have been derivatized with sulfhydryl-selective reagents such as maleimide, vinyl sulfone, or thiols may be used.
  • PEG derivatives which may be used according to the invention to generate PEGylated antibodies can be found in the Nektar Catalog (available on the world wide web at nektar.com).
  • Nektar Catalog available on the world wide web at nektar.com.
  • derivitized forms of PEG may be used according to the invention to facilitate attachment of the PEG polymer to an antibody polypeptide.
  • PEG derivatives useful in the invention include, but are not limited to PEG-succinimidyl succinate, urethane linked PEG, PEG phenylcarbonate, PEG succinimidyl carbonate, PEG-carboxymethyl azide, dimethylmaleic anhydride PEG, PEG dithiocarbonate derivatives, PEG-tresylates (2,2,2- trifluoroethanesolfonates), mPEG imidoesters, and other as described in Zalipsky and Lee, (1992) ("Use of functionalized poly(ethylene glycol)s for modification of peptides" in PolvfEthylene Glycol) Chemistry: Biotechnical and Biomedical Applications, J. Milton Harris, Ed., Plenum Press, NY).
  • the invention provides an anti-CD40L antibody single variable domain composition comprising an antibody single variable domain and PEG polymer wherein the ratio of PEG polymer to antibody single variable domain is a molar ratio of at least 0.25:1. In a further embodiment, the molar ratio of PEG polymer to antibody single variable domain is 0.33:1 or greater. In a still further embodiment the molar ratio of PEG polymer to antibody single variable domain is 0.5:1 or greater.
  • the invention also provides dual-specific ligands comprising immunoglobulin single variable domains which each have different specificities; that is, the first and the second epitopes bound by the dual-specific ligand are preferably different.
  • a "dual-specific ligand" refers to 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.
  • Dual-specific ligands may be, or be part of, polypeptides, proteins or nucleic acids, which may be naturally occurring or synthetic.
  • the ligand of the invention may bind an epiotpe or antigen and act as an antagonist or agonist (eg, EPO receptor agonist).
  • the epitope binding domains of the ligand in one embodiment have the same epitope specificity, and may for example simultaneously bind their epitope when multiple copies of the epitope are present on the same antigen.
  • these epitopes are provided on different antigens such that the ligand can bind the epitopes and bridge the antigens.
  • epitopes and antigens may be for instance human or animal proteins, cytokines, cytokine receptors, enzymes co-factors for enzymes or DNA binding proteins.
  • Suitable cytokines and growth factors include but are not limited to: ApoE, Apo-SAA, BDNF 5 Cardiotrophin-1, EGF, EGF receptor, ENA-78, Eotaxin, Eotaxin-2, Exodus-2, EpoR, FGF-acidic, FGF-basic, fibroblast growth factor-10, FLT3 ligand, Fractalkine (CX3C), GDNF, G-CSF, GM-CSF, GF- ⁇ l, insulin, IFN- ⁇ , IGF-I, IGF-II, IL-I ⁇ , IL-l ⁇ , IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL- 8 (72 a.a.), IL-8 (77 a.a.), IL-9, IL-IO, IL-11, IL-12, IL-13, IL-15, IL-16, IL-17, IL-18 (IGIF), Inhibin ⁇ , Inhibin ⁇
  • Cytokine receptors include receptors for the foregoing cytokines, e.g. IL-I Rl; IL-6R; IL-IOR; IL- 18R, as well as receptors for cytokines set forth in Annex 2 or Annex 3 and also receptors disclosed in Annex 2 and 3. It will be appreciated that this list is by no means exhaustive. Where the multispecific ligand binds to two epitopes (on the same or different antigens), the antigen(s) may be selected from this list.
  • variable domains are derived from an antibody directed against the first and/or second antigen or epitope.
  • variable domains are derived from a repertoire of single variable antibody domains.
  • the repertoire is a repertoire that is not created in an animal or a synthetic repertoire.
  • the single variable domains are not isolated (at least in part) by animal immunisation.
  • the single domains can be isolated from a na ⁇ ve library.
  • the invention provides a multi-specific ligand comprising a first epitope binding domain having a first epitope binding specificity and a non- complementary second epitope binding domain having a second epitope binding specificity.
  • the first and second binding specificities may be the same or different.
  • the present invention provides a closed conformation multi-specific ligand comprising a first epitope binding domain having a first epitope binding specificity and a non-complementary second epitope binding domain having a second epitope binding specificity wherein the first and second binding specificities are capable of competing for epitope binding such that the closed conformation multi- specific ligand cannot bind both epitopes simultaneously.
  • the invention provides open conformation ligands comprising non-complementary binding domains, wherein the domains are specific for a different epitope on the same target. Such ligands bind to targets with increased avidity.
  • the invention provides multivalent ligands comprising non- complementary binding domains specific for the same epitope and directed to targets which comprise multiple copies of said epitope.
  • ligands according to the invention can be configured to bind individual epitopes with low affinity, such that binding to individual epitopes is not therapeutically significant; but the increased avidity resulting from binding to two epitopes provides a therapeutic benefit.
  • epitopes may be targeted which are present individually on normal cell types, but present together only on abnormal or diseased cells, such as tumour cells. In such a situation, only the abnormal or diseased cells are effectively targeted by the bispecific ligands according to the invention.
  • Ligand specific for multiple copies of the same epitope, or adjacent epitopes, on the same target may also be trimeric or polymeric
  • ligands comprising three, four or more non-complementary binding domains.
  • ligands may be constructed comprising three or four
  • VH domains or V L domains VH domains or V L domains.
  • ligands are provided which bind to multisubunit targets, wherein each binding domain is specific for a subunit of said target.
  • the ligand may be dimeric, trimeric or polymeric.
  • the invention also includes a dual specific ligand comprising a first immunoglobulin single variable domain having a binding specificity to a first antigen and a second single variable domain having a binding activity to a second antigen, wherein the first antigen is CD40L and the second single variable domain is an Antigen Presenting Cell surface antigen or a T cell surface antigen.
  • the Antigen Presenting Cell (APC) surface antigen can be selected from one of the group consisting of dendritic cell surface antigens, activated macrophage surface antigens, activated B cell surface antigens, co-stimulatory signal pathway surface antigens, and MHC, such as MHC II alpha or beta.
  • the (APC) surface antigen may be selected from the group consisting of CD28, Inducible costimulatory molecule (ICOS), CD27, CD30, OX40, CD45, CD69, CD3, CD70, Inducible costimulatory molecule ligand (ICOSL), OX40L, CD80, CD86, HVEM (Herpes Virus Entry Mediator), and LIGHT, but is preferably one of CD28, Inducible costimulatory molecule (ICOS), CD27, CD30, OX40, CD45, CD69, or CD3.
  • the surface antigen is preferably a B7 gene surface antigen such as B7-2 or B7-1.
  • Dendritic cell surface antigens are known in the art and can include but are not limited to ICAM-I, ICAM-2, LFA-I, LFA-3, DEC205, MHC class I, MHC class II, B7-1, and B7-2.
  • Activated macrophage surface antigens include, but are not limited to, TNF receptor, CD40, MHC class I and II, and B7 molecules.
  • Activated B-cell surface antigens are known in the art (e.g., including but not limited to CD20 and CD86)and further described above (see, for example, Janeway et al. ⁇ 1999, Immunobiologv, Garland Publishing NY, NY).
  • the multi -specific ligands according to the above aspects of the invention are obtainable by the method comprising the steps of: a) selecting a first epitope binding domain by its ability to bind to a first epitope,
  • first epitope binding domain and the second epitope binding domains are non-complementary immunoglobulin variable domains, as herein defined. That is either V H " V H O ⁇ V L -V L variable domains.
  • Chelating dAbs in particular may be prepared according to a preferred aspect of the invention, namely the use of anchor dAbs, in which a library of dimeric, trimeric or multimeric dAbs is constructed using a vector which comprises a constant dAb upstream or downstream of a linker sequence, with a repertoire of second, third and further dAbs being inserted on the other side of the linker.
  • linkers may be avoided, for example by the use of non- covalent bonding or natural affinity between binding domains such as V H and V ⁇ .
  • the invention accordingly provides a method for preparing a multimeric ligand comprising the steps of:
  • the first and second epitopes are adjacent such that a multimeric ligand is capable of binding to both epitopes simultaneously. This provides the ligand with the advantages of increased avidity if binding. Where the epitopes are the same, the increased avidity is obtained by the presence of multiple copies of the epitope on the target, allowing at least two copies to be simultaneously bound in order to obtain the increased avidity effect.
  • At least one epitope binding domain comprises a non- immunoglobulin 'protein scaffold' or 'protein skeleton' as herein defined.
  • Suitable non-immunoglobulin protein scaffolds include but are not limited to any of those selected from the group consisting of: SpA, fibronectin, GroEL and other chaperones, lipocallin, CCTLA4 and affibodies, as set forth above.
  • the epitope binding domains are attached to a 'protein skeleton'.
  • a protein skeleton according to the invention is an immunoglobulin skeleton.
  • the term 'immunoglobulin skeleton' refers to a protein which comprises at least one immunoglobulin fold and which acts as a nucleus for one or more epitope binding domains, as defined herein.
  • 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.
  • Linking of the skeleton to the epitope binding domains may be achieved at the polypeptide level, that is after expression of the nucleic acid encoding the skeleton and/or the epitope binding domains. Alternatively, the linking step may be performed at the nucleic acid level.
  • Methods of linking a protein skeleton according to the present invention, to the one or more epitope binding domains include the use of protein chemistry and/or molecular biology techniques which will be familiar to those skilled in the art and are described herein.
  • the dual- or multispecific ligand may comprise a first domain capable of binding a target molecule, and a second domain capable of binding a molecule or group which extends the half-life of the ligand.
  • the molecule or group may be a bulky agent, such as HSA or a cell matrix protein.
  • the phrase "molecule or group which extends the half-life of a ligand" refers to a molecule or chemical group which, when bound by a dual-specific ligand as described herein increases the in vivo half-life of such dual specific ligand when administered to an animal, relative to a ligand that does not bind that molecule or group.
  • the closed conformation multispecific ligand may be capable of binding the target molecule only on displacement of the half-life enhancing molecule or group.
  • a closed conformation multispecific ligand is maintained in circulation in the bloodstream of a subject by a bulky molecule such as HSA.
  • HSA bulky molecule
  • Ligands according to any aspect of the present invention may advantageously dissociate from their cognate target(s) with a K d of 30OnM to 5pM (ie, 3 x 10 "7 to 5 x 10 "12 M), preferably 5OnM to20pM, or 5nM to 20OpM or InM to 10OpM, 1 x 10 '7 M or less, 1 x
  • the K d rate constand is defined as K 0f f/K OI1 .
  • the invention provides a dAb monomer(or dual specific ligand comprising such a dAb) that binds to serum albumin (SA) with a K d of InM to 500 ⁇ M (ie, x 10 '9 to 5 x 10 "4 ), preferably 10OnM to lO ⁇ M.
  • SA serum albumin
  • the affinity (eg Ka and/or K Off 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 lO ⁇ M
  • the second dAb binds its target with an affinity of lOOpM
  • the serum albumin is human serum albumin (HSA).
  • the first dAb (or a dAb monomer) binds SA (eg, HSA) with a K d of approximately 50, preferably 70, and more preferably 100, 150 or 200 nM.
  • SA eg, HSA
  • the invention moreover provides dimers, trimers and polymers of the aforementioned dAb monomers, in accordance with the foregoing aspect of the present invention.
  • Ligands according to the invention can be linked to an antibody Fc region, comprising one or both of C H 2 and Cjp 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 according to the invention may be free of an Fc domain.
  • the present invention provides one or more nucleic acid molecules encoding at least a dual- or multispecific ligand as herein defined.
  • the ligand is a closed conformation ligand. In another embodiment, it is an open conformation ligand.
  • the multispecific ligand may be encoded on a single nucleic acid molecule; alternatively, each epitope binding domain may be encoded by a separate nucleic acid molecule. Where the ligand is encoded by a single nucleic acid molecule, the domains may be expressed as a fusion polypeptide, or may be separately expressed and subsequently linked together, for example using chemical linking agents. Ligands expressed from separate nucleic acids will be linked together by appropriate means.
  • the nucleic acid may further encode a signal sequence for export of the polypeptides from a host cell upon expression and may be fused with a surface component of a filamentous bacteriophage particle (or other component of a selection display system) upon expression.
  • Leader sequences which may be used in bacterial expression and/or phage or phagemid display, include pelB, stll, ompA, phoA, bla and pelA.
  • the present invention provides a vector comprising nucleic acid according to the present invention.
  • the present invention provides a host cell transfected with a vector according to the present invention.
  • Expression from such a vector may be configured to produce, for example on the surface of a bacteriophage particle, epitope binding domains for selection. This allows selection of displayed domains and thus selection of 'multispecific ligands' using the method of the present invention.
  • 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 VH-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).
  • variable domains of immunoglobulins and in particular antibody V H domains, may be employed as appropriate.
  • dual specific ligands can be in "closed” conformations in solution.
  • a “closed” configuration is that in which the two domains (for example V H and V L ) are present in associated form, such as that of an associated V H -V L pair which fo ⁇ ns an antibody binding site.
  • scFv may be in a closed conformation, depending on the arrangement of the linker used to link the V H and V L domains. If this is sufficiently flexible to allow the domains to associate, or rigidly holds them in the associated position, it is likely that the domains will adopt a closed conformation.
  • V H domain pairs and V L domain pairs may exist in a closed conformation. Generally, this will be a function of close association of the domains, such as by a rigid linker, in the ligand molecule. Ligands in a closed conformation will be unable to bind both the molecule which increases the half-life of the ligand and a second target molecule. Thus, the ligand will typically only bind the second target molecule on dissociation from the molecule which increases the half-life of the ligand.
  • V H /VH, V L /VL or V H /VL dimers without linkers provides for competition between the domains.
  • Ligands according to the invention may moreover be in an open conformation. In such a conformation, the ligands will be able to simultaneously bind both the molecule which increases the half-life of the ligand and the second target molecule.
  • variable domains in an open configuration are (in the case of V H -V L pairs) held far enough apart for the domains not to interact and form an antibody binding site and not to compete for binding to their respective epitopes.
  • VH/V H or V L /V L dimers the domains are not forced together by rigid linkers. Naturally, such domain pairings will not compete for antigen binding or form an antibody binding site.
  • Fab fragments and whole antibodies will exist primarily in the closed conformation, although it will be appreciated that open and closed dual specific ligands are likely to exist in a variety of equilibria under different circumstances. Binding of the ligand to a target is likely to shift the balance of the equilibrium towards the open configuration.
  • certain ligands according to the invention can exist in two conformations in solution, one of which (the open form) can bind two antigens or epitopes independently, whilst the alternative conformation (the closed form) can only bind one antigen or epitope; antigens or epitopes thus compete for binding to the ligand in this conformation.
  • the open form of the dual specific ligand may thus exist in equilibrium with the closed form in solution, it is envisaged that the equilibrium will favor the closed form; moreover, the open form can be sequestered by target binding into a closed conformation.
  • certain dual specific ligands of the invention are present in an equilibrium between two (open and closed) conformations.
  • Dual specific ligands according to the invention may be modified in order to favor an open or closed conformation.
  • stabilisation of V R -V L interactions with disulphide bonds stabilises the closed conformation.
  • linkers used to join the domains, including VR domain and V L domain pairs may be constructed such that the open from is favoured; for example, the linkers may sterically hinder the association of the domains, such as by incorporation of large amino acid residues in opportune locations, or the designing of a suitable rigid structure which will keep the domains physically spaced apart. Characterisation of the dual-specific ligand.
  • binding of the dual-specific ligand to its specific antigens or epitopes can be tested by methods which will be familiar to those skilled in the art and include ELISA. In a preferred embodiment of the invention binding is tested using monoclonal phage ELISA.
  • Phage ELISA may be performed according to any suitable procedure: an exemplary protocol is set forth below.
  • 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. It is also desirable to screen soluble antibody fragments for binding to antigen or epitope, and this can also be undertaken by ELISA using reagents, for example, against a C- or N- terminal tag (see for example Winter et al. (1994) Ann. Rev. Immunology 12, 433-55 and references cited therein.
  • 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.
  • an antibody is herein defined as an antibody (for example IgG, IgM, IgA, IgA, IgE) or fragment (Fab, Fv, disulphide linked Fv, scFv, diabody) which comprises at least one heavy and a light chain variable domain, at least two heavy chain variable domains or at least two light chain variable domains. It may be at least partly 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).
  • the dual-specific ligand comprises at least one single heavy chain variable domain of an antibody and one single light chain variable domain of an antibody, or two single heavy or light chain variable domains.
  • the ligand may comprise a V H /VL pair, a pair of VH domains or a pair of V L domains.
  • the first and the second variable domains of such a ligand may be on the same polypeptide chain. Alternatively they may be on separate polypeptide chains. In the case that they are on the same polypeptide chain they may be linked by a linker, which is preferentially a peptide sequence, as described above.
  • the first and second variable domains may be covalently or non-covalently associated.
  • the covalent bonds may be disulphide bonds.
  • 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.
  • variable domains are 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.
  • the 'dual-specific ligand' is a single chain Fv fragment. In an alternative embodiment of the invention, the 'dual-specific ligand' consists of a Fab format. In a further aspect, the present invention provides nucleic acid encoding at least a 'dual-specific ligand' as herein defined.
  • both antigens or epitopes may bind simultaneously to the same antibody molecule. Alternatively, they may compete for binding to the same antibody molecule. For example, where both epitopes are bound simultaneously, both variable domains of a dual specific ligand are able to independently bind their target epitopes.
  • the one variable domain is capable of binding its target, but not at the same time as the other variable domain binds its cognate target; or the first variable domain is capable of binding its target, but not at the same time as the second variable domain binds its cognate target.
  • variable regions may be derived from antibodies directed against target antigens or epitopes. Alternatively they may be derived from a repertoire of single antibody domains such as those expressed on the surface of filamentous bacteriophage. Selection may be performed as described below.
  • nucleic acid molecules and vector constructs required for the performance of the present invention may be constructed and manipulated as set forth in standard laboratory manuals, such as Sambrook et a (1989) Molecular Cloning: A Laboratory Manual, Cold Spring Harbor, USA.
  • nucleic acids useful in the present invention is typically carried out in recombinant vectors.
  • the present invention provides a vector comprising nucleic acid encoding at least a 'dual-specific ligand' as herein defined.
  • 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 "polylinker") 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. Since the replication of vectors encoding a ligand according to the present invention is most conveniently performed in E. coli, an E.
  • co//-selectable marker for example, the ⁇ -lactamase gene that confers resistance to the antibiotic ampicillin, is of use.
  • E. coli plasmids such as pBR.322 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 As described above, the preferred
  • 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. origin of replication (for double stranded replication) and also- a phage origin of replication (for production of single-stranded DNA). The manipulation and expression of such vectors is well known in the art (Hoogenboom and Winter (1992) supra; Nissim et al. (1994) supra).
  • 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, KNfA or protein, in situ hybridisation, immunocytochernistry or sequence analysis of nucleic acid or protein molecules. Those skilled in the art will readily envisage how these methods may be modified, if desired.
  • two or more non-complementary epitope binding domains are linked so that they are in a closed conformation as herein defined.
  • they may be further attached to a skeleton which may, as a alternative, or on addition to a linker described herein, facilitate the formation and/or maintenance of the closed conformation of the epitope binding sites with respect to one another.
  • the monomeric anti-CD40L antibody single variable domain polypeptides of the invention may be constructed using scaffold or skeleton frameworks as discussed herein.
  • 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 5 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 in addition to those based on immunoglobulin domains, may also be based on protein scaffolds or skeletons other than immunoglobulin domains.
  • natural bacterial receptors such as SpA have 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 (Aff ⁇ body, Bromma, Sweeden).
  • Suitable, scaffolds include lipocallin and CTLA4, as described in van den Beuken et al, J. MoI. Biol. (2001) 310, 591-601, and scaffolds such as those described in WO0069907 (Medical Research Council), which are based for example on the ring structure of bacterial GroEL or other chaperone polypeptides.
  • Other non-immunoglobulin based scaffolds which may be used according to the invention include those based on the LDL receptor class A, EGF domain monomers and mutimers, and scaffolds available from Biorexis (King of Prussia, PA) or Avidia (Mountain View, CA).
  • Other non-immunoglobulin scaffolds which may be used are described, for example, in WO05/040229, WO04/044011, and US20050089932
  • 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 crystallographic 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).
  • the ligands of the present invention are advantageously selected and/or assembled from libraries of domains, such as libraries of VH domains and/or libraries of V L domains. Moreover, the ligands of the invention may themselves be provided in the form of libraries.
  • libraries of ligands and/or domains are 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 chose residues for diversification which do not affect the canonical structure.
  • lt 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.
  • Vj 1 domain encodes a different range of canonical structures for the Ll, L2 and L3 loops and that V ⁇ and Yx domains can pair with any V R 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 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.
  • 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. It is more preferable, that 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.
  • the incidence of the different main- chain conformations for each of the antigen binding loops may be considered separately.
  • 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%.
  • Hl - CS 1 (79% of the expressed repertoire), H2 - CS 3 (46%), Ll - CS 2 of V ⁇ (39%), L2 - CS 1 (100%),
  • V H segment 3-23 DP- 47
  • J H segment JH4b the V ⁇ segment 02/012
  • J ⁇ segment J ⁇ l the J ⁇ segment J ⁇ l.
  • 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.
  • ligands according to the invention or 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. Sc?. 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. Sci. USA, 89: 4457; Nissim et al.
  • the binding site for the target is most often the antigen binding site.
  • the invention provides libraries of or for the assembly of antibody ligands in which only those 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. For example, in L2 it is known that positions 50 and 53 are diverse in naturally occurring antibodies and are observed to make contact with the antigen. In contrast, 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. This represents a significant improvement in terms of the functional diversity required to create a range of antigen binding specificities.
  • 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 hypermutation spreads diversity to regions at the periphery of the antigen binding site that are highly conserved in the primary repertoire (see Tomlinson et a (1996) J MoI. Biol, 256: 813).
  • This complementarity has probably evolved as an efficient strategy for searching sequence space and, although apparently unique to antibodies, it can easily be applied to other polypeptide repertoires.
  • the residues which are varied are a subset of those that form the binding site for the target. Different (including overlapping) subsets of residues in the target binding site are diversified at different stages during selection, if desired.
  • an initial 'naive' repertoire is 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-determined 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.
  • the invention provides two different naive repertoires of binding domains for the construction of ligands, or a naive library of ligands, in which some or all of the residues in the antigen binding site are varied.
  • 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, Wl, H98, L50, L53, L91, L92, L93, L94 and L96.
  • residues that are diversified during the recombination process include, but are not limited to: H31, H33. H35, H95, H96, H97, H98, L30, L31, L32, L34 and L96. All the residues listed above as suitable for diversification in these libraries are known to make contacts in one or more antibody-antigen complexes. Since in both libraries, not all of the residues in the antigen binding site are varied, additional diversity is incorporated during selection by varying the remaining residues, if it is desired to do so. It shall be apparent to one skilled in the art that any subset of any of these residues (or additional residues which comprise the antigen binding site) can be used for the initial and/or subsequent diversification of the antigen binding site.
  • 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 VH, V K and Vx 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%).
  • the codons (AGT)(AGC)T 5 (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.
  • the PEGylated monovalent anti-CD40L antibodies as described herein confer a distinct advantage over non-PEGylated antibody polypeptides, in that the
  • PEGylated antibody molecules will have a greatly prolonged in vivo half life.
  • the increased half-life of the molecules described herein is conferred by the increased hydrodynamic size of the antibody polypeptide resulting from the attachment of PEG polymer(s). More specifically, it is believed that two parameters play an important role in determining the serum half-life of PEGylated antibody polypeptides.
  • the first criterion is the nature and size of the PEG attachment, i.e., if the polymer used is simply a linear chain or a branched/forked chain, wherein the branched/forked chain gives rise to a longer half-life.
  • the second is the location of the PEG moiety or moieties on the antibody polypeptide in the final format and how many "free" unmodified PEG arms the molecule has.
  • the resulting hydrodynamic size of the PEGyI ated antibody polypeptide reflects the serum half-life of the molecule. Accordingly, the larger the hydrodynamic size of the PEGylated molecule, the greater the serum half life.
  • a monovalent anti-CD40L antibody polypeptide as described herein is stabilized in vivo by fusion with a moiety, such as PEG, that increases the hydrodynamic size of the antibody polypeptide.
  • a moiety such as PEG
  • Methods for pharmacokinetic anafysis and determination of half-life will be familiar to those skilled in the art. Details may be found in Kenneth et al: Chemical Stability of Pharmaceuticals: A Handbook for Pharmacists and in Peters et al, Pharmacokinetc analysis: A Practical Approach (1996). Reference is also made to "Pharmacokinetics", M Gibaldi & D Perron, published by Marcel Dekker, 2 nd Rev. ex edition (1982), which describes pharmacokinetic parameters such as t alpha and t beta half lives and area under the curve (AUC).
  • the half life of a PEGylated antibody polypeptide as described herein is increased by 10%, 20%, 30%, 40%, 50% or more relative to a non- PEGylated dAb (wherein the antibody polypeptide of the PEGylated antibody polypeptide and non-PEGylated antibody polypeptide are the same).
  • Increases in the range of 2x, 3x, 4x, 5x, 7x, 1Ox, 20x, 3Ox, 4Ox, and up to 5Ox or more of the half life are possible.
  • increases in the range of up to 30x, 40x, 5Ox, 6Ox, 7Ox, 8Ox, 9Ox, 10Ox, 150x of the half life are possible.
  • Half lives (tl4 alpha and t 1 /. beta) and AUC can be determined from a curve of serum concentration of ligand against time.
  • the WiriNonlin analysis package (available from Pharsight Corp., Mountain View, CA 94040, USA) can be used, for example, to model the curve.
  • a first phase the alpha phase
  • a second phase (beta phase) is the terminal phase when the ligand has been distributed and the serum concentration is decreasing as the ligand is cleared from the patient.
  • the t ⁇ half life is the half life of the first phase and the t ⁇ half life is the half life of the second phase.
  • a dAb-containing composition e.g., a dAb-effector group composition, having a t ⁇ half-life in the range of 0.25 hours to 6 hours or more.
  • the lower end of the range is 30 minutes, 45 minutes, 1 hour, 1.3 hours, 2 hours, 3 hours, 4 hours, 5 hours, 6 hours, 7 hours, 10 hours, 11 hours or 12 hours.
  • a dAb containing composition will have a t ⁇ half- life in the range of up to and including 12 hours.
  • the upper end of the range is 11, 10, 9, 8, 7, 6, or 5 hours.
  • An example of a suitable range is 1.3 to 6 hours, 2 to 5 hours or 3 to 4 hours.
  • the present invention provides a dAb containing composition comprising a ligand according to the invention having a t ⁇ half-life in the range of 1 - 170 hours or more.
  • the lower end of the range is 2.5 hours, 3 hours, 4 hours, 5 hours, 6 hours, 7 hours, 8 hours, 9 hours, 10 hours, 11 hours or 12 hours.
  • a dAb containing composition e.g. a dAb-effector group composition has a t ⁇ half-life in the range of up to and including 21 days.
  • the upper end of the range is 12 hours, 24 hours, 2 days, 3 days, 5 days, 10 days, 15 days, or 20 days.
  • a dAb containing composition according to the invention will have a t ⁇ half-life in the range 2-100 hours, 4-80 hours, and 10-40 hours. In a further embodiment, it will be in the range 12-48 hours.
  • the present invention provides a dAb containing composition comprising a ligand according to the invention having a half-life in the range of 1-170 hours or more.
  • the lower end of the range is 1.3 hours, 2.5 hours, 3 hours, 4 hours, 5 hours, 6 hours, 7 hours, 8 hours, 9 hours, 10 hours, 11 hours or 12 hours.
  • a dAb containing composition e.g. a dAb-effector group composition has a half-life in the range of up to and including 21 days.
  • the upper end of the range is 12 hours, 24 hours, 2 days, 3 days, 5 days, 10 days, 15 days, or 20 days.
  • the present invention provides a dAb containing composition
  • a dAb containing composition comprising a ligand according to the invention having an AUC value (area under the curve) in the range of 1 mg.min/ml or more.
  • the lower end of the range is 5, 10, 15, 20, 30, 100, 200 or 300 mg.min/ml.
  • a ligand or composition according to the invention has an AUC in the range of up to 600 mg.min/ml.
  • the upper end of the range is 500, 400, 300, 200, 150, 100, 75 or 50 mg.min/ml.
  • a ligand according to the invention will have an AUC in the range selected from the group consisting of the following: 15 to 150 mg.min/ml, 15 to 100 mg.min/ml, 15 to 75 mg.min/ml, and 15 to 50 mg.min/ml.
  • the ligands according to the invention are capable of binding to one or more molecules which can increase the half-life of the ligand in vivo.
  • molecules are polypeptides which occur naturally in vivo and which resist degradation or removal by endogenous mechanisms which remove unwanted material from the organism.
  • the molecule which increases the half-life of the organism may be selected from the following:
  • Proteins from the extracellular matrix for example collagen, 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, eg type 1 collagen (accounting for 90% of body collagen) found in bone, skin, tendon, ligaments, cornea, internal organs or type II collagen found in cartilage, invertebral disc, notochord, vitreous humour of the eye.
  • Proteins found in blood including:
  • Plasma proteins such as fibrin, ⁇ -2 macroglobulin, serum albumin, fibrinogen A, fibrinogen B, serum amyloid protein A, heptaglobin, profilin, ubiquitin, uteroglobulin and ⁇ -2-microglobulin;
  • Enzymes and inhibitors such as plasminogen, lysozyme, cystatin C, alpha- 1- antitrypsin and pancreatic trypsin inhibitor.
  • Plasminogen is the inactive precursor of the trypsin-like serine protease plasmin. It is normally found circulating through the blood stream. When plasminogen becomes activated and is converted to plasmin, it unfolds a potent enzymatic domain that dissolves the fibrinogen fibers that entgangle the blood cells in a blood clot. This is called fibrinolysis.
  • Immune system proteins such as IgE, IgG, IgM.
  • Transport proteins such as retinol binding protein, ⁇ -1 microglobulin.
  • Defensins such as beta-defensin 1, Neutrophil defensins 1,2 and 3.
  • Proteins found at the blood brain barrier or in neural tissues such as melanocortin receptor, myelin, ascorbate transporter.
  • Transferrin receptor specific ligand-neuropharmaceutical agent fusion proteins see US5977307; brain capillary endothelial cell receptor, transferrin, transferrin receptor, insulin, insulin-like growth factor 1 (IGF 1) receptor, insulin-like growth factor 2 (IGF 2) receptor, insulin receptor. Proteins localised to the kidney, such as polycystin, type IV collagen, organic anion transporter Kl, Heymann's antigen.
  • Proteins localised to the liver for example alcohol dehydrogenase, G250.
  • Proteins localised to the lung such as secretory component (binds IgA).
  • HSP 27 Proteins localised to the Heart, for example HSP 27. This is associated with dilated cardiomyopathy.
  • Proteins localised to the skin for example keratin.
  • Bone specific proteins such as bone morphogenic proteins (BMPs), which are a subset of the transforming growth factor ⁇ superfamily that demonstrate osteogenic activity. Examples include BMP-2, -4, -5, -6, -7 (also referred to as osteogenic protein (OP-I) and -8 (OP-2).
  • Tumour specific proteins including human trophoblast antigen, herceptin receptor, oestrogen receptor, cathepsins eg cathepsin B (found in liver and spleen).
  • Disease-specific proteins such as antigens expressed only on activated T-cells: including
  • LAG-3 lymphocyte activation gene
  • osteoprotegerin ligand OPGL
  • OX40 a member of the TNF receptor family, expressed on activated T cells and the only costimulatory T cell molecule known to be specifically up-regulated in human T cell leukaemia virus type-I (HTLV-I)-producing cells.
  • FGF-I acidic fibroblast growth factor
  • FGF-2 basic fibroblast growth factor
  • 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) only occurs when as a result of trauma, disease or injury and therefore in vivo, extracellular HSPs trigger a response from the immune system that will fight infection and disease.
  • a dual specific which binds to extracellular HSP can be localised to a disease site.
  • Brambell receptor also known as FcRB
  • This Fc receptor has two functions, both of which are potentially useful for delivery
  • Ligands according to the invention may designed to be specific for the above targets without requiring any increase in or increasing half life m vivo.
  • ligands according to the invention can be specific for targets selected from the foregoing which are tissue-specific, thereby enabling tissue-specific targeting of the dual specific ligand, or a dAb monomer that binds a tissue-specific therapeutically relevant target, irrespective of any increase in half-life, although this may result.
  • the ligand or dAb monomer targets kidney or liver, this may redirect the ligand or dAb monomer to an alternative clearance pathway in vivo (for example, the ligand may be directed away from liver clearance to kidney clearance).
  • Polypeptides useful for increasing half-life include, but are not limited to those shown in Annex I.
  • a further advantage of the present invention is that the PEGylated dAbs and dAb multimers described herein possess increased stability to the action of proteases.
  • dAbs are generally intrinsically stable to the action of proteases.
  • pepsin In the presence of pepsin, however, many dAbs are totally degraded at pH 2 because the protein is unfolded under the acid conditions, thus making the protein more accessible to the protease enzyme.
  • the present invention provides PEGylated dAb molecules, including dAb multimers, wherein it is believed that the PEG polymer provides protection of the polypeptide backbone due the physical coverage of the backbone by the PEG polymer, thereby preventing the protease from gaining access to the polypeptide backbone and cleaving it.
  • a PEGylated dAb having a higher hydrodynamic size e.g., 200 to 500 kDa
  • the larger hydrodynamic size will confirm a greater level of protection from protease degradation than a PEGylated dAb having a lower hydrodynamic size.
  • a PEG- or other polymer-linked antibody single variable domain monomer or multimer is degraded by no more than 10% when exposed to one or more of pepsin, trypsin, elastase, chymotrypsin, or carboxypeptidase, wherein if the protease is pepsin then exposure is carried out at pH 2.0 for 30 minutes, and if the protease is one or more of trypsin, elastase, chymotrypsin, or carboxypeptidase, then exposure is carried out at pH 8.0 for 30 minutes.
  • a PEG- or other polymer-linked dAb monomer or multimer is degraded by no more than 10% when exposed to pepsin at pH 2.0 for 30 minutes, preferably no more than 5%, and preferably not degraded at all.
  • a PEG- or other polymer-linked dAb multimer e.g., hetero- or homodimer, trimer, tetramer, octamer, etc.
  • a PEG- or other polymer-linked dAb multimer e.g., hetero- or homodimer, trimer, tetramer, octamer, etc.
  • a PEG- or other polymer-linked dAb monomer or multimer is degraded by no more than 10% when exposed to trypsin, elastase, chymotrypsin, or carboxypeptidase at pH 8.0 for 30 minutes, preferably no more than 5%, and preferably not degraded at all.
  • a PEG- or other polymer-linked dAb multimer e.g., hetero- or homodimer, trimer, tetramer, octamer, etc.
  • a PEG- or other polymer-linked dAb multimer e.g., hetero- or homodimer, trimer, tetramer, octamer, etc.
  • the relative ratios of protease rantibody single variable domain polypeptide may be altered according to the invention to achieve the desired level of degradation as described above.
  • the ratio or protease to antibody single variable domain may be from about 1:30, to about 10:40, to about 20:50, to about 30:50, about 40:50, about 50:50, about 50:40, about 50:30, about 50:20, about 50:10, about 50: 1, about 40:1, and about 30:1.
  • the present invention provides a method for decreasing the degradation of an antibody single variable domain comprising linking an antibody single variable domain monomer or multimer to a PEG polymer according to any of the embodiments described herein.
  • the antibody single variable domain is degraded by no more than 10% in the presence of pepsin at pH2.0 for 30 minutes.
  • a PEG-linked dAb multimer is degraded by no more than 5%, and preferably not degraded at all in the presence of pepsin at pH 2.0 for 30 minutes.
  • the antibody single variable domain is degraded by no more than 10% when exposed to trypsin, elastase, chymotrypsin, or carboxypeptidase at pH 8.0 for 30 minutes, preferably no more than 5%, and preferably not degraded at all.
  • Degradation of PEG-linked dAb monomers and multimers according to the invention may be measured using methods which are well known to those of skill in the art. For example, following incubation of a PEG-linked dAb with pepsin at pH 2.0 for 30 minutes, or with trypsin, elastase, chymotrypsin, or carboxypeptidase at pH 8.0 for 30 minutes, the dAb samples may be analyzed by gel filtration, wherein degradation of the dAb monomer or multimer is evidenced by a gel band of a smaller molecular weight than an un-degraded (i.e., control dAb not treated with pepsin, trypsin, chymotrypsin, elastase, or carboxypeptidase) dAb.
  • an un-degraded i.e., control dAb not treated with pepsin, trypsin, chymotrypsin, elastase, or carboxypeptida
  • Molecular weight of the dAb bands on the gel may be determined by comparing the migration of the band with the migration of a molecular weight ladder (see Figure 5). Other methods of measuring protein degradation are known in the art and may be adapted to evaluate the PEG-linked dAb monomers and multimers of the present invention.
  • Pharmaceutical Compositions Dosage and Administration
  • the antibody polypeptides of the invention can be incorporated into pharmaceutical compositions suitable for administration to a subject.
  • the pharmaceutical composition comprises a monovalent anti-CD40L antibody polypeptide and a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable carrier includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like that are physiologically compatible.
  • pharmaceutically acceptable carrier excludes tissue culture medium comprising bovine or horse serum.
  • pharmaceutically acceptable carriers include one or more of water, saline, phosphate buffered saline, dextrose, glycerol, ethanol and the like, as well as combinations thereof.
  • compositions as described herein may be in a variety of forms. These include, for example, liquid, semi-solid and solid dosage forms, such as liquid solutions (e.g., injectable and infusible solutions), dispersions or suspensions, tablets, pills, powders, liposomes and suppositories. The preferred form depends on the intended mode of administration and therapeutic application.
  • Typical preferred compositions are in the form of injectable or infusible solutions, such as compositions similar to those used for passive immunization of humans with other antibodies.
  • the preferred mode of administration is parenteral (e.g., intravenous, subcutaneous, intraperitoneal, intramuscular).
  • compositions typically must be sterile and stable under the conditions of manufacture and storage.
  • the composition can be formulated as a solution, microemulsion, dispersion, liposome, or other ordered structure suitable to high drug concentration.
  • Sterile injectable solutions can be prepared by incorporating the active compound in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filter sterilization.
  • dispersions are prepared by incorporating the active compound into a sterile vehicle that contains a basic dispersion medium and the required other ingredients from those enumerated above.
  • the preferred methods of preparation are vacuum drying and freeze-drying that yields a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • the proper fluidity of a solution can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
  • the antibody polypeptides described herein can be administered by a variety of methods known in the art, although for many therapeutic applications, the preferred route/mode of administration is intravenous injection or infusion.
  • the polypeptide can also be administered by intramuscular or subcutaneous injection.
  • the route and/or mode of administration will vary depending upon the desired results.
  • the active compound may be prepared with a carrier that will protect the compound against rapid release, such as a controlled release formulation, including implants, transdermal patches, and microencapsulated delivery systems.
  • Single immunoglobulin variable domains and other relatively small monovalent antibody polypeptides are well suited for formulation as extended release preparations due, in part, to their small size - the number of moles per dose can be significantly higher than the dosage of, for example, full sized antibodies.
  • Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid.
  • Prolonged absorption of injectable compositions can be brought about by including in the composition an agent that delays absorption, for example, monostearate salts and gelatin. Many methods for the preparation of such formulations are patented or generally known to those skilled in the art.
  • a monovalent anti-CD40L antibody polypeptide can be orally administered, for example, with an inert diluent or an assimilable edible carrier.
  • the compound (and other ingredients, if desired) may also be enclosed in a hard or soft shell gelatin capsule, compressed into tablets, or incorporated direct! ⁇ ' into the individual's diet
  • the compounds may be incorporated with excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, and the like.
  • a monovalent anti-CD40L antibody polypeptide is coformulated with and/or coadministered with one or more additional therapeutic agents.
  • a monovalent anti-CD40L antibody polypeptide can be coformulated and/or coadministered with one or more additional antibodies that bind other targets (e.g., antibodies that bind other cytokines or that bind cell surface molecules), or, for example, one or more cytokines.
  • Such combination therapies may utilize lower dosages of the administered therapeutic agents, thus avoiding possible toxicities or complications associated with the various monotherapies.
  • compositions of the invention can include a
  • therapeutically effective amount or a “prophylactically effective amount” of a monovalent anti-CD40L antibody polypeptide.
  • a “therapeutically effective amount” refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic result.
  • a therapeutically effective amount of the antibody polypeptide can vary according to factors such as the disease state, age, sex, and weight of the individual, and the ability of the monovalent anti-CD40L antibody polypeptide to elicit a desired response in the individual.
  • a therapeutically effective amount is also one in ⁇ vhich any toxic or detrimental effects of the antibody or antibody portion are outweighed by the therapeutically beneficial effects.
  • prophylactically effective amount refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired prophylactic result. Typically, because a prophylactic dose is used in subjects prior to or at an earlier stage of disease, the prophylactically effective amount will be less than the therapeutically effective amount.
  • Dosage regimens may be adjusted to provide the optimum desired response
  • Dosage unit form refers to physically discrete units suited as unitary dosages for the mammalian subjects to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier.
  • a non-limiting range for a therapeutically or prophylactically effective amount of a monovalent anti-CD40L antibody polypeptide is 0.1-20 mg/kg, more preferably 1-10 mg/kg. It is to be noted that dosage values can vary with the type and severity of the condition to be alleviated. It is to be further understood that for any particular subject, specific dosage regimens should be adjusted over time according to the individual need and the professional judgment of the administering clinician.
  • the efficacy of treatment with a monovalent anti-CD40L antibody polypeptide as described herein is judged by the skilled clinician on the basis of improvement in one or more symptoms or indicators of the disease state or disorder being treated.
  • An improvement of at least 10% (increase or decrease, depending upon the indicator being measured) in one or more clinical indicators is considered "effective treatment," although greater improvements are preferred, such as 20%, 30%, 40%, 50%, 75%, 90%, or even 100%, or, depending upon the indicator being measured, more than 100% (e.g., two-fold, three-fold, ten-fold, etc., up to and including attainment of a disease-free state.
  • Indicators can be physical measurements, e.g., enzyme, cytokine, growth factor or metabolite levels, rate of cell growth or cell death, or the presence or amount of abnormal cells.
  • One can also measure, for example, differences in the amount of time between flare-ups of symptoms of the disease or disorder (e.g., for remitting/relapsing diseases, such as multiple sclerosis).
  • non-physical measurements such as a reported reduction in pain or discomfort or other indicator of disease status can be relied upon to gauge the effectiveness of treatment.
  • CDAI Crohn's Disease Activity Index
  • prophylaxis performed using a composition as described herein is “effective” if the onset or severity of one or more symptoms is delayed or reduced by at least 10%, or abolished, relative to such symptoms in a similar individual (human or animal model) not treated with the composition.
  • the monovalent anti-CD40L antibody polypeptides described herein must bind human CD40L, where one is to evaluate its effect in an animal model system, the polypeptide must cross-react with one or more antigens in the animal model system, preferably at high affinity.
  • One of skill in the art can readily determine if this condition is satisfied for a given animal model system and a given monovalent anti-CD40L antibody polypeptide. If this condition is satisfied, the efficacy of the monovalent anti-CD40L antibody polypeptide can be examined by administering it to an animal model under conditions which mimic a disease state and monitoring one or more indicators of that disease state for at least a 10% improvement.
  • Monovalent anti-CD40L antibody polypeptides as described herein are useful for the treatment of autoimmune disorders in which CD40/CD40L signaling is inappropriately active. There are several animal models in which the therapeutic efficacy of a given monovalent anti-CD40L antibody polypeptide can be assessed, as discussed below.
  • SLE Systemic Lupus Erythematosis
  • Anti-CD40L antibody treatment prevents the development of lupus-like nephritis in NZB/NZW and SNFl SLE mice.
  • Treatment of SNFl mice with anti- CD40L antibody reverses established nephritis and preserves kidney function. See, e.g., Mohan et al., 1995, J. Immunol. 154: 1470-1480; Early et al., 1996, J. Immunol.
  • Anti-CD40L blocks the development of joint inflammation, serum antibody titers to collagen, the infiltration of inflammatory cells into the synovial tissue, ant the erosion of cartilage and bone in collagen-induced arthritis. See, e.g., Durie et al., 1993, Science 261: 132; and Chess, 2001, supra.
  • the non-obese diabetic (NOD) mouse spontaneously develops T cell dependent autoimmune diabetes.
  • Anti-CD40L prevents the development of renal rejection of fully allogeneic grafts in mice. Moreover, the survival of renal allografts transplanted into nephrectomized rehsus monkeys is typically prolonged by anti-CD40L therapy alone.
  • anti CD40L therapy has prevented graft rejection of skin, islet cells and cardiac transplants as well as GVHD in rodents. See, e.g., Kirk et al., 1997, Proc. Natl. Acad. Sci. U.S.A. 94: 8789-8794; Parker et al., 1995, Proc. Natl. Acad. Sci. U.S.A. 92: 9560; Larsen et al., 1996, Transplantation 61 : 4; and Chess, 2001, supra.
  • Anti-CD40L antibody polypeptides as described herein are useful for the treatment or prevention of diseases or disorders in which inappropriate activation of a CD40L/CD40-mediated pathway is involved.
  • autoimmune diseases frequently involve inappropriate regulation or activity of CD40L/CD40 pathways.
  • Administration of an anti-CD40L antibody polypeptide as described herein to an individual suffering from such a disease, can reduce one or more symptoms of the disease.
  • Non-limiting examples of diseases for which the antibody polypeptides described herein can be therapeutically useful include Systemic Lupus Erythematosus (SLE), Idiotypic Thrombocytopenic Purpura (ITP), transplant rejection, Crohn's Disease, Inflammatory Bowel Disease (IBD), colitis, asthma/allergy, atherosclerosis, Myasthenia Gravis, immune response to recombinant drug products, e.g., factor VII in hemophilia, Multiple Sclerosis, Psoriasis, Rheumatoid Arthritis, Ankylosing Spondylitis, Coronary Heart Disease, and Diabetes, including Type 1 and/or Type 2 Diabetes.
  • SLE Systemic Lupus Erythematosus
  • IDP Idiotypic Thrombocytopenic Purpura
  • IBD Inflammatory Bowel Disease
  • colitis asthma/allergy
  • atherosclerosis e.g., atherosclerosis
  • Myasthenia Gravis
  • the anti-CD40L antibody polypeptides described herein are additionally useful in the way that generally any antibody preparation is useful, e.g., for in vivo imaging or diagnostic uses, in vitro diagnostic uses, etc. For these and other uses it may be desirable to. label the anti-CD40L antibody polypeptides, e.g i5 with a fluorescent, colorimetric, enzymatic or radioactive label. Methods of labeling antibody polypeptides are well known in the art.
  • Biotinylation of CD40L was performed by incubating CD40L (0.5mg/ml) with EZ-LinkTM Sulfo-NHS-LC-Biotin [Sulfosuccinimidyl-6-(biotinamido)hexanoate] (Pierce) at a molar ratio of 5:1 on ice for 2 hours according to the product instructions.
  • the biotinylation reaction mixture was then dialysed against 3 exchanges of PBS (100Ox sample volume) in a Slide-A-Lyzer ® Dialysis Cassette at 4 0 C to remove the unincorporated biotinylation reagent.
  • biotinylated-CD40L was tested by receptor binding assay for binding to CD40/Fc to confirm its biological activity. Quality of biotin-CD40L was also monitored by analysing on a NuPaGE 4-12% Bis-Tris gel and detected by Simply Blue Safe-Stain (Invitrogen) ( Figure Ia), and western-blotting by probing with Streptavidin-HRP ( Figure Ib). The biotinylated-CD40L was further analysed by mass spectrometry with the majority of CD40L subunits containing 1 or 2 biotin moieties (data not shown).
  • the Domain Antibody (dAb) libraries are based on a single human framework for the V H (DP47 and JH4b) and for the VK (DPK9 and JKl) with side chain diversity incorporated at positions in the antigen binding site that make contact with antigen in known molecular structures and mirror residues diversified in the human antibody repertoire.
  • the antibodies are displayed as fusion proteins covalently linked to the N - terminus of the Fd -phage protein pill, using the phage vector pDOM4 (Fd- Tet) with encodes the Fd phage genome with dAb expression under the control of the gene-HI promoter.
  • the dAb cassette consists of (5' to 3'): eukaryotic leader sequence, dAb, myc tag, gill.
  • the vector contains both the Ml 3 and colEl origins of replication and is selectable using tetracycline.
  • the V H and V ⁇ libraries each have a calculated size of over 1x10 10 molecules. Reagents, equipment and sources from which they are available are provided in Table 2.
  • the antigen concentration used for the first round of selection was 60 nM; the antigen concentration was decreased to 6 nM for round 2, and to 0.6 nM for round 3.
  • Biotech were prepared by washing once in 1 ml of PBS containing 0.1% Tween-20 followed by a second wash in 1 ml of PBS. The beads were then blocked in 1 ml of PBS containing 2% MarvelTM in a 2 ml eppendorf tube at room temperature on a rotating wheel for 1 h.
  • the tube containing the blocked streptavi din -coated magnetic beads was placed into a magnetic holder, enabling capture of the magnetic beads.
  • the supernatant was removed and the beads resuspended in the antigen/phage mix. This mixture was rotated for 10 min to allow for bead capture of phage/antigen complexes.
  • the beads were captured using a magnetic holder and repeatedly washed 19 times using 1 ml of PBS containing 0.1% Tween-20, followed by a final wash of 1 ml PBS.
  • the eppendorf tubes were changed following washing steps 3, 9, 15 and 19 to minimise background phage carryover.
  • the washed beads were then recaptured and all washing solution removed.
  • the phage were eluted through resuspension in 500 ⁇ l of trypsin solution (50 ⁇ l of 10 mg/ml trypsin stock solution added to 450 ⁇ l PBS, freshly diluted) and rotated for 10 min at room temperature.
  • the eluted phage were recovered by capturing the beads using the magnetic holder and the liquid containing the eluted phage recovered.
  • the eluted phage were used to infect E. coli TGl to prepare phage for a further round of selection.
  • the eluted phage (250 ⁇ l) were mixed with 1.75 ml of log phase E. coli TGl (OD 6O o between 0.3 and 0.6) and infection allowed to occur for 30 min at 37°C without shaking.
  • the infected E. coli TGl culture was centrifuged at 11,600 g in a micro centrifuge for 1 min at room temperature.
  • the pelleted bacteria were resuspended in 100 ⁇ l of 2xTY and plated on regular 9 cm diameter plates containing TYE supplemented with 15 ⁇ g/ml tetracycline. Plates were grown at 37°C overnight.
  • the 50 ml culture was grown at 37°C for 16 to 24 hours with shaking at 250 rpm.
  • the culture was centrifuged at 3,300 g for 15 min to pellet the bacteria.
  • the phage were then precipitated from the supernatant through the addition of 10 ml of PEG/NaCl to 40 ml of clarified supernatant.
  • the phage/PEG solution was mixed and incubated on ice for at least 1 h.
  • To pellet the phage the solution was centrifuged at 3,300 g for 30 min at 4 0 C. The supernatant was decanted and any remaining supernatant removed by aspiration.
  • the resulting phage pellet was resuspended in 2 ml PBS and centrifuged at
  • E. coli cells infected with the enriched dAb displaying fd-phage populations were obtained. An aliquot of these cells was used to prepare phage DNA and the enriched V-genes excised by digestion using the restriction endonucleases, Sail and Notl. The purified V-genes were ligated into the corresponding sites of pD0M5 (expression vector derived from pUC119 with LacZ promoter, eukaryotic leader, dAb cloning site, myc tag), and the ligated DNA used to electro-transform E. coli HB2151 cells which were grown overnight on agar plates containing the antibiotic carbenicillin. The resulting colonies were induced to express dAb protein either as 200 ⁇ l microcultures or 50 ml cultures. The resulting dAb was analysed for inhibitory activity using the CD40L receptor binding assay.
  • pDOM4 DNA was purified from the cell pellet obtained from a 50 ml overnight E. coli culture using the QIAfilter Plasmid Midi DNA purification kit from Qiagen, following the manufacturer's instructions.
  • the dAb genes were excised from the pDOM4 vector by mixing: 10 ⁇ l of 10x SaR buffer; 1 ⁇ l of 10Ox BSA; 20 ⁇ g of purified DNA fragment; 2.5 ⁇ l of SaR enzyme (10 U/ ⁇ l); 2.5 ⁇ l of Notl enzyme (10 U/ ⁇ l); the digestion mix was made up to a final volume of 100 ⁇ l using sterile water. The digestion mix was incubated for 5 hours at 37°C.
  • the digested DNA samples were electrophoresed on a 1.5% agarose gel and the band corresponding to the dAb V-genes (-324 bp to 372 bp) was excised from the gel.
  • the dAb gene DNA was purified from the gel slice using the QIAquick Gel Extraction kit from Qiagen, following the manufacturer's instructions.
  • the expression vector pDOM5 was digested with Sail and Notl as follows: 10 ⁇ l of 1Ox SaR buffer; 1 ⁇ l of 10Ox BSA; 20 ⁇ g of plasmid pD0M5; 1.5 ⁇ l of SaR enzyme (10 U/ ⁇ l); 1.5 ⁇ l of Notl enzyme (10 U/ ⁇ l); the digestion mix was made up to a final volume of 100 ⁇ l using sterile water. The digestion mix was incubated for 2 hours at 37°C. The digested vector fragment was purified using the QIAquick PCR Purification Kit.
  • digested pDOM5 and digested dAb genes were ligated by mixing: 2 ⁇ l of 1 Ox T4 DNA ligase buffer; 400 ng of digested pD0M5 vector; 100 ng of digested dAb genes;
  • T4 DNA ligase 400 U/ ⁇ l
  • the ligation mix was made up to 20 ⁇ l with sterile water.
  • the ligation mixes were incubated for 2 hours at 25 0 C.
  • Dilutions of the cells from 10 ' ° to IO '3 were plated on regular 9 cm diameter plates containing TYE supplemented with 5% glucose and 50 ⁇ g/ml carbenicillin. The cells are incubated overnight at 37°C in an inverted position. Reagents, equipment and sources from which they are available are provided in Table 3.
  • a 96 well transfer device was used to transfer between 1-5 ⁇ l of the bacterial culture into a fresh 96 well culture plate containing 100 ⁇ l per well of 2xTY supplemented with 0.1% glucose and 50 ⁇ g/ml carbenicillin.
  • the bacterial cells were pelleted by centrifugation at 1,800 g for 10 min at 4 0 C. The supernatant (containing expressed dAb) was then analysed to determine if dAbs were capable of inhibiting binding of CD40L to CD40-Fc fusion in a receptor binding assay.
  • dAb protein for analysis, 50 ml cultures were used for induction.
  • a single colony of the desired dAb (for example DOM-24) grown on TYE plates was inoculated into 10 ml 2xTY supplemented with 5% glucose and 50 ⁇ g/ml carbenicillin in a 30 ml universal tube and grown overnight at 37°C with shaking at 250 rpm.
  • Five hundred microlitres of the overnight culture was added into 50 ml of 2xTY supplemented with 0.1% glucose and 50 ⁇ g/ml carbenicillin and grown, at 37 0 C with shaking at 250 rpm.
  • the OD 6 oo of the culture was monitored regularly in comparison with sterile 2xTY and at an OD 6 oo of 0.9 the culture was induced by the addition of 1 M IPTG to a final concentration of 1 mM.
  • the inoculated culture was incubated at 3O 0 C with shaking at 250 rpm overnight.
  • the next day the culture was centrifuged at 6000 g for 15 min at 4°C and the clarified supernatant mixed with 100 ⁇ l of protein-A streamline or protein-L agarose (pre- washed with 5 mM MgSO 4 ) overnight at 4 0 C.
  • the supernatant/bead mixture was then centrifuged at 180 g at 4 0 C for 2 minutes.
  • the supernatant was decanted and the retained beads washed with 10 ml of PBS containing 0.5M NaCl.
  • the bead solution was transferred into a 96 well Whatman filter plate and the beads washed once with 400 ⁇ l of PBS containing 0.5M NaCl 5 then once with 400 ⁇ l of PBS, followed by centrifugation for 2 minutes at 180 g after each washing step.
  • dAb protein was eluted using 70 ⁇ l of 0.1 M glycine (pH 2.0) and the solution neutralised by the addition of 40 ⁇ l of 1 M Tris-HCl (pH 8.0). The purified dAb concentration was determinate by OD 280 .
  • CD40L assay was used to measure the binding of CD40L to CD40 and the ability of binding entities (eg, monvalent antibody fragments such a dAbs) to block this interaction, as described below and shown schematically in Figure 7.
  • binding entities eg, monvalent antibody fragments such a dAbs
  • a 96 well Nunc Maxisorp assay plate was coated overnight at 4 0 C with 100 ⁇ l per well of recombinant human CD40/Fc (R&D Systems) at 0.5 ⁇ g/ml in carbonate buffer.
  • the plate was washed 3 times with 300 ⁇ l of 0.05% Tween/PBS and 3 times with 300 ⁇ l of PBS using a Tecan plate washer.
  • the wells were blocked using 200 ⁇ l of PBS containing 2% (w/v) BSA and incubated for a minimum of 1 h at room temperature.
  • the wells were washed as above, then 50 ⁇ l of purified dAb protein (or unpurified supernatant containing dAb from a micro-culture expression) was added to each well.
  • the plate was washed as described previously and 100 ⁇ l biotinylated anti- CD40L antibody, 0.5 ⁇ g/ml in diluent, was added and incubated for 1 hr at room temperature. The plate was washed as described above, then 100 ⁇ l HRP conjugated anti-biotin antibody (1:5000 dilution in diluent) added to each well and the plate incubated for 1 hr at room temperature. The plate was washed again as described above using a Tecan plate washer and the assay developed using 100 ⁇ l of SureBlue 1-Component TMB MicroWell Peroxidase solution (the plate was left at room temperature for up to 20 min). The reaction was stopped by the addition of 100 ⁇ l 1 M hydrochloric acid.
  • the OD 4 50nm of the plate was assayed within 30 minutes of acid addition.
  • the OD 45 o nm is proportional to the amount of bound streptavidin-HRP conjugate, therefore the greater the degree of dAb inhibition the lower the OD 450 n m of the resulting signal.
  • Reagents, equipment and sources from which they are available are provided in Table 6. Controls
  • Receptor binding data for the most potent inhibitors is summarised in Figures 2, 3, and 4, and in Table 7, below.
  • Table 8, below provides DNA and translated amino acid sequence of unique dAbs identified in the receptor binding assay as inhibiting CD40L binding to CD40.
  • Figure 2 shows a dose response receptor binding assay(RBA) readout, analysing the inhibition of CD40L binding to CD40-Fc by dAbs DOM-10, -20, -27, - 30, -31, -62, -77, titrated from 1 ⁇ M down to lO pM.
  • dAbs DOM-20, -30, and -31 are the most potent, with IC 50 values of approximately 8 nM.
  • Figure 3 shows a dose response receptor binding assay readout, analysing the inhibition of CD40L binding to CD40-Fc by dAbs DOM-4 and DOM-5, titrated from 1 ⁇ M down to 500 pM.
  • the IC 50 values for dAbs DOM-5 and DOM-4 are approximately 3 nM and 100 nM respectively.
  • Figure 4 shows a dose response receptor binding assay readout, analysing the inhibition of CD40L binding to CD40-Fc by dAb DOM-24, titrated from 100 nM down to 0.5 pM. The data were curve-fitted using GraphPad Prism software.
  • Table 8 Summary of dAbs exhibiting a ranee of CD40L inhibitory ICso values as determined using the CD40L / CD40-Fc receptor inhibition assay.
  • AACTAATAAG (SEQ ID NO: 115)
  • GCATCCAAAC GACCAGTGGC AGAGCTCG SEQ ID NO: 125
  • CTCCACGTCG ACMCCTCAG ACCCCCTCCG AACCATGTCG GACCCCCCAG GGACGCAGAG AGGACACGTC GGAGGCCTAA GTGGAAACCA TTAATACGCT
  • GCAGTGGATC CAACCATGGT CGTCTTTGGT CCCTTTCGGG GATTCGAGGA CTAGATAAAA CCAAGGATAA ACGTTTCACC CCAGGGTAGT GCAAAGTCAC CGTCACCTAG

Abstract

The invention relates to antibody polypeptides that monovalently bind CD40L. Antibody polypeptides that are monovalent for binding of CD40L can inhibit CD40L activity while avoiding potential undesirable effects that can occur with antibodies capable of divalent or multivalent binding of CD40L. In one aspect, a monovalent anti-CD40L antibody polypeptide consists of or comprises a single immunoglobulin variable domain that specifically binds and antagonizes the activity of CD40L, preferably without substantially agonizing CD40 and/or CD40L activity. In another aspect, the monovalent anti-CD40L antibody polypeptide is a human antibody polypeptide. The invention further encompasses methods of antagonizing CD40/CD40L interactions in an individual and methods of treating diseases or disorders involving CD40/CD40L interactions, the methods involving administering a monovalent anti-CD40L antibody polypeptide to the individual.

Description

COMPOSITIONS MONOVALENT FOR CD40L BINDING AND METHODS
OF USE
BACKGROUND OF THE INVENTION
CD40 is a 50 IcD cell surface glycoprotein molecule expressed on the surface of mature and immature B cells, macrophages, follicular dendritic cells, thymic epithelium, normal basal epithelium, and some tumor-derived cell lines. The CD40 molecule is a member of the TNF receptor family, and has important signaling functions leading to a variety of downstream effects in various cell types. Early studies showed that cross-linking of CD40 on the B cell surface with an antibody resulted in B cell proliferation and activation. Antibody cross linking of CD40 in the presence of IL-4 induces proliferation and class switching in vitro, B cell aggregation via LFA-I (Gordon et al., 1988, J. Immunol. 140: 1425), and serine/threonine and tyrosine phosphorylation of a number of intracellular substrates (Gordon et al., 1988, supra; Uckun et al., 1991, J. Biol. Chem. 266:17478). Anti-CD40 monoclonal antibodies also prime B cells to proliferate in response to agents such as PMA (Gordon et al., 1987, Eur. J. Immunol. 17: 1535) and anti-CD20 antibody (Clark & Ledbetter, 1986, Proc. Natl. Acad. Sci. U.S.A. 83: 4494).
The receptor homology of CD40 and the antibody cross-linking studies showing a central role for CD40 in B cell activation prompted the search for a natural ligand. A mutant of the Jurkat T cell line was found to constitutively activate human B cells to secrete immunoglobulin (Yellin et al., 1991, J. Immunol. 147: 3389-3395). A monoclonal antibody, termed 5c8, was raised which specifically reacted with the mutant line, but not with the parental Jurkat cell line. The 5c8 antibody immunoprecipitated a 30 kD (more accurately, 29.3 kD, 261 amino acids) cell surface polypeptide and was found to specifically inhibit the B cell helper function of the mutant cell line. (Lederman et al., 1992, J. Exp. Med., 175: 1091-1101; Lederman et al., 1992, J. Immunol. 149: 3817-3826; Lederman et al., 1993, Curr. Opin. Immunol. 5: 439.444;). The 30 kD polypeptide ligand of the 5c8 antibody was termed T-BAM, for T-B-cell Activating Molecule. A second line of studies used molecular cloning techniques to identify polypeptides that specifically bind the CD40 molecule. cDNA clones for a specific ligand of CD40 were identified in a CD40 binding assay and alternately termed CD40 Ligand (CD40L), gp39, CD154, or TRAP (Graf et al., 1992, Eur. J. Immunol. 22: 3191-3194; Armitage et al., 1992, Nature 357: 80-82; and Aruffo et al., 1993, Cell 72: 291-300). Subsequently, the CD40L clone was found to have the same structure as T-BAM (Covey et al., 1994, MoI. Immunol. 31 : 471-484). Human CD40L protein shows 82.8% and 77.4% identity at the nucleic acid and amino acid levels, respectively, to a similar protein isolated from murine EL4 thymoma cells. Both of these proteins are ligands for CD40 cell surface antigen expressed on resting B cells. CD40L has also been described as IMD3, a protein involved in hyper-IgM immunodeficiency syndrome.
The human gene encoding CD40L maps to chromosome Xq26.3-q27. The gene contains five exons. Deletions, point mutations and frameshift mutations clustering within a limited region of the CD40L extracellular domain have been found to be the basis of a rare X-linked immunodeficiency syndrome (Hyper-IgM immunodeficiency syndrome, HIGMl) characterized by recurrent bacterial infections, very low or absent IgG, IgA and IgE, and normal to increased IgM and IgD serum levels. Causally-related mutations have been found to consist of clustered deletions arising by splice-donor mutations with exon skipping, splice-acceptor mutations with utilization of a cryptic splice site, and deletion/insertion events with the creation of a new splice site.
CD40L is expressed on activated, but not resting CD4+ T cells, and was found to play a particularly important role in the humoral immune response, being linked to B cell proliferation, antibody and cytokine production, and cell viability. In vivo, deletion or mutation of CD40L leads to severe immunodeficiency, both in mice and in humans, characterized by hypogammaglobulinemia and T cell deficits in cell- mediated immunity (Chess, C, 2001, in Therapeutic Immunology, 2nd edition, Austen, K.F., Burakoff, S., Rosen, F. and Strom, T., eds., Blackwell Sciences, pp. 441-456). Human CD4+ T cells infected by HIVl, which causes severe dysfunction of cellular immunity, but paradoxically results in intense polyclonal activation of B cells, do not express CD40L. Gene and cell surface expression of the CD40L by activated T cells has been shown to be depressed in a subgroup of patients with common variable immunodeficiency (CVI). Thus, inefficient signaling via CD40 may be responsible, at least in part, for the failure of B cell differentiation in these patients.
The functional consequences of CD40L binding to CD40 include, for example, a) rescuing B cells from apoptosis induced by Fas or cross-linking of IgM, b) induction of the co-stimulator molecules CD80 (B7-1) and CD86 (B7-2) which interact with CD28 and CD 152 (CTLA-4) on the surface of activated T cells; c) increased expression of other cell surface activation molecules including CD23, CD54, CD95 and lymphotoxin-a; and d) inducing immunoglobulin class switching (see Chess, supra, and references 25, 44, and 47-60 cited therein). CD40L binding to CD40 also augments the antigen-presenting functions of dendritic cells, inducing maintenance of high levels of MHC class II antigens and upregulation of accessory molecules including CD58 (LF A-3). CD40L induces cytokine production and tumoricidal activity in peripheral blood monocytes. CD40L also co-stimulates the proliferation of activated T cells, and the co-stimulation is accompanied by the production of IFN-γ, TNF-α and IL2. The expression of CD40L on murine T-helper cells and CD4+ T cells is inhibited by IFN-γ, and is inhibited on T-helper-type 2 cells by TGF-β.
CD40L upregulates the expression of CD54 by cultured Hodgkin and Reed- Sternberg cells. The increased CD54 surface expression is accompanied by increased shedding of surface-bound CD54.
CD40L has also been suggested to be important in the induction of tolerance - CD80 and CD86, which are upregulated by CD40L, interact with CD28 to provide essential co-stimulation of T cells, in concert with T cell receptor activation, that results in full activation of T cells. In the absence of CD80 and CD86-triggered activation of CD28, anergy or tolerance occurs as a consequence of antigen triggering (Linsley & Ledbetter, 1993, Ann. Rev. Immunol. 11: 191-212; Jenkins et al., 1993, Curr Opin. Immunol. 5: 361-367; and Boussiotis et al., 1996, Immunol. Rev. 153: 5- 26).
The CD40L/CD40 pathway has been implicated in the in vivo priming of CD8+ cytotoxic T lymphocytes (CTSs) by CD4+ T cells. As noted, CD40L expressed on the surface of activated CD4+ T cells interacts with CD40 expressed on dendritic cells, inducing the dendritic cells to express more MHC, and signaling through CD40 can replace the requirement for CD4+ T-helper cells in priming CD 8+ CTL responses. Blockade of CD40L inhibits CTL priming, emphasizing the vital role of CD40L/CD40 interactions in CTL priming by helper T cells (Ridge et al., 1998, Nature 393: 474-478; Schoenberger et al., 1998, Nature 393: 480-483; Bennett et al., 1998, Nature 393: 478-480).
CD40L can also mediate functional interactions of CD4+ T cells with other cells that express CD40, such as fibroblasts, synovial cells and endothelial cells (Yellin et al., 1995, J. Leuko. Biol. 58: 209-216; Yellin et al., 1995, J. Exp. Med. 182: 1857-1864). CD40L induces the expression of CD54 (ICAM-I) and CD106 (VCAM- 1) by fibroblasts, as well as increasing fibroblast IL-6, collagenase and collagen production and inducing fibroblast proliferation. Thus, CD40L/CD40 interactions may be involved in the induction of fibrosis associated with autoimmunity and immune responses.
CD40L interaction with CD40 induces endothelial cells to express CD62E (E- selectin), ICAM-I and VCAM-I. The upregulation of these adhesion molecules may be involved in the binding of inflammatory cells to vascular endothelium and the subsequent migration of the inflammatory cells to sites of inflammation. CD40L blockade retards the migration of leukocytes through endothelial cell barriers. In animal models of autoimmunity, antibodies to CD40L interfere with the accumulation of inflammatory cells at the site of inflammation.
CD40/CD40L interactions have been implicated in diseases having an immune or autoimmune connection. Animal models of immune-related disease in which the CD40L/CD40 pathway has been demonstrated to play a role in the pathology include, for example, murine models of systemic lupus erythematosis (Lupus or SLE; see, e.g., Kalled et al., 1998, J. Immunol. 160: 2158-2165), arthritis (collagen-induced arthritis, see, e.g., Durie et al., 1993, Science 261: 1328-1330), multiple sclerosis (experimental autoimmune encephalomyelitis, EAE; see, e.g., Howard et al., 1999, J. Clin . Invest. 103: 281-290), autoimmune thyroiditis (experimental autoimmune thyroiditis, EAT; see, e.g., Caryanniotis et al., 1997, Immunology 90: 421-426), colitis (hapten-induced colitis; see, e.g., Stuber et al., 1996, J. Exp. Med. 183: 693-698), atherosclerosis and coronary artery disease (see, e.g., Mach et al., 1998, Nature 394: 200-203), and allograft rejection (see, e.g., Parker et al., 1995, Proc. Natl. Acad. Sci. U.S.A. 92: 9560-9564; Kirk et al., 1997, Proc. Natl. Acad. Sci. U.S.A. 94: 8789-8794; Larsen et al., 1996, Nature 381 : 434-438 and Blazar et al., 1997, J. Immunol. 158: 29-39).
CD40L antibody trials for treatment of human immune-related diseases include studies in patients with Lupus (see, e.g., Huang et al., 2002, Arthritis Rheum. 46: 1554-1562). A phase I trial demonstrated that anti-CD40L humanized monoclonal antibody (IDEC-131) is safe and well tolerated by patients with Lupus (Davis et al., 2001, J. Rheumatol. 28: 95-101). A phase II study with the IDEC-131 antibody showed improvement in clinical symptoms, but efficacy of the drug over placebo controls was not demonstrated (Kalunian et al., 2002, Arthritis Rheum. 46: 3251-3258). In a phase II study with BG9588 anti-CD40L antibody, clinical efficacy was demonstrated, but the study was terminated due to the occurrence of thromboembolic events (Boumpas et al., 2003, Arthritis Rheum. 48: 719-727).
U.S. Patent Nos. 5,474,771 (Lederman et al.) and 5,876,950 (Siadak et al.) disclose murine monoclonal antibodies specific for different epitopes of human gp39. WO95/06666 (Noelle & Foy) discloses murine anti-gp39 antibodies.
U.S. Patent No. 6,328,964 (Noelle & Claassen) discloses methods for the treatment of multiple sclerosis using gp39-specific antibodies.
U.S. Patent No. 5,747,037 (Noelle et al.), and EP0721469B1 (Ledbetter et al.) and its U.S. counterpart U.S. 5,869,049 disclose anti-human monoclonal (mouse) antibodies specific for gp39. U.S. Patent No. 5,876,718 (Noelle et al.) discloses methods of inducing T cell non-responsiveness to transplanted tissues and of treating graft-versus-host disease with anti-gp39 monoclonal (mouse) antibodies. EP0742721B1 (Noelle et al.) discloses methods of inhibiting a 'humoral immune response to a thymus-dependent antigen that use anti-gp39 monoclonal (mouse) antibodies. U.S. Patent No. 6,375,950 describes methods for inducing T cell unresponsiveness to donor tissue or organs in a transplant recipient through use of anti-gp39 monoclonal (murine) antibodies.
EP1005372B1 (De Boer et al.) describes methods for the selective killing of autoreactive CD40L+ T cells using anti-CD40L monoclonal (mouse) antibody-toxin fusion proteins.
U.S. Patent No. 6,340,459 (Yellin et al.) describes the use of murine anti gp39 monoclonal antibody 5c8 for the treatment or prevention of reperfusion injury.
EP0831906B1 (Claassen et al.) describes methods for the treatment of T cell- mediated tissue destruction in autoimmune diseases such as multiple sclerosis using anti-gp39 monoclonal (mouse) antibodies. Antibodies used in therapeutic approaches in the prior art have been divalent antibodies of murine origin.
A number of smaller antigen binding fragments of naturally occurring antibodies have been identified following protease digestion. These include, for example, the "Fab fragment" (VL-CL-CH1-VH), 'Tab' fragment" (a Fab with the heavy chain hinge region) and "F(ab')2 fragment" (a dimer of Fab' fragments joined by the heavy chain hinge region). Recombinant methods have been used to generate even smaller antigen-binding fragments, referred to as "single chain Fv" (variable fragment) or "scFv," consisting of VL and VH joined by a synthetic peptide linker. While the antigen binding unit of a naturally-occurring antibody (e.g., in humans and most other mammals) is generally known to be comprised of a pair of V regions (VL/VH), camelid species express a large proportion of fully functional, highly specific antibodies that are devoid of light chain sequences. The camelid heavy chain antibodies are found as homodimers of a single heavy chain, dimerized via their constant regions. The variable domains of these camelid heavy chain antibodies are referred to as VHH domains and retain the ability, when isolated as fragments of the VH chain, to bind antigen with high specificity ((Hamers-Casterman et al., 1993, Nature 363: 446-448; Gahroudi et al., 1997, FEBS Lett. 414: 521-526). Antigen binding single VH domains have also been identified from, for example, a library of murine VH genes amplified from genomic DNA from the spleens of immunized mice and expressed in E. coli (Ward et al., 1989, Nature 341 : 544-546). Ward et al. named the isolated single VH domains "dAbs," for "domain antibodies." The term "dAb" will refer herein to an antibody single variable domain (VH or VL) polypeptide that specifically binds antigen. A "dAb" binds antigen independently of other V domains; however, as the term is used herein, a "dAb" can be present in a homo- or heteromultimer with other VH or VL domains where the other domains are not required for antigen binding by the dAb, i.e., where the dAb binds antigen independently of the additional VH or VL domains. Antibody single variable domains, for example, VHH, are the smallest antigen- binding antibody unit known. For use in therapy, human antibodies are preferred, primarily because they are not as likely to provoke an immune response when administered to a patient. As noted above, isolated non-camelid VH domains tend to be relatively insoluble and are often poorly expressed. Comparisons of camelid VHH with the VH domains of human antibodies reveals several key differences in the framework regions of the camelid VHH domain corresponding to the VH/VL interface of the human VH domains. Mutation of these residues of human VH3 to more closely resemble the VHH sequence (specifically GIy 44→Glu, Leu 45→Arg and Trp 47-→-Gly) has been performed to produce "camelized" human VH domains that retain antigen binding activity (Davies & Riechmann, 1994, FEBS Lett. 339: 285-290) yet have improved expression and solubility. (Variable domain amino acid numbering used herein is consistent with the Kabat numbering convention (Kabat et al., 1991, Sequences of Immunological Interest, 5th ed. U.S. Dept. Health & Human Services, Washington, D.C.)) WO 03/035694 (Muyldermans) reports that the Trp 103-→Arg mutation improves the solubility of non-camelid VH domains. Davies & Riechmann (1995, Biotechnology N-. Y. 13: 475-479) also report production of a phage-displayed repertoire of camelized human VH domains and selection of clones that bind hapten with affinities in the range of 100-400 nM, but clones selected for binding to protein antigen had weaker affinities.
While many antibodies and their derivatives are useful for diagnosis and therapy, the ideal pharmacokinetics of antibodies, are often not achieved for a particular application. In order to provide improvement in the pharmacokinetics of antibody molecules, the present invention provides single domain variable region polypeptides that are linked to polymers which provide increased stability and half- life. The attachment of polymer molecules (e.g., polyethylene glycol; PEG) to proteins is well established and has been shown to modulate the pharmacokinetic properties of the modified proteins. For example, PEG modification of proteins has been shown to alter the in vivo circulating half-life, antigenicity, solubility, and resistance to proteolysis of the protein (Abuchowski et al., J Biol. Chem. 1977, 252:3578; Nucci et al., Adv. Drug Deliver)' Reviews 1991, 6:133; Francis et al., Pharmaceutical Biotechnology Vol. 3 (Borchardt, R. T. ed.); and Stability of Protein Pharmaceuticals: in vivo Pathways of Degradation and Strategies for Protein Stabilization 1991 pp235-263, Plenum, NY).
Both site-specific and random PEGylation of protein molecules is known in the art (See, for example, Zalipsky and Lee, Polvfethylene glycol") Chemistry: Biotechnical and Biomedical Applications 1992, pp 347-370, Plenum, NY; Goodson and Katre, 1990, Bio/Technology, 8:343; Hershfield et al., 1991, PNAS 88:7185). More specifically, random PEGylation of antibody molecules has been described at lysine residues and thiolated derivatives (Ling and Mattiasson, 1983, Immunol Methods 59: 327; Wilkinson et al., 1987, Immunol Letters, 15: 17; Kitamura et al., 1991, Cancer Res. 51:4310; Delgado et al., 1996 Br. J. Cancer, 73: 175; Pedley et al., 1994, Br. J. Cancer, 70:1126).
SUMMARY OF THE INVENTION
The invention relates to antibody polypeptides that monovalently bind
CD40L. Because of the clear importance of CD40L in the production of antibodies, the CD40/CD40L interaction and pathways present important targets for the development of therapeutic approaches for the treatment of diseases and disorders that involve inappropriate or excessive antibody responses, such as autoimmune diseases. Antibody polypeptides that are monovalent for binding of CD40L can inhibit CD40L activity, including binding and activation of CD40 on the B cell surface and downstream effects, while avoiding potential undesirable effects that can occur with antibodies capable of divalent or multivalent binding of CD40L. Monovalent anti-CD40L antibody polypeptides can also be applied to any of a number of uses for which standard divalent antibodies are also used, e.g., in vivo imaging and diagnosis.
In one aspect, the antibody polypeptide consists of or comprises a single immunoglobulin variable domain that specifically binds and antagonizes the activity of CD40L, preferably without substantially agonizing CD40 and/or CD40L activity. In another aspect, because human antibodies will avoid the generation of an immune response to the antibodies when administered to human subjects for the treatment or prevention of disease, the antibody polypeptide is a human antibody polypeptide that monovalently binds CD40L, preferably without substantially agonizing CD40 and/or CD40L activity.
In summary then, in one embodiment, the invention provides an antibody polypeptide, preferably a human antibody polypeptide, that is monovalent for binding to CD40L (gp39).
In one embodiment, the human antibody polypeptide dissociates from human
CD40L with a Kd in the range of 50 nM to 20 pM, inclusive, as measured by surface plasmon resonance. For example, the Kd for human CD40L can be 25 nM to 20 pM,
10 nM to 20 pM, 5 nm to 20 pM, 1 nM to 20 pM, 0.5 nM to 20 pM, 0.1 nM to 20 pM, 0.1 nM to 50 nM, 75 pM to 20 pM or even 50 pM to 20 pM.
Unless otherwise stated, all ranges described herein are inclusive of the specific endpoints. In another embodiment, the antibody polypeptide inhibits the binding of CDAQh to CD40.
In another embodiment, the binding of the antibody polypeptide to CD40L does not substantially agonize CD40 and/or CD40L activity.
In another embodiment, the human antibody polypeptide inhibits the binding of CD40 to CD40L, and does not substantially agonize signaling by CD40.
In another embodiment, the binding of the antibody polypeptide to CD40L does not substantially induce JNK phosphorylation in Jurkat T-cells.
In another embodiment, the binding of the antibody polypeptide to CD40L does not substantially induce IFN-γ secretion by Jurkat T-cells co-stimulated with anti-CD3 antibody.
In another embodiment, the presence of the antibody polypeptide in a standard platelet aggregation assay does not result in aggregation of more than 25% over the aggregation observed in a negative control assay performed without the addition of antibody.
In another embodiment, the human antibody polypeptide comprises a single immunoglobulin variable domain that binds CD40L. In a preferred embodiment, the single immunoglobulin variable domain is a VH or a VL domain.
In another embodiment, the antibody polypeptide is selected from the group consisting of a dAb, a FAb, an scFv, an Fv, or a disulfide-bonded Fv.
In another embodiment, the human antibody polypeptide is PEG-linked. In one embodiment, the PEG is covalently linked to the human antibody polypeptide. In one preferred embodiment, the PEG-linked human antibody polypeptide has a hydrodynamic size of at least 24 kD. In another preferred embodiment, the PEG is linked to the antibody polypeptide at a cysteine or lysine residue. In another preferred embodiment, the total PEG size is from 20 to 60 IcD, inclusive. In another preferred embodiment, the PEG-linked human antibody pofypeptide has a hydrodynamic size of at least 200 IcD.
In one embodiment, the antibody polypeptide has an increased in vivo half-life relative to the same antibody polypeptide composition lacking polyethylene glycol.
In another embodiment, the tα-half life of the antibody polypeptide composition is increased by 10% or more. In another embodiment, the tα-half life of the antibody polypeptide composition is increased by 50% or more. In another embodiment, the tα-half life of the antibody polypeptide composition is increased by 2X or more. In another embodiment, the tα-half life of the antibody polypeptide composition is increased by 5X or more, e.g., 1OX, 15X, 2OX, 25X, 3OX, 4OX, or more. In another embodiment, the tα-half life of the antibody polypeptide composition is increased by 5OX or more.
In another embodiment, the PEG-linked antibody polypeptide has a tα half- life of 0.25 to 6 hours, inclusive. In another embodiment, the tα half-life is in the range of 30 minutes to 12 hours, inclusive. In another embodiment, the tα-half life of the antibody polypeptide composition is in the range of 1 to 6 hours.
In another embodiment, the tβ-half life of the antibody polypeptide composition is increased by 10% or more. In another embodiment, the tβ-half life of the antibody polypeptide composition is increased by 50% or more. In another embodiment, the tβ-half life of the antibody polypeptide composition is increased by 2X or more. In another embodiment, the tβ-half life of the antibody polypeptide composition is increased by 5X or more, e.g., 10X, 15X, 20X, 25X, 30X, 4OX, .or more. In another embodiment, the tβ-half life of the antibody polypeptide composition is increased by 5OX or more.
In another embodiment, the antibody polypeptide composition has a tβ half- life of 1 to 170 hours, inclusive. In another embodiment, the tβ-half life is in the range of 12 to 48 hours, inclusive. In another embodiment, the tβ-half life is in the range of 12 to 26 hours, inclusive.
In addition, or alternatively to the above criteria, the present invention provides a dAb containing composition comprising a ligand according to the invention having an AUC value (area under the curve) in the range of 1 mg.min/ml or more. In one embodiment, the lower end of the range is 5, 10, 15, 20, 30, 100, 200 or
300 mg.min/ml. In addition, or alternatively, a ligand or composition according to the invention has an AUC in the range of up to 600 mg.min/ml. In one embodiment, the upper end of the range is 500, 400, 300, 200, 150, 100, 75 or 50 mg.min/ml. Advantageously a ligand according to the invention will have an AUC in the range selected from the group consisting of the following: 15 to 150 mg.min/ml, 15 to 100 mg.min/ml, 15 to 75 mg.min/ml, and 15 to 50 mg.min/ml.
In another embodiment, the antibody polypeptides described herein can be linked to human serum albumin (HSA), which also has the effect of increasing the in vivo half life of the molecule. The human serum albumin coding sequences can be obtained by PCR using primers derived from the cDNA sequence available at GenBank Accession No. NM000477. Such coding sequences can be fused to the coding sequence for a monovalent anti-CD40L antibody polypeptide as described herein, and the fusion can be expressed by one of skill in the art.
In another embodiment, the tα-half life of the HSA-linked human antibody polypeptide composition is increased by 10% or more.
In another embodiment, the tα-half life of the HSA-linked human antibody polypeptide composition is in the range of 0.25 hours to 6 hours.
In another embodiment, the tβ-half life of the HSA-linked human antibody polypeptide composition is increased by 10% or more.
In another embodiment, the tβ-half life of the HSA-linked human antibody polypeptide composition is in the range of 12 to 48 hours. In another embodiment, the human antibody polypeptide comprises an amino acid sequence selected from the group consisting of SEQ ID NOs 7-82 and 246-360.
In another embodiment, the human antibody polypeptide inhibits binding of
CD40L to CD40 with an IC50 in the range of 20 pM to 1.5 μM, inclusive; IC5O for inhibition of CD40L binding to CD40 in any embodiment described herein is preferably measured as described herein in Example 6. The IC50 can preferably be in the range of 20 pM to 1 μM, 20 pM to 900 nM, 20 pM to 800 nM, 20 pM to 700 nM,
20 pM to 600 nM, 20 pM to 500 nM, 20 pM to 400 nM, 20 pM to 300 nM, 20 pM to
200 nM, 20 pM to 100 nM, or 20 pM to 50 nM. Further acceptable or preferred ranges include, for example, 50 pM to 1 μM, 100 pM to 500 nM, 125 pM to 250 nM,
150 pM to 200 nM, 150 pM to 100 nM and 200 pM to 50 nM.
In another embodiment, the antibody polypeptide is fused to a second antibody polypeptide which binds a ligand other than CD40L. In a preferred embodiment, the antibody polypeptide which binds a ligand other than CD40L binds a ligand selected from the group consisting of HSA, TNFα, IL-I, IL-2, IL-4, IL-6, IL- 8, IL-12, IL-18, IFN-γ, CD2, CD4, CD8, CTLA4, LFAl, LFA3, VLA4, CD80 (B7- 1), CD28, CD86 (B7-2), and CTLA-4.
In another embodiment, the human antibody polypeptide is free of an Fc domain. The limits of an Fc domain are set out in Kabat et al. (1991, Sequences of Immunological Interest, 5th ed. U.S. Dept. Health & Human Services, Washington, D. C; incorporated herein by reference). In the alternative, an Fc domain consists of the CH2-CH3 regions, optionally including a hinge region linked to the CH2. In a preferred embodiment, the human antibody polypeptide does not mediate platelet aggregation in a standard platelet aggregation assay.
The invention further encompasses a human antibody polypeptide which has an amino acid sequence at least 85% identical to a sequence selected from the group consisting of SEQ ID NOs 7-82 and 246-360, which antibody polypeptide specifically and monovalently binds CD40L. The invention further encompasses an antigen-binding polypeptide, the polypeptide comprising a single immunoglobulin variable domain which specifically and monovalent])' binds CD40L. Recited differently, the invention further encompasses a polypeptide comprising a moiety which specifically binds CD40L, which moiety consists of a single immunoglobulin variable domain.
In one embodiment, the polypeptide consists of a human single immunoglobulin variable domain.
In another embodiment, the polypeptide has a Kd for human CD40L in the range of 50 nM to 20 pM, inclusive, as determined by surface plasmon resonance. For example, the Kd for human CD40L can be 25 nM to 20 pM, 10 nM to 20 pM, 5 nm to 20 pM, 1 nM to 20 pM, 0.5 nM to 20 pM, 0.1 nM to 20 pM, 75 pM to 20 pM or even 50 pM to 20 pM.
In another embodiment, the polypeptide inhibits the binding of CD40L to CD40.
In another embodiment, the polypeptide inhibits the binding of CD40 to
CD40L and has an IC50 in the range of 20 pM to 1.5 μM, inclusive. For example, the IC50 can be in the range of 20 pM to 1 μM, 20 pM to 900 nM, 20 pM to 800 nM, 20 pM to 700 nM, 20 pM to 600 nM, 20 pM to 500 nM, 20 pM to 400 nM, 20 pM to 300 nM, 20 pM to 200 nM, 20 pM to 100 nM, or 20 pM to 50 nM. Further acceptable or preferred ranges include, for example, 50 pM to 1 μM, 100 pM to 500 nM, 125 pM to 250 nM, 150 pM to 200 nM, 150 pM to 100 nM and 200 pM to 50 nM.
In another embodiment, the binding of the polypeptide to CD40L does not substantially agonize CD40 and/or CD40L activity.
In another embodiment, the binding of the polypeptide to CD40L does not substantially induce JNK phosphorylation in Jurkat T-cells. In another embodiment, the binding of the polypeptide to CD40L does not substantially induce IFN-γ secretion by Jurkat T-cells co-stimulated with anti-CD3 antibody.
In another embodiment, the presence of the antibody polypeptide in a standard platelet aggregation assay does not result in aggregation more than 25% over the aggregation observed in a negative control assay lacking antibody polypeptide.
In another embodiment, the single immunoglobulin variable domain is a human single immunoglobulin variable domain.
In another embodiment, the single immunoglobulin variable domain is a VH or a VL domain.
In one embodiment, the polypeptide is PEG-linked. In one embodiment, the PEG is covalently linked. In one preferred embodiment, the PEG-linked antigen- binding polypeptide has a hydrodynamic size of at least 24 kD. In another preferred embodiment, the PEG is linked to the antigen-binding polypeptide at a cysteine or lysine residue. In another preferred embodiment, the total PEG size is from 20 to 60 kD, inclusive. In another preferred embodiment, the PEG-linked antigen-binding polypeptide has a hydrodynamic size of at least 200 kD.
In another embodiment, the PEG-linked polypeptide has an increased in vivo half-life relative to the same polypeptide composition lacking linked polyethylene glycol. In another embodiment, the tα-half life of the polypeptide composition is increased by 10% or more. In another embodiment, the tα-half life of the polypeptide composition is increased by 50% or more. In another embodiment, the tα-half life of the polypeptide composition is increased by 2X or more. In another embodiment, the tα-half life of the polypeptide composition is increased by 5X or more, e.g., 10X, 15X, 2OX, 25X, 3OX, 4OX, or more. In another embodiment, the tα-half life of the polypeptide composition is increased by 5OX or more. In another embodiment, the PEG-linked antibody polypeptide has a toe half- life of 0.25 to 6 hours, inclusive. In another embodiment, the tα half-life is in the range of 30 minutes to 12 hours, inclusive. In another embodiment, the tα -half life of the polypeptide composition is in the range of 1 to 6 hours. In another embodiment, the tβ-half life of the polypeptide composition is increased by 10% or more. In another embodiment, the tβ-half life of the polypeptide composition is increased by 50% or more. In another embodiment, the tβ-half life of the polypeptide composition is increased by 2X or more. In another embodiment, the tβ-half life of the polypeptide composition is increased by 5X or more, e.g., 10X, 15X, 2OX, 25X, 3OX, 4OX, .or more. In another embodiment, the tβ-half life of the polypeptide composition is increased by 5OX or more.
In another embodiment, the antibody polypeptide composition has a tβ half- life of 1 to 170 hours, inclusive. In another embodiment, the tβ-half life is in the range of 12 to 48 hours, inclusive. In another embodiment, the tβ-half life is in the range of 12 to 26 hours, inclusive.
In another embodiment, the composition has an AUC value (area under the curve) in the range of 1 mg.min/ml or more. In one embodiment, the lower end of the range is 5, 10, 15, 20, 30, 100, 200 or 300 mg.min/ml. In addition, or alternatively, a ligand or composition according to the invention has an AUC in the range of up to 600 mg.min/ml. In one embodiment, the upper end of the range is 500, 400, 300, 200, 150, 100, 75 or 50 mg.min/ml. Advantageously a ligand according to the invention will have an AUC in the range selected from the group consisting of the following: 15 to 150 mg.min/ml, 15 to 100 mg.min/ml, 15 to 75 mg.min/ml, and 15 to 50 mg.min/ml. In another embodiment, the antibody polypeptide is linked to human serum albumin (HSA). In another embodiment, the antibody polypeptide has an increased in vivo half-life relative to the same polypeptide composition lacking linked HSA. In another embodiment, the antibody polypeptide has a tα-half life that is increased by 10% or more relative to a molecule lacking linked HSA. In another embodiment, the tα-half life of the polypeptide composition is in the range of 0.25 minutes to 6 hours. In another embodiment, the tβ-half life of the polypeptide composition is increased by 10% or more. In another embodiment, the tβ-half life is in the range of 12 to 48 hours.
In another embodiment, the antigen-binding polypeptide comprises an amino acid sequence selected from the group consisting of SEQ ID NOs 7-82 and 246-360.
In another embodiment, the antigen-binding polypeptide is free of an Fc domain.
In another aspect, the invention encompasses an immunoglobulin variable domain polypeptide which has an amino acid sequence at least 85% identical to a sequence selected from the group consisting of SEQ ID NOs 7-82 and 246-360, which polypeptide specifically and monovalently binds CD40L.
In one embodiment, the immunoglobulin variable domain polypeptide antagonizes the binding of CD40L to CD40.
In another embodiment, the immunoglobulin variable domain polypeptide inhibits the binding of CD40 to CD40L and has an IC50 in the range of 20 pM to 1.5 μM, inclusive. For example, the IC50 can be in the range of 20 pM to 1 μM, 20 pM to 900 nM, 20 pM to 800 nM, 20 pM to 700 nM, 20 pM to 600 nM, 20 pM to 500 nM, 20 pM to 400 nM, 20 pM to 300 nM, 20 pM to 200 nM, 20 pM to 100 nM, or 20 pM to 50 nM. Further acceptable or preferred ranges include, for example, 50 pM to 1 μM, 100 pM to 500 nM, 125 pM to 250 nM, 150 pM to 200 nM, 150 pM to 100 nM and 200 pM to 50 nM.
In another embodiment, the immunoglobulin variable domain polypeptide inhibits the interaction of CD40 with CD40L, but does not substantially agonize intracellular signaling by CD40. In a preferred embodiment, the binding of the polypeptide to CD40L does not substantially induce JNK phosphorylation in Jurkat
T-cells. In another preferred embodiment, the binding of the polypeptide to CD40L does not substantially induce IFN-γ secretion by Jurkat T-cells co-stimulated with anti-CD3 antibody. In another preferred embodiment, the binding of the antibody polypeptide to CD40L does not substantially induce platelet aggregation in a platelet aggregation assay.
In another embodiment, the antigen-binding polypeptide further comprises a second antibody polypeptide which binds a ligand other than CD40L. In a preferred embodiment, the second antibody polypeptide binds a ligand selected from the group consisting of HSA, TNFα, IL-I, IL-2, IL-4, IL-6, IL-δ, IL-12, IL-18, IFN-γ, CD2, CD4, COS, CTLA4, LFAl, LFA3 and VLA4.
In one embodiment, the invention relates to an antibody polypeptide comprising an immunoglobulin variable domain which specifically and monovalently binds CD40L (e.g., an anti-CD40L dAb, FAb, an scFv, an Fv, or a disulfide-bonded Fv), and which 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 sequence of one or more of said framework regions collectively comprises 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.
In one embodiment, the amino acid sequences of FWl, FW2, FW3 and FW4 of the anti-CD40L variable domain or dAb 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. In a further embodiment, the amino acid sequences of FWl, FW2 and FW3 of the anti-CD40L variable domain or dAb are the same as the amino acid sequences of corresponding framework regions encoded by human germline antibody gene segments. In a further embodiment of the foregoing, the human germline antibody gene segment can be selected from the group consisting of DP47, DP45. DP48 and DPK9.
The invention further encompasses a method of antagonizing the binding of CD40 to CD40L in an individual, the method comprising administering a monovalent anti-CD40L antibody polypeptide as described Herein to the individual, wherein the polypeptide antagonizes the binding of CD40 to CD40L in the individual.
The invention further encompasses a method of antagonizing an activity of CD40 or CD40L in an individual, the method comprising administering a monovalent anti-CD40L antibody polypeptide as described herein to the individual, wherein the polypeptide antagonizes an activity of CD40 or CD40L or both.
The invention further encompasses a composition comprising an extended release formulation comprising a monovalent anti-CD40L antibody polypeptide, preferably, but not limited to, a polypeptide comprising a single immunoglobulin variable domain that binds CD40L. In one embodiment, the single immunoglobulin variable domain is a non-human mammalian single immunoglobulin variable domain. In another embodiment, the single immunoglobulin variable domain is a human single immunoglobulin variable domain.
The invention further encompasses a method of treating or preventing a disease or disorder mediated by CD40L in an individual in need of such treatment, the method comprising administering to the individual a therapeutically effective amount of a composition comprising a monovalent anti-CD40L antibody polypeptide, preferably a composition comprising a single human immunoglobulin variable domain that binds CD40L. In one embodiment, the disease or disorder is an autoimmune disease or disorder.
The invention further encompasses a method of treating or preventing a symptom of systemic lupus erythematosus (SLE) in an individual, the method comprising administering a monovalent anti-CD40L antibody polypeptide to said individual in an amount effective to treat or prevent a symptom of SLE. The invention further encompasses a method of reducing or alleviating a symptom of a disease such as systemic lupus erythematosis, multiple sclerosis, rheumatoid arthritis, allograft rejection, xenograft rejection, and Diabetes, including insulin-dependent Type I Diabetes.
The invention further encompasses an antibody polypeptide that is monovalent for binding to CD40L, wherein the antibody polypeptide comprises a universal framework.
In one embodiment, the universal framework comprises a VH framework selected from the group consisting of DP47, DP45 and DP38, and/or the VL framework is DPK9.
In another embodiment, the antibody polypeptide comprises a generic ligand binding site. In another embodiment, the generic ligand binding site binds a generic ligand selected from the group consisting of protein A, protein L and protein G.
In another embodiment, the antibody polypeptide comprises a variable domain having 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 the framework regions collectively comprises up to 5 amino acid differences relative to the amino acid sequence of the corresponding framework region encoded by a human germline antibody gene segment.
In another embodiment, the antibody polypeptide comprises a variable domain, wherein the amino acid sequences of FWl, FW2, FW3 and FW4 are the same as the amino acid sequences of corresponding framework regions encoded by a human germline antibody gene segment, or the antibody 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 the human germline antibody gene segment. In another embodiment, the antibody polypeptide comprises an antibody variable domain comprising FWl, FW2 and FW3 regions, and the amino acid sequence of said FWl, FW2 and FW3 are the same as the amino acid sequences of corresponding framework regions encoded by human germline antibody gene segments. In another embodiment, the human germline antibody gene segment is selected from the group consisting of DP47, DP45, DP48 and DPK9.
The invention includes an antibody single variable domain polypeptide that binds to CD40L, wherein the polypeptide has an amino acid sequence that is identical to the amino acid sequence of DOM8-24, or differs from the amino acid sequence of DOM8-24 at no more than 25 amino acid positions and has a sequence that is at least 80% homologous to the sequence of DOM8-24. In one embodiment, the antibody single variable domain polypeptide differs form the amino acid sequence of DOM8- 24 at 25 or fewer amino acid positions, 20 or fewer amino acid positions, 15 or fewer amino acid positions, 10 or fewer amino acid positions, 5 or fewer amino acid positions, 2 or fewer amino acid positions, or as few as one amino acid position. In a further embodiment, the antibody single variable domain polypeptide is at least 80% homologous to the sequence of DOM8-24, for example, at least 85% homologous, at least 90% homologous, at least 95% homologous, and up to and including 96%, 97%, 98%, or 99% homologous.
The invention includes an antibody single variable domain polypeptide that binds to CD40L, wherein the polypeptide has an amino acid sequence that is identical to the amino acid sequence of DOM8-24, or differs from the amino acid sequence of DOM8-24 at no more than 25 amino acid positions and has a CDRl sequence that is at least 50% homologous to the CDRl sequence of DOM8-24, or has a CDR2 sequence that is at least 50% homologous to the CDR2 sequence of DOM8-24, or has a CDR3 sequence that is at least 50% homologous to the CDR3 sequence of DOM8- 24.
The invention also includes an antibody single variable domain polypeptide that binds CD40L, wherein the dAb has an amino acid sequence that is identical to the amino acid sequence of DOM8-24, or differs from the amino acid sequence of DOM8-24 at no more than 25 amino acid positions and has a CDRl sequence that is at least 50% homologous to the CDRl sequence of DOM8-24 and has a CDR2 sequence that is at least 50% homologous to the CDR2 sequence of DOM8-24 .
The invention also includes an antibody single variable domain polypeptide that binds CD40L, wherein the dAb has an amino acid sequence that is identical to the amino acid sequence of DOM8-24, or differs from the amino acid sequence of DOM8-24 at no more than 25 amino acid positions and has a CDR2 sequence that is at least 50% homologous to the CDR2 sequence of DOM8-24 and has a CDR3 sequence that is at least 50% homologous to the CDR3 sequence of DOM8-24 .
The invention also includes an antibody single variable domain polypeptide that binds CD40L, wherein the dAb has an amino acid sequence that is identical to the amino acid sequence of DOM8-24, or differs from the amino acid sequence of DOM8-24 at no more than 25 amino acid positions and has a CDRl sequence that is at least 50% homologous to the CDRl sequence of DOM8-24 and has a CDR3 sequence that is at least 50% homologous to the CDR3 sequence of DOM8-24 .
The invention also includes an antibody single variable domain polypeptide that binds CD40L, wherein the dAb has an amino acid sequence that is identical to the amino acid sequence of DOM8-24, or differs from the amino acid sequence of DOM8-24 at no more than 25 amino acid positions and has a CDRl sequence that is at least 50% homologous to the CDRl sequence of DOM8-24 and has a CDR2 sequence that is at least 50% homologous to the CDR2 sequence of DOM8-24 and has a CDR3 sequence that is at least 50% homologous to the CDR3 sequence of DOM8-24.
In one embodiment, the antibody single variable domain polypeptide that binds to CD40L, if not identical in sequence to that of DOM8-24, differs form the amino acid sequence of DOM8-24 at 25 or fewer amino acid positions, 20 or fewer amino acid positions, 15 or fewer amino acid positions, 10 or fewer amino acid positions, 5 or fewer amino acid positions, 2 or fewer amino acid positions, or as few as one amino acid position.
The invention also includes a CDAQiL antagonist having a CDRl sequence that is at least 50% homologous to the CDRl sequence of DOM8-24.
The invention also includes a CD40L antagonist having a CDR2 sequence that is at least 50% homologous to the CDR2 sequence of DOM8-24.
The invention also includes a CD40L antagonist having a CDR3 sequence that is at least 50% homologous to the CDR3 sequence of DOM8-24.
The invention also includes a CD40L antagonist having a CDRl sequence that is at least 50% homologous to the CDRl sequence of DOM8-24 and a CDR2 sequence that is at least 50% homologous to the CDR2 sequence of DOM8-24.
The invention also includes a CD40L antagonist having a CDR2 sequence that is at least 50% homologous to the CDR2 sequence of DOM8-24 and a CDR3 sequence that is at least 50% homologous to the CDR3 sequence of DOM8-24.
The invention also includes a CD40L antagonist having a CDRl sequence that is at least 50% homologous to the CDRl sequence of DOM8-24 and a CDR3 sequence that is at least 50% homologous to the CDR3 sequence of DOM8-24.
The invention also includes a CD40L antagonist having a CDRl sequence that is at least 50% homologous to the CDRl sequence of DOM8-24 and a CDR2 sequence that is at least 50% homologous to the CDR2 sequence of DOM8-24 and a
CDR3 sequence that is at least 50% homologous to the CDR3 sequence of DOM8-24.
In one embodiment the CD40L antagonist inhibits the binding of CD40 to
CD40L, and/or inhibits an activity of CD40 and/or CD40L, and/or results in no more than 25% platelet aggregation in a platelet aggregation assay. In one embodiment, the antagonist results in platelet aggregation of 25% or less, 20% or less, 15% or less,
10% or less, 5% or less, and as little as zero platelet aggregation. The invention" also includes a dual specific ligand comprising a first immunoglobulin single variable domain having a binding specificity to a first antigen and a second single variable domain having a binding activity to a second antigen, wherein the first antigen is CD40L and binding of the second single variable domain to the second antigen acts to increase the half-life of the ligand in vivo. In one embodiment, the dual specific ligand is a four chain IgG immunoglobulin.
In one embodiment, the four chain IgG comprises two dual specific ligands, said dual specific ligands being different in their variable domains.
The invention also includes a dual specific ligand comprising an anti-human CD40L dAb and an anti-SA dAb.
In one embodiment, the dAbs are Camelid VHH domains.
In one embodiment of the dual specific ligand, either (i) the first and second immunoglobulin variable domains are heavy chain variable domains; or (ii) the first and the second immunoglobulin variable domains are light chain variable domains.
In one embodiment, the ligand is provided as an IgG immunoglobulin comprising four heavy chain single variable domains or four light chain single variable domains. The heavy chain can comprise Camelid VHH domains.
In a further embodiment of the dual specific ligand, the first and second domains bind independently, such that the dual specific ligand may simultaneously bind both the first and second antigens.
In one embodiment of the dual specific ligand, the first single variable domain has a dissociation constant (Kd) of 1x10'8 M or less for human CD40L, and a KOff rate constant of IxIO"3 s"1 or less, as determined by surface plasmon resonance.
In one embodiment of the dual specific ligand, the second single variable domain is specific for serum albumin (SA) and has a dissociation constant (Kd) of InM to 500μm for SA, as determined by surface plasmon resonance. In a further embodiment, the second domain binds SA in a standard ligand binding assay with an IC50 of InM to 500μM. The second single variable domain may be specific for SA, and comprise the amino acid sequence of MSA-] 6 or a sequence that is at least 80% homologous thereto. Alternatively, the second single variable domain may be specific for SA, and comprise the amino acid sequence of MSA-26 or a sequence that is at last 80% homologous thereto.
In one embodiment of the dual specific ligand, the anti-CD40L variable domain or dAb comprises a universal framework. The anti-CD40L variable domain or dAb may also comprise a VH framework selected from the group consisting of DP47, DP45 and DP38; or a VL framework which is DPK9. In a further embodiment, the dual specific ligand or dAb can comprise a binding site for a generic ligand.
In one embodiment, the generic ligand binding site is selected from the group consisting of protein A, protein L and protein G binding site.
In one embodiment of the dual specific ligand, the anti-CD40L variable domain or dAb 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 sequence of one or more of said framework regions collectively comprises 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.
In one embodiment, the amino acid sequences of FWl, FW2, FW3 and FW4 of the anti-CD40L variable domain or dAb 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. In one embodiment, the amino acid sequences of said FWl, FW2 and FW3 of the anti-CD40L variable domain or 'dAb are the same as the amino acid sequences of corresponding framework regions encoded by human germline antibody gene segments. The human germline antibody gene segments are preferably selected from the group consisting of DP47, DP45, DP48 and DPK9.
The invention also includes a method for producing a dual specific ligand as described herein, comprising a first immunoglobulin single variable domain having a binding specificity for CD40L and a second single immunoglobulin single variable domain having a binding specificity for a protein which increases the half-life of the ligand in vivo, the method comprising the steps of:selecting a first variable domain by its ability to bind CD40L; selecting a second variable domain by its ability to bind to said protein; combining the variable domains; and selecting the ligand by its ability to bind to CD40L and said protein.
In one embodiment, the first variable domain is selected for binding to CD40L in absence of a complementary variable domain.
The invention also includes nucleic acid encoding a dual specific ligand described herein. The nucleic acid may comprise the nucleic acid sequence of MS A- 16 or a sequence that is at least 80% homologous thereto, or alternatively may comprise, the nucleic acid sequence of MSA-26 or a sequence that is at least 70% homologous thereto. The nucleic acid may be incorporated into a vector, which may be incorporated into a host cell.
The invention also includes a pharmaceutical composition comprising a dual specific ligand as described herein and a pharmaceutically acceptable excipient, carrier or diluent.
The invention also includes a dAb monomer specific for CD40L, which monomer has a dissociation constant (Ka) of 1x10"8 M or less for human CD40L, and a Koff rate constant of 1x10"3 s"1 or less, as determined by surface plasmon resonance. In one embodiment, the dAb monomer specific for CD40L has a dissociation constant (Kd) of 1x10"7 M or less, as determined by surface plasmon resonance.
In one embodiment, the dAb monomer has binding specificity to CD40L with a dissociation constant (Kd) of IxIO"8 M or less, as determined by surface plasmon resonance.
In one embodiment, the dAb monomer has binding specificity to CD40L with a dissociation constant (Kd) of 5OnM to 2OpM, as determined by surface plasmon resonance.
In one embodiment, the monomer inhibits binding of CD40 to CD40L with an IC50 of 5OnM or less.
In a further embodiment, the dAb monomer has binding specificity to CD40L with a KOff rate constant of 1x10"3 s"1 or less, 1x10"4 s"1 or less, 1x10"5 s"1 or less, or 1x10"6 s"1 or less,as determined by surface plasmon resonance.
In one embodiment, the dAb monomer neutralizes CD40L in a standard assay with an ND50 of 5OnM or less.
In invention also includes a dual specific ligand comprising first and second heavy chain single variable domains, or first and second light chain single variable domains, wherein the first variable domain is an anti-CD40L dAb monomer.
In one embodiment, the second variable domain has binding specificity for an antigen other than CD40L.
In a further embodiment, the second variable domain has binding specificity for an antigen selected from the group consisting of EPO receptor, ApoE, Apo-SAA, BDNF5 Cardiotrophin-1, EGF, EGF receptor, ENa-78, Eotaxin, Eotaxin-2, Exodus-2, EpoR, FGF-acidic, FGF-basic, fibroblast growth factor- 10, FLT3 ligand, Fractalkine (CX3C), GDNF, G-CSF, GM-CSF, GF-bl, insulin, IFN-g, IGF-I, IGF-II, IL-Ia, U-Ib, IL-2, 11-3, IL-4, IL-5, IL-6, IL-7, IL-8 (72 a.a.), IL-8 (77 a.a.), IL-9, IL-10, IL-11, IL- 12, IL-13, IL-15, EL-16, IL-17, IL-18 (IGIF), Inhibin a, Inhibin b, IP-10, keratinocyte growth factor-2 (KGF-2), KGF, Leptin, LIF, Lymphotactin. Mullerian inhibitory substance, monocyte colony inhibitor)' factor, monocyte attractant protein, M-CSF, MDC (67 a.a.), MDC (69 a.a.), MCP-I (MCAF), MCP-2, MCP-3, MCP-4, MDC (67 a.a.), MIG, MIP-Ia, MIP-Ib5 MIP-Sa, MIP-3b, MIP-4, myeloid progenitor inhibitor factor-1 (MPIF-I), NAP -2, Neurturin, Nerve growth factor, b-NGF, NT-3, NT-4, Oncostatin M, PDGF-AA, PDGF-AB, PDGF-BB, PF-4, RANTES, SDFIa, DFGIb SCF, SCGF, stem cell factor, (SCF), TARC, TGF-a, TGF-b, TGF-b2,< TGF-b3, tumour necrosis factor (TNF), TNF-as TNF-b, TNF receptor I, TNF receptor II, TNIL-I, TPO, VEGF, VEGF receptor 1, VEGF receptor 2, VEGF receptor-3, GCP-2, GRO/MGSA, GRO-b, GRO-g, HCCl, 1-309, HER 1, HER 2, HER 3, HER 4, TACE recognition site, TNF BP-I, TNF BP-II, and an antigen disclosed in Annex 2 or 3.
The invention also includes an antibody polypeptide that antagonizes or inhibits the binding of DOM8-24 to CD40L, or an antibody polypeptide that binds to the same epitope of CD40L bound by DOM8-24.
The invention also includes a dual specific ligand comprising a . first immunoglobulin single variable domain having a binding specificity to a first antigen and a second single variable domain having a binding activity to a second antigen, wherein the first antigen is CD40L and the second single variable domain is an Antigen Presenting Cell surface antigen or a T cell surface antigen. The Antigen Presenting Cell surface antigen can be selected from one of the group consisting of dendritic cell surface antigens, activated macrophage surface antigens, activated B cell surface antigens, co-stimulatory signal pathway surface antigens, and MHC.
In one embodiment, the MHC is class II, and the class II can be alpha or beta.
The Antigen Presenting Cell surface antigen may be selected from the group consisting of CD28, Inducible costimulatory molecule (ICOS), CD27, CD30, OX40, CD45, CD69, CD3, CD70, Inducible costimulatory molecule ligand (ICOSL)5 OX40L, CD80, CD86, HVEM (Herpes Virus Entry Mediator), and LIGHT, but is preferably one of CD28, Inducible costimulatory molecule (ICOS), CD27, CD30, OX40, CD45, CD69, or CD3. The surface antigen is preferably a B7 gene surface antigen such as B7-2 or B7-1.
Definitions:
As used herein, the term "human" when applied to an antibody polypeptide or to an immunoglobulin variable domain means that the polypeptide has a sequence derived from a human immunoglobulin. A sequence is "derived from" a human immunoglobulin coding sequence when the sequence is either: a) isolated from a human individual or from cells or a cell line from a human individual; b) isolated from a library of cloned human antibody gene sequences (or a library of human antibody V domain sequences); or c) when a cloned human antibody gene sequence (or a cloned human V region sequence (including, e.g., a germline V gene segment)) was used to generate one or more diversified sequences that were then selected for binding to a desired target antigen. The term "human" as applied herein to an antibody polypeptide or to an immunoglobulin variable domain does not encompass an immunoglobulin from another species, e.g., mouse, camel, etc., that has been "humanized" through grafting of human constant region sequences onto an antibody polypeptide (i.e., replacing non-human constant regions with human constant regions) or through grafting of human V region framework sequences onto an immunoglobulin variable domain from a non-human mammal (i.e., replacing non-human framework regions of a V domain with human framework regions).
At a minimum, a human variable domain has at least 85% amino acid similarity (including, for example, 87%, 90%, 93%, 95%, 97%, 99% or higher similarity) to a naturally-occurring human immunoglobulin variable domain sequence.
As used herein, the term "domain" refers to 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. By "single immunoglobulin variable domain" is meant a folded polypeptide domain which comprises a sequence characteristic of immunoglobulin variable domains and which specifically binds an antigen (e.g., dissociation constant of 500 nM or less). A "single immunoglobulin variable domain" therefore includes complete antibody variable domains as well as 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 a dissociation constant of 500 nM or less (e.g., 450 nM or less, 400 nM or less, 350 nM or less, 300 nM or less, 250 nM or less, 200 nM or less, 150 nM or less, 100 nM or less) and the target antigen specificity of the full- length domain. Where necessary or in case of any doubt, the numbering convention and boundaries set forth by Kabat et al. (1991, supra) are applicable to immunoglobulin variable and constant domains referred to herein. An antibody single variable domain polypeptide, as used herein refers to a mammalian single immunoglobulin variable domain polypeptide, preferably human, but also includes rodent (for example, as disclosed in WO00/29004, the contents of which are incorporated herein in their entirety) or camelid VHH dAbs. Camelid dAbs are antibody 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: VHH- VHH molecules are about 10x smaller than IgG molecules, and as single polypeptides, they are very stable, resisting extreme pH and temperature conditions. Moreover, camelid antibody single variable domain polypeptides are resistant to the action of proteases. Camelid antibodies are described in, for example, U.S. Pat. Nos. 5,759,808; 5,800,988; 5,840,526; 5,874,541; 6,005,079; and 6,015,695, the contents of each of which are incorporated herein in their entirety. Camelid VHH antibody single variable domain polypeptides useful according to the invention include a class of camelid antibody single variable domain polypeptides having human-like sequences, wherein the class is characterized in that the VHH domains carry an amino acid from the group consisting of glycine, alanine, valine, leucine, isoleucine, proline, phenylalanine, tyrosine, tryptophan, methionine, serine, threonine, asparagine, or glutamine at position 45, such as for example L45, and further comprise a tryptophan at position 103 according to the Kabat numbering. Humanized camelid VHH polypeptides are taught for example in WO04/041862, the teachings of which are incorporated herein in their entirety. It will be understood by one of skill in the art that naturally occurring camelid antibody single variable domain polypeptides may be modified according to the teachings of WO04/041862 (e.g., amino acid substitutions at positions 45 and 103) to generate humanized camelid VHH polypeptides. Also included in the present invention are antibody single variable domain polypeptides which are nurse shark VHH- Nurse shark dAbs are antibody single variable domain polypeptides derived from the nurse shark, that comprise heavy chain antibodies naturally devoid of light chain: VHH- Nurse Shark VHH dAbs are described, for example, in Greenberg et al. (Nature 374 pp 168- 173 1995) and U.S. 20050043519.
The phrase "single immunoglobulin variable domain polypeptide" encompasses not only an isolated single immunoglobulin variable domain polypeptide, but also larger polypeptides that comprise a monomer of a single immunoglobulin variable domain polypeptide sequence. A "domain antibody" or
"dAb" is equivalent to a "single immunoglobulin variable domain polypeptide" as the term is used herein. With regard to a single immunoglobulin variable domain polypeptide, the binding to antigen, e.g., CD40L, is mediated by the single immunoglobulin V domain without a requirement for a complementary V domain.
According to the invention, the terms "antibody single variable domain polypeptide", "antibody single variable domain", "single antibody variable domain", and "single immunoglobulin variable domain" are understood to be equivalent. As used herein, the phrase "sequence characteristic of immunoglobulin variable domains" refers to an amino acid sequence that is homologous, over 20 or more, 25 or more, 30 or more, 35 or more, 40 or more, 45 or more, or even 50 or more contiguous amino acids, to a sequence comprised by an immunoglobulin variable domain sequence. Sequences similar or homologous (e.g., at least about 70% sequence identity) to the sequences disclosed herein are also part of the invention. In some embodiments, 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. At the nucleic acid level, the sequence identity can be about 70%, 75%, 80%, 85%, 90%, 91%, 92%,
93%, 94%, 95%, 96%, 97%, 98%, 99% or higher. Alternatively, 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.
As used herein, the terms "homology" or "similarity" refer to the degree with which two nucleotide or amino acid sequences structurally resemble each other. As used herein, sequence "similarity" is a measure of the degree to which amino acid sequences share similar amino acid residues at corresponding positions in an alignment of the sequences. Amino acids are similar to each other where their side chains are similar. Specifically, "similarity" encompasses amino acids that are conservative substitutes for each other. A "conservative" substitution is any substitution that has a positive score in the blosum62 substitution matrix (Hentikoff and Hentikoff, 1992, Proc. Natl. Acad. Sci. USA 89: 10915-10919). By the statement "sequence A is n% similar to sequence B" is meant that n% of the positions of an optimal global alignment between sequences A and B consists of identical amino acids or conservative substitutions. Optimal global alignments can be performed using the following parameters in the Needleman-Wunsch alignment algorithm:
For polypeptides: Substitution matrix: blosum62.
Gap scoring function: -A -B*LG, where A=I l (the gap penalty), B=I (the gap length penalty) and LG is the length of the gap. For nucleotide sequences:
Substitution matrix: 10 for matches, 0 for mismatches. Gap scoring function: -A -B*LG where A=50 (the gap penalty), B=3
(the gap length penalty) and LG is the length of the gap. Typical conservative substitutions are among Met, VaI, Leu and lie; among Ser and Thr; among the residues Asp, GIu and Asn; among the residues GIn, Lys and Arg; or aromatic residues Phe and Tyr.
As used herein, two sequences are "homologous" or "similar" to each other where they have at least 70%, 80%, or 85% sequence similarity to each other, including, e.g., 90%, 95%, 97%, 99% or even 100% sequence similarity, when aligned using either the Needleman-Wunsch algorithm or the "BLAST 2 sequences" algorithm described by Tatusova & Madden, 1999, FEMS Microbiol Lett. 174:247-
250. Where amino acid sequences are aligned using the "BLAST 2 sequences algorithm," the Blosum 62 matrix is the default matrix.
As used herein, the teπns "inhibit," "inhibits" and "inhibited" refer to a decrease in a given measurable activity (e.g., binding activity) by at least 10% relative to a reference. Where inhibition is desired, such inhibition is preferably at least 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or more, up to and including 100%, i.e., complete inhibition or absence of the given activity. One way that inhibition of CD40L binding to CD40 is measured is as described in Example 6 herein. As used herein, the term "substantially inhibits" refers to a decrease in a given measurable activity (e.g., the binding of CD40L to CD40) by at least 50% relative to a reference. For example, "substantially inhibits" refers to a decrease in a given measurable activity of at least 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% and up to and including 100% relative to a reference. As used herein, "inhibits the binding", with reference to the binding of an antibody polypeptide binding to CD40L, or CD40 binding to CD40L, refers to a decrease in binding by at least 10% relative to a reference. "Inhibits the binding" preferably refers to a decrease in binding of at least 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or more, up to and including 100%.
As used herein, the terms "activate," "activates" and "activated" refer to an increase in a given measurable activity by at least 5% relative to a reference, for example, at least 10%, 25%, 50%, 75% or even 100%. As used herein, the term "antagonist" refers to an agent that inhibits at least one activity mediated by CD40L. inhibits the binding of CD40 to CD40L, and/or results in no more than 25% platelet activation and/or aggregation in a platelet aggregation assay or platelet activation assay as described herein, and preferably results in 25% or less platelet activation and/or aggregation, 20% or less, 15% or less, 10% or less, 5% or less, and as little as zero platelet activation and/or aggregation. An activity is "antagonized" if the activity (i.e., CD40L mediated activity, binding of CD40 or CD40L, or platelet activation and/or aggregation) is reduced by at least 10%, and preferably at least 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 97% or even 100% (i.e., no activity) in the presence, relative to the absence of an antagonist. An antagonist as the term is used herein preferably comprises a single immunoglobulin variable domain that binds monovalently to CD40L.
As used herein, the term "agonist" refers to an agent that activates at least one activity mediated by CD40L, either alone or when combined with another co- stimulus, relative to a reference. An activity is "agonized" if the activity is increased by at least 10%, e.g., 50%, in the presence, relative to the absence of an agonist.
As used herein, the term "epitope" refers to a unit of structure conventionally bound by an immunoglobulin VVVL 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 immunoglobulin variable domain, an epitope represents the unit of structure bound by a single variable domain in isolation. That is, the binding site is provided by one, single immunoglobulin variable domain.
As used herein, the term "extended release" or the equivalent terms "controlled release" or "slow release" refer to drug formulations that release active drug, such as a polypeptide drug, over a period of time following administration to an individual. Extended release of polypeptide drugs, which can occur over a range of desired times, e.g., minutes, hours, days, weeks or longer, depending upon the drug formulation, is in contrast to standard formulations in which substantially the entire dosage unit is available for immediate absorption or immediate distribution via the bloodstream. Preferred extended release formulations result in a level of circulating drug from a single administration that is sustained, for example, for 8 hours or more, 12 hours or more, 24 hours or more, 36 hours or more, 48 hours or more, 60 hours or more, 72 hours or more 84 hours or more, 96 hours or more, or even, for example, for 1 week or 2 weeks or more, for example, 1 month or more,
As used herein, a "CD40L activity" is an activity involving or resulting from the binding of CD40L to CD40, and includes, but is not limited to binding to CD40 (assayed, for example, according to the method described in Example 6), activation of Jun-N-terminal Kinase (JNK), the induction of T cells to produce and secrete cytokines including, for example, IL-IO, IFN-γ and TNF-α, and the mediation of platelet activation and/or aggregation. Assays for these activities are provided herein below.
As used herein, the term "does not substantially agonize" means that a given agent, e.g., an anti-CD40L antibody polypeptide, does not activate one or more of the CD40L activities including Jun-N-terminal kinase activation (phosphorylation) in Jurkat T cells and induction of IFN-γ production or secretion in anti-CD3-stimulated Jurkat T cells, as the term "activate" is defined herein. As used herein, "does not substantially agonize" means that the agent does not activate more than 20% of the activity which is activated by CD40 binding to CD40L, preferably, the agent does not activate more than 10%, 8%, 5%, 3%, or more than 2% or less, including zero activation, of the activity which is activated by CD40 binding to CD40L.
As used herein, the term "antibody polypeptide" refers to a polypeptide which either is an antibody or is a part of an antibody, modified or unmodified, which retains the ability to specifically bind antigen. Thus, the term antibody polypeptide includes an antigen-binding heavy chain, light chain, heavy chain-light chain dimer, Fab fragment, F(ab')2 fragment, dAb, or an Fv fragment, including a single chain Fv (scFv). The phrase "antibody polypeptide" is intended to encompass recombinant fusion polypeptides that comprise an antibody polypeptide sequence that retains the ability to specifically bind antigen in the context of the fusion. As used herein, the term "monovalent" means that a given antibody polypeptide or single immunoglobulin variable domain polypeptide can bind only a single molecule of its target. Naturally-occurring antibodies are generally divalent, in that they have two functional antigen-binding arms, each comprising a VH and a VL domain. Where steric hindrance is not an issue, a divalent antibody can bind two separate molecules of the same antigen. In contrast, a "monovalent" antibody has the capacity to bind only one such antigen molecule. As the term is used herein, a "monovalent" antibody can also comprise more than one antigen binding site, e.g., two antigen bindiαg sites, but the binding sites must be for different antigens, such that the antibody can only bind one molecule of CD40L at a time. The antigen- binding domain of a monovalent antibody can comprise a VH and a VL domain, but preferably comprises only a single immunoglobulin variable domain, i.e., a VH or a VL domain, that has the capacity to bind CD40L without the need for a corresponding VL or VH domain, respectively. A monovalent antibody lacks the capacity to cross link molecules of a single antigen.
As used herein, the term "standard platelet aggregation assay" means the assay described in the section herein below, entitled "Platelet Aggregation Assaj'."
As used herein, the terms "VH domain" and "VL domain" refer to immunoglobulin variable regions as defined by Kabat et al. (supra), which is incorporated herein by reference.
.As used herein, "linked" refers to the attachment of a polymer moiety, such as PEG to an amino acid residue of an antibody polypeptide. Attachment of a PEG polymer to an amino acid residue of an antibody polypeptide, e.g., an anti-CD40L dAb, is referred to as "PEGylation" and may be achieved using several PEG attachment moieties including, but not limited to N-hydroxylsuccinimide (NHS) active ester, succinimidyl propionate (SPA), maleimide (MAL), vinyl sulfone (VS), or thiol. A PEG polymer, or other polymer, can be linked to an antibody polypeptide at either a predetermined position, or may be randomly linked to the an antibody polypeptide molecule. It is preferred, however, that the PEG polymer be linked to an antibody polypeptide at a predetermined position. A PEG polymer may be linked to any residue in the an antibody polypeptide, however, it is preferable that the polymer is linked to either a lysine or cysteine, which is either naturally occurring in the antibody polypeptide, or which has been engineered into the antibody polypeptide, for example, by mutagenesis of a naturally occurring residue in the antibody polypeptide to either a cysteine or lysine. PEG-linkage can also be mediated through a peptide linker attached to an antibody polypeptide. That is, the PEG moiety can be attached to a peptide linker fused to an antibody polypeptide, where the linker provides the site, e.g., a free cysteine or lysine, for PEG attachment. As used herein, "linked" can also refer to the association of two or more antibody polypeptides, e.g., dAb monomers, to form a dimer, trimer, tetramer, or other multimer. Antibody polypeptide monomers can be linked to form a multimer by several methods known in the art, including, but not limited to, expression of the antibody polypeptide monomers as a fusion protein, linkage of two or more monomers via a peptide linker between monomers, or by chemically joining monomers after translation, either to each other directly, or through a linker by disulfide bonds, or by linkage to a di-, tri- or multivalent linking moiety (e.g., a multi-arm PEG). While dAb multimers are specifically contemplated herein, e.g., in the context of dual- or multi-specific antibody polypeptide constructs, it is emphasized that for any given antibody polypeptide construct, the construct should only be able to bind one molecule of CD40L, i.e., the constructs can have only one CD40L-binding element, and cannot cross link CD40L.
As used herein, "polymer" refers to a macromolecule made up of repeating monomeric units, and can refer to a synthetic or naturally occurring polymer such as an optionally substituted straight or branched chain polyalkylene, polyalkenylene, or polyoxyalkylene polymer or a branched or unbranched polysaccharide. A "polymer" as used herein, specifically refers to an optionally substituted or branched chain poly(ethylene glycol), poly(propylene glycol), or polyvinyl alcohol) and derivatives thereof. As used herein, "PEG" or "PEG polymer" refers to polyethylene glycol, and more specifically can refer to a derivitized form of PEG, including, but not limited to N-hydroxylsuccinimide (NHS) active esters of PEG such as succinimidyl propionate, benzotriazole active esters, PEG derivatized with maleimide, vinyl sulfones, or thiol groups. Particular PEG formulations can include PEG-O-CH2CH2CH2-CO2-NHS; PEG-O-CH2-NHS; PEG-O-CH2CH2-CO2-NHS; PEG-S-CH2CH2-CO-NHS; PEG- O2CNH-CH(R)-CO2-NHS; PEG-NHCO-CH2CH2-CO-NHS; and PEG-O-CH2-CO2- NHS; where R is (CH?)4)NHCO2(mPEG). PEG polymers useful in the invention may be linear molecules, or may be branched wherein multiple PEG moieties are present in a single polymer. Some particularly preferred PEG conformations that are useful in the invention include, but are not limited to the following:
Figure imgf000041_0001
mPEG-MAL mPEG2-MAL
Figure imgf000041_0002
mPEG2-(MAL)2
Figure imgf000041_0003
As used herein, a "sulfhydryl-selective reagent" is a reagent which is useful for the attachment of a PEG polymer to a thiol-containing amino acid. Thiol groups on the amino acid residue cysteine are particularly useful for interaction with a sulfhydryl-selective reagent. Sulfhydryl-selective reagents which are useful for such attachment include, but are not limited to maleimide, vinyl sulfone, and thiol. The use of sulfhydryl-selective reagents for coupling to cysteine residues is known in the art and may be adapted as needed according to the present invention (See Eg., Zalipsky, 1995, Bioconjug. Chem. 6:150; Greenwald et al., 2000, Crit. Rev. Ther. Drug Carrier Syst. 17:101; Herman et al., 1994, Macromol. Chem. Phys. 195:203).
The attachment of PEG or another agent, e.g., HSA, to an antibody polypeptide or to a single immunoglobulin variable domain polypeptide as described herein will preferably not impair the ability of the polypeptide to specifically bind CD40L. That is, the PEG-linked antibody polypeptide or single immunoglobulin variable domain polypeptide will retain its binding activity relative to a non-PEG- linked counterpart. As used herein, "retains activity" refers to a level of activity of a PEG-linked antibody polypeptide which is at least 10% of the level of activity of a non-PEG-linked antibody polypeptide, preferably at least 20%, 30%, 40%, 50%, 60%, 70%, 80% and up to 90%, preferably up to 95%, 98%, and up to 100% of the activity of a non-PEG-linked antibody polypeptide comprising the same antigen- binding domain or domains. More specifically, the activity of a PEG-linked antibody polypeptide compared to a non-PEG linked antibody variable domain should be determined on an antibody polypeptide molar basis; that is equivalent numbers of moles of each of the PEG-linked and non-PEG-linked antibody polypeptides should be used in each trial. In determining whether a particular PEG-linked antibody polypeptide "retains activity", it is preferred that the activity of a PEG-linked antibody polypeptide be compared with the activity of the same antibody polypeptide in the absence of PEG.
As used herein, the term "in vivo half-life" refers to the time taken for the serum concentration of a ligand (e.g., a single immunoglobulin variable domain) 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 anti CD40L antibody polypeptides or single immunoglobulin variable domain polypeptides described herein can be stabilized in vivo and their half-life increased by binding to molecules, such as PEG, which resist degradation and/or clearance or sequestration. The half-life of an antibody polypeptide is increased if its functional activity persists, in vivo, for a longer period than a similar antibody polypeptide which is not linked to a PEG polymer. Typically, the half life of a PEGylated antibody polypeptide is increased by 10%, 20%, 30%, 40%, 50% or more relative to a non-PEGylated antibody polypeptide. Increases in the range of 2x, 3x, 4x, 5x, 10x, 2Ox, 3Ox, 40x, 5Ox or more of the half life are possible. Alternatively, or in addition, increases in the range of up to 3Ox, 4Ox, 50x, 60x, 70x, 80x, 9Ox5 10Ox, 15Ox of the half life are possible. According to the invention, a PEG-linked antibody single variable domain has a half- life of between 0.25 and 170 hours, preferably between 1 and 100 hours, more preferably between 30 and 100 hours, and still more preferably between 50 and 100 hours, and up to 170, 180, 190, and 200 hours or more.
As used herein, "resistant to degradation" or "resists degradation" with respect to a PEG or other polymer-linked antibody polypeptide monomer or multimer means that the PEG- or other polymer-linked antibody polypeptide monomer or multimer is degraded by no more than 10% when exposed to pepsin at pH 2.0 for 30 minutes and preferably not degraded at all.
As used herein, "hydrodynamic size" refers to the apparent size of a molecule (e.g., a protein molecule) based on the diffusion of the molecule through an aqueous solution. The diffusion, or motion of a protein through solution can be processed to derive an apparent size of the protein, where the size is given by the "Stokes radius" " or "hydrodynamic radius" of the protein particle. The "hydrodynamic size" of a protein depends on both mass and shape (conformation), such that two proteins having the same molecular mass may have differing hydrodynamic sizes based on the overall conformation of the protein. Hydrodynamic size is measured, for example, by size exclusion chromatography. The hydrodynamic size of a PEG-linked antibody polypeptide, e.g., a single immunoglobulin variable domain (including antibody variable domain multimers as described herein), can be in the range of 24 kDa to 500 kDa; 30 to 500 kDa; 40 to 500 kDa; 50 to 500 IcDa; 100 to 500 kDa; 150 to 500 IcDa; 200 to 500 IcDa; 250 to 500 kDa; 300 to 500 IcDa; 350 to 500 IcDa; 400 to 500 IcDa and 450 to 500 IcDa. Preferably the hydrodynamic size of a PEGylated antibody polypeptide of the invention is 30 to 40 kDa; 70 to 80 IcDa or 200 to 300 IcDa. Where a single immunoglobulin variable domain polypeptide is desired for use in imaging applications, the polypeptide should have a hydrodynamic size of between 50 and 100 IcDa. Alternatively, where a single immunoglobulin variable domain polypeptide is desired for therapeutic applications, the polypeptide preparation should have a hydrodynamic size of greater than 200 kDa.
As used herein, the term "IC50" refers to the concentration of an inhibitor necessary to inhibit a given activity by 50%. IC50 is determined by assaying a given activity, e.g., binding of CD40L to CD40, in the presence of varying amounts of the inhibitor (e.g., monovalent anti-CD40L antibody polypeptide), and plotting the inhibitor concentration versus the activity being targeted. Binding of CD40L to CD40 is measured herein by the method described in Example 6. Alternatively, SPR can be used.
As used herein, the term "fused to an antibody polypeptide" means that a polypeptide is fused to a given antibody through use of recombinant DNA techniques. Thus, an antibody "fused to" another polypeptide, e.g., to another antibody of different binding specificity, does not exist in nature and is generated through recombinant means. The term "fused to an antibody polypeptide" also encompasses the linkage of a polypeptide to a given antibody polypeptide through, for example, disulfide or other chemical linkages, where the fused polypeptide is not naturally found fused to the antibody polypeptide. Recombinant and chemical methods of fusing a polypeptide to another polypeptide, e.g., to an antibody, are well known in the art.
As used herein, the term "Fc domain" refers to the constant region antibody sequences comprising CH2 and CH3 constant domains as delimited according to Kabat et al., supra. The Fc portion of the heavy chain polypeptide has the ability to self-associate, a function which facilitates the formation of divalent antibodies. The term "lacks an Fc domain" means that a given antibody polypeptide lacks at least the portion of an immunoglobulin Fc domain (as such domains are defined according to Kabat et al., supra) sufficient to mediate the dimerization of Fc-containing antibody polypeptides. Dimerization of Fc-containing antibody polypeptides is measured, for example, by chromatographic methods or by surface plasmon resonance. An antibody polypeptide lacking an Fc domain avoids Fc-platelet interactions and therefore avoids induction of platelet aggregation.
As used herein "treat", "reduce", "prevent", or "alleviate" as it relates to a symptom of disease refer to a decrease of the a symptom by at least 10% based on a a clinically measurable parameter, or by at least one point on a clinically-accepted scale of disease or symptom severity. As used herein, the term "symptom of systemic lupus erythematosus" refers to any of the clinically relevant symptoms of SLE known to those of skill in the art. Non-limiting examples include the accumulation of IgG autoantibodies (e.g., against nuclear antigens such as chromatin, snRHPs (especially Ul, Sm, Ro/SSA and La/SSB), phospholipids and cell surface molecules), hemolytic anemia, thrombocytopenia, leukopenia, glomerulonephritis, vasculitis, arthritis, and serositis). A reduction in such a symptom is a reduction by at least 10% in a clinically measurable parameter, or by at least one point on a clinically-accepted scale of disease severity.
As used herein, the phrase "specifically binds" refers to the binding of an antigen by an immunoglobulin variable domain with a dissociation constant (Kd) of 1 μM or lower as measured by surface plasmon resonance analysis using, for example, a BIAcore™ surface plasmon resonance system and BIAcoreT kinetic evaluation software (e.g., version 2.1). The affinity or Kd for a specific binding interaction is preferably about 500 nM or lower, more preferably about 300 nM or lower.
As used herein, a "generic ligand" is a ligand that binds a substantial proportion of functional members in a given repertoire, e.g., in a phage display library. Thus, the same generic ligand can bind many members of the repertoire regardless of their target ligand specificities. In general, the presence of a functional generic ligand binding site indicates that the repertoire member is expressed and folded correctly. Thus, binding of the generic ligand to its binding site provides a method for preselecting functional polypeptides from a repertoire of polypeptides. Generic ligands include, for example, Protein A, Protein G and Protein L.
As used herein, the term "universal framework" refers to a single antibody framework sequence corresponding to the regions of an antibody conserved in sequence as defined by Kabat (supra) 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.
BRIEF DESCRIPTION OF THE FIGURES
Figure 1 shows gel analysis of the quality of biotin-labeled CD40L used in the screening procedures described herein, (a) 1 μg of non-bio tinylated-CD40L (Lane 1) and 0.3 μg of biotin-CD40L (Lane 2) were analysed on SDS-PAGE and detected by Simply Blue Safe-Stain, (b) 0.1 μg of biotin-CD40L (Lane 1) and 0.02 μg of biotin- CD40L (Lane 2) were detected by Western-blot probing with 1 :5000 Streptavidin- HRP.
Figure 2 shows a graphical representation of a dose response receptor binding assay (RBA) readout, analysing the inhibition of CD40L binding to CD40-Fc by dAbs DOM-IO, -20, -27, -30, -31, -62, -77, titrated from 1 μM down to 10 pM. dAbs DOM-20, -30, and -31 are the most potent with IC50 values of approximately 8 nM.
Figure 3 shows a graphical representation of a dose response receptor binding assay readout, analysing the inhibition of CD40L binding to CD40-Fc by dAbs DOM- 4. and DOM-5, titrated from 1 μM down to 500 pM. The IC50 values for dAbs DOM- 5 and DOM-4 are approximately 3 nM and 100 nM respectively.
Figure 4 shows a graphical representation of a dose response receptor binding assay readout, analysing the inhibition of CD40L binding to CD40-Fc by dAb DOM- 24, titrated from 100 nM down to 0.5 pM. The data were curve-fitted using GraphPad Prism software.
Figure 5 shows the sequence of the VH framework based on germline sequence DP47 - JH4b (SEQ E) NO: 1, amino acid sequence; SEQ E) NO: 2, nucleotide sequence - both sense and antisense strands are shown - SEQ E) NO: 2 is the top strand (sense) and SEQ E) NO: 476 is the lower strand (anti-sense)). HCDRs
1 -3 are indicated by underlining.
Figure 6 shows the sequence of the Vκ framework based on germline sequence DPK9 - Jκl (SEQ E) NO: 3, amino acid sequence; SEQ E) NO: 4, nucleotide sequence — both sense and antisense strands are shown - SEQ E) NO: 4 is the top strand (sense) and SEQ E) NO: 477 is the lower strand (anti-sense)). LCDRs
1-3 are indicated by underlining.
Figure 7 shows a schematic representation of the CD40L binding assay used herein, e.g., in Example 6.
Figure 8 shows various GASl secretion signal peptide coding sequences.
GAS wt: The natural occurring sequence in yeast. GAS E.Coli: The nucleotide sequence according to optimal E. CoIi codon usage (Wada et al. 1992 NAR 20 p 2111). GAS leader AT: AT rich nucleotide sequence. AU nucleotide sequences encode the same amino acid sequence. Yellow (light grey in greyscale) indicates nucleotides that are similar for all sequences. Blue (dark grey in greyscale) indicates nucleotides that are similar to the wt sequence. White (white in greyscale) indicates nucleotides that are different from the wt sequence. Figure 9 shows the results of -a receptor binding assay which demonstrates the affinity of PEGylated DOM8-24cys with either 3OK PEG MAL or 40K PEG2-MAL.
Figure 10 shows the results of an assay to assess the simultaneous binding of a dual specific dimer to HSA and CD40L (shaded bars). Binding to control BSA antigen is also shown (solid bars).
Figure 11 shows the results of an assay to assess the simultaneous binding of a dual specific Fab to HSA and CD40L (shaded bars). Binding to control skimmed mild powder antigen is also shown (solid bars).
45a Figure 12 shows the results of FACS analysis of the inhibitory effect of monomeric DOM-24 (grey dotted line). Control stimulated cells ae shown as the solid black line and a control dAb is shown as the grey solid line.
Figure 13 shows the results of FACS analysis of the inhibitory effect of the Vk dAb DOM-116 (dotted line). Control stimulated cells are shown as the solid black line and a control dAb is shown as the grey solid line.
DETAILED DESCRIPTION
The invention provides antibody polypeptides that are monovalent for binding to CD40L. Monovalency for CD40L binding removes the possibility for cross- linking that occurs with prior art antibodies, and which plays a role in undesirable side effects observed with anti-CD40L monoclonal antibodies. Further, while not wishing to be limited to any specific mechanism or theory, because antibody polypeptides monovalent for CD40L cannot cross link CD40L, the possibility is eliminated that cross-linked CD40L may in turn cross-link cell surface CD40 and result in agonism of CD40 signaling activity. Thus, in a preferred aspect, the anti- CD40L antibodies disclosed herein not only inhibit or antagonize the binding of CD40L to CD40, they do not substantially agonize CD40 and/or CD40L activity.
In one aspect, the antibodies monovalent for CD40L binding are human antibody polypeptides. Human antibody polypeptides can be administered to human patients while largely avoiding the anti-antibody immune response often provoked by the administration of antibodies from other species, e.g., mouse. While murine antibodies can be "humanized" by grafting human constant domains onto the murine antigen-binding domains, human antibodies as disclosed herein are produced without the need for laborious and time-consuming genetic manipulation of a murine antibody sequence.
Monovalent antibody polypeptides:
The heavy and light polypeptide chains of antibodies comprise variable (V) regions that directly participate in antigen interactions, and constant (C) regions that provide structural support and function in non-antigen-specific interactions with immune effectors. The antigen binding domain of a conventional antibody is comprised of two separate domains: a heavy chain variable domain (VH) and a light chain variable domain (VL: which can be either Vκ or Vx). The antigen binding site itself is formed by six polypeptide loops: three from the VH domain (Hl, H2 and H3) and three from the VL domain (Ll, L2 and L3). In vivo, a diverse primary repertoire of V genes that encode the VH and VL domains is produced by the combinatorial rearrangement of gene segments. C regions include the light chain C regions (referred to as CL regions) and the heavy chain C regions (referred to as CHI, CH2 and CH3 regions). A naturally-occurring antibody generally comprises two antigen binding domains and is therefore divalent.
A number of smaller antigen binding fragments of naturally occurring antibodies have been identified following protease digestion. These include, for example, the "Fab fragment" (VL-CL-CH1-VH), "Fab' fragment" (a Fab with the heavy chain hinge region), and "F(ab')2 fragment" (a dimer of Fab' fragments joined by the heavy chain hinge region). Recombinant methods have been used to generate such fragments and to generate even smaller antigen-binding fragments, e.g., those referred to as "single chain Fv" (variable fragment) or "scFv," consisting of VL and VH joined by a synthetic peptide linker (VL-linker-Vn). Fab fragments, Fab' fragments and scFv fragments are monovalent for antigen binding, as they each comprise only one antigen binding domain comprising one VR/VL dimer. Even smaller monovalent antibody fragments are the "domain antibodies," or "dAbs," which comprise only a single immunoglobulin variable domain, e.g., VH or VL, that alone specifically binds antigen, i.e., without the need for a complementary VL or VH domain, respectively.
The term "dAb" will refer herein to a single immunoglobulin variable domain
(VH or VL) polypeptide that specifically binds antigen. A- "dAb" binds antigen independently of other V domains; however, a "dAb" can be present in a homo- or heteromultimer with other VH or VL domains where the other domains are not required for antigen binding by the dAb, i.e., where the dAb binds antigen independently of the additional VH or VL domains. The preparation of single immunoglobulin variable domains is described and exemplified herein below.
Monovalent antibody polypeptides can be generated in several different ways. For example, the nucleic acid sequence encoding heavy and light chains of an antibody known to bind CD40L can be manipulated to generate a number of different antibody polypeptides that are monovalent for CD40L binding. Thus, given the sequences encoding the heavy and light chain polypeptides that constitute an antibody and standard molecular cloning methodologies, one can generate monovalent antigen- binding polypeptide constructs such as Fab fragments, scFv, dAbs, or even bispecific antibodies (i.e., antibodies that comprise two different antigen-binding moieties and can therefore bind two separate antigens, preferably simultaneously) that are monovalent for CD40L.
Thus, one means of generating monovalent antibody polypeptides specific for CD40L is to amplify and express the VH and VL regions of the heavy chain and light chain gene sequences isolated, for example, from a hybridoma (e.g., a mouse hybridoma) that expresses anti-CD40L monoclonal antibody. The boundaries of VH and VL domains are set out by Kabat et al. (1991, supra). The information regarding the boundaries of the VH and VL domains of heavy and light chain genes is used to design PCR primers that amplify the V domain from a heavy or light chain coding sequence encoding an antibody known to bind CD40L. The amplified V domains are inserted into a suitable expression vector, e.g., pHEN-1 (Hoogenboom et al., 1991, Nucleic Acids Res. 19: 4133-4137) and expressed, e.g., as a fusion of the VH and VL in an scFv or other suitable monovalent format. The resulting polypeptide is then screened for high affinity monovalent binding to CD40L. For all aspects of the present invention, screening for binding is performed as known in the art or as described herein below.
Alternatively, library screening methods can be used to identify monovalent CD40L-specific binding proteins. Phage display technology (see, e.g., Smith, 1985, Science 228: 1315; Scott & Smith, 1990, Science 249: 386; McCafferty et al., 1990, Nature 348: 552) provides an approach for the selection of antibody polypeptides which bind a desired target from among large, diverse repertoires of antibody polypeptides. These phage-antibody libraries can be grouped into two categories: natural libraries which use rearranged V genes harvested from human B cells (Marks et al., 1991, J. MoI. Biol., 222: 581; Vaughan et al., 1996, Nature Biotech., 14: 309) or synthetic libraries whereby germline V gene segments or other antibody polypeptide coding sequences are 'rearranged' in vitro (Hoogenboom & Winter, 1992, J. MoI. Biol., 227: 381; Nissim et al., 1994, EMBO J., 13: 692; Griffiths et al., 1994, EMBO J., 13: 3245; De Kruif et al., 1995, J. MoI. Biol., 248: 97) or where synthetic CDRs are incorporated into a single rearranged V gene (Barbas et al., 1992. Proc. Natl. Acad. Sci. USA, 89: 4457). Methods involving genetic display packages (e.g., phage display, polysome display) are well-suited for the selection of monovalent CD40L-specifϊc antibody constructs because they generally express only monovalent fragments, rather than whole, divalent antibodies, on the display packages. Methods for the preparation of phage display libraries displaying various antibody fragments are described in the preceding references. Such methods are also described, for example, in U.S. Patent No. 6,696,245, which is incorporated herein by reference. The methods described in the '245 patent generally involve the randomization of selected regions of immunoglobulin gene coding regions, in particular VH and VL coding regions, while leaving other regions non-randomized (see below). The '245 patent also describes the generation of scFv constructs comprising individually randomized VH and VL domains.
The VH gene is produced by the recombination of three gene segments, VH, D and JH- In humans, there are approximately 51 functional VH segments (Cook and Tomlinson (1995) Immunol Today 16: 237), 25 functional D segments (Corbett et al. (1997) J. MoI. Biol. 268: 69) and 6 functional JH segments (Ravetch et al. (1981) Cell 27: 583), depending on the haplotype. The VH segment encodes the region of the polypeptide chain which forms the first and second antigen binding loops of the VH domain (Hl and H2), while the VH, D and JH segments combine to form the third antigen binding loop of the VH domain (H3). The VL §ene ιs produced by the recombination of only two gene segments, VL and JL. In humans, there are approximately 40 functional Vκ segments (Schable and Zachau (1993) Biol. Chem. Hoppe-Seyler 374: 1001), 31 functional Vχ segments (Williams et al. (1996) J. MoI. Biol. 264: 220; Kawasaki et al. (1997) Genome Res. 7: 250), 5 functional Jκ segments (Hieter et al. (1982) J. Biol. Chem. 257: 1516) and 4 functional Jχ segments (Vasicek and Leder (1990) J. Exp. Med. 172: 609), depending on the haplotype. The VL segment encodes the region of the polypeptide chain which forms the first and second antigen binding loops of the VL domain (Ll and L2), while the VL and JL segments combine to form the third antigen binding loop of the VL domain (L3). Antibodies selected from this primary repertoire are believed to -be sufficiently diverse to bind almost all antigens with at least moderate affinity. High affinity antibodies are produced in vivo by "affinity maturation" of the rearranged genes, in which point mutations are generated and selected by the immune system on the basis of improved binding.
Analysis of the structures and sequences of antibodies has shown that five of the six antigen binding loops (Hl, H2, Ll, L2, L3) possess 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). The main-chain conformations are determined by (i) the length of the antigen binding loop, and (ii) particular residues, or types of residue, at certain key positions in the antigen binding loop and the antibody framework. Analysis of the loop lengths and key residues has enabled the prediction of the main-chain conformations of Hl, H2, Ll, L2 and L3 encoded by the majority of human antibody sequences (Chothia et al. (1992) J. MoI. Biol. 227: 799; Tomlinson et al. (1995) EMBO J. 14: 4628; Williams et al. (1996) J. MoI. Biol. 264: 220). Although the 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. While, in one approach, diversity can be added to S3'nthetic repertoires at any site in the CDRs of the various antigen-binding loops, this 'approach results in a greater proportion of V domains that do not properly fold and therefore contribute to a lower proportion of molecules with the potential to bind antigen. An understanding of the residues contributing to the main chain conformation of the antigen-binding loops permits the identification of specific residues to diversify in a synthetic repertoire of VH or VL domains. That is, diversity is best introduced in residues that are not essential to maintaining the main chain conformation. As an example, for the diversification of loop L2, the conventional approach would be to diversify all the residues in the corresponding CDR (CDR2) as defined by Kabat et al. (1991, supra), some seven residues. However, for L2, it is known that positions 50 and 53 are diverse in naturally occurring antibodies and are observed to make contact with the antigen. The preferred approach would be to diversify only those two residues in this loop. This represents a significant improvement in terms of the functional diversity required to create a range of antigen binding specificities.
Immunoglobulin polypeptide libraries can advantageously be designed to be based on predetermined variable domain main chain conformation. Such libraries may be constructed as described in International Patent Application WO 99/20749, the contents of which are incorporated herein by reference. Thus, in one aspect, an antibody polypeptide comprises the amino acid sequence of a given human germline V region gene segment, e.g., VH germline gene segment DP-47, or Vκ germline gene segment DPK9. Such variable region polypeptides can be used for the production of scFvs or Fabs, e.g., an scFv or Fab comprising (i) an antibody heavy chain variable domain (VH), or antigen binding fragment thereof, which comprises the amino acid sequence of germline VH segment DP-47 and (ii) an antibody light chain variable domain (VL), or antigen binding fragment thereof, which comprises the amino acid sequence of germline Vκ segment DPK9. Diversification of sequences within the context of the selected heavy and light chain germline gene segments, e.g., DP-47, DPK 9, DP45, DP38, etc. can generate a repertoire of diverse immunoglobulin coding sequences. One approach to diversification is described below in the context of generating a library of diversified dAb or scFv sequences. These variable region polypeptides can also be expressed as dAbs and screened for high affinity binding to CD40L. The repertoire can be cloned into or generated in a vector suitable for phage display, e.g., a lambda or filamentous bacteriophage display vector and is then screened for binding to a given target antigen, e.g., CD40.L.
Preparation of Human Single Immunoglobulin Variable Domain Polypeptides:
A single immunoglobulin variable domain is a folded polypeptide domain which comprises sequences characteristic of immunoglobulin variable domains and which specifically binds an antigen (e.g., dissociation constant of 500 nM or less), and which binds antigen as a single variable domain; that is, there is one binding site provided by a single immunoglobulin variable domain without any complementary variable domain. A single immunoglobulin variable domain therefore includes complete antibody variable domains as well as 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 a dissociation constant of 500 nM or less (e.g., 450 nM or less, 400 nM or less, 350 nM or less, 300 nM or less, 250 nM or less, 200 nM or less, 150 nM or less, 100 nM or less) and the target antigen specificity of the full- length domain. Preferably an antibody single variable domain useful in the invention is selected from the group of VH and VL, including VkapPa and Viambda- The single immunoglobulin variable domains of use herein are preferably "human" as that term is defined herein.
Preparation of Single Immunoglobulin Variable Domains:
Single immunoglobulin variable domains are prepared in a number of ways. For each of these approaches, well-known methods of preparing (e.g., amplifying, mutating, etc.) and manipulating nucleic acid sequences are applicable.
One means of preparing single immunoglobulin variable domains is to amplify and express the VH or VL region of a heavy chain or light chain gene for a
52 cloned antibody known to bind the desired antigen. That is, the VH or VL domain of a known anti-CD40L antibody coding region can be amplified and expressed as a single domain (or as a fusion of a single domain) and evaluated for binding to CD40L. The boundaries of VH and VL domains are set out by Kabat et al. (1991, supra). The information regarding the boundaries of the VH and VL domains of heavy and light chain genes is used to design PCR primers that amplify the V domain from a cloned heavy or light chain coding sequence encoding an antibody known to bind CD40L. The amplified V domain is inserted into a suitable expression vector, e.g., pHEN-1 (Hoogenboom et al., 1991, Nucleic Acids Res. 19: 4133-4137) and expressed, either alone or as a fusion with another polypeptide sequence.
In a preferred approach, a repertoire of VH or VL domains, preferably human VH or VL domains, is screened by, for example, phage display, panning against the desired antigen. Methods for the construction of bacteriophage display libraries and lambda phage expression libraries are well known in the art, and taught, for example, by: McCafferty et al., 1990, Nature 348: 552; Kang et al., 1991, Proc. Natl. Acad. Sci. U.S.A., 88: 4363; Clackson et al., 1991, Nature 352: 624; Lowman et al., 1991, Biochemistry 30: 10832; Burton et al., 1991, Proc. Natl. Acad. Sci U.S.A. 88: 10134; Hoogenboom et al., 1991, Nucleic Acids Res. 19: 4133; Chang et al.,1991, J. Immunol. 147: 3610; Breitling et al., 1991, Gene 104: 147; Marks et al., 1991, J. MoI. Biol. 222: 581; Barbas et al., 1992, Proc. Natl. Acad. Sci. U.S.A. 89: 4457; Hawkins and Winter (1992) J. Immunol., 22: 867; Marks et al. (1992) J. Biol. Chem., 267: 16007; and Lerner et al. (1992) Science, 258: 1313. Fab phage display libraries are taught, for example, by U.S. 5,922,545. scFv phage libraries are taught, for example, by Huston et al., 1988, Proc. Natl. Acad. Sci U.S.A. 85: 5879-5883; Chaudhary et al., 1990, Proc. Natl. Acad. Sci U.S.A. 87: 1066-1070; McCafferty et al., 1990, supra; Clackson et al., 1991, supra; Marks et al., 1991, supra; Chiswell et al., 1992, Trends Biotech. 10: 80; and Marks et al., 1992, supra. Various embodiments of scFv libraries displayed on bacteriophage coat proteins have been described. Refinements of phage- display approaches are also known, for example as described in WO96/06213 and WO92/01047 (Medical Research Council et al.) and WO97/08320 (Morphosys, supra). The repertoire of VH or VL domains can be a naturally-occurring repertoire of immunoglobulin sequences or a synthetic repertoire. A naturally-occurring repertoire is one prepared, for example, from immunoglobulin-expressing cells harvested from one or more individuals. Such repertoires can be "naive," i.e., prepared, for example, from human fetal or newborn immunoglobulin-expressing cells, or rearranged, i.e., prepared from, for example, adult human B cells. Natural repertoires are described, for example, by Marks et al., 1991, J. MoI. Biol. 222: 581 and Vaughan et al., 1996, Nature Biotech. 14: 309. If desired, clones identified from a natural repertoire, or any repertoire, for that matter, that bind the target antigen are then subjected to mutagenesis and further screening in order to produce and select variants with improved binding characteristics.
Synthetic repertoires of single immunoglobulin variable domains are prepared by artificially introducing diversity into a cloned V domain. Synthetic repertoires are described, for example, by Hoogenboom & Winter, 1992, J. MoI. Biol. 227: 381; Barbas et al., 1992, Proc. Natl. Acad. Sci. U.S.A. 89: 4457; Nissim et al., 1994, EMBO J. 13: 692; Griffiths et al, 1994, EMBO J. 13: 3245; DeKriuf et al., 1995, J. MoI. Biol. 248: 97; and WO 99/20749.
In one aspect, synthetic variable domain repertoires are prepared in VH or VK backgrounds, based on artificially diversified germline VH or VK sequences. For example, the VH domain repertoire can be based on cloned germline VH gene segments V3-23/DP47 (Tomlinson et al., 1992, J. MoI. Biol. 227: 7768) and JH4b. The Vκ domain repertoire can be based, for example, on germline Vκ gene segments O2/O12/DPK9 (Cox et al., 1994, Eur. J. Immunol. 24: 827) and Jκl. Diversity is introduced into these or other gene segments by, for example, PCR mutagenesis. Diversity can be randomly introduced, for example, by error prone PCR (Hawkins, et al., 1992, J. MoI. Biol. 226: 889) or chemical mutagenesis. As discussed above, however it is preferred that the introduction of diversity is targeted to particular residues. It is further preferred that the desired residues are targeted by introduction of the codon NNK using mutagenic primers (using the IUPAC nomenclature, where N = G, A, T or C, and K = G or T), which encodes all amino acids and the TAG stop codon. Other codons which achieve similar ends are also of use, including the NNN codon (which leads to the production of the additional stop codons TGA and TAA), DVT codon ((A/G/T) (A/G/QT ), DVC codon ((A/G/T)(A/G/C)C), and DVY codon ((A/G/T)(A/G/C)(C/T). The DVT codon encodes 22% serine and 11% tyrosine, asgpargine, glycine, alanine, aspartate, threonine and cysteine, which most closely mimics the distribution of amino acid residues for the antigen binding sites of natural human antibodies. Repertoires are made using PCR primers having the selected degenerate codon or codons at each site to be diversified. PCR mutagenesis is well known in the art.
In one aspect, diversity is introduced into the sequence of human germline VH gene segments V3-23/DP47 (Tomlinson et al., 1992, J. MoI. Biol. 227: 7768) and JH4b using the NNK codon at sites H30, H31, H33, H35, H50, H52, H52a, H53, H55, H56, H58, H95, H97 and H98, corresponding to diversity in CDRs 1, 2 and 3, with the numbering as used in U.S. 6,696,245.
In another aspect, diversity is also introduced into the sequence of human germline VH gene segments V3-23/DP47 and JH4b, for example, using the NNK codon at sites H30, H31, H33, H35, H50, H52, H52a, H53, H55, H56, H58, H95, H97, H98, H99, HlOO, HlOOa and HlOOb, corresponding to diversity in CDRs 1, 2 and 3,with the numbering as used in U.S. 6,696,245.
In another aspect, diversity is introduced into the sequence of human germline
Vκ gene segments O2/O12/DPK9 and Jκl, for example, using the NNK codon at sites L30, L31, L32, L34, L50, L53, L91, L92, L93, L94 and L96, corresponding to diversity in CDRs 1, 2 and 3,with the numbering as used in U.S. 6,696,245.
Diversified repertoires are cloned into phage display vectors as known in the art and as described, for example, in WO 99/20749. In general, the nucleic acid molecules and vector constructs required for the performance of the present invention are available in the art and are 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 in the present invention is typicalfy carried out in recombinant vectors. As used herein, '"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 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, as is typical of vectors in which repertoire (or pre-repertoire) members of the invention are carried, a gene expression vector is employed. A vector of use according to the invention is selected to accommodate a polypeptide coding sequence of a desired size, typically from 0.25 kilobase (kb) to 40 kb 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 "polylinker") site, an origin of replication and at least one selectable marker gene. If a 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 polypeptide repertoire member 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. Typically in cloning vectors, 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. Such 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. Generally, 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. Advantageously, a cloning or expression vector also contains 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.
Because the replication of vectors according to the present invention is most conveniently performed in E. coli, an E. coli-selectable marker, for example, the β- lactamase gene that confers resistance to the antibiotic ampicillin, is of use. These can be obtained from E. coli plasmids, such as pBR322 or a pUC plasmid such as pUC18 or pUC19.
Expression vectors usually contain a promoter that is recognized by the host organism and is operably linked to the coding sequence of interest. Such a promoter may be inducible or constitutive. The term "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 β- 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-Dalgarno sequence operably linked to the coding sequence.
In libraries or repertoires as described herein, the preferred vectors are expression vectors that enable the expression of a nucleotide sequence corresponding to a polypeptide library member. Thus, selection is performed by separate propagation and expression of a single clone expressing the polypeptide library member or by use of any selection display system. As described above, a preferred selection display system uses bacteriophage display. Thus, phage or phagemid vectors can be used. Preferred vectors are phagemid vectors, which have an E. coli origin of replication (for double stranded replication) and also a phage origin of replication (for production of single-stranded DNA). The manipulation and expression of such vectors is well known in the art (Hoogenboom and Winter (1992) supra; Nissim et al. (1994) supra). Briefly, the vector contains a β-lactamase or other selectable marker 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 tags (for detection), optionally, one or more TAG stop codons and the phage protein pill. In one embodiment, the vector encodes, rather than the pelB leader sequence, a eukaryotic GASl leader sequence which serves to direct the secretion of the fusion polypeptide to the periplasmic space in E. coli or to the medium in eukaryotic cell systems. Using various suppressor and non-suppressor strains of E. coli and with the addition of glucose, iso-propyl thio-β-D-galactoside (PTG) or a helper phage, such as VCS Ml 3, 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.
An example of a preferred vector is the pHENl phagemid vector (Hoogenboom et al., 1991, Nucl. Acids Res. 19: 4133-4137; sequence is available, e.g., as SEQ ID NO: 7 in WO 03/031611), in which the production of pill fusion protein is under the control of the LacZ promoter, which is inhibited in the presence of glucose and induced with IPTG. When grown in suppressor strains of E. coli, e.g., TGl, the gene III fusion protein is produced and packaged into phage, while growth in non-suppressor strains, e.g., HB2151, permits the secretion of soluble fusion protein into the bacterial periplasm and into the culture medium. Because the expression of gene III prevents later infection with helper phage, the bacteria harboring the phagemid vectors are propagated in the presence of glucose before infection with VCSM 13 helper phage for phage rescue.
Construction of vectors according to the invention employs conventional ligation techniques. Isolated vectors or DNA fragments are cleaved, tailored, and re- ligated in the form desired to generate the required vector. If desired, sequence analysis to confirm that the correct sequences are present in the constructed vector is performed using standard methods. 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. 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 hybridization, immunocytochemistry or sequence analysis of nucleic acid or protein molecules. Those skilled in the art will readily envisage how these methods may be modified, if desired.
Screening Single Immunoglobulin Variable Domains for Antigen Binding:
Following expression of a repertoire of single immunoglobulin variable domains on the surface of phage, selection is performed by contacting the phage repertoire with immobilized target antigen (e.g., CD40L and/or an epitope bound by DOM8-24), washing to remove unbound phage, and propagation of the bound phage, the whole process frequently referred to as "panning." This process is applicable to the screening of single immunoglobulin variable domains as well as other antibody fragments that can be expressed on a display library, e.g., scFv, Fab, etc. Alternatively, phage are pre-selected for the expression of properly folded member variants by panning against an immobilized generic ligand (e.g., protein A or protein L) that is only bound by folded members. This has the advantage of reducing the proportion of non-functional members, thereby increasing the proportion of members likely to bind a target antigen. Pre-selection with generic ligands is taught in WO 99/20749. The screening of phage antibody libraries is generally described, for example, by Harrison et al., 1996, Meth. Enzymol. 267: 83-109.
Screening is commonly performed using purified antigen immobilized on a solid support, for example, plastic tubes or wells, or on a chromatography matrix, for example Sepharoseτ (Pharmacia). Screening or selection can also be performed on complex antigens, such as the surface of cells (Marks et al., 1993, BioTechnology 11: 1145; de Kruif et al., 1995, Proc. Natl. Acad. Sci. U.S.A. 92: 3938). Another alternative involves selection by binding biotinylated antigen in solution, followed by capture on streptavidin-coated beads.
In a preferred aspect, panning is performed by immobilizing antigen (generic or specific) on tubes or wells in a plate, e.g., Nunc MAXISORP™ immunotube 8 well strips. Wells are coated with 150 μl of antigen (100 μg/ml in PBS) and incubated overnight. The wells are then washed 3 times with PBS and blocked with 400 μl PBS-2% skim milk (2%MPBS) at 370C for 2 hr. The wells are rinsed 3 times with PBS and phage are added in 2%MPBS. The mixture is incubated at room temperature for 90 minutes and the liquid, containing unbound phage, is removed. Wells are rinsed 10 times with PBS-0.1% tween 20, and then 10 times with PBS to remove detergent. Bound phage are eluted by adding 200 μl of freshly prepared 100 mM triethylamine, mixing well and incubating for 10 min at room temperature. Eluted phage are transferred to a tube containing 100 μl of IM Tris-HCl, pH 7.4 and vortexed to neutralize the triethylamine. Exponentially-growing E. coli host cells (e.g., TGl) are infected with, for example, 150 ml of the eluted phage by incubating for 30 min at 370C. Infected cells are spun down, resuspended in fresh medium and plated in top agarose. Phage plaques are eluted or picked into fresh cultures of host cells to propagate for analysis or for further rounds of selection. One or more rounds of plaque purification are performed' if necessary to ensure pure populations of selected phage. Other screening approaches are described by Harrison et al., 1996, supra. Following identification of phage expressing a single immunoglobulin variable domain that binds a desired target, if a phagemid vector such as pHENl has been used, the variable domain fusion proteins are easily produced in soluble form by infecting non-suppressor strains of bacteria, e.g., HB2151 that permit the secretion of soluble gene III fusion protein. If a GAS 1 secretion signal peptide is encoded by the vector, the fusion polypeptide can be secreted by eukaryotic (e.g., yeast or mammalian) or prokaryotic (e.g., E. coli) ceils. Alternatively, the V domain sequence can be sub-cloned into an appropriate expression vector to produce soluble protein according to methods known in the art.
Purification and Concentration of Single Immunoglobulin Variable Domains:
Single immunoglobulin variable domain polypeptides or other monovalent antibody polypeptides secreted into the periplasmic space or into the medium of bacteria are harvested and purified according to known methods (Harrison et al., 1996, supra). Skerra & Pluckthun (1988, Science 240: 1038) and Breitling et al. (1991, Gene 104: 147) describe the harvest of antibody polypeptides from the periplasm,, and Better et al. (1988, Science 240: 1041) describes harvest from the culture supernatant. For some antibody polypeptides, purification can also be achieved by binding to generic ligands, such as protein A or Protein L. Alternatively, the variable domains can be expressed with a peptide tag, e.g., the Myc, HA or 6X- His tags, which facilitates purification by affinity chromatography.
If necessary, monovalent anti-CD40L antibody polypeptides are concentrated by any of several methods well known in the art, including, for example, ultrafiltration, diafiltration and tangential flow filtration. The process of ultrafiltration uses semi-permeable membranes and pressure to separate molecular species on the basis of size and shape. The pressure is provided by gas pressure . or by centrifugation. Commercial ultrafiltration products are widely available, e.g., from Millipore (Bedford, MA; examples include the Centricon™ and Microcon™ concentrators) and Vivascience (Hannover, Germany; examples include the Vivaspin™ concentrators). By selection of a molecular weight cutoff smaller than the target polypeptide (usually 1/3 to 1/6 the molecular weight of the target polypeptide, although differences of as little as 10 kD can be used successfully), the polypeptide is retained when solvent and smaller solutes pass through the membrane. Thus, a molecular weight cutoff of about 5 kD is useful for concentration of anti- CD40L single immunoglobulin variable domain polypeptides described herein.
Diafiltration, which uses ultrafiltration membranes with a "washing" process, is used where it is desired to remove or exchange the salt or buffer in a polypeptide preparation. The polypeptide is concentrated by the passage of solvent and small solutes through the membrane, and remaining salts or buffer are removed by dilution of the retained polypeptide with a new buffer or salt solution or water, as desired, accompanied by continued ultrafiltration. In continuous diafiltration, new buffer is added at the same rate that filtrate passes through the membrane. A diafiltration volume is the volume of polypeptide solution prior to the start of diafiltration - using continuous diafiltration, greater than 99.5% of a fully permeable solute can be removed by washing through six diafiltration volumes with the new buffer. Alternatively, the process can be performed in a discontinuous manner, wherein the sample is repeatedly diluted and then filtered back to its original volume to remove or exchange salt or buffer and ultimately concentrate the polypeptide. Equipment for diafiltration and detailed methodologies for its use are available, for example, from Pall Life Sciences (Ann Arbor, MI) and Sartorius AG/Vivascience (Hannover, Germany).
Tangential flow filtration (TFF), also known as "cross-flow filtration," also uses ultrafiltration membrane. Fluid containing the target polypeptide is pumped tangentially along the surface of the membrane. The pressure causes a portion of the fluid to pass through the membrane while the target polypeptide is retained above the filter. In contrast to standard .ultrafiltration, however, the retained molecules do not accumulate on the surface of the membrane, but are carried along by the tangential flow. The solution that does not pass through the filter (containing the target polypeptide) can be repeatedly circulated across the membrane to achieve the desired degree of concentration. Equipment for TFF and detailed methodologies for its use are available, for example, from Millipore (e.g., the ProFlux M12™ Benchtop TFF system and the Pellicon™ systems), Pall Life Sciences (e.g., the Minim™ Tangential Flow Filtration system).
Protein concentration is measured in a number of ways that are well known in the art. These include, for example, amino acid analysis, absorbance at 280 inn, the "Bradford" and "Lowry" methods, and SDS-PAGE. The most accurate method is total hydrolysis followed by amino acid analysis by HPLC, concentration is then determined then comparison with the known sequence of the single immunoglobulin variable domain polypeptide. While this method is the most accurate, it is expensive and time-consuming. Protein determination by measurement of UV absorbance at 280 nm faster and much less expensive, yet relatively accurate and is preferred as a compromise over amino acid analysis. Absorbance at 280 nm was used to determine protein concentrations reported in the Examples described herein.
"Bradford" and "Lowry" protein assays (Bradford, 1976, Anal. Biochem. 72: 248-254; Lowry et al.,1951, J. Biol. Chem. 193: 265-275) compare sample protein concentration to a standard curve most often based on bovine serum albumin (BSA). These methods are less accurate, tending to undersetimate the concentration of single immunoglobulin variable domains. Their accuracy could be improved, however, by using a VH or Vκ single domain polypeptide as a standard.
An additional protein assay method is the bicinchoninic acid assay described in U.S. Patent No. 4,839,295 (incorporated herein by reference) and marketed by Pierce^ Biotechnology (Rockford, IL) as the "BCA Protein Assay" (e.g., Pierce Catalog No. 23227).
The SDS-PAGE method uses gel electrophoresis and Coomassie Blue staining in comparison to known concentration standards, e.g., known amounts of a single immunoglobulin variable domain polypeptide. Quantitation can be done by eye or by densitometry.
Single human immunoglobulin variable domain antigen-binding polypeptides described herein retain solubility at high concentration (e.g., at least 4.8 mg (~400 μM) in aqueous solution (e.g., PBS), and preferably at least 5 mg/ml (-417 μM), 10 mg/ml (-833 μM), 20 mg/ml (-1.7 mM), 25 mg/ml (-2.3 mM), 30 mg/ml (-2.5 mM), 35 mg/ml (-2.9 mM), 40 mg/ml (-3.3 mM), 45 mg/ml (-3.75 mM), 50 mg/ml (-4.2 mM), 55 mg/ml (-4.6 mM), 60 mg/ml (-5.0 mM), 65 mg/ml (-5.4 mM), 70 mg/ml (-5.8 mM), 75 mg/ml (-6.3 mM), 100 mg/ml (-8.33 mM), 150 mg/ml (-12.5 mM), 200 mg/ml (-16.7 mM), 240 mg/ml (-20 mM) or higher). One structural feature that promotes high solubility is the relatively small size of the single immunoglobulin variable domain polypeptides. A full length conventional four chain antibody, e.g., IgG is about 150 kD in size. In contrast, single immunoglobulin variable domains, which all have a general structure comprising 4 framework (FW) regions and 3 CDRs, have a size of approximately 12 IcD, or less than 1/10 the size of a conventional antibody. Similarly, single immunoglobulin variable domains are approximately 1A the size of an scFv molecule (-26 IcD), and approximately 1/5 the size of a Fab molecule (-60 kD). It is preferred that the size of a single immunoglobulin variable domain-containing structure disclosed herein is 100 kD or less, including structures of, for example, about 90 kD or less, 80 kD or less, 70 kD or less, 60 IcD or less, 50 IcD or less, 40 kD or less, 30 kD or less, 20 kD or less, down to and including about 12 kD, or a single immunoglobulin variable domain in isolation.
The solubility of a polypeptide is primarily determined by the interactions of the amino acid side chains with the surrounding solvent. Hydrophobic side chains tend to be localized internally as a polypeptide folds, away from the solvent- interacting surfaces of the polypeptide. Conversely, hydrophilic residues tend to be localized at the solvent-interacting surfaces of a polypeptide. Generally, polypeptides having a primary sequence that permits the molecule to fold to expose more hydrophilic residues to the aqueous environment are more soluble than one that folds to expose fewer hydrophilic residues to the surface. Thus, the arrangement and number of hydrophobic and hydrophilic residues is an important determinant of solubility. Other parameters that determine polypeptide solubility include solvent pH, temperature, and ionic strength. In a common practice, the solubility of polypeptides can be maintained or enhanced by the addition of glycerol (e.g., -10% v/v) to the solution. As discussed above, specific amino acid residues have been identified in conserved residues of human VH domains that vary in the VH domains of camelid species, which are generally more soluble than human VH domains. These include, for example, GIy 44 (GIu in camelids), Leu 45 (Arg in camelids) and Trp 47 (GIy in camelids). Amino acid residue 103 of VH is also implicated in solubility, with mutation from Trp to Arg tending to confer increased VH solubility.
In preferred aspects of the invention, single immunoglobulin variable domain polypeptides are based on the DP47 germline VH gene segment or the DPK9 germline Vκ gene segment. Thus, these germline gene segments are capable, particularly when diversified at selected structural locations described herein, of producing specific binding single immunoglobulin variable domain polypeptides that are highly soluble. In particular, the four framework regions, which are preferably not diversified, can contribute to the high solubility of the resulting proteins.
It is expected that a single human immunoglobulin variable domain that is highly homologous to one having a known high solubility will also tend to be highly soluble. Thus, as one means of prediction or recognition that a given single immunoglobulin variable domain would have the high solubility recited herein, one can compare the sequence of a single immunoglobulin variable domain polypeptide to one or more single immunoglobulin variable domain polypeptides having known solubility. Thus, when a single immunoglobulin variable domain polypeptide is identified that has high binding affinity but unknown solubility, comparison of its amino acid sequence with that of one or more (preferably more) human single immunoglobulin variable domain polypeptides known to have high solubility (e.g., a dAb sequence disclosed herein) can permit prediction of its solubility. While it is not an absolute predictor, where there is a high degree of similarity to a known highly soluble sequence, e.g., 90-95% or greater similarity, and particularly where there is a high degree of similarity with respect to hydrophilic amino acid residues, or residues likely to be exposed at the solvent interface, it is more likely that a newly identified binding polypeptide will have solubility similar to that of the known highly soluble sequence. Molecular modeling software can also be used to predict the solubility of a polypeptide sequence relative to that of a polypeptide of known solubility. For example, the substitution or addition of a hydrophobic residue at the solvent-exposed surface, relative to a molecule of known solubility that has a less hydrophobic or even hydrophilic residue exposed in that position is expected to decrease the relative solubility of the polypeptide. Similarly, the substitution or addition of a more hydrophilic residue at such a location is expected to increase the relative solubility. That is, a change in the net number of hydrophilic or hydrophobic residues located at the surface of the molecule (or the overall hydrophobic or hydrophilic nature of the surface-exposed residues) relative to a single immunoglobulin variable domain polypeptide structure with known solubility can predict the relative solubility of a single immunoglobulin variable domain polypeptide.
Alternatively, or in conjunction with such prediction, one can determine limits of a single immunoglobulin variable domain polypeptide's solubility by simply concentrating the polypeptide.
Affinity Determination:
Isolated single immunoglobulin variable domain- and antibody polypeptide- containing polypeptides as described herein preferably have affinities (dissociation constant, Ka, = K0ff/Kon) of at least 500 nM or less, and preferably at least 400 nM-50 pM, 300 nM-50 pM, 200 nM - 50 pM, and more preferably at least 100 nM - 50 pM, 75 nM - 50 pM, 50 nM - 50 pM, 25 nM - 50 pM, 10 nM - 50 pM, 5 nM- 50 pM, 1 nM - 50 pM, 950 pM - 50 pM, 900 pM - 50 pM, 850 pM - 50 pM, 800 pM - 50 pM, 750 pM - 50 pM, 700 pM - 50 pM, 650 pM - 50 pM, 600 pM - 50 pM, 550 pM - 50 pM, 500 pM - 50 pM, 450 pM - 50 pM, 400 pM - 50 pM, 350 pM - 50 pM, 300 pM - 50 pM, 250 pM - 50 pM, 200 pM - 50 pM, 150 pM - 50 pM, 100 pM - 50 pM, 90 pM - 50 pM, 80 pM - 50 pM, 70 pM - 50 pM, 60 pM - 50 pM, or even as low as 50 pM.
The antigen-binding affinity of an antibody polypeptide, e.g., a single immunoglobulin variable domain polypeptide or other monovalent antibody polypeptide, can be conveniently measured by SPR using the BIAcore system (Pharmacia Biosensor, Piscataway, NJ.). In this method, antigen is coupled to the BIAcore chip at known concentrations, and variable domain polypeptides are introduced. Specific binding between the variable domain polypeptide and the immobilized antigen results in increased protein concentration on the chip matrix and a change in the SPR signal. Changes in SPR signal are recorded as resonance units (RU) and displayed with respect to time along the Y axis of a sensorgram. Baseline signal is taken with solvent alone (e.g., PBS) passing over the chip. The net difference between baseline signal and signal after completion of antibody polypeptide injection represents the binding value of a given sample. To determine the off rate (KOff), on rate (K0n) and dissociation rate (Kd) constants, BIAcore kinetic evaluation software (e.g., version 2.1) is used.
Thus, SPR can be used to monitor antagonism of CD40L binding to CD40 by a monovalent anti-CD40L antibody preparation by measuring the displacement or inhibition of binding of CD40L to CD40 caused the monovalent antibody preparation. SPR can also be used to monitor the dimerization, or preferably, the lack of dimerization, occurring via Fc region in antibody preparations as described herein.
High affinity is dependent upon the complementarity between a surface of the antigen and the CDRs of the antibody or antibody fragment. Complementarity is determined by the type and strength of the molecular interactions possible between portions of the target and the CDR, for example, the potential ionic interactions, van der Waals attractions, hydrogen bonding or other interactions that can occur. CDR3 tends to contribute more to antigen binding interactions than CDRs 1 and 2, probably due to its generally larger size, which provides more opportunity for favorable surface interactions. (See, e.g., Padlan et al., 1994, MoL Immunol. 31: 169-217; Chothia & Lesk, 1987, J. MoL Biol. 196: 904-917; and Chothia et al., 1985, J. MoL Biol. 186: 651-663.) High affinity indicates antibody polypeptide/antigen pairings that have a high degree of complementarity, which is directly related to the structures of the variable domain and the target. In one aspect, a monovalent anti-CD40L antibody polypeptide, e.g., a single immunoglobulin variable domain polypeptide, is linked to another antibody polypeptide to form a heterodimer in which each individual antibody polypeptide is capable of binding a different cognate antigen. Fusing antibody polypeptides, such as single immunoglobulin variable domains, as heterodimers, wherein each monomer binds a different target antigen, can produce a dual-specific ligand capable, for example, of bridging the respective target antigens. Such dual specific ligands may be used to target cytokines and other molecules which cooperate synergistically in therapeutic situations in the body of an organism. Thus, there is provided a method for synergising the activity of two or more cytokines, comprising administering a dual specific antibody heterodimer capable of binding to the two or more cytokines.
Non-limiting examples of second targets for anti-CD40L dual specific antibody polypeptides include the following: TNF-α; IL-I; IL-2; IL-4; IL-6; IL-8; IL- 12; IL-18; IFN-γ; CD2; CD4; CD8; CTLA4; LFAl; LFA3, VLA4, CD80, B7-1, CD28, CD86, B7-2, and CTLA-4. In particular, second targets useful according to the invention include CD80, B7-1, CD28, CD86, B7-2, and CTLA-4. These targets are thought to be involved in a co-stimulatory pathway critical for T-cell activation (termed, co-stimulatory signal pathway antigens). This pathway includes activation of the molecule CD28 on the surface of T cells. This molecule can receive a costimulatory signal delivered by a ligand on B cells or other APCs. Ligands for CD28 include members of the B7 family of B lymphocyte activation antigens, such as B7-1 and/or B7-2 (Freedman, A. S. et al. (1987) J. Immunol. 137, 3260-3267; Freeman, G. J. et al. (1989) J. Immunol. 143, 2714-2722; Freeman, G. J. et al. (1991) J. Exp. Med. 174, 625-631; Freeman, G. J. et al. (1993) Science 262, 909-911; Azuma, M. et al. (1993) Nature 366, 76-79; Freeman, G. J. et al. (1993) J. Exp. Med. 178, 2185-2192). B7-1 and B7-2 are also ligands for another molecule, CTLA4, present on the surface of activated T cells. Accordingly, the present invention contemplates that members of the CD28 signalling pathway may be useful second targets for the dual-specific format anti-CD40L antibody polypeptides.
Homologous sequences:
68 The invention encompasses anti-CD40L antibody polypeptides, e.g., CD40L- binding single immunoglobulin variable domain clones, and clones with substantial sequence similarity or homology to them that also bind target antigen with high affinity. As used herein, "substantial" sequence similarity or homology is at least 85% similarity or homology.
Calculations of "homology" or "sequence identity" between two sequences (the terms are equivalent and 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). In a preferred embodiment, 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. The amino acid residues or nucleotides at corresponding amino acid positions or nucleotide positions are then compared. When 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.
As used herein, sequence "similarity" is a measure of the degree to which amino acid sequences share similar amino acid residues at corresponding positions in an alignment of the sequences. Amino acids are similar to each other where their side chains are similar. Specifically, "similarity" encompasses amino acids that are conservative substitutes for each other. A "conservative" substitution is any substitution that has a positive score in the blosum62 substitution matrix (Hentikoff and Hentikoff, 1992, Proc. Natl. Acad. Sci. USA 89: 10915-10919). By the statement "sequence A is n% similar to sequence B" is meant that n% of the positions of an optimal global alignment between sequences A and B consists of identical amino acids or conservative substitutions. Optimal global alignments can be performed using the following parameters in the Needleman-Wunsch alignment algorithm: For polypeptides:
Substitution matrix: blosum62.
Gap scoring function: -A -B*LG, where A=I l (the gap penalty), B=I (the gap length penalty) and LG is the length of the gap.
For nucleotide sequences: Substitution matrix: 10 for matches, 0 for mismatches.
Gap scoring function: -A -B*LG where A=50 (the gap penalty), B=3 (the gap length penalty) and LG is the length of the gap.
Typical conservative substitutions are among Met, VaI, Leu and lie; among Ser and Thr; among the residues Asp, GIu and Asn; among the residues GIn, Lys and Arg; or aromatic residues Phe and Tyr. In calculating the degree (most often as a percentage) of similarity between two polypeptide sequences, one considers the number of positions at which identity or similarity is observed between corresponding amino acid residues in the two polypeptide sequences in relation to the entire lengths of the two molecules being compared.
Alternatively, the BLAST (Basic Local Alignment Search Tool) algorithm is employed for sequence alignment, with parameters set to default values. The BLAST algorithm "BLAST 2 Sequences" is available at the world wide web site ("www") of the National Center for Biotechnology Information (".ncbi"), of the National Library of Medicine (".nlm") of the National Institutes of Health ("nih") of the U.S. government (".gov"), in the "/blast/" directory, sub-directories "bl2seq/bl2.html." This algorithm aligns two sequences for comparison and is described by Tatusova & Madden, 1999, FEMS Microbiol Lett. 174:247-250.
An additional measure of homology or similarity is the ability to hybridize under highly stringent hybridization conditions. Thus, a first sequence encoding a single immunoglobulin variable domain polypeptide is substantially similar to a second coding sequence if the first sequence hybridizes to the second sequence (or its complement) under highly stringent hybridization conditions (such as those described by Sambrook et al., Molecular Cloning, Laboratory Manuel, Cold Spring, Harbor Laboratory press, New York). "Highly stringent hybridization conditions" refer to hybridization in 6X SSC at about 45°C, followed by one or more washes in 0.2X SSC, 0.1% SDS at 65°C. "Very highly stringent hybridization conditions" refer to hybridization in 0.5M sodium phosphate, 7% SDS at 65°C, followed by one or more washes at 0.2X SSC, 1% SDS at 650C.
Assays for CD40L Activities:
It is preferred that a monovalent anti-CD40L antibody polypeptides as described herein bind to CD40L yet do not substantially agonize CD40 signaling. Activation of the CD40L/CD40 pathway manifests a number of different outcomes that can be measured in order to assess the effect of a given monovalent anti-CD40L antibody polypeptide on the activity of the pathway. However, for the assessment of the antagonist or agonist function of monovalent anti-CD40L antibody polypeptides described herein, at least one of the following CD40L assays can be used:
1) Activation of Jun-N-Terminal Kinase (JNK):
Stimulation of T-lymphocytes via CD40L induces strong activation of JNK. The ability of a monovalent anti-CD40L antibody polypeptide to activate this signaling pathway is measured as follows. Human leukemic Jurkat cells are stimulated with a positive control agonistic anti-CD40L antibody (2 μg/ml monoclonal anti-human or anti-mouse gp39/CD40L antibody (Pharmingen, San Diego, CA, USA) or isotype matched hamster or mouse immunoglobulins (Dianova, Hamburg, Germany)), monovalent anti-CD40L antibody polypeptide, or a negative control irrelevant antibody as described by Brenner et al., 1997, FEBS Lett. 417: 301- 306, which is incorporated herein by reference. The cells are lysed and the extract assayed for phosphorylated JNK via colorimetric assay (e.g., Titerzyme™ colorimetric (EIA) phospho-JNKl/2 immunoassay kit, by Assay Designs Inc., Catalog # 900-106). An increase in phospho-JNK (e.g., by 5% or more) for anti- CD40L-stimulated cells over non-stimulated cells indicates agonism of CD40L activity by the antibody polypeptide.
2. Induction of Cytokine Secretion:
Co-stimulation of T cells with anti-CD3 Ab and CD40L has been shown to upregulate the production of IL-IO, IFN-γ and TNF-α by those cells. The ability of a monovalent anti-CD40L antibody polypeptide to activate this signaling pathway is measured as follows. Human leukemic Jurkat T cells (or freshly isolated CD4+ T cells) are plated into 96 well plates containing immobilized anti-CD3 antibody. The cells are then cultured for 72 hours in the presence of a positive control agonistic anti- CD40L antibody, CD40L, monovalent anti-CD40L antibody polypeptide, or a negative control irrelevant antibody, as described by Blair et al., 2000, J. Exp. Med. 191 : 651-660. IFN-γ (or IL-10 of TNF-α) is then quantitated in the supernatant by sandwich ELISA using an IFN-g standard to generate a standard curve from which all other unknowns can be calculated. An increase in IFN-g (e.g., by 5% or more) for anti-CD40L-stimulated cells over non-stimulated cells indicates agonism by the antibody polypeptide.
3. Platelet Aggregation Assay:
Divalent anti-CD40L antibodies tend to cause platelet aggregation, which is likely associated with the thromboembolic events observed in clinical trials of divalent anti-CD40L antibodies in the prior art. Monovalent anti-CD40L antibody polypeptides as described herein preferably do not substantially mediate or agonize CD40L-mediated platelet aggregation. With regard tothis aspect, the "standard platelet aggregation assay" is as follows:
Platelets are prepared at 2.5x105/ml and left stirring in a 500-Ca lumi- aggregometer (or its equivalent, e.g., a Platelet Aggregation Profiler (BioData, Horsham, PA)). Platelets are partially activated by the addition of a dilution series of 0.1-10 μM ADP (the lOμM ADP induces aggregation, and is used as a positive control - lower concentrations activate platelets but do not induce aggregation). CD40L mediated platelet aggregation is stimulated by addition of either anti-CD40L monoclonal antibodies (i.e., divalent monoclonal antibodies, available from, e.g., Pharmingen, San Diego, CA, USA) or soluble CD40/Fc fusion protein (available from R&D Systems). The reaction is allowed to proceed for between 3 and 5 minutes. Stimulation of platelet aggregation above the mininimal aggregation/activation achieved with ADP alone is plotted against stimulating anti- CD40L or CD40/Fc concentration. The percentage of platelet aggregation is measured by the change in light transmittance following addition of antibody polypeptide being tested or positive control peptide. A value greater than that observed for negative control lacking antibody and amounting to 25% or more of the positive control value (divalent anti-CD40L or CD40/Fc fusion) is considered to be indicative of induction of platelet aggregation.
Other methods to assess platelet aggregation and/or activation, including events which precede aggregation, or which are downstream from platelet aggregation, include assays which determine various indicators of platelet activation, and are known in the art. For example, platelet activation (and, thus, CD40/CD40L activity) can be determined by assaying for CD62P expression in platelets (e.g., using anti-CD26P antibodies and flow cytometry), assaying for monocyte-platelet- conjugate formation, assaying for platelet closure time under high shear conditions
(e.g., using a PFA-100, Dade Behrϊng, Newark, DE), and assaying for platelet dense granule release. Methods for performing such assays are known in the art and can be found, for example, in Langer et al., 2005 Thromb. Haemost. 93: 1137-46.
PEGylation of monovalent anti-CD40L antibody polypeptides
The present invention provides PEGylated monovalent anti-CD40L antibody polypeptides which have increased half-life and preferably also resistance to degradation without a loss in activity (e.g., binding affinity) relative to non- PEGylated antibody polypeptides. Monovalent anti-CD40L antibody polypeptides according to this aspect can be coupled, using methods known in the art to polymer molecules (preferably PEG) useful for achieving the increased half-life and degradation resistance properties encompassed by the present invention. Polymer moieties which can be utilized in the invention can be synthetic or naturally occurring and include, but are not limited to straight or branched chain polyalkylene, polyalkenylene or polyoxyalkylene polymers, or a branched or unbranched polysaccharide such as a homo- or heteropolysaccharide. Preferred examples of synthetic polymers which may be used in the invention include straight or branched chain poly(ethylene glycol) (PEG), poly(propylene glycol), or poly(vinyl alcohol) and derivatives or substituted forms thereof. Particularly preferred substituted polymers useful in the invention include substituted PEG, including methoxy(polyethylene glycol). Naturally occurring polymer moieties which may be used according to the invention in addition to or in place of PEG include lactose, amylose, dextran, or glycogen, as well as derivatives thereof which would be recognized by one of skill in the art. Derivatized forms of polymer molecules of the invention include, for example, derivatives which have additional moieties or reactive groups present therein to permit interaction with amino acid residues of the dAb polypeptides described herein. Such derivatives include N- hydroxylsuccinimide (NHS) active esters, succinimidyl propionate polymers, and sulfhydryl-selective reactive agents such as maleimide, vinyl sulfone, and thiol. Particularly preferred derivatized polymers include, but are not limited to PEG polymers having the formulae: PEG-O-CH2CH2CH2-CO2-NHS; PEG-O-CH2-NHS; PEG-O-CH2CH2-CO2-NHS; PEG-S-CH2CH2-CO-NHS; PEG-O2CNH-CH(R)-CO2- NHS; PEG-NHCO-CH2CH2-CO-NHS; and PEG-O-CH2-CO2-NHS; where R is (CH2)4)NHCO2(mPEG). PEG polymers useful in the invention may be linear molecules, or may be branched wherein multiple PEG moieties are present in a single polymer. Some particularly preferred PEG derivatives which are useful in the invention include, but are not limited to the following:
Figure imgf000078_0001
mPEG-MAL mPEG2-MAL
mPEG — CONH
Figure imgf000078_0002
mPEG-(MAL)2 multi-arm PEG
Figure imgf000078_0003
Figure imgf000079_0001
and
The reactive group (e.g., MAL, NHS, SPA, VS, or Thiol) may be attached directly to the PEG polymer or may be attached to PEG via a linker molecule.
The size of polymers useful in the invention can be in the range of between 500 Da to 60 kDa, for example, between 1000 Da and 60 kDa, 10 kDa and 60 kDa, 20 kDa and 60 kDa, 30 kDa and 60 kDa, 40 kDa and 60 kDa, and up to between 50 kDa and 60 kDa. The polymers used in the invention, particularly PEG, can be straight chain polymers or can possess a branched conformation. Depending on the combination of molecular weight and conformation, the polymer molecules useful in the invention, when attached to a monovalent anti-CD40L antibody polypeptide, will yield a molecule having an average hydrodynamic size of between 24 and 500 kDa. The hydrodynamic size of a polymer molecule used herein refers to the apparent size of a molecule (e.g., a protein molecule) based on the diffusion of the molecule through an aqueous solution. The diffusion, or motion of a protein through solution can be processed to derive an apparent size of the protein, where the size is given by the Stokes radius or hydrodynamic radius of the protein particle. The "hydrodynamic size" of a protein depends on both mass and shape (conformation), such that two proteins having the same molecular mass may have differing hydrodynamic sizes based on the overall conformation of the protein. The hydrodynamic size of a PEG- linked monovalent anti-CD40L antibody polypeptide, e.g., an anti-CD40L single immunoglobulin variable domain as described herein, can be in the range of 24 IdDa to 500 IdDa; 30 to 500 IcDa; 40 to 500 kDa; 50 to 500 kDa; 100 to 500 IvDa; 150 to 500 IdDa; 200 to 500 kDa; 250 to 500 IdDa; 300 to 500 IdDa; 350 to 500 IdDa; 400 to 500 IdDa and 450 to 500 kDa. Preferably the hydrodynamic size of a PEGylated antibody polypeptide as described herein is 30 to 40 IdDa; 70 to 80 IdDa or 200 to 300 IdDa. The size of a polymer molecule attached to a monovalent anti-CD40L antibody polypeptide may thus be varied depending upon the desired application. For example, where the PEGylated antibody polypeptide is intended to leave the circulation and enter into peripheral tissues, it is desirable to keep the size of the attached polymer low to facilitate extravazation from the blood stream. Alternatively, where it is desired to have the PEGylated antibody polypeptide remain in the circulation for a longer period of time, a higher molecular weight polymer can be used (e.g., a 30 to 60 kDa polymer).
The polymer (PEG) molecules useful in the invention can be attached to antibody polypeptides using methods that are well known in the art. The first step in the attachment of PEG or other polymer moieties to an antibody polypeptide is the substitution of the hydroxyl end-groups of the PEG polymer by electrophile- containing functional groups. Particularly, PEG polymers are attached to either cysteine or lysine residues present in the antibody polypeptide. The cysteine and lysine residues can be naturally occurring, or can be engineered into the antibody polypeptide molecule. For example, cysteine residues can be recombinantly engineered at the C-terminus of antibody polypeptides, or residues at specific solvent accessible locations in the antibody polypeptide can be substituted with cysteine or lysine. In a preferred embodiment, a PEG moiety is attached to a cysteine residue which is present in the hinge region at the C-terminus of an antibody polypeptide.
In a further preferred embodiment a PEG moiety or other polymer is attached to a cysteine or lysine residue which is either naturally occurring at or engineered into the N-terminus of antibody single variable domain polypeptide of the invention. In a still further embodiment, a PEG moiety or other polymer is attached to an antibody single variable domain according to the invention at a cysteine or lysine residue (either naturally occurring or engineered) which is at least 2 residues away from (e.g., internal to) the C- and/or N-terminus of the antibody single variable domain polypeptide.
In one embodiment, the PEG polymer(s) is attached to one or more cysteine or lysine residues present in a framework region (FWs) and one or more heterologous CDRs of a single immunoglobulin variable domain. CDRs and framework regions (e.g., CDR1-CDR3 and FW1-FW4) are those regions of an immunoglobulin variable domain as defined in the Kabat database of Sequences of Proteins of Immunological Interest (Kabat et al., 1991, supra). In a preferred embodiment, a PEG polymer is linked to a cystine or lysine residue in the VH framework segment DP47, or the Vk framework segment DPK9. Cysteine and/or lysine residues of DP47 which may be linked to PEG according to the invention include the cysteine at positions 22, or 96 and the lysine at positions 43, 65, 76, or 98 of SEQ ID NO: 1 (Figure 5). Cysteine and/or lysine residues of DPK9 which may be linked to PEG according to the invention include the cysteine residues at positions 23, or 88 and the lysine residues at positions 39, 42, 45, 103, or 107 of SEQ ID NO: 3 (Figure 6). In addition, specific cysteine or lysine residues may be linked to PEG in the VH canonical framework region DP38, or DP45.
In addition, specific solvent accessible sites in the antibody molecule which are not naturally occurring cysteine or lysine residues may be mutated to a cysteine or lysine for attachment of a PEG polymer. Solvent accessible residues in any given antibody, e.g., a dAb, can be determined using methods known in the art such as analysis of the crystal structure of the antibody polypeptide. For example, using the solved crystal structure of the VH dAb HEL4 (SEQ ID NO: 3; a dAb that binds hen egg lysozyme), the residues Gln-13, Pro- 14, GIy- 15, Pro-41, Gly-42, Lys-43, Asp-62,
Lys-65, Arg-87, Ala-88, Glu-89, GIn-112, Leu-115, Thr-117, Ser-119, and Ser-120 have been identified as being solvent accessible, and according to the present invention would be attractive candidates for mutation to cysteine or lysine residues for the attachment of a PEG polymer. In addition, using the solved crystal structure of the Vk dummy dAb (SEQ ID NO: 4), the residues VaI- 15, Pro-40, Gly-41, Ser-56, Gly-57, Ser-60, Pro-80, Glu-81, GIn- 100, Lys-107, and Arg-108 have been identified as being solvent accessible, and according to the present invention would be attractive candidates for mutation to cysteine or lysine residues for the attachment of a PEG polymer. In one embodiment of the invention, a PEG polymer is linked to multiple solvent accessible cysteine or lysine residues, or to solvent accessible residues which have been mutated to a cysteine or lysine residue. Alternatively, only one solvent accessible residue is linked to PEG, either where the particular antibody polypeptide only possesses one solvent accessible cysteine or lysine (or residue modified to a cysteine or lysine) or where a particular solvent accessible residue is selected from among several such residues for PEGylation.
Primary amino acid sequence of HEL4 (SEQ ID NO: 5).
1 EVQLLESGGG LVQPGGSLRL SCAASGFRIS DEDMGWVRQA PGKGLEWVSS
51 IYGPSGSTYY ADSVKGRPTI SRDNSKNTLY LQMNSLRAED TAVYYCASAL
101 EPLSEPLGFW GQGTLVTVSS
Primary amino acid sequence of V^ dummy (SEQ ID NO: 6).
1 DIQMTQSPSS LSASVGDRVT ITCRASQSIS SYLNWYQQKP
GKAPKLLIΎA 51 ASSLQSGVPS RFSGSGSGTD FTLTISSLQP EDFATYYCQQ
SYSTPNTFGQ 101 GTKVEIKR-
Several PEG attachment schemes which are useful in the invention are provided by the company Nektar (SanCarlos, CA). For example, where attachment of PEG or other polymer to a lysine residue is desired, active esters of PEG polymers which have been derivatized with N-hydroxylsuccinimide, such as succinimidyl propionate may be used. Where attachment to a cysteine residue is intended, PEG polymers which have been derivatized with sulfhydryl-selective reagents such as maleimide, vinyl sulfone, or thiols may be used. Other examples of specific embodiments of PEG derivatives which may be used according to the invention to generate PEGylated antibodies can be found in the Nektar Catalog (available on the world wide web at nektar.com). In addition, several derivitized forms of PEG may be used according to the invention to facilitate attachment of the PEG polymer to an antibody polypeptide. PEG derivatives useful in the invention include, but are not limited to PEG-succinimidyl succinate, urethane linked PEG, PEG phenylcarbonate, PEG succinimidyl carbonate, PEG-carboxymethyl azide, dimethylmaleic anhydride PEG, PEG dithiocarbonate derivatives, PEG-tresylates (2,2,2- trifluoroethanesolfonates), mPEG imidoesters, and other as described in Zalipsky and Lee, (1992) ("Use of functionalized poly(ethylene glycol)s for modification of peptides" in PolvfEthylene Glycol) Chemistry: Biotechnical and Biomedical Applications, J. Milton Harris, Ed., Plenum Press, NY).
In one embodiment, the invention provides an anti-CD40L antibody single variable domain composition comprising an antibody single variable domain and PEG polymer wherein the ratio of PEG polymer to antibody single variable domain is a molar ratio of at least 0.25:1. In a further embodiment, the molar ratio of PEG polymer to antibody single variable domain is 0.33:1 or greater. In a still further embodiment the molar ratio of PEG polymer to antibody single variable domain is 0.5:1 or greater.
Dual-specific Ligands
The invention also provides dual-specific ligands comprising immunoglobulin single variable domains which each have different specificities; that is, the first and the second epitopes bound by the dual-specific ligand are preferably different. As used herein a "dual-specific ligand" refers to 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. For example, 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. Dual-specific ligands may be, or be part of, polypeptides, proteins or nucleic acids, which may be naturally occurring or synthetic. In this respect, the ligand of the invention may bind an epiotpe or antigen and act as an antagonist or agonist (eg, EPO receptor agonist). The epitope binding domains of the ligand in one embodiment have the same epitope specificity, and may for example simultaneously bind their epitope when multiple copies of the epitope are present on the same antigen. In another embodiment, these epitopes are provided on different antigens such that the ligand can bind the epitopes and bridge the antigens. One skilled in the art will appreciate that the choice of epitopes and antigens is large and varied. They may be for instance human or animal proteins, cytokines, cytokine receptors, enzymes co-factors for enzymes or DNA binding proteins. Suitable cytokines and growth factors include but are not limited to: ApoE, Apo-SAA, BDNF5 Cardiotrophin-1, EGF, EGF receptor, ENA-78, Eotaxin, Eotaxin-2, Exodus-2, EpoR, FGF-acidic, FGF-basic, fibroblast growth factor-10, FLT3 ligand, Fractalkine (CX3C), GDNF, G-CSF, GM-CSF, GF- βl, insulin, IFN-γ, IGF-I, IGF-II, IL-I α, IL-lβ, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL- 8 (72 a.a.), IL-8 (77 a.a.), IL-9, IL-IO, IL-11, IL-12, IL-13, IL-15, IL-16, IL-17, IL-18 (IGIF), Inhibin α, Inhibin β, IP-IO, keratinocyte growth factor-2 (KGF-2), KGF, Leptin, LIF, Lymphotactin, Mullerian inhibitory substance, monocyte colony inhibitory factor, monocyte attractant protein, M-CSF, MDC (67 a.a.), MDC (69 a.a.), MCP-I (MCAF), MCP-2, MCP-3, MCP-4, MDC (67 a.a.), MDC (69 a.a.), MIG, MIP-Ia, MlP-lβ, MIP-3α, MIP-3β, MIP-4, myeloid progenitor inhibitor factor-1 (MPIF-I), NAP-2, Neurturin, Nerve growth factor, β-NGF, NT-3, NT-4, Oncostatin M, PDGF-AA, PDGF-AB, PDGF-BB, PF-4, RANTES, SDFlα, SDFlβ, SCF, SCGF, stem cell factor (SCF), TARC, TGF-α, TGF-β, TGF-β2, TGF-β3, tumour necrosis factor (TNF), TNF-α, TNF-β, TNF receptor I, TNF receptor II, TNIL-I, TPO, VEGF, VEGF receptor 1, VEGF receptor 2, VEGF receptor 3, GCP-2, GRO/MGSA, GRO-β, GRO-γ, HCCl, 1-309, HER 1, HER 2, HER 3, HER 4, TACE recognition site, TNF BP-I and TNF BP-II, as well as any target disclosed in Annex 2 or Annex 3 hereto, whether in combination as set forth in the Annexes, in a different combination or individually. Cytokine receptors include receptors for the foregoing cytokines, e.g. IL-I Rl; IL-6R; IL-IOR; IL- 18R, as well as receptors for cytokines set forth in Annex 2 or Annex 3 and also receptors disclosed in Annex 2 and 3. It will be appreciated that this list is by no means exhaustive. Where the multispecific ligand binds to two epitopes (on the same or different antigens), the antigen(s) may be selected from this list.
In one embodiment of the second configuration of the invention, the variable domains are derived from an antibody directed against the first and/or second antigen or epitope. In a preferred embodiment the variable domains are derived from a repertoire of single variable antibody domains. In one example, the repertoire is a repertoire that is not created in an animal or a synthetic repertoire. In another example, the single variable domains are not isolated (at least in part) by animal immunisation. Thus, the single domains can be isolated from a naϊve library.
In another aspect, the invention provides a multi-specific ligand comprising a first epitope binding domain having a first epitope binding specificity and a non- complementary second epitope binding domain having a second epitope binding specificity. The first and second binding specificities may be the same or different.
In a further aspect, the present invention provides a closed conformation multi-specific ligand comprising a first epitope binding domain having a first epitope binding specificity and a non-complementary second epitope binding domain having a second epitope binding specificity wherein the first and second binding specificities are capable of competing for epitope binding such that the closed conformation multi- specific ligand cannot bind both epitopes simultaneously.
In a still further aspect, the invention provides open conformation ligands comprising non-complementary binding domains, wherein the domains are specific for a different epitope on the same target. Such ligands bind to targets with increased avidity. Similarly, the invention provides multivalent ligands comprising non- complementary binding domains specific for the same epitope and directed to targets which comprise multiple copies of said epitope. In a similar aspect, ligands according to the invention can be configured to bind individual epitopes with low affinity, such that binding to individual epitopes is not therapeutically significant; but the increased avidity resulting from binding to two epitopes provides a therapeutic benefit. In a particular example, epitopes may be targeted which are present individually on normal cell types, but present together only on abnormal or diseased cells, such as tumour cells. In such a situation, only the abnormal or diseased cells are effectively targeted by the bispecific ligands according to the invention.
Ligand specific for multiple copies of the same epitope, or adjacent epitopes, on the same target (known as chelating dAbs) may also be trimeric or polymeric
(tertrameric or more) ligands comprising three, four or more non-complementary binding domains. For example, ligands may be constructed comprising three or four
VH domains or VL domains.
Moreover, ligands are provided which bind to multisubunit targets, wherein each binding domain is specific for a subunit of said target. The ligand may be dimeric, trimeric or polymeric.
The invention also includes a dual specific ligand comprising a first immunoglobulin single variable domain having a binding specificity to a first antigen and a second single variable domain having a binding activity to a second antigen, wherein the first antigen is CD40L and the second single variable domain is an Antigen Presenting Cell surface antigen or a T cell surface antigen. The Antigen Presenting Cell (APC) surface antigen can be selected from one of the group consisting of dendritic cell surface antigens, activated macrophage surface antigens, activated B cell surface antigens, co-stimulatory signal pathway surface antigens, and MHC, such as MHC II alpha or beta.
The (APC) surface antigen may be selected from the group consisting of CD28, Inducible costimulatory molecule (ICOS), CD27, CD30, OX40, CD45, CD69, CD3, CD70, Inducible costimulatory molecule ligand (ICOSL), OX40L, CD80, CD86, HVEM (Herpes Virus Entry Mediator), and LIGHT, but is preferably one of CD28, Inducible costimulatory molecule (ICOS), CD27, CD30, OX40, CD45, CD69, or CD3.
The surface antigen is preferably a B7 gene surface antigen such as B7-2 or B7-1.
Dendritic cell surface antigens are known in the art and can include but are not limited to ICAM-I, ICAM-2, LFA-I, LFA-3, DEC205, MHC class I, MHC class II, B7-1, and B7-2. Activated macrophage surface antigens include, but are not limited to, TNF receptor, CD40, MHC class I and II, and B7 molecules. Activated B-cell surface antigens are known in the art (e.g., including but not limited to CD20 and CD86)and further described above (see, for example, Janeway et al.} 1999, Immunobiologv, Garland Publishing NY, NY).
Preferably, the multi -specific ligands according to the above aspects of the invention are obtainable by the method comprising the steps of: a) selecting a first epitope binding domain by its ability to bind to a first epitope,
b) selecting a second epitope binding domain by its ability to bind to a second epitope,
c) combining the epitope binding domains; and d) selecting the closed conformation multispecific ligand by its ability to bind to said first second epitope and said second epitope..
Advantageously the first epitope binding domain and the second epitope binding domains are non-complementary immunoglobulin variable domains, as herein defined. That is either VH" VH VL-VL variable domains.
Chelating dAbs in particular may be prepared according to a preferred aspect of the invention, namely the use of anchor dAbs, in which a library of dimeric, trimeric or multimeric dAbs is constructed using a vector which comprises a constant dAb upstream or downstream of a linker sequence, with a repertoire of second, third and further dAbs being inserted on the other side of the linker. In alternative methodologies, the use of linkers may be avoided, for example by the use of non- covalent bonding or natural affinity between binding domains such as VH and Vκ. The invention accordingly provides a method for preparing a multimeric ligand comprising the steps of:
(a) providing a vector comprising a nucleic acid sequence encoding a single binding domain specific for a first epitope on a target;
(b) providing a vector encoding a repertoire comprising second binding domains specific for a second epitope on said target, which epitope can be the same or different to the first epitope, said second epitope being adjacent to said first epitope; and
(c) expressing said first and second binding domains; and
(d) isolating those combinations of first and second binding domains which combine together to produce a target-binding dimer.
The first and second epitopes are adjacent such that a multimeric ligand is capable of binding to both epitopes simultaneously. This provides the ligand with the advantages of increased avidity if binding. Where the epitopes are the same, the increased avidity is obtained by the presence of multiple copies of the epitope on the target, allowing at least two copies to be simultaneously bound in order to obtain the increased avidity effect.
In an alternative embodiment of the above aspect of the second configuration of the invention, at least one epitope binding domain comprises a non- immunoglobulin 'protein scaffold' or 'protein skeleton' as herein defined. Suitable non-immunoglobulin protein scaffolds include but are not limited to any of those selected from the group consisting of: SpA, fibronectin, GroEL and other chaperones, lipocallin, CCTLA4 and affibodies, as set forth above. According to the above aspect of the second configuration of the invention, advantageously, the epitope binding domains are attached to a 'protein skeleton'. Advantageously, a protein skeleton according to the invention is an immunoglobulin skeleton.
According to the present invention, the term 'immunoglobulin skeleton' refers to a protein which comprises at least one immunoglobulin fold and which acts as a nucleus for one or more epitope binding domains, as defined herein.
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.
Linking of the skeleton to the epitope binding domains, as herein defined may be achieved at the polypeptide level, that is after expression of the nucleic acid encoding the skeleton and/or the epitope binding domains. Alternatively, the linking step may be performed at the nucleic acid level. Methods of linking a protein skeleton according to the present invention, to the one or more epitope binding domains include the use of protein chemistry and/or molecular biology techniques which will be familiar to those skilled in the art and are described herein.
Advantageously, the dual- or multispecific ligand may comprise a first domain capable of binding a target molecule, and a second domain capable of binding a molecule or group which extends the half-life of the ligand. For example, the molecule or group may be a bulky agent, such as HSA or a cell matrix protein. As used herein, the phrase "molecule or group which extends the half-life of a ligand" refers to a molecule or chemical group which, when bound by a dual-specific ligand as described herein increases the in vivo half-life of such dual specific ligand when administered to an animal, relative to a ligand that does not bind that molecule or group. Examples of molecules or groups that extend the half-life of a ligand are described hereinbelow. In a preferred embodiment, the closed conformation multispecific ligand may be capable of binding the target molecule only on displacement of the half-life enhancing molecule or group. Thus, for example, a closed conformation multispecific ligand is maintained in circulation in the bloodstream of a subject by a bulky molecule such as HSA. When a target molecule is encountered, competition between the binding domains of the closed conformation multispecific ligand results in displacement of the HSA and binding of the target. Molecules which increase half-life are discussed in further detail above.
Ligands according to any aspect of the present invention, as well as dAb monomers useful in constructing such ligands, may advantageously dissociate from their cognate target(s) with a Kd of 30OnM to 5pM (ie, 3 x 10"7 to 5 x 10"12M), preferably 5OnM to20pM, or 5nM to 20OpM or InM to 10OpM, 1 x 10'7 M or less, 1 x
10"8 M or less, 1 x 1O-9 M or less, 1 x 10"10 M or less, 1 x 10"1 ' M or less; and/or a Koff rate constant of 5 x 10"1 to 1 x 10"7 S"1, preferably 1 x 10"2 to 1 x 10
Figure imgf000090_0001
or 5 x 10"3 to 1 x 10"5 S"1, or 5 x 10'1 S"1 or less, or 1 x 10"2 S'1 or less, or 1 x 10'3 S"1 or less, or 1 x
10"4 S'1 or less, or 1 x 10~5 S"1 or less, or 1 x 10'6 S"1 or less as determined by surface plasmon resonance. The Kd rate constand is defined as K0ff/KOI1.
Furthermore, the invention provides a dAb monomer(or dual specific ligand comprising such a dAb) that binds to serum albumin (SA) with a Kd of InM to 500μM (ie, x 10'9 to 5 x 10"4), preferably 10OnM to lOμM. Preferably, for a dual specific ligand comprising a first anti-SA dAb and a second dAb to another target, the affinity (eg Ka 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. For example, the first dAb binds SA with an affinity of approximately lOμM, while the second dAb binds its target with an affinity of lOOpM, Preferably, the serum albumin is human serum albumin (HSA).
In one embodiment, the first dAb (or a dAb monomer) binds SA (eg, HSA) with a Kd of approximately 50, preferably 70, and more preferably 100, 150 or 200 nM.
The invention moreover provides dimers, trimers and polymers of the aforementioned dAb monomers, in accordance with the foregoing aspect of the present invention.
Ligands according to the invention, including dAb monomers, dimers and trimers, can be linked to an antibody Fc region, comprising one or both of CH2 and Cjp domains, and optionally a hinge region. For example, vectors encoding ligands linked as a single nucleotide sequence to an Fc region may be used to prepare such polypeptides. Alternatively, ligands according to the invention may be free of an Fc domain.
In a further aspect, the present invention provides one or more nucleic acid molecules encoding at least a dual- or multispecific ligand as herein defined. In one embodiment, the ligand is a closed conformation ligand. In another embodiment, it is an open conformation ligand. The multispecific ligand may be encoded on a single nucleic acid molecule; alternatively, each epitope binding domain may be encoded by a separate nucleic acid molecule. Where the ligand is encoded by a single nucleic acid molecule, the domains may be expressed as a fusion polypeptide, or may be separately expressed and subsequently linked together, for example using chemical linking agents. Ligands expressed from separate nucleic acids will be linked together by appropriate means.
The nucleic acid may further encode a signal sequence for export of the polypeptides from a host cell upon expression and may be fused with a surface component of a filamentous bacteriophage particle (or other component of a selection display system) upon expression. Leader sequences, which may be used in bacterial expression and/or phage or phagemid display, include pelB, stll, ompA, phoA, bla and pelA.
In a further aspect of the second configuration of the invention the present invention provides a vector comprising nucleic acid according to the present invention.
In a yet further aspect, the present invention provides a host cell transfected with a vector according to the present invention.
Expression from such a vector may be configured to produce, for example on the surface of a bacteriophage particle, epitope binding domains for selection. This allows selection of displayed domains and thus selection of 'multispecific ligands' using the method of the present invention.
Combining single variable domains
Domains useful in the invention, once selected using methods exemplified above, 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. One linker example is a (GIy4 Ser)n linker, where n=l to 8, eg, 2, 3, 4, 5 or 7. The linkers used in diabodies, which are less flexible, may also be employed (Holliger et al, (1993) PNAS (USA) 90:6444-6448).
In one embodiment, 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 VH~VH>VL"VL or VH-VL 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 stabilizing variable domains of immunoglobulins, and in particular antibody VH domains, may be employed as appropriate.
In accordance with the present invention, dual specific ligands can be in "closed" conformations in solution. A "closed" configuration is that in which the two domains (for example VH and VL) are present in associated form, such as that of an associated VH-VL pair which foπns an antibody binding site. For example, scFv may be in a closed conformation, depending on the arrangement of the linker used to link the VH and VL domains. If this is sufficiently flexible to allow the domains to associate, or rigidly holds them in the associated position, it is likely that the domains will adopt a closed conformation.
Similarly, VH domain pairs and VL domain pairs may exist in a closed conformation. Generally, this will be a function of close association of the domains, such as by a rigid linker, in the ligand molecule. Ligands in a closed conformation will be unable to bind both the molecule which increases the half-life of the ligand and a second target molecule. Thus, the ligand will typically only bind the second target molecule on dissociation from the molecule which increases the half-life of the ligand.
Moreover, the construction of VH/VH, VL/VL or VH/VL dimers without linkers provides for competition between the domains.
Ligands according to the invention may moreover be in an open conformation. In such a conformation, the ligands will be able to simultaneously bind both the molecule which increases the half-life of the ligand and the second target molecule. Typically, variable domains in an open configuration are (in the case of VH-VL pairs) held far enough apart for the domains not to interact and form an antibody binding site and not to compete for binding to their respective epitopes. In the case of VH/VH or VL/VL dimers, the domains are not forced together by rigid linkers. Naturally, such domain pairings will not compete for antigen binding or form an antibody binding site.
Fab fragments and whole antibodies will exist primarily in the closed conformation, although it will be appreciated that open and closed dual specific ligands are likely to exist in a variety of equilibria under different circumstances. Binding of the ligand to a target is likely to shift the balance of the equilibrium towards the open configuration. Thus, certain ligands according to the invention can exist in two conformations in solution, one of which (the open form) can bind two antigens or epitopes independently, whilst the alternative conformation (the closed form) can only bind one antigen or epitope; antigens or epitopes thus compete for binding to the ligand in this conformation.
Although the open form of the dual specific ligand may thus exist in equilibrium with the closed form in solution, it is envisaged that the equilibrium will favor the closed form; moreover, the open form can be sequestered by target binding into a closed conformation. Preferably, therefore, certain dual specific ligands of the invention are present in an equilibrium between two (open and closed) conformations.
Dual specific ligands according to the invention may be modified in order to favor an open or closed conformation. For example, stabilisation of VR-VL interactions with disulphide bonds stabilises the closed conformation. Moreover, linkers used to join the domains, including VR domain and VL domain pairs, may be constructed such that the open from is favoured; for example, the linkers may sterically hinder the association of the domains, such as by incorporation of large amino acid residues in opportune locations, or the designing of a suitable rigid structure which will keep the domains physically spaced apart. Characterisation of the dual-specific ligand.
The binding of the dual-specific ligand to its specific antigens or epitopes (e.g., CD40L and/or an epitope bound by DOM8-24) can be tested by methods which will be familiar to those skilled in the art and include ELISA. In a preferred embodiment of the invention 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. It is also desirable to screen soluble antibody fragments for binding to antigen or epitope, and this can also be undertaken by ELISA using reagents, for example, against a C- or N- terminal tag (see for example Winter et al. (1994) Ann. Rev. Immunology 12, 433-55 and references cited therein.
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.
Structure of "Dual-specific ligands'.
As described above, an antibody is herein defined as an antibody (for example IgG, IgM, IgA, IgA, IgE) or fragment (Fab, Fv, disulphide linked Fv, scFv, diabody) which comprises at least one heavy and a light chain variable domain, at least two heavy chain variable domains or at least two light chain variable domains. It may be at least partly 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). In a preferred embodiment of the invention the dual-specific ligand comprises at least one single heavy chain variable domain of an antibody and one single light chain variable domain of an antibody, or two single heavy or light chain variable domains. For example, the ligand may comprise a VH/VL pair, a pair of VH domains or a pair of VL domains.
The first and the second variable domains of such a ligand may be on the same polypeptide chain. Alternatively they may be on separate polypeptide chains. In the case that they are on the same polypeptide chain they may be linked by a linker, which is preferentially a peptide sequence, as described above.
The first and second variable domains may be covalently or non-covalently associated. In the case that they are covalently associated, the covalent bonds may be disulphide bonds.
In the case that the 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.
Where 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.
In one embodiment of the invention the 'dual-specific ligand' is a single chain Fv fragment. In an alternative embodiment of the invention, the 'dual-specific ligand' consists of a Fab format. In a further aspect, the present invention provides nucleic acid encoding at least a 'dual-specific ligand' as herein defined.
One skilled in the art will appreciate that, depending on the aspect of the invention, both antigens or epitopes may bind simultaneously to the same antibody molecule. Alternatively, they may compete for binding to the same antibody molecule. For example, where both epitopes are bound simultaneously, both variable domains of a dual specific ligand are able to independently bind their target epitopes.
Where the domains compete, the one variable domain is capable of binding its target, but not at the same time as the other variable domain binds its cognate target; or the first variable domain is capable of binding its target, but not at the same time as the second variable domain binds its cognate target.
The variable regions may be derived from antibodies directed against target antigens or epitopes. Alternatively they may be derived from a repertoire of single antibody domains such as those expressed on the surface of filamentous bacteriophage. Selection may be performed as described below.
In general, the nucleic acid molecules and vector constructs required for the performance of the present invention may be constructed and manipulated as set forth in standard laboratory manuals, such as Sambrook et a (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.
Thus in a further aspect, the present invention provides a vector comprising nucleic acid encoding at least a 'dual-specific ligand' as herein defined.
As used herein, 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 "polylinker") 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. Typically in cloning vectors, 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. Such 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. Generally, 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.
Advantageously, 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. Since the replication of vectors encoding a ligand according to the present invention is most conveniently performed in E. coli, an E. co//-selectable marker, for example, the β-lactamase gene that confers resistance to the antibiotic ampicillin, is of use. These can be obtained from E. coli plasmids, such as pBR.322 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. The term "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 β- 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. Thus, 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. As described above, the preferred
' selection display system is bacteriophage display. Thus, 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. One example are phagemid vectors which have an E. coli. origin of replication (for double stranded replication) and also- a phage origin of replication (for production of single-stranded DNA). The manipulation and expression of such vectors is well known in the art (Hoogenboom and Winter (1992) supra; Nissim et al. (1994) supra). Briefly, the vector contains a β-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. Thus, using various suppressor and non-suppressor strains of E. coli and with the addition of glucose, iso-propyl thio-β-D-galactoside (IPTG) or a helper phage, such as VCS Ml 3, 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. 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, KNfA or protein, in situ hybridisation, immunocytochernistry or sequence analysis of nucleic acid or protein molecules. Those skilled in the art will readily envisage how these methods may be modified, if desired.
Structure of ligands
According to one aspect of the invention, two or more non-complementary epitope binding domains are linked so that they are in a closed conformation as herein defined. Advantageously, they may be further attached to a skeleton which may, as a alternative, or on addition to a linker described herein, facilitate the formation and/or maintenance of the closed conformation of the epitope binding sites with respect to one another. Alternatively, the monomeric anti-CD40L antibody single variable domain polypeptides of the invention may be constructed using scaffold or skeleton frameworks as discussed herein.
(I) Skeletons
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 Fv5 scFv, Fab or F(ab')2 molecules. Those skilled in the art will be aware that this list is not intended to be exhaustive.
(II) Protein scaffolds
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. Advantageously, an epitope binding domain according to the present invention comprises three CDRs. Suitable protein scaffolds, in addition to those based on immunoglobulin domains, may also be based on protein scaffolds or skeletons other than immunoglobulin domains. For example natural bacterial receptors such as SpA have 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 (Affϊbody, Bromma, Sweeden). Details of suitable procedures are described in WO 98/58965. Other suitable, scaffolds include lipocallin and CTLA4, as described in van den Beuken et al, J. MoI. Biol. (2001) 310, 591-601, and scaffolds such as those described in WO0069907 (Medical Research Council), which are based for example on the ring structure of bacterial GroEL or other chaperone polypeptides. Other non-immunoglobulin based scaffolds which may be used according to the invention include those based on the LDL receptor class A, EGF domain monomers and mutimers, and scaffolds available from Biorexis (King of Prussia, PA) or Avidia (Mountain View, CA). Other non-immunoglobulin scaffolds which may be used are described, for example, in WO05/040229, WO04/044011, and US20050089932
Scaffolds for use in Constructing Ligands
i. Selection of the main-chain conformation
The members of the immunoglobulin superfamily all share a similar fold for their polypeptide chain. For example, although antibodies are highly diverse in terms of their primary sequence, comparison of sequences and crystallographic 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. (1992) J. MoI. Biol., 227: 799; Tomlinson et al. (1995) EMBO J., 14: 4628; Williams et al. (1996) J. MoI. Biol, 264: 220). Although the 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).
The ligands of the present invention are advantageously selected and/or assembled from libraries of domains, such as libraries of VH domains and/or libraries of VL domains. Moreover, the ligands of the invention may themselves be provided in the form of libraries. In one aspect of the present invention, libraries of ligands and/or domains are designed in which certain loop lengths and key residues have been chosen to ensure that the main-chain conformation of the members is known. Advantageously, 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 chose residues for diversification which do not affect the canonical structure. lt is known that, in the human Vκ domain, the Ll loop can adopt one of four canonical structures, the L2 loop has a single canonical structure and that 90% of human Vκ domains adopt one of four or five canonical structures for the L3 loop (Tomlinson et al. (1995) supra); thus, in the Vκ domain alone, different canonical structures can combine to create a range of different main-chain conformations. Given that the Vj1 domain encodes a different range of canonical structures for the Ll, L2 and L3 loops and that Vκ and Yx domains can pair with any VR 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. However, 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. Even more surprisingly, 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. Thus, in a preferred aspect, the 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. Accordingly, in a preferred aspect of the invention, 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. It is more preferable, that 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.
In designing ligands or libraries thereof the incidence of the different main- chain conformations for each of the antigen binding loops may be considered separately. For 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. Typically, its observed incidence is above 35% (i.e. between 35% and 100%) and, ideally, above 50% or even above 65%. 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. In human antibodies, 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 Vκ (39%), L2 - CS 1 (100%),
L3 - CS 1 of Vκ (36%) (calculation assumes a κ:λ ratio of 70:30, Hood et al. (1967) Cold Spring Harbor Symp. Quant. Biol., 48: 133). For H3 loops that have canonical structures, a CDR3 length (Kabat eϊ at. (1991) Sequences of proteins of immunological interest, U.S. Department of Health and Human Services) of seven residues with a salt-bridge from residue 94 to residue 101 appears to be the most common. There are at least 16 human antibody sequences in the EMBL data library with the required H3 length and key residues to form this conformation and at least two crystal! ographic structures in the protein data bank which can be used as a basis for antibody modelling (2cgr and Itet). The most frequently expressed germline gene segments that this combination of canonical structures are the VH segment 3-23 (DP- 47), the JH segment JH4b, the Vκ segment 02/012 (DPK9) and the Jκ segment Jκl. VH 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.
Alternatively, instead of choosing the single main-chain conformation based on the natural occurrence of the different main-chain conformations for each of the binding loops in isolation, the natural occurrence of combinations of main-chain conformations is used as the basis for choosing the single main-chain conformation. In the case of antibodies, for example, 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. Here, it is preferable that the chosen conformation , is commonplace in naturally occurring antibodies and most preferable that it observed most frequently in the natural repertoire. Thus, in human antibodies, for example, when natural combinations of the five antigen binding loops, Hl, H2, Ll, L2 and L3, are considered, 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.
ii. Diversification of the canonical sequence
Having selected several known main-chain conformations or, preferably a single known main-chain conformation, ligands according to the invention or 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.
Various methods have been reported for introducing such diversity. Error- prone PCR (Hawkins et al. (1992) J. MoI Biol, 226: 889), chemical mutagenesis (Deng et al. (1994) J. Biol. Chem., 269: 9533) or bacterial mutator strains (Low et al. (1996) J MoI. Biol, 260: 359) can be used to introduce random mutations into the genes that encode the molecule. Methods for mutating selected positions are also well known in the art and include the use of mismatched oligonucleotides or degenerate oligonucleotides, with or without the use of PCR. For example, several synthetic antibody libraries have been created by targeting mutations to the antigen binding loops. The 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. Sc?. 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. Sci. USA, 89: 4457; Nissim et al. (1994) EMBO J, 13: 692; Griffiths et al (1994) EMBO J, 13: 3245; De Kruif et al. (1995) J. MoI Biol, 248: 97). Such diversification has been extended to include some or all of the other antigen binding loops (Crameri et al (1996) Nature Med., 2: 100; Riechmann et al. (1995) Bio/Technology, 13: 475; Moiphosys, WO97/08320, supra). Since loop randomisation has the potential to create approximately more than 1015 structures for H3 alone and a similarly large number of variants for the other five loops, it is not feasible using current transformation technology or even by using cell free systems to produce a library representing all possible combinations. For example, in one of the largest libraries constructed to date, 6 x 1010 different antibodies, which is only a fraction of the potential diversity for a library of this design, were generated (Griffiths et al (1994) supra).
In a preferred embodiment, only those residues which are directly involved in creating or modifying the desired function of the molecule are diversified. For many molecules, the function will be to bind a target and therefore diversity should be concentrated in the target binding site, while avoiding changing residues which are crucial to the overall packing of the molecule or to maintaining the chosen main-chain conformation.
Diversification of the canonical sequence as it applies to antibody domains
In the case of the ligands of the invention, the binding site for the target is most often the antigen binding site. Thus, in a highly preferred aspect, the invention provides libraries of or for the assembly of antibody ligands in which only those 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. For example, in L2 it is known that positions 50 and 53 are diverse in naturally occurring antibodies and are observed to make contact with the antigen. In contrast, 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. This represents a significant improvement in terms of the functional diversity required to create a range of antigen binding specificities.
In nature, 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. 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 a (1996) J MoI. Biol, 256: 813). This complementarity has probably evolved as an efficient strategy for searching sequence space and, although apparently unique to antibodies, it can easily be applied to other polypeptide repertoires. The residues which are varied are a subset of those that form the binding site for the target. Different (including overlapping) subsets of residues in the target binding site are diversified at different stages during selection, if desired.
In the case of an antibody repertoire, an initial 'naive' repertoire is created where some, but not all, of the residues in the antigen binding site are diversified. As used herein in this context, the term "naive" refers to antibody molecules that have no pre-determined 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.
The invention provides two different naive repertoires of binding domains for the construction of ligands, or a naive library of ligands, in which some or all of the residues in the antigen binding site are varied. 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). Those residues which are diversified include, but are not limited to, H50, H52, H52a, H53, H55, H56, H58, H95, H96, Wl, H98, L50, L53, L91, L92, L93, L94 and L96. In the "somatic" library, diversity is restricted to residues that are diversified during the recombination process (junctional diversity) or are highly somatically mutated). Those residues which are diversified include, but are not limited to: H31, H33. H35, H95, H96, H97, H98, L30, L31, L32, L34 and L96. All the residues listed above as suitable for diversification in these libraries are known to make contacts in one or more antibody-antigen complexes. Since in both libraries, not all of the residues in the antigen binding site are varied, additional diversity is incorporated during selection by varying the remaining residues, if it is desired to do so. It shall be apparent to one skilled in the art that any subset of any of these residues (or additional residues which comprise the antigen binding site) can be used for the initial and/or subsequent diversification of the antigen binding site.
In the construction of libraries for use in the invention, 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. Using the IUPAC nomenclature, 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 VH, VK and Vx 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. Since it is preferable to mimic this distribution of amino acids, 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. By comparing the amino acid profile encoded by all combinations of degenerate codons (with single, double, triple and quadruple degeneracy in equal ratios at each position) with the natural amino acid use it is possible to calculate the most representative codon. The codons (AGT)(AGC)T5 (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. Preferably, therefore, libraries are constructed using either the DVT, DVC or DVY codon at each of the diversified positions.
Increased Half-life
In vivo, the PEGylated monovalent anti-CD40L antibodies as described herein confer a distinct advantage over non-PEGylated antibody polypeptides, in that the
PEGylated antibody molecules will have a greatly prolonged in vivo half life.
Without being bound to one particular theory, it is believed that the increased half-life of the molecules described herein is conferred by the increased hydrodynamic size of the antibody polypeptide resulting from the attachment of PEG polymer(s). More specifically, it is believed that two parameters play an important role in determining the serum half-life of PEGylated antibody polypeptides. The first criterion is the nature and size of the PEG attachment, i.e., if the polymer used is simply a linear chain or a branched/forked chain, wherein the branched/forked chain gives rise to a longer half-life. The second is the location of the PEG moiety or moieties on the antibody polypeptide in the final format and how many "free" unmodified PEG arms the molecule has. The resulting hydrodynamic size of the PEGyI ated antibody polypeptide, as estimated, for example, by size exclusion chromatography, reflects the serum half-life of the molecule. Accordingly, the larger the hydrodynamic size of the PEGylated molecule, the greater the serum half life.
Increased half-life is useful in vivo applications of immunoglobulins, especially antibodies and most especially antibody fragments of small size. Such fragments (Fvs, Fabs, scFvs, dAbs) suffer from rapid clearance from the body; thus, while they are able to reach most parts of the body rapidly, and are quick to produce and easier to handle, their in vivo applications have been limited by their only brief persistence in vivo.
In one aspect, a monovalent anti-CD40L antibody polypeptide as described herein is stabilized in vivo by fusion with a moiety, such as PEG, that increases the hydrodynamic size of the antibody polypeptide. Methods for pharmacokinetic anafysis and determination of half-life will be familiar to those skilled in the art. Details may be found in Kenneth et al: Chemical Stability of Pharmaceuticals: A Handbook for Pharmacists and in Peters et al, Pharmacokinetc analysis: A Practical Approach (1996). Reference is also made to "Pharmacokinetics", M Gibaldi & D Perron, published by Marcel Dekker, 2nd Rev. ex edition (1982), which describes pharmacokinetic parameters such as t alpha and t beta half lives and area under the curve (AUC).
Typically, the half life of a PEGylated antibody polypeptide as described herein is increased by 10%, 20%, 30%, 40%, 50% or more relative to a non- PEGylated dAb (wherein the antibody polypeptide of the PEGylated antibody polypeptide and non-PEGylated antibody polypeptide are the same). Increases in the range of 2x, 3x, 4x, 5x, 7x, 1Ox, 20x, 3Ox, 4Ox, and up to 5Ox or more of the half life are possible. Alternatively, or in addition, increases in the range of up to 30x, 40x, 5Ox, 6Ox, 7Ox, 8Ox, 9Ox, 10Ox, 150x of the half life are possible.
Half lives (tl4 alpha and t1/. beta) and AUC can be determined from a curve of serum concentration of ligand against time. The WiriNonlin analysis package (available from Pharsight Corp., Mountain View, CA 94040, USA) can be used, for example, to model the curve. In a first phase (the alpha phase) the ligand is undergoing mainly distribution in the patient, with some elimination. A second phase (beta phase) is the terminal phase when the ligand has been distributed and the serum concentration is decreasing as the ligand is cleared from the patient. The tα half life is the half life of the first phase and the tβ half life is the half life of the second phase. "Half-life" as used herein, unless otherwise noted, refers to the overall half-life of an antibody single variable domain of the invention determined by non-compartment modeling (as contrasted with biphasic modeling, for example). Beta half-life is a measurement of the time it takes for the amount of dAb monomer or multimer to be cleared from the mammal to which it is administered. Thus, advantageously, the present invention provides a dAb-containing composition, e.g., a dAb-effector group composition, having a tα half-life in the range of 0.25 hours to 6 hours or more. In one embodiment, the lower end of the range is 30 minutes, 45 minutes, 1 hour, 1.3 hours, 2 hours, 3 hours, 4 hours, 5 hours, 6 hours, 7 hours, 10 hours, 11 hours or 12 hours. In addition or alternatively, a dAb containing composition will have a tα half- life in the range of up to and including 12 hours. In one embodiment, the upper end of the range is 11, 10, 9, 8, 7, 6, or 5 hours. An example of a suitable range is 1.3 to 6 hours, 2 to 5 hours or 3 to 4 hours. Advantageously, the present invention provides a dAb containing composition comprising a ligand according to the invention having a tβ half-life in the range of 1 - 170 hours or more. In one embodiment, the lower end of the range is 2.5 hours, 3 hours, 4 hours, 5 hours, 6 hours, 7 hours, 8 hours, 9 hours, 10 hours, 11 hours or 12 hours. In addition, or alternatively, a dAb containing composition, e.g. a dAb-effector group composition has a tβ half-life in the range of up to and including 21 days. In one embodiment, the upper end of the range is 12 hours, 24 hours, 2 days, 3 days, 5 days, 10 days, 15 days, or 20 days. Advantageously a dAb containing composition according to the invention will have a tβ half-life in the range 2-100 hours, 4-80 hours, and 10-40 hours. In a further embodiment, it will be in the range 12-48 hours. In a further embodiment still, it will be in the range 12-26 hours. The present invention provides a dAb containing composition comprising a ligand according to the invention having a half-life in the range of 1-170 hours or more. In one embodiment, the lower end of the range is 1.3 hours, 2.5 hours, 3 hours, 4 hours, 5 hours, 6 hours, 7 hours, 8 hours, 9 hours, 10 hours, 11 hours or 12 hours. In addition, or alternatively, a dAb containing composition, e.g. a dAb-effector group composition has a half-life in the range of up to and including 21 days. In one embodiment, the upper end of the range is 12 hours, 24 hours, 2 days, 3 days, 5 days, 10 days, 15 days, or 20 days.
In addition, or alternatively to the above criteria, the present invention provides a dAb containing composition comprising a ligand according to the invention having an AUC value (area under the curve) in the range of 1 mg.min/ml or more. In one embodiment, the lower end of the range is 5, 10, 15, 20, 30, 100, 200 or 300 mg.min/ml. In addition, or alternatively, a ligand or composition according to the invention has an AUC in the range of up to 600 mg.min/ml. In one embodiment, the upper end of the range is 500, 400, 300, 200, 150, 100, 75 or 50 mg.min/ml. Advantageously a ligand according to the invention will have an AUC in the range selected from the group consisting of the following: 15 to 150 mg.min/ml, 15 to 100 mg.min/ml, 15 to 75 mg.min/ml, and 15 to 50 mg.min/ml.
The ligands according to the invention, including, mono-, dual- and multi- specific, in one configuration thereof, are capable of binding to one or more molecules which can increase the half-life of the ligand in vivo. Typically, such molecules are polypeptides which occur naturally in vivo and which resist degradation or removal by endogenous mechanisms which remove unwanted material from the organism. For example, the molecule which increases the half-life of the organism may be selected from the following:
Proteins from the extracellular matrix; for example collagen, 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, eg type 1 collagen (accounting for 90% of body collagen) found in bone, skin, tendon, ligaments, cornea, internal organs or type II collagen found in cartilage, invertebral disc, notochord, vitreous humour of the eye.
Proteins found in blood, including:
Plasma proteins such as fibrin, α-2 macroglobulin, serum albumin, fibrinogen A, fibrinogen B, serum amyloid protein A, heptaglobin, profilin, ubiquitin, uteroglobulin and β-2-microglobulin;
Enzymes and inhibitors such as plasminogen, lysozyme, cystatin C, alpha- 1- antitrypsin and pancreatic trypsin inhibitor. Plasminogen is the inactive precursor of the trypsin-like serine protease plasmin. It is normally found circulating through the blood stream. When plasminogen becomes activated and is converted to plasmin, it unfolds a potent enzymatic domain that dissolves the fibrinogen fibers that entgangle the blood cells in a blood clot. This is called fibrinolysis.
Immune system proteins, such as IgE, IgG, IgM.
Transport proteins such as retinol binding protein, α-1 microglobulin.
Defensins such as beta-defensin 1, Neutrophil defensins 1,2 and 3.
Proteins found at the blood brain barrier or in neural tissues, such as melanocortin receptor, myelin, ascorbate transporter.
Transferrin receptor specific ligand-neuropharmaceutical agent fusion proteins (see US5977307); brain capillary endothelial cell receptor, transferrin, transferrin receptor, insulin, insulin-like growth factor 1 (IGF 1) receptor, insulin-like growth factor 2 (IGF 2) receptor, insulin receptor. Proteins localised to the kidney, such as polycystin, type IV collagen, organic anion transporter Kl, Heymann's antigen.
Proteins localised to the liver, for example alcohol dehydrogenase, G250.
, Blood coagulation factor X
αl antitrypsin
HNF lα
Proteins localised to the lung, such as secretory component (binds IgA).
Proteins localised to the Heart, for example HSP 27. This is associated with dilated cardiomyopathy.
Proteins localised to the skin, for example keratin.
Bone specific proteins, such as bone morphogenic proteins (BMPs), which are a subset of the transforming growth factor β superfamily that demonstrate osteogenic activity. Examples include BMP-2, -4, -5, -6, -7 (also referred to as osteogenic protein (OP-I) and -8 (OP-2).
Tumour specific proteins, including human trophoblast antigen, herceptin receptor, oestrogen receptor, cathepsins eg cathepsin B (found in liver and spleen).
Disease-specific proteins, such as 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 the only costimulatory T cell molecule known to be specifically up-regulated in human T cell leukaemia virus type-I (HTLV-I)-producing cells.) See
J Immunol 2000 JuI l;165(l):263-70; Metalloproteases (associated with arthritis/cancers), including CG6512 Drosophila, human paraplegin, human FtsH, human AFG3L2, murine ftsH; 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-a (TGF a), tumor necrosis factor-alpha (TNF-α), angiogenin, interleulάn-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), fractalkine.
Stress proteins (heat shock proteins)
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) only occurs when as a result of trauma, disease or injury and therefore in vivo, extracellular HSPs trigger a response from the immune system that will fight infection and disease. A dual specific which binds to extracellular HSP can be localised to a disease site.
Proteins involved in Fc transport
Brambell receptor (also known as FcRB):
This Fc receptor has two functions, both of which are potentially useful for delivery
The functions are
(1) The transport of IgG from mother to child across the placenta
(2) the protection of IgG from degradation thereby prolonging its serum half life of IgG. It is thought that the receptor recycles IgG from endosome.
See Holliger et al, Nat Biotechnol 1997 Jul;15(7):632-6.
Other proteins involved in Fc transport include the neonatal Fc receptor (FcRn) described in Gastinel et al., 1992, PNAS 89:638; and Roopenian et al., 2003 J. Immunol. 170:3528. Ligands according to the invention may designed to be specific for the above targets without requiring any increase in or increasing half life m vivo. For example, ligands according to the invention can be specific for targets selected from the foregoing which are tissue-specific, thereby enabling tissue-specific targeting of the dual specific ligand, or a dAb monomer that binds a tissue-specific therapeutically relevant target, irrespective of any increase in half-life, although this may result. Moreover, where the ligand or dAb monomer targets kidney or liver, this may redirect the ligand or dAb monomer to an alternative clearance pathway in vivo (for example, the ligand may be directed away from liver clearance to kidney clearance).
Polypeptides useful for increasing half-life include, but are not limited to those shown in Annex I.
Increased Protease Stability
A further advantage of the present invention is that the PEGylated dAbs and dAb multimers described herein possess increased stability to the action of proteases. Depending on the assay conditions, dAbs are generally intrinsically stable to the action of proteases. In the presence of pepsin, however, many dAbs are totally degraded at pH 2 because the protein is unfolded under the acid conditions, thus making the protein more accessible to the protease enzyme. The present invention provides PEGylated dAb molecules, including dAb multimers, wherein it is believed that the PEG polymer provides protection of the polypeptide backbone due the physical coverage of the backbone by the PEG polymer, thereby preventing the protease from gaining access to the polypeptide backbone and cleaving it. In a preferred embodiment a PEGylated dAb having a higher hydrodynamic size (e.g., 200 to 500 kDa) is generated according to the invention, because the larger hydrodynamic size will confirm a greater level of protection from protease degradation than a PEGylated dAb having a lower hydrodynamic size. In one embodiment, a PEG- or other polymer-linked antibody single variable domain monomer or multimer is degraded by no more than 10% when exposed to one or more of pepsin, trypsin, elastase, chymotrypsin, or carboxypeptidase, wherein if the protease is pepsin then exposure is carried out at pH 2.0 for 30 minutes, and if the protease is one or more of trypsin, elastase, chymotrypsin, or carboxypeptidase, then exposure is carried out at pH 8.0 for 30 minutes. In a preferred embodiment, a PEG- or other polymer-linked dAb monomer or multimer is degraded by no more than 10% when exposed to pepsin at pH 2.0 for 30 minutes, preferably no more than 5%, and preferably not degraded at all. In a further preferred embodiment, a PEG- or other polymer-linked dAb multimer (e.g., hetero- or homodimer, trimer, tetramer, octamer, etc.) of the invention is degraded by less than 5%, and is preferably not degraded at all in the presence of pepsin at pH 2.0 for 30 minutes. In a preferred embodiment, a PEG- or other polymer-linked dAb monomer or multimer is degraded by no more than 10% when exposed to trypsin, elastase, chymotrypsin, or carboxypeptidase at pH 8.0 for 30 minutes, preferably no more than 5%, and preferably not degraded at all. In a further preferred embodiment, a PEG- or other polymer-linked dAb multimer (e.g., hetero- or homodimer, trimer, tetramer, octamer, etc.) of the invention is degraded by less than 5%, and is preferably not degraded at all in the presence of trypsin, elastase, chymotrypsin, or carboxypeptidase at pH 8.0 for 30 minutes.
The relative ratios of protease rantibody single variable domain polypeptide may be altered according to the invention to achieve the desired level of degradation as described above. For example the ratio or protease to antibody single variable domain may be from about 1:30, to about 10:40, to about 20:50, to about 30:50, about 40:50, about 50:50, about 50:40, about 50:30, about 50:20, about 50:10, about 50: 1, about 40:1, and about 30:1.
Accordingly, the present invention provides a method for decreasing the degradation of an antibody single variable domain comprising linking an antibody single variable domain monomer or multimer to a PEG polymer according to any of the embodiments described herein. According to this aspect of the invention, the antibody single variable domain is degraded by no more than 10% in the presence of pepsin at pH2.0 for 30 minutes. In particular, a PEG-linked dAb multimer is degraded by no more than 5%, and preferably not degraded at all in the presence of pepsin at pH 2.0 for 30 minutes. In an alternate embodiment, the antibody single variable domain is degraded by no more than 10% when exposed to trypsin, elastase, chymotrypsin, or carboxypeptidase at pH 8.0 for 30 minutes, preferably no more than 5%, and preferably not degraded at all.
Degradation of PEG-linked dAb monomers and multimers according to the invention may be measured using methods which are well known to those of skill in the art. For example, following incubation of a PEG-linked dAb with pepsin at pH 2.0 for 30 minutes, or with trypsin, elastase, chymotrypsin, or carboxypeptidase at pH 8.0 for 30 minutes, the dAb samples may be analyzed by gel filtration, wherein degradation of the dAb monomer or multimer is evidenced by a gel band of a smaller molecular weight than an un-degraded (i.e., control dAb not treated with pepsin, trypsin, chymotrypsin, elastase, or carboxypeptidase) dAb. Molecular weight of the dAb bands on the gel may be determined by comparing the migration of the band with the migration of a molecular weight ladder (see Figure 5). Other methods of measuring protein degradation are known in the art and may be adapted to evaluate the PEG-linked dAb monomers and multimers of the present invention. Pharmaceutical Compositions. Dosage and Administration
The antibody polypeptides of the invention can be incorporated into pharmaceutical compositions suitable for administration to a subject. Typically, the pharmaceutical composition comprises a monovalent anti-CD40L antibody polypeptide and a pharmaceutically acceptable carrier. As used herein, "pharmaceutically acceptable carrier" includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like that are physiologically compatible. The term "pharmaceutically acceptable carrier" excludes tissue culture medium comprising bovine or horse serum. Examples of pharmaceutically acceptable carriers include one or more of water, saline, phosphate buffered saline, dextrose, glycerol, ethanol and the like, as well as combinations thereof. In many cases, it will be preferable to include isotonic agents, for example, sugars, polyalcohols such as mannitol, sorbitol, or sodium chloride in the composition. Pharmaceutically acceptable substances include minor amounts of auxiliary substances such as wetting or emulsifying agents, preservatives or buffers, which enhance the shelf life or effectiveness of the antibody polypeptide. The compositions as described herein may be in a variety of forms. These include, for example, liquid, semi-solid and solid dosage forms, such as liquid solutions (e.g., injectable and infusible solutions), dispersions or suspensions, tablets, pills, powders, liposomes and suppositories. The preferred form depends on the intended mode of administration and therapeutic application. Typical preferred compositions are in the form of injectable or infusible solutions, such as compositions similar to those used for passive immunization of humans with other antibodies. The preferred mode of administration is parenteral (e.g., intravenous, subcutaneous, intraperitoneal, intramuscular).
Therapeutic compositions typically must be sterile and stable under the conditions of manufacture and storage. The composition can be formulated as a solution, microemulsion, dispersion, liposome, or other ordered structure suitable to high drug concentration. Sterile injectable solutions can be prepared by incorporating the active compound in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filter sterilization. Generally, dispersions are prepared by incorporating the active compound into a sterile vehicle that contains a basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum drying and freeze-drying that yields a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof. The proper fluidity of a solution can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
The antibody polypeptides described herein can be administered by a variety of methods known in the art, although for many therapeutic applications, the preferred route/mode of administration is intravenous injection or infusion. The polypeptide can also be administered by intramuscular or subcutaneous injection. As will be appreciated by the skilled artisan, the route and/or mode of administration will vary depending upon the desired results. In certain embodiments, the active compound may be prepared with a carrier that will protect the compound against rapid release, such as a controlled release formulation, including implants, transdermal patches, and microencapsulated delivery systems. Single immunoglobulin variable domains and other relatively small monovalent antibody polypeptides are well suited for formulation as extended release preparations due, in part, to their small size - the number of moles per dose can be significantly higher than the dosage of, for example, full sized antibodies. Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Prolonged absorption of injectable compositions can be brought about by including in the composition an agent that delays absorption, for example, monostearate salts and gelatin. Many methods for the preparation of such formulations are patented or generally known to those skilled in the art. See, e.g., Sustained and Controlled Release Drug Delivery Systems, J. R. Robinson, ed., Marcel Dekker, Inc., New York, 1978. Additional methods applicable to the controlled or extended release of polypeptide agents such as the monovalent antibody polypeptides disclosed herein are described, for example, in U.S. Patent Nos. 6,306,406 and 6,346,274, as well as, for example, in U.S. Patent Application Nos. US20020182254 and US20020051808, all of which are incorporated herein by reference.
In certain embodiments, a monovalent anti-CD40L antibody polypeptide can be orally administered, for example, with an inert diluent or an assimilable edible carrier. The compound (and other ingredients, if desired) may also be enclosed in a hard or soft shell gelatin capsule, compressed into tablets, or incorporated direct!}' into the individual's diet For oral therapeutic administration, the compounds may be incorporated with excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, and the like. To administer a compound of the invention by other than parenteral administration, it may be necessary to coat the compound with, or co-administer the compound with, a material to prevent its inactivation. Additional active compounds can also be incorporated into the compositions. In certain embodiments, a monovalent anti-CD40L antibody polypeptide is coformulated with and/or coadministered with one or more additional therapeutic agents. For example, a monovalent anti-CD40L antibody polypeptide can be coformulated and/or coadministered with one or more additional antibodies that bind other targets (e.g., antibodies that bind other cytokines or that bind cell surface molecules), or, for example, one or more cytokines. Such combination therapies may utilize lower dosages of the administered therapeutic agents, thus avoiding possible toxicities or complications associated with the various monotherapies.
The pharmaceutical compositions of the invention can include a
"therapeutically effective amount" or a "prophylactically effective amount" of a monovalent anti-CD40L antibody polypeptide. A "therapeutically effective amount" refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic result. A therapeutically effective amount of the antibody polypeptide can vary according to factors such as the disease state, age, sex, and weight of the individual, and the ability of the monovalent anti-CD40L antibody polypeptide to elicit a desired response in the individual. A therapeutically effective amount is also one in λvhich any toxic or detrimental effects of the antibody or antibody portion are outweighed by the therapeutically beneficial effects. A "prophylactically effective amount" refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired prophylactic result. Typically, because a prophylactic dose is used in subjects prior to or at an earlier stage of disease, the prophylactically effective amount will be less than the therapeutically effective amount.
Dosage regimens may be adjusted to provide the optimum desired response
(e.g., a therapeutic or prophylactic response). For example, a single bolus may be administered, several divided doses may be administered over time or the dose may be proportionally reduced or increased as indicated by the exigencies of the therapeutic situation. It is advantageous to formulate parenteral compositions in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the mammalian subjects to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier.
A non-limiting range for a therapeutically or prophylactically effective amount of a monovalent anti-CD40L antibody polypeptide is 0.1-20 mg/kg, more preferably 1-10 mg/kg. It is to be noted that dosage values can vary with the type and severity of the condition to be alleviated. It is to be further understood that for any particular subject, specific dosage regimens should be adjusted over time according to the individual need and the professional judgment of the administering clinician.
The efficacy of treatment with a monovalent anti-CD40L antibody polypeptide as described herein is judged by the skilled clinician on the basis of improvement in one or more symptoms or indicators of the disease state or disorder being treated. An improvement of at least 10% (increase or decrease, depending upon the indicator being measured) in one or more clinical indicators is considered "effective treatment," although greater improvements are preferred, such as 20%, 30%, 40%, 50%, 75%, 90%, or even 100%, or, depending upon the indicator being measured, more than 100% (e.g., two-fold, three-fold, ten-fold, etc., up to and including attainment of a disease-free state. Indicators can be physical measurements, e.g., enzyme, cytokine, growth factor or metabolite levels, rate of cell growth or cell death, or the presence or amount of abnormal cells. One can also measure, for example, differences in the amount of time between flare-ups of symptoms of the disease or disorder (e.g., for remitting/relapsing diseases, such as multiple sclerosis). Alternatively, non-physical measurements, such as a reported reduction in pain or discomfort or other indicator of disease status can be relied upon to gauge the effectiveness of treatment. Where non-physical measurements are made, various clinically acceptable scales or indices can be used, for example, the Crohn's Disease Activity Index, or CDAI (Best et al., 1976, Gastroenterology 70: 439), which combines both physical indicators, such as hematocrit and the number of liquid or very soft stools, among others, with patient-reported factors such as the severity of abdominal pain or cramping and general well-being, to assign a disease score.
As the term is used herein, "prophylaxis" performed using a composition as described herein is "effective" if the onset or severity of one or more symptoms is delayed or reduced by at least 10%, or abolished, relative to such symptoms in a similar individual (human or animal model) not treated with the composition.
Whereas the monovalent anti-CD40L antibody polypeptides described herein must bind human CD40L, where one is to evaluate its effect in an animal model system, the polypeptide must cross-react with one or more antigens in the animal model system, preferably at high affinity. One of skill in the art can readily determine if this condition is satisfied for a given animal model system and a given monovalent anti-CD40L antibody polypeptide. If this condition is satisfied, the efficacy of the monovalent anti-CD40L antibody polypeptide can be examined by administering it to an animal model under conditions which mimic a disease state and monitoring one or more indicators of that disease state for at least a 10% improvement.
Animal Models:
Monovalent anti-CD40L antibody polypeptides as described herein are useful for the treatment of autoimmune disorders in which CD40/CD40L signaling is inappropriately active. There are several animal models in which the therapeutic efficacy of a given monovalent anti-CD40L antibody polypeptide can be assessed, as discussed below.
Systemic Lupus Erythematosis (SLE):
Anti-CD40L antibody treatment prevents the development of lupus-like nephritis in NZB/NZW and SNFl SLE mice. Treatment of SNFl mice with anti- CD40L antibody reverses established nephritis and preserves kidney function. See, e.g., Mohan et al., 1995, J. Immunol. 154: 1470-1480; Early et al., 1996, J. Immunol.
157: 3159-3164; Railed et al., 1998, J..Immunol. 160: 2158-2165 ,and Chess, 2001, "Blockade of the CD40L/CD40 Pathway," in Therapeutic Immunology 2nd Edition, Austen, Burakof, Rosen and Strom, Eds., Blackwell Sciences (Pubs.), pp 441-456.
Multiple Sclerosis:
Specific blockade of CD40L at the time of immunization markedly suppresses the incidence, mortality, day of onset, and clinical scores of experimental autoimmune encephalomyelitis (EAE) in BlOPlL and (PLJ x SJL)Fl mice induced by either myelin basic protein or PLP myelin antigens. See, for example, Gerritse, 1996, Proc. Natl. Acad. Sci. U.S.A. 93: 2494; Grewal et al., 1996, Science 273: 186; Laman et al., 1998, Mult. Scler. 4: 14; and Chess, 2001, supra.
Rheumatoid Arthritis :
Anti-CD40L blocks the development of joint inflammation, serum antibody titers to collagen, the infiltration of inflammatory cells into the synovial tissue, ant the erosion of cartilage and bone in collagen-induced arthritis. See, e.g., Durie et al., 1993, Science 261: 132; and Chess, 2001, supra.
Insulin-dependent Type I Diabetes Models:
The non-obese diabetic (NOD) mouse spontaneously develops T cell dependent autoimmune diabetes. Anti-CD40L monoclonal antibody treatment of 3 to 4 week old NOD females (the age at which insulitis typically begins) completely prevented the insulitis and diabetes. Cytokine analysis revealed a dramatic decrease in IFN-g and IL-2 release without a concomitatnt increase in IL-4 production by T cells from anti-CD40L-treated mice. See, e.g., Balasa et al., 1997, J. Immunol. 159: 1420; and Chess, 2001, supra.
Inhibition of Allograft and Xenograft Transplant Rejection:
Anti-CD40L prevents the development of renal rejection of fully allogeneic grafts in mice. Moreover, the survival of renal allografts transplanted into nephrectomized rehsus monkeys is typically prolonged by anti-CD40L therapy alone.
Similarly, anti CD40L therapy has prevented graft rejection of skin, islet cells and cardiac transplants as well as GVHD in rodents. See, e.g., Kirk et al., 1997, Proc. Natl. Acad. Sci. U.S.A. 94: 8789-8794; Parker et al., 1995, Proc. Natl. Acad. Sci. U.S.A. 92: 9560; Larsen et al., 1996, Transplantation 61 : 4; and Chess, 2001, supra.
Uses of Monovalent Anti-CD40L Antibody Polypeptides
Anti-CD40L antibody polypeptides as described herein are useful for the treatment or prevention of diseases or disorders in which inappropriate activation of a CD40L/CD40-mediated pathway is involved. In particular, autoimmune diseases frequently involve inappropriate regulation or activity of CD40L/CD40 pathways. Administration of an anti-CD40L antibody polypeptide as described herein to an individual suffering from such a disease, can reduce one or more symptoms of the disease. Non-limiting examples of diseases for which the antibody polypeptides described herein can be therapeutically useful include Systemic Lupus Erythematosus (SLE), Idiotypic Thrombocytopenic Purpura (ITP), transplant rejection, Crohn's Disease, Inflammatory Bowel Disease (IBD), colitis, asthma/allergy, atherosclerosis, Myasthenia Gravis, immune response to recombinant drug products, e.g., factor VII in hemophilia, Multiple Sclerosis, Psoriasis, Rheumatoid Arthritis, Ankylosing Spondylitis, Coronary Heart Disease, and Diabetes, including Type 1 and/or Type 2 Diabetes.
The anti-CD40L antibody polypeptides described herein are additionally useful in the way that generally any antibody preparation is useful, e.g., for in vivo imaging or diagnostic uses, in vitro diagnostic uses, etc. For these and other uses it may be desirable to. label the anti-CD40L antibody polypeptides, e.gi5 with a fluorescent, colorimetric, enzymatic or radioactive label. Methods of labeling antibody polypeptides are well known in the art.
EXAMPLES
Example 1. Biotinylation of Recombinant CD40L Recombinant human soluble CD40L (PeproTech) was biotinylated and used during phage selections. Reagents, equipment and sources from which they are available are provided in Table 1.
Biotinylation of CD40L was performed by incubating CD40L (0.5mg/ml) with EZ-Link™ Sulfo-NHS-LC-Biotin [Sulfosuccinimidyl-6-(biotinamido)hexanoate] (Pierce) at a molar ratio of 5:1 on ice for 2 hours according to the product instructions. The biotinylation reaction mixture was then dialysed against 3 exchanges of PBS (100Ox sample volume) in a Slide-A-Lyzer® Dialysis Cassette at 40C to remove the unincorporated biotinylation reagent.
The biotinylated-CD40L was tested by receptor binding assay for binding to CD40/Fc to confirm its biological activity. Quality of biotin-CD40L was also monitored by analysing on a NuPaGE 4-12% Bis-Tris gel and detected by Simply Blue Safe-Stain (Invitrogen) (Figure Ia), and western-blotting by probing with Streptavidin-HRP (Figure Ib). The biotinylated-CD40L was further analysed by mass spectrometry with the majority of CD40L subunits containing 1 or 2 biotin moieties (data not shown).
Table 1.
Figure imgf000127_0001
Figure imgf000128_0001
Example 2. Phage Selections using Biotinylated Antigen
The Domain Antibody (dAb) libraries are based on a single human framework for the VH (DP47 and JH4b) and for the VK (DPK9 and JKl) with side chain diversity incorporated at positions in the antigen binding site that make contact with antigen in known molecular structures and mirror residues diversified in the human antibody repertoire. The antibodies are displayed as fusion proteins covalently linked to the N - terminus of the Fd -phage protein pill, using the phage vector pDOM4 (Fd- Tet) with encodes the Fd phage genome with dAb expression under the control of the gene-HI promoter. The dAb cassette consists of (5' to 3'): eukaryotic leader sequence, dAb, myc tag, gill. The vector contains both the Ml 3 and colEl origins of replication and is selectable using tetracycline. The VH and Vκ libraries each have a calculated size of over 1x1010 molecules. Reagents, equipment and sources from which they are available are provided in Table 2.
Approximately 1x1011 phage from the each of the Domantis dAb libraries were incubated in a final volume of 1 ml PBS containing 2% Marvel™ at room temperature for 1 h. Biotinylated antigen was added to the blocked phage such that the phage antigen mixture had a final concentration of 2% Marvel™ in PBS. The antigen concentration used for the first round of selection was 60 nM; the antigen concentration was decreased to 6 nM for round 2, and to 0.6 nM for round 3. The antigen/phage mix was incubated for 1 h at room temperature with rotation at ~40 rpm.
For each selection, 100 μl of streptavi din-coated paramagnetic beads (Dynal
Biotech) were prepared by washing once in 1 ml of PBS containing 0.1% Tween-20 followed by a second wash in 1 ml of PBS. The beads were then blocked in 1 ml of PBS containing 2% Marvel™ in a 2 ml eppendorf tube at room temperature on a rotating wheel for 1 h.
The tube containing the blocked streptavi din -coated magnetic beads was placed into a magnetic holder, enabling capture of the magnetic beads. The supernatant was removed and the beads resuspended in the antigen/phage mix. This mixture was rotated for 10 min to allow for bead capture of phage/antigen complexes.
The beads were captured using a magnetic holder and repeatedly washed 19 times using 1 ml of PBS containing 0.1% Tween-20, followed by a final wash of 1 ml PBS. The eppendorf tubes were changed following washing steps 3, 9, 15 and 19 to minimise background phage carryover.
The washed beads were then recaptured and all washing solution removed. The phage were eluted through resuspension in 500 μl of trypsin solution (50 μl of 10 mg/ml trypsin stock solution added to 450 μl PBS, freshly diluted) and rotated for 10 min at room temperature. The eluted phage were recovered by capturing the beads using the magnetic holder and the liquid containing the eluted phage recovered. The eluted phage were used to infect E. coli TGl to prepare phage for a further round of selection.
The eluted phage (250 μl) were mixed with 1.75 ml of log phase E. coli TGl (OD6Oo between 0.3 and 0.6) and infection allowed to occur for 30 min at 37°C without shaking. The infected E. coli TGl culture was centrifuged at 11,600 g in a micro centrifuge for 1 min at room temperature. The pelleted bacteria were resuspended in 100 μl of 2xTY and plated on regular 9 cm diameter plates containing TYE supplemented with 15 μg/ml tetracycline. Plates were grown at 37°C overnight.
After overnight growth, 2 ml of 2xTY containing 15% glycerol was added to the culture plates and cells loosened with a spreader, ensuring the cells were thoroughly mixed. Two millilitres of the culture were recovered by pipetting into a cryo-vial, from which 50 μl was used to inoculate 50 ml of 2xTY supplemented with 15 μg/ml tetracycline. The remaining cells in the cryo-vial were stored at -8O0C.
The 50 ml culture was grown at 37°C for 16 to 24 hours with shaking at 250 rpm.
Following overnight growth, the culture was centrifuged at 3,300 g for 15 min to pellet the bacteria. The phage were then precipitated from the supernatant through the addition of 10 ml of PEG/NaCl to 40 ml of clarified supernatant. The phage/PEG solution was mixed and incubated on ice for at least 1 h. To pellet the phage, the solution was centrifuged at 3,300 g for 30 min at 40C. The supernatant was decanted and any remaining supernatant removed by aspiration.
The resulting phage pellet was resuspended in 2 ml PBS and centrifuged at
11,600 g for 10 min in a micro centrifuge to remove any remaining bacterial debris. The supernatant was filtered through a 0.45 μm filter (Sartorius, Minisart). The resuspended phage solution was used for the next round of selection.
Table 2
Figure imgf000132_0001
Figure imgf000133_0001
Example 3: Cloning Enriched Phage Selection Outputs into the Soluble dAb Expression Vector pDOMS
Following the second and third rounds of selection, E. coli cells infected with the enriched dAb displaying fd-phage populations were obtained. An aliquot of these cells was used to prepare phage DNA and the enriched V-genes excised by digestion using the restriction endonucleases, Sail and Notl. The purified V-genes were ligated into the corresponding sites of pD0M5 (expression vector derived from pUC119 with LacZ promoter, eukaryotic leader, dAb cloning site, myc tag), and the ligated DNA used to electro-transform E. coli HB2151 cells which were grown overnight on agar plates containing the antibiotic carbenicillin. The resulting colonies were induced to express dAb protein either as 200 μl microcultures or 50 ml cultures. The resulting dAb was analysed for inhibitory activity using the CD40L receptor binding assay.
Following selection of phage, pDOM4 DNA was purified from the cell pellet obtained from a 50 ml overnight E. coli culture using the QIAfilter Plasmid Midi DNA purification kit from Qiagen, following the manufacturer's instructions. The dAb genes were excised from the pDOM4 vector by mixing: 10 μl of 10x SaR buffer; 1 μl of 10Ox BSA; 20 μg of purified DNA fragment; 2.5 μl of SaR enzyme (10 U/μl); 2.5 μl of Notl enzyme (10 U/μl); the digestion mix was made up to a final volume of 100 μl using sterile water. The digestion mix was incubated for 5 hours at 37°C.
The digested DNA samples were electrophoresed on a 1.5% agarose gel and the band corresponding to the dAb V-genes (-324 bp to 372 bp) was excised from the gel. The dAb gene DNA was purified from the gel slice using the QIAquick Gel Extraction kit from Qiagen, following the manufacturer's instructions.
The expression vector pDOM5 was digested with Sail and Notl as follows: 10 μl of 1Ox SaR buffer; 1 μl of 10Ox BSA; 20 μg of plasmid pD0M5; 1.5 μl of SaR enzyme (10 U/μl); 1.5 μl of Notl enzyme (10 U/μl); the digestion mix was made up to a final volume of 100 μl using sterile water. The digestion mix was incubated for 2 hours at 37°C. The digested vector fragment was purified using the QIAquick PCR Purification Kit. The digested pDOM5 and digested dAb genes were ligated by mixing: 2 μl of 1 Ox T4 DNA ligase buffer; 400 ng of digested pD0M5 vector; 100 ng of digested dAb genes;
1 μl of T4 DNA ligase (400 U/μl); the ligation mix was made up to 20 μl with sterile water. The ligation mixes were incubated for 2 hours at 250C.
Two microlitres of the ligation mix was transferred to the bottom of a pre- chilled (on ice) 0.2 cm electroporation cuvette to which 100 μl of electrocompetent E. coli HB2151 cells were added. The DNA / cell mixture was incubated on ice for 1-2 min, then electroporated at 2.5 kV (25 μF, 200 Ω). One millilitre of 2xTY was immediately added to the cuvette and the cells gently resuspended. The resuspended cells were transferred to a 14 ml disposable culture tube and incubated for 1 hour at 37°C with shaking at 250 rpm. Dilutions of the cells from 10'° to IO'3 were plated on regular 9 cm diameter plates containing TYE supplemented with 5% glucose and 50 μg/ml carbenicillin. The cells are incubated overnight at 37°C in an inverted position. Reagents, equipment and sources from which they are available are provided in Table 3.
Table
Figure imgf000136_0001
Example 4. Microwell Expression of Soluble dAbs
Following cloning of the selected phage dAb outputs into pDOM5, individual bacterial colonies were inoculated as microwell cultures and induced using IPTG to express dAb protein which was analysed for inhibitory activity using the CD40L receptor binding assay. Reagents, equipment and sources from which they are available are provided in Table 4.
Individual bacterial colonies were carefully picked to ensure that contamination from neighbouring colonies was avoided. The picked colonies were used to inoculate 96 well cell culture plates containing 100 μl per well of 2xTY supplemented with 5% glucose and 50 μg/ml carbenicillin. The lids were placed on the cell culture plates which were incubated overnight in a HiGro orbital shaker (GeneMachines, 935 Washington St, San Carlos, CA 94070, USA) under a humidified atmosphere at 370C with shaking at 450 rpm (4 mm shaking orbital diameter), with gas (30% O2 + 70% N2) pulsed for 10 seconds every 5 minutes at a flow rate of 5 SLPM (standard litres per minute). [These plates are referred to as Master Plates].
Following overnight growth, a 96 well transfer device was used to transfer between 1-5 μl of the bacterial culture into a fresh 96 well culture plate containing 100 μl per well of 2xTY supplemented with 0.1% glucose and 50 μg/ml carbenicillin.
The freshly inoculated plates were incubated at 370C for 3 to 4 h (shaking at
450 rpm, gas (30% O2 + 70% N2) pulsed for 10 seconds every 5 minutes at a flow rate of 5 SLPM) until the culture OD6oo reached approximately 1.0. The cultures were then induced by the addition of 100 μl per well of 2xTY containing 50 μg/ml carbenicillin and 2 mM IPTG (final IPTG concentration of 1 mM) and incubated overnight at 3O0C with shaking at 450 rpm, with gas (30% O2 + 70% N2) pulsed for 10 seconds every 5 minutes at a flow rate of 5 SLPM. [These plates are referred to a Induction Plates]. Glycerol stocks of the original Master Plates were made by the addition of 100 μl per well of 2xTY containing 50% sterile glycerol. These plates were stored at -8O0C.
Following overnight incubation of the Induction Plates, the bacterial cells were pelleted by centrifugation at 1,800 g for 10 min at 40C. The supernatant (containing expressed dAb) was then analysed to determine if dAbs were capable of inhibiting binding of CD40L to CD40-Fc fusion in a receptor binding assay.
Table 4
Figure imgf000138_0001
Example 5. Expression of dAb in E. coli at 50 ml
To generate greater quantities of dAb protein for analysis, 50 ml cultures were used for induction. A single colony of the desired dAb (for example DOM-24) grown on TYE plates was inoculated into 10 ml 2xTY supplemented with 5% glucose and 50 μg/ml carbenicillin in a 30 ml universal tube and grown overnight at 37°C with shaking at 250 rpm. Five hundred microlitres of the overnight culture was added into 50 ml of 2xTY supplemented with 0.1% glucose and 50 μg/ml carbenicillin and grown, at 370C with shaking at 250 rpm. The OD6oo of the culture was monitored regularly in comparison with sterile 2xTY and at an OD6oo of 0.9 the culture was induced by the addition of 1 M IPTG to a final concentration of 1 mM. The inoculated culture was incubated at 3O0C with shaking at 250 rpm overnight. The next day, the culture was centrifuged at 6000 g for 15 min at 4°C and the clarified supernatant mixed with 100 μl of protein-A streamline or protein-L agarose (pre- washed with 5 mM MgSO4) overnight at 40C. The supernatant/bead mixture was then centrifuged at 180 g at 40C for 2 minutes. The supernatant was decanted and the retained beads washed with 10 ml of PBS containing 0.5M NaCl. The bead solution was transferred into a 96 well Whatman filter plate and the beads washed once with 400 μl of PBS containing 0.5M NaCl5 then once with 400 μl of PBS, followed by centrifugation for 2 minutes at 180 g after each washing step. dAb protein was eluted using 70 μl of 0.1 M glycine (pH 2.0) and the solution neutralised by the addition of 40 μl of 1 M Tris-HCl (pH 8.0). The purified dAb concentration was determinate by OD280.
Reagents, equipment and sources from which they are available are provided in Table 5
Table 5
Equipment/ Reagent Suggested or Instrument setting, required supplier reagent preparation
Figure imgf000140_0001
Figure imgf000141_0001
Example 6: CD40L Receptor Binding Assay
The CD40L assay was used to measure the binding of CD40L to CD40 and the ability of binding entities (eg, monvalent antibody fragments such a dAbs) to block this interaction, as described below and shown schematically in Figure 7. (The soluble proteins from R&D Systems are CD40/Fc = homodimer and CD40L = homotrimer).
A 96 well Nunc Maxisorp assay plate was coated overnight at 40C with 100 μl per well of recombinant human CD40/Fc (R&D Systems) at 0.5 μg/ml in carbonate buffer. The plate was washed 3 times with 300 μl of 0.05% Tween/PBS and 3 times with 300 μl of PBS using a Tecan plate washer. The wells were blocked using 200 μl of PBS containing 2% (w/v) BSA and incubated for a minimum of 1 h at room temperature. The wells were washed as above, then 50 μl of purified dAb protein (or unpurified supernatant containing dAb from a micro-culture expression) was added to each well. To each well 50 μl of CD40L, at 6 ng/ml in diluent (for a final concentration of 3 ng/ml), was also added and the plate incubated for 1 hr at room temperature.
The plate was washed as described previously and 100 μl biotinylated anti- CD40L antibody, 0.5 μg/ml in diluent, was added and incubated for 1 hr at room temperature. The plate was washed as described above, then 100 μl HRP conjugated anti-biotin antibody (1:5000 dilution in diluent) added to each well and the plate incubated for 1 hr at room temperature. The plate was washed again as described above using a Tecan plate washer and the assay developed using 100 μl of SureBlue 1-Component TMB MicroWell Peroxidase solution (the plate was left at room temperature for up to 20 min). The reaction was stopped by the addition of 100 μl 1 M hydrochloric acid. The OD450nm of the plate was assayed within 30 minutes of acid addition. The OD45onm is proportional to the amount of bound streptavidin-HRP conjugate, therefore the greater the degree of dAb inhibition the lower the OD450nm of the resulting signal. Reagents, equipment and sources from which they are available are provided in Table 6. Controls
The following controls were included:
0 ng/ml CD40L (diluent only)
3 ng/ml CD40L
ng/ml CD40L with 1 μg/ml anti-CD40L antibody
Table 6
Figure imgf000143_0001
Figure imgf000144_0001
Example 7: Results
Receptor binding data for the most potent inhibitors is summarised in Figures 2, 3, and 4, and in Table 7, below. Table 8, below, provides DNA and translated amino acid sequence of unique dAbs identified in the receptor binding assay as inhibiting CD40L binding to CD40.
Figure 2 shows a dose response receptor binding assay(RBA) readout, analysing the inhibition of CD40L binding to CD40-Fc by dAbs DOM-10, -20, -27, - 30, -31, -62, -77, titrated from 1 μM down to lO pM. dAbs DOM-20, -30, and -31 are the most potent, with IC50 values of approximately 8 nM.
Figure 3 shows a dose response receptor binding assay readout, analysing the inhibition of CD40L binding to CD40-Fc by dAbs DOM-4 and DOM-5, titrated from 1 μM down to 500 pM. The IC50 values for dAbs DOM-5 and DOM-4 are approximately 3 nM and 100 nM respectively.
Figure 4 shows a dose response receptor binding assay readout, analysing the inhibition of CD40L binding to CD40-Fc by dAb DOM-24, titrated from 100 nM down to 0.5 pM. The data were curve-fitted using GraphPad Prism software.
Table 7
Figure imgf000145_0001
Figure imgf000146_0001
Table 8: Summary of dAbs exhibiting a ranee of CD40L inhibitory ICso values as determined using the CD40L / CD40-Fc receptor inhibition assay.
The DNA and translated amino acid sequence of unique dAbs identified in the receptor binding assay as inhibiting CD40L binding to CD40 are detailed below:
DOM-2 SEQ ID NO: 7
GRSGTGCAGC
• M B V E Q A P G K Y Y A D K G R
DOM-4 SEQID NO: 8
TTAATACTCT
ACTGCACCcA
- F T I S R B N S K H T L Y L Q M N S I1 R A E D T A V Y Y C A.
201 GTTCACCATC
GAAACCTAAT
DOM-5 SEQID NO: 9
E V Q L I E S G G G L V Q P G G S L R L S C A A S G F T F D G Y E M 1 GAGGTGCAGC TGTIGGAGTC TGGGGGAGGC TTGGTACAGC CTGGGGGGTC CCTGCGTCTC TCCTGTGCAG CCTCCGGATT CACCTTTGAT GGGTATGAGA
CTCCACGTCG ACAACCTCAG CCCATACTCT
I T E D G T S T Y Y A D S V K G R - ioi TGGCGTGGGT
DOM-7 SEQ ID NO: 10
JXJ AACATACTCT
JU K A G
35
40
DOM-8 SEQID NO: 11
DOM-10 SEQID NO: 12
CAAGTGGTAG AGGGCGCTGT TAAGGTTCTT GTGCGACATA GACGTTTACT TGTCGGACGC ACGGCTCCTE TGGCGCCATA TAATGACACG CTTTTTCTGC
CCTGAGGAGT C (SEQ ID NO: 104) GGACTCCTCA G (SEQ ID NO: 105)
DOM-Il SEQ ID NO: 13 E V Q l L E S G G G L V Q P G G S L R L S C A A S G F T P G D
Y E M '
1 GAGGTGCAGC
GATTATGAGA CTCCACGTCG ACAACCTCAG ACCCCCTCCG AACCATGTCG GACCCCCCAG GGACGCAGAG AGGACACGTC GGAGGCCTAA GΓGGAAACCA CTAATACTCT
S W V R Q A P G K G L E W V S G I D G E G S D T Y Y A. D S V K G R - 101 TGAGTTGGGT TGAAGGGCCG
ACTCAACCCA ACTTCCCGGC
' F T I S R D N S K H T L Y L Q M M S L R A E D T A V Y Y C A K P G
ZOl GTTCACCATC ATTCCAAGM.
GAAACCGGGG
CAAGTGGTAG AGGGCGCTGT TAAGGTTCTT GTGCGACATA GACGTTTACT TGTCGGACGC ACGGCTCCTA TGGCGCCATA TAATGACACG CTTTGGCCCC
R S F D Y W G Q G T L V T V S S
301 AGGAGTTTTG TCTCGAGC (SEQ ID NO: 106)
TCCTCAAAAC AGTCCCTTGG AGAGCTCG (SEQ ID NO: 107)
DOM-12 SEQ ID NO: 14
E V Q L L E S G G G L V Q P G G S L R L S C A A S G F T F R L Y E M ' 1 GAGGTGCAGC TGTTGGAGTC TGGGGGAGGC TTGGTACAGC CTGGGGGGTC CCTGCGTCTC TCCTGTGCAG CCTCCGGATT CACCTTTAGG TTGTATGAGA
CTCCACGTCG ACAACCTCAG ACCCCCTCCG AACCATGTCG GACCCCCCAG SGACGCAGAG AGGACACGTC GGAGGCCTAA GTGGAAATCC AACATACTCT
K D L
DOM-13 SEQID NO: 15
K G R -
GAGACGAGTG ETCCGATTTC TGAGAATTTT GACTACTGGG GTCAGGGAAC CCTGGTCACC GTCTCGSGC (SEQ ID NO: 110) CTCTGCTCAC CAGAGCTCG (SEQ ID NO: 111)
DOM-14 SEQ ID NO: 16
E V O l L E S G G G L V Q P G G S I R l S C A A S G F T F K S Y D M -
1 GAGGTGCAGC
TCTTATGATA CTCCACGTCG
AGAATACTAT
T W V R Q A P G K G L E W V S S I M A S G D D T Y Y A D S V K G R - 101 TGACGTGGGT CCAGGGAAGG
TGAAGGGCCG
ACTGCACCCA CAGATCTCAC ACTTCCCGGC
- F T l S R D N S K N T L Y L Q M N S L R A E D T A V Y Y C A
K W D
201 GTTCACCATC
GAAATGGGAT CAAGTGGTAG CTTTACCCTA
R D F D Y W G Q G T L V T V S S
CGGGATTTTG TCTCGAGC (SEQ ID NO: 112) GCCCTAAAAC AGTCCCTTSG AGAGCTCG (SEQ ID NO: 113)
DOM-15 SEQID NO: 17
E V Q L L E S G G ' G L V Q F G G S L R L S C A A S G F T F E E Y V M - 1 GAGGTGCAGC TGTTGGAGTC TGGGGGAGGC TTGGTACAGC CTGGGGGGTC CCTGCGTCTC TCCTGTGCAG CCTCCGGATT CACCTTTGAG
GAGTATGITA
CTCCACGTCG ACAACCTCAG
CTCATACAAT
- S W V R Q A P G K G L E W V S T I S P I G L T T Y Y A D S V
K G R -
101 TGTCGTGGGT
TGAAGGGCCG ACAGCACCCA GGCGGTCCGA GGTCCCTTCC CAGACCTCAC CCAGAGTTGA TAAAGAGGAT AACCAGACTG ATGTATGATG CGTCTGAGGC ACTTCCCGGC
- F T I S R D H S K K T L Y L Q M H S L R A E D T A V Y Y C A E F P
201 GTTCACCATC
GGAATTTCCT
CAAGTGGTAG CCTTAAAGGA
L I I L P D F D Y K G Q G T L V T V S S
301 TTGATTATTC (SEQ ID NO: 114)
AACTAATAAG (SEQ ID NO: 115)
DOM-16 SEQID NO; 18
E V Q L L E S G G G L V Q P G G S L R L S C A A. S G F T F M E Y A M 1 GAGGTGCAGC . GAGTATGCGA
CTCCACGTCG ACA1CCTCAG CTCATACGCT
- I M V R Q A P G K G L E K V S I I S P L G L S T Y Y A D E V K G R -
101 TGATTTGGGT
TGAAGGGCCG
ACTAAACCCA GGCGGTCCGA GGTCCCTTCC CAGATCTCAC CCAGAGTTAA TAAAGAGGCG AACCAAACAG ATGTATGATG CGTCTGAGGC ACTTCCCGGC
- F T I S R D N S K H T L Y L Q M H S L R A E D T A V Y Y C A K Y Q
201 GTTCACCATC TCCCGCGACA ATTCCAAGAf. CACGCTGTAT CTGCAAATGA ACAGCCTGCG TGCCGAGGAC ACCGCGGTAT ATTACTGTGC
GAAATATCAG CAAGTGGTAG AGGGCGCTGT TAAGGTTCTT GTGCGACATA GACGTTTACT TGTCGGACGC ACGGCTCCTG TGGCGCCATA TAATGACACG
CTTTATAGTC
D S S D S Q Y T H F D Y M G Q G T L V T V S S 301 GATTCGTCTG GTCTCGAGC (SEQ ID NO: 116)
CTAAGCAGAC CAGAGCTCG (SEQ ID NO: 117) DOM-17 SEQ ID NO: 19
DOM-18 SEQID NO: 20
DOM-19 SEQID NO: 21
P L F D Y B G Q G T L V T V S S
301 CCTCTTTTTG TCTCGAGC .(SEQ ID NO: 122) GGAGAAAAAC AGAGCTCG (SEQ ID HO: 123)
DOM-20 SEQID NO: 22
E V Q L E S G G G L V Q P G G S L R L Y Q M -
TGAAGGGCCG
R R F D Y W G Q G T L V T V S S
301 CGTAGGTTTG TCTCGAGC (SEQ ID NO: 124)
GCATCCAAAC GACCAGTGGC AGAGCTCG (SEQ ID NO: 125)
DOM-21 SEQID NO: 23
E V Q L L E S G G G L V Q P G G S L R L S C A A E G F T F A K Y E M -
1 GAGGTGCAGC TGTTGGAGTC TGGGGGAGGC TTGGTACAGC CTGGGGGGTC CCTGCGTCTC TCCTGTGCAG CCTCCGGATT CACCTTTGCG
AATTATGAGA CTCCACGTCG ACAACCTCAG
TTAATACTCT
G W A R Q A P G K G L E W V S V I S E W G Y S T Y Y A D S A K G R - 101 TGGGGTGGGC CCGCCAGGCT CCAGGGAAGG GTCTAGAGTG GGTCTCAGTT ATTTCTGAGT GGGGTTATTC TACATACTAC GCAGACTCCG
CGAAGGGCCG
ACCCCACCCG
GCTTCCcGGC
- F T I S R D K S K N T L Y L Q M N S L R A E D T A V Y Y C A.
K L V
201 GTTCACCATC TCCCGCGACA ATTCCAAGAA CACGCTGTAT CTGCAAATGA ACAGCCTGCG TGCCGAGGAC ACCGCGGTAT ATTACTGTGC
GAAACTTGTG CAAGTGGTAG AGGGCGCTGT TAAGGTTCTT GTGCGACATA GACGTTTACT TGTCGGACGC ACGGCTCCTG TGGCGCCATA TAATGACACG CTTTGAACAC
DOM-22 SEQ ID NO: 24
DOM-23 SEQID NO: 25
DOM-24 SEQID NO: 26
DOM-25 SEQID NO: 27 Y E M -
DOM-26 SEQID NO: 28
CΓTTGGACTA
DOM-27 SEQ ID NO : 29
201 GTTCACCATC TCCCGCGACA ATTCCAAGAA CACGCTGTAT CTGCAAATGA ACAGCCTGCG TGCCGAGGAC ACCGCGGTAT ATTACTGTGC GAAACCGGAT CAAGTGGTAG AGGGCGCTGT TAAGGTTCTT GTGCGACATA GACGTTTACT TGTCGGACGC ACGGCTCCTG TGGCGCCATA TAATGACACG CTTTGGCCTA
DOM-28 SEQID NO: 30
GAAACCGGGG
DOM-29 SEQID NO: 31
DOM-30 SEQID NO: 32
DOM-31 SEQID NO: 33 Y Q M -
DOM-32 SEQID NO: 34
E V Q L L V Q P G G S L R L S C A A G F T F E Y Q M -
DOM-33 SEQID NO: 35
CAAGCCGGTT G T K V E I K R
GGGACCAAGG ACGG (SEQ ID NO: 150) CCCTGGTTCC TGCC ISEQ ID HO; 151)
DOM-34 SEQID NO: 36
Z D
30 301 GGGACCAAGG TGGGAATCAA ACGG (SEQ ID NO: 152) CCCTGGTTCC ACCCTTAGTT TGCC (SEQ ID NO: 153)
DOM-35 SEQ ID NO : 37
GAACCATGGT CGTCTTTGGT CCCTTTCGGG GATTCGAGGA CTAGATAAAA AGAAGGTAAP. ACGTTTCACC CCAGGGTAGT GCAAAGTCAC CGTCACCTAG
201 TGGGACAGAT TTCACTCTCA CCATCAGCAG TCTGCAACCT GAAGATTTTG CTACGTACTA CTGTCAACAG TATATGGATA TTCCTATTAC
DOM-36 SEQ ID NO : 38 H L E -
1 GACATCCAGA ΓGACCCAGTC TCCATCCTCC CTGTCTGCAT CTGTAGGAGA CCGTGTCACC ATCACTTGCC GGGCAAGTCA GGATATTGAT
0
DOM-37 SEQID NO: 39
U S G S -
5
DOM-38 SEQID NO: 40 U N L E -
DOM-39 SEQID NO: 41
DOM-40 SEQID NO: 42
DOM-41 SEQ ID NO: 43
E V Q L L E S G G G L V Q P G G S L R L S C A A S G F T F F E Y E M -
GAGGTGCAGC GAGTATGAGA
CTCCACGTCG CTCATACTCT
- T W V R Q A P G K G I E IJ V S S I A H D G S T T Y Y A D S V
K G R '
101 TGACGTGGGT
TGAAGGGCCG ACTGCACCCA ACTTCCCGGC
• F T I S R D H S K H T L Y L Q M K S L R A E D T A V Y Y C A K P D
201 GTTCACCATC GAAACCTGAT
CAAGTGGTAG CTTTGGACTA
R Q F D Y W G Q G T L V T V S S 301 CGGCAGTTTG ACTACTGGGG TCTCGASC (SEQ ID NO: 166)
GCCGTCAAAC AGAGCTCG (SEQ ID NO: 157)
DOM-42 SEQID NO: 44 E V Q L L E S G G G L V Q P G G S L R L S C A A S G F T F G P
Y E M -
1 GAGGTGCAGC TGTTGGAGTC TGGGGGAGGC TTGGTACAGC CTGGGGGGTC CCTGCGTCTC TCCTGTGCAG CCTCCGGATT CACCTTTGGT
CCGTATGAGA CTCCACGTCG ACAACCTCAG GGCATACTCT
• T W V R Q A P G K G L E W V S S I V G D G L D T Y Y A D S V K G R - 101 TGACTTGGGT GCAGACTCC5 TGAAGGGCCG
ACTGAACCCA ACTTCCCGGC
DOM-43 SEQID NO: 45 E V O L L E S G G G L V Q P G G S L R L S C A A S G F T F A S Y E M -
1 GAGGTGCAGc
ACCGCACCCA
ACTTCCCGGC
F T I E R D N S K N T L Y L Q M N S L R A E D S A V Y Y C A K P D 201 GTTCACCATC TCCCGCGACA ATTCCAAGAA CACGCTGTAT CTGCAAATGA ACAGCCTGCG TGCCGAGGAC TCCGCGGTAT ATTACTGTGC
GAAACCTGAT
CAAGTGGTAG AGGGCGCTGT TAAGGTTCTT GTGCGACATA GACGTTTACT TGTCGGACGC ACGGCTCCTG AGGCGCCATA TAATGACACG CTTTGGACTA
R A F D Y W G Q G T L V T V S S
301 AGGGCTTTTG CTGGTCACCG TCTCGAGC ISEQ ID NO: 170)
TCCCGAAAAC TGATGACCCC AGAGCTCG (SEQ ID NO: 171)
DOM-44 SEQ ID NO: 46 E V Q L L E S G G G L V Q P G G S L R L S C A A S G F T F T S Y E M -
GAGGTGCAGC TCTTATGAGA
CTCCACGTCG
AGAATACTCT
G W V R Q A P G K G L E W V S S I E P T G I T T Y Y A D E V K G R - 101 TGGGGTGGGT
TGAAGGSCCG
ACCCCACCCA ACTTCCCGGC
- F T I S E D N S K N T L Y L Q M N S L R A E D T A V Y Y C A
K P H
201 GTTCACCATC
GAAACCTCAT CAAGTGGTAG TGTCGGACGC ACGGCTCCTG CTTTGGAGTA
F T E L G F D Y B G Q G T L V T V S S
301 TTTACTGAGC TGGTCACCGT CTCGAGC (SEQ ID NO: 172)
AAATGACTCG AACCAAAACT GAGCTCG (SEQ ID NO: 173)
DOM-45 SEQ ID NO: 47
E V Q L L E S G G G L V Q P G G S L E L S C A A S G F T F G N Y A M - 1 GAGGTGCAGC TGTTGGAGTC TGGGGGAGGC TTGGTACAGC CTGGGGGGTC CCTGCGTCTC TCCTGTGCAG CCTCCGG ATT CACCTTTGGT
AATTATGCGA
CTCCACGTCG ACMCCTCAG ACCCCCTCCG AACCATGTCG GACCCCCCAG GGACGCAGAG AGGACACGTC GGAGGCCTAA GTGGAAACCA TTAATACGCT
- A W V R Q A P G K G L E W V S K I G A Q G L H T Y Y A G S V
K G R -
101 TGGCGTGGGT
TGAAGGGCCG ACCGCACCCA ACTTCCCGGC
- F T I S B B N S K N T l Y L Q M N S L R A E D T A V Y Y C A K Q T 201 GTTCACCATC TCCCGCGACA ATTCCAAGAA CACGCTGTAT CTGCAAATGA ACAGCCTGCG TGCCGAGGAC ACCGCGGTAT ATTACTGTGC GAAACAGACG
DOM-46 SEQID NO: 48
GAAAGAGGCT
DOM-47 SEQID NO: 49
CCCTGGTTCC TGCC (SEQ ID NO: 179)
DOM-48 SEQID NO: 50 S L S -
GATTCGTTAA
G T K V E I K R
301 GGGACCAAGG TGGAAATCAA ACGG (SEQ ID NO: 180) CCCTGGTTCC ACCTTTAGTT TGCC (SEQ ID NO: 181)
DOM-49 SEQID NO: 51 D V Q K T C S P E S L S A S V G D P V T I T C K A S Q M I G D S L S '
GACGTCCAGA GATTCGTTAA
CTGCAGGTCT CTAAGCAATT
W Y Q Q K P G K A P K L L I Y F G E Y L Q S G V P S R F S G S G S - ioi GTTGGTACCA GCAGAAACCA GGGAAAGCCC CTAAGCTCCT GATCTATTTT GGTTCCTATT TGCAAAGTGG GGTCCCATCA CGTTTCAGTG
GCAGTGGATC
CAACCATGGT CGTCACCTAG
- G T B F T L T I S S L Q P E D F A T Y Y C Q Q Y M S A P S T
F G Q
201 TGGGACAGAT
GTTCGGCCAA ACCCTGTCTAAAGTGAGAGT CAAGCCGGTT
G T K V E I K R
301 GGGACCAAGG ACGG (SEQ ID NO: 182)
CCCTGGTTCC ACCTTTAGTT TGCC (SEQ ID NO: 183)
DOM-50 SEQID NO; 52
D I Q M T Q S P S S L S A S V G D R V T I T C R A S Q W I G D S L E 1 GACATCCAGA
GATTCGTTAA
CTGTAGGTCT CTAAGCAATT
- W Y Q Q K P G K A P K L L I Y F G S Y L Q S G V P S R F S G
S G S -
101 GTTGGTACCA GCAGAAACCA GGGAAAGCCC CTAAGCTCCT GATCTATTTT GGTTCCTATT TGCAAAGTGG GGTCCCATCA CGTTTCAGTG
GCAGTGGATC CAACCATGGT CGTCTTTGGT CCCTTTCGGG GATTCGAGGA CTAGATAAAA CCAAGGATAA ACGTTTCACC CCAGGGTAGT GCAAAGTCAC CGTCACCTAG
- G T D F T L T I S S L Q P E D S A T Y Y C O Q Y Q Y V P S T F G Q 201 TGGGACAGAT TTCACTCTCA CCATCAGCAG TCTGCAACCT GAAGATTCTG CTACGTACTA CTGTCAACAG TATCAGTATG TTCCTTCTAC GTTCGGCCAA ACCCTGΓCTA AAGΓGAGAGT GGTAGTCGTC AGACGTTGGA CTTCTAAGAC GATGCATGAT GACAGTTGTC ATAGTCATAC AAGGAAGATG CAAGCCGGTT
G T K V E I K Q
GGGACCAAGG ACAG (SEQ ID NO. IB4) CCCTGGTTCC TGTC (SEQ ID NO: 185)
DOM-51 SEQID NO; 53
DOM-52 SEQID NO: 54
DOM-53 SEQID NO: 55 G L R -
G T K V E I K R
301 GGGACCAAGG TGGAAATCAA ACGG (SEQ ID MO: 190) CCCTGGTTCC ACCTTTAGTT TGCC (SEQ ID NO: ISl)
DOM-54 SEQID NO: 56 CGCATATCCT
K T l
201 GTTCACCATC
GAAAACTTTG CAAGTGGTAG CTTTTGAAAC
DOM-55 SEQID NO: 57
E V Q L L E S G G G L V Q P G G S L R L S C A A S G F T P A R Y E M - 1 GAGGTGCAGC TGTTGGAGTC TGGGGGAGGC TTGGTACAGC CTGGGGGGTC CCTGCGTCTC TCCTGTGCAG CCTCCGGATT CACCTTTGCG
CGGTATGAGA
CTCCACGTCG ACAACCTCAG GCCATACTCT
- G M V R Q A P G K G L E K V S R I T A Q G L G T Y Y A D S V
K G R -
101 TGGGGTGGGT
TGAAGGGCCG ACCCCACCCA CSTCTGAGGC ACTTCCCGGC
DOM-56 SEQ ID NO: 58
E V Q L L E S G G G L V O P G G B L B L S C A A S G F T F W D Y T M '
1 GAGGTGCAGC
GATTATACTA
CTCCACGTCG CTAATATGAT
- G W V R Q A P G K G L E W V S W I H G T G G Q T Y Y A D S V
K G R -
101 TGGGTTGGGT
TGAAGGGCCG ACCCAACCCA ACTTCCCGGC
- P T I S R D N S K H T L Y L Q M H S L R A E D T A V Y Y C A K A L 201 GTTCACCATC ATTACTGTGC GAAAGCTTTG
CAAGTGGTAG TGTCGGACGC
CTTTCGAAAC
A D R S G G V V E F D Y W G Q G T L V T V S S 301 GCTGATASGA GTCTCGAGC (SEQ ID HO: 196)
CGACTATCCT CAGAGCTCG (SEQ ID KO: 197)
DOM-57 SEQID NO: 59 E V Q L L E S G G G L V Q P G G S L R L S C A A S G F T F S E
Y D M -
1 GAGGTGCAGC TGTTGGAGTC TGGGGGAGSC TTGGTACAGC CTGGGGGGTC CCTGCGTCTC TCCTGTGCAG CCTCCGGATT CACCTTTTCT
GAGTATGATA CTCCACGTCG ACAACCTCAG CTCATACTAT
• Y M V R Q A P G K G L E W V S W I D T D G G D T Y Y A D S V K G R - 101 TGTATTGGGT TGAAGGGCCG
ACATAACCCA ACTTCCCGGC • F T I S E D N S K K T L Y L O M W S L R A E D T A V Y Y C A K P G
GTTCACCATC GAAACCTGGT
CAAGTGGTAG CTTTGGACCA
L K F D Y H G Q G T L V T V S S
301 CTGAAGTTTG TCTCGAGC (SEQ ID NO: 19B)
10 GACTTCAAAC AGAGCTCG [SEQ ID NO: 199)
DOM-58 SEQID NO: 60
1 « E V Q L L E S G G G L V Q P G G S L R L S C A A S G F T F E V
IJ Y T M -
1 GAGGTGCAGC
GTTTATACTA
CTCCACGTCG CAAATATGAT 0
• A W V R Q A P G K G L E W V S T I D E S G R D T Y Y A D S V K G R - I ioi TGGCGTGGGT
TGAAGGGCCG 5 ACCGCACCCA
ACTTCCCGGC
F T I S R D N S K N T L Y L Q M N E L R A E D T A V Y Y C A K P G 0 201 GTTCACCATC TCCCGCGACA ATTCCAAGAA CACGCTGTAT CTGCAAATGA ACAGCCTGCG TGCCGAGGAC ACCGCGGTAT ATTACTGTGC
GAAACCTGGT
CAAGTGGTAG AGGGCGCTGT TAAGGTTCTT GTGCGACATA GACGTTTACT TGTCGGACGC ACGGCTCCTG TGGCGCCATA TAATGACACG CTTTGGACCA
5 V W F D Y W G Q G T L V T V S S
301 GTTTGGTTTG TCTCGAGC (SEQ ID NO: 200)
CAAACCAAAC AGAGCTCG (SEQ ID NO: 201)
0 DOM-59 SEQID NO: 61
DOM-61 SEQID NO: 62
CAAGTGGTAG AGGGCGCTGT TAAGGTTCTT GTGCGACATA GACGTTTACT TGTCGGACGC ACGGCTCCTG TGGCGCCATA TAATGACACG CCTTATACAC
G M R W N E F D Y W G O G T L V T V S S
GGGATGCGTT (SEQ ID NO: 204) CCCTACGCAA (SEQ ID NO: 205)
DOM-62 SEQID NO: 63
E V Q L L E S G G G L V Q P G G S L R L S C A A S G F T F S S Y E M -
GAGGTGCAGC AGTTATGAGA
CTCCACGTCG TCAATACTCT
10 - A W V R Q A P G K G L E K V Ξ S I T S L G T S T Y Y A D S V
K G R -
101 TGGCTTGGGT
TGAAGGGCCG
1 c ACCGAACCCA
1 J ACTTCCCGGC
- F T l S R D N S K H T L Y L Q M H S L R A E D T A V Y Y C A K P G
_Λ 201 GTTCACCATC
Z U GAAACCGGGT
CAAGTGGTAG CTTTGGCCCA
R K F D Y W G Q G T L V T V S S
25 301 AGGAAGTTTG TCTCGAGC (SEQ ID NO: 206)
TCCTTCAAAC AGAGCTCG (EEQ ID NO: 207)
DOM-65 SEQ ID NO: 64
30 E V Q L L E S G G G L V Q P G G S L R L S C A A S G F T F N E
Y E M -
1 GAGGTGCAGC TGTTGGAGTC TGGGGGAGGC TTGGTACAGC CTGGGGGGTC CCTGCGTCTC TCCTGTGCAG CCTCCGGATT CACCTTTAAT
GAGTATGAGA
„ _ CTCCACGTCG ACAACCTCAG
JJ CTCATACTCT
• T K V R Q A P G K G L E K V S T I T S E G S G T Y Y A D S V K G R -
. „ 101 TGACGTGGGT
4U TAAAGGGCCG
ACTGCACCCA ATTTCCCGGC
DOM-66 SEQID NO: 65 E V Q L L E S G G G L V Q P G G S L R L S C A A S G F T F S D Y E M -
1 GAGGTGCAGC
GATTATGAGA
ACAACACCCA TAATGATCAC TCCCAGTAAG
ACTTCCCGGC
- F T I E R D K S K N T L Y L Q M H S L R A E D T A V Y Y C A K P G 201 GTTCACCATC TCCCGCGACA ATTCCAAGAA CACGCTGTAT CTGCAAATGA ACAGCCTGCG TGCCGAGGAC ACCGCGGTAT ATTACTGTGC
GAAACCTGGG
CAAGTGGTAG AGGGCGCTGT TAAGGTTCTT GTGCGACATA GACGTTTACT TGTCGGACGC ACGGCTCCTG TGGCGCCATA TAATGACACG cTTTGGACCc
T S F D Y W G Q G T L V T V S S
301 ACTTCGTTTG TCTCGAGC (SEQ ID NO: 210)
TGAAGCAAAC AGAGCTCG (SEQ ID NO: 211)
DOM-67 SEQID NO: 66
DOM-68 SEQID NO: 67
186 DOM-69 SEQ, ID NO: 68
E V Q L L E S G G G L V O P G G S L R L S C A A S G F T F L D Y G M -
GAGGTGCAGC GATTATGGTA
CTCCACGTCG CTAATACCAT
- A W V R Q A P G K G L E H V S A I S P L G L S T Y Y A D S V
K S R -
101 TGGCTTGGGT
TGAAGAGCCG ACCGAACCCA ACTTCTCGGC
- F T I E R D N S K N T L Y L Q M K S L R A E D T A. V Y Y C A K E V 201 GTTCACCATC GAAAGAGGTG
CAAGTGGTAG CTTTCTCCAC
R V G R G V H P P K F D Y M G Q G T L V T V S S 301 AGGGTGGGTA GGGGTCA.GGG GC (SEQ ID NO: 216)
TCCCACCCAT CG (SEQ ID NO: 217)
DOM-70 SEQ ID NO: 69 E V Q L L E S G G G L V Q P G G S I R L S C A A S G F T F E N
Y A M -
1 GAGGTGCAGC TGTTGGAGTC TGGGGGAGGC TTGGTACAGC CTGGGGGGTC CCTGCGTCTC TCCTGTGCAG CCTCCGGATT CACCTTTGAG
AATTATGCTA CTCCACGTCG ACAACCTCAG TTAATACGAT
• S W V R Q A P G K G L E W V S T I A P L G V P T Y Y A D S V K G R - 101 TGTCGTGGGT TGAAGGGCCG
ACAGCACCCA ACTTCCCGGC
DOM-71 SEQID NO: 70 Y P M -
DOM-72 SEQID NO: 71 E V C l I E S G G G I V Q P G G S L R L S C A A S G F T F E A Y P M
GAGGTGCAGC CACCTTTGAG GCGTATCCTA
CTCCACGTCG GACCCCCCAG
CGCATAGSAT
• S K V R Q A P G K G L E K V S E I S F L G L W T Y Y A D S V K G R - 101 TGTCGTGSGT GACMACTAC
TGAAGGGCCG
ACAGCACCCA GGCGGTCCGA ACTTCCCGGC
' F T I S R D N S K N T L Y L Q M N S L R A E D T A V Y Y C A
K L S
201 GTTCACCATC ATTCCAAGAA
GAAACTTAGT CAAGTGGTAG AGGGCGCTGT TAAGGTTCTT GTGCGACATA GACGTTTACT TGTCGGACGC ACGGCTCCTG TGGCGCCATA TAATGACACG CTTTGAATCA
A G A E T H V Y R l F D Y W G Q G T L V T V S S
301 GCTGGGGCGG GC (SEQ ID KO: 222)
CGACCCCGCC CG (SEQ ID NO: 223)
DOM-73 SEQID NO: 72
E V Q L L E S G G G L V Q P G G S L R L S C A A S G F T F S K Y D M ' 1 GAGGTGCAGC TGTTGGAGTC TGGGGGAGGC TTGGTACAGC CTGGGGGGTC CCTGCGTCTC TCCTGTGCAG CCTCCGGATT CACCTJ1TrCT
AAGTATGATA
CTCCACGTCG ACAACCTCAG ACCCCCTCCG AACCATGTCG GACCCCCCAG GGACGCAGAG AGGACACGTC GGAGGCCTAA GTGGAAAAGA TTCATACTAT
- S W V R Q A P G K G L E W V S T I L B D G I T T Y Y A D S V
K G R -
101 TGTCTTGGGT
TGAAGGGCCG ACAGAACCCA GGCGGTCCGA GGTCCCTTCC CAGATCTCAC CCAGAGTTGA TAAGACCTCC TACCAGACTG ATGTATGATG CGTCTGAGGC ACTTCCCGGC
- F T I S R D M S K N T L Y L Q M N S L R A E D T A V Y Y C A K P G 201 GTTCACCATC TCCCGCGACA ATTCCAAGAA CACGCTGTAT CTGCAAATGA ACAGCCTGCG TGCCGAGGAC ACCGCGGTAT ATTACTGTGC GAAACCGGGG
DOM-74 SEQID NO: 73
DOM-75 SEQID NO: 74
K G R -
DOM-77 SEQID NO: 75
- A W V R Q A P G K G L E W V S T I S P L G I S T Y Y A D S V K G R -
DOM-78 SEQID NO: 76
DOM-80 SEQID NO: 77
DOM-81 SEQ ID NO: 78
E V Q L I E S G G G I V Q P G G F I R l S C A A S G F T F E L Y F M -
GAGGTGCAGC
TTGTATCCGA
CTCCACGTCG GGAGGCCTAS.
AAcATAGGCT
K G R -
101 TGGCGTGGGT
TGAAGGGCCG ACCGCACCCA ACTTCCCGGC
- F T I S R D M S K N T L Y L Q M H S L R A E D T A V Y Y C A K G H 201 GTTCACCATC GAAAGGGCAT
CAAGTGGTAG CTTTCCCGTA
E G S Y T P R S A F D Y K G Q G T L V T V S S 301 GAGGGGTCGT GTCTCGAGC (SEQ ID NO: 236)
CTCCCCAGCA GGACCAGTGG CAGAGCTCG 8SEQ ID NO: 237)
DOM-82 SEQID NO: 79 E V Q L L E S G G G L V Q P G G S L R L S C A A S G F T F V A
Y P M -
1 GAGGTGCAGC TGTTGGAGTC TGGGGGAGGC TTGGTACAGC CTGGGGGGTC CCTGCGTCTC TCCTGTGCAG CCTCCGGATT CACCTTTGTG
GCGTATCCTA CTCCACGTCG ACAACCTCAG CGCATAGGAT
A W V R Q A P G K G L E K V S T I A P L G G N T Y Y A D S V K G R - 101 TGGCGTGGGT CCGCCAGGCT CCAGGGAAGG GTCTGGAGTG GGTCTCAACT ATTGCGCCTC TGGGTGGTAA TACATACTAC GCAGACTCCG TGAAGGGCCG
ACCGCACCCA GSCGGTCCGA GGTCCCTTCC CAGACCTCAC CCAGAGTTGA TAACGCGGAG ACCCACCATT ATGTATGATG CGTCTGAGGC ACTTCCCGGC cAAGTGGTAG
DOM-83 SEQID NO: 80 Y Q M -
DOM-84 SEQ ID NO: 81 E V Q l L E S G G G L V Q P G G S L R L S C A A E G F T F R Q Y Q M -
GAGGTGCAGC TTGGTACAGC
CAGTACCAGA
CTCCACGTCG GTCATGGTCT
• A W A R Q A P G K G L E W V S T I A S D G V S T Y Y A D S V K G R - 101 TGGCTTGGGC
TGAAGGGCCG
ACCGAACCCG TAACGCAGCC TACCACAAAG ACTTCCCGGC
- F T l S R D H S K H T L Y L Q M N S L R A E D T A V Y Y C A
K V G
201 GTTCACCATC
GAAAGTTGGT CAAGTGGTAG CTTTCAACCA
R D F D Y W G Q G T L V T V S S
301 CGTGATTTTG TCTCGAGC (SEQ ID NO: 242)
GCACTAAAAC AGAGCTCG (SEQ ID NO: 243)
DOM-116 SEQID NO: 82
D I Q M T Q S P S S L S A S V G D R V T I T C R A S Q P I G P D L L - 1 GACATCCAGA GGGCAAGTCA. GCCTA-TTGGT
CCTGATTTAC
CTGTAGGTCT GGACTAAATG
- W Y Q Q K P G K A P K L L I Y Q T E I L Q S G V P S R F S G
S G S -
101 TGTGGTACCA
GCAGTGGATC ACACCATGGT CGTCACCTAG
- G T D F T L T I S S L Q P E D F A T Y Y C Q Q Y W A F P V T F G Q 201 TGGGACAGAT GTTCGGCCAA CCCTGGTTCC ACCTTTAGTT TGCC (SBQ ID NO: 245) —
DOM-85 - SEQ ID NO.: 246
E V Q L L E S G G G L V Q P G G S L R L S C A A S G F T F E Q Y D M -
1 GAGGTGCAGC TGTTGGAGTC TGGGGGAGGC TTGGTACAGC CTGGGGGGTC CCTGCGTCTC TCCTGTGCAG CCTCCGGATT CACCTTTGAG CAGTATGATA
R W V R Q A P G K G L E W V S W I D E A G H E T Y Y A D S V K G R -
101 TGAGGTGGGT CCGCCAGGCT CCAGGGAAGG GTCTAGAGTG GGTCTCATGG ATTGATGAGG CGGGTCATGA GACATACTAT GCAGACTCCG TGAAGGGCCG ' F T I S E D N S R N T L Y L Q M N S L R A E D T A V Y Y
C A K G M
201 GTTCACCATC TCCCGCGACA ATTCCAGGAA CACGCTGTAT CTGCAAATGA ACAGCCTGCG TGCCGAGGAC ACCGCGGTAT ATTACTGTGC GAAAGGGATG
D G F D Y W G Q G T L V T V S S 301 GATGGGTTTG ACTACTGGGG TCAGGGAACC CTGGTCACCG TCTCGAGC — SEQ ID NO. : 361.
DOM-86 - SEQ ID NO.: 247
D I Q M T Q S P S S L S A S V G D R V T I T C R A S Q D I G D A L F - i GACATCCAGA TGACCCAGTC TCCATCCTCC CTGTCTGCAT CTGTAGGAGA CCGTGTCACC ATCACTTGCC GGGCAAGTCA
GGATATTGGG GATGCTTTAT
W Y Q Q K P G K A P K L L I Y Y S S M L Q S G V P S H F S G G G S -
101 TTTGGTACCA GCAGAAACCA GGGAAAGCCC CTAAGCTCCT GATCTAΓTAT TCTTCCATGT TGCAAAGTGG GGTCCCATCA CGTTTCAGTG GCGGTGGATC
- G T D F T L T I S S L Q P E D F A T Y Y C Q Q R H E T P A T F G Q
201 TGGGACAGAT TTCACTCTCA CCATCAGCAG TCTGCAACCT GAAGATTTTG CTACGTACTA CTGTCAACAG CGGCATAGTA CTCCTGCTAC GTTCGGCCAA G T K V E I K R
301 GGGACCAAGG TGGARATCAA ACGG ~ SEQ ID NO. : 362
DOM-87 - SEQ ID NO.: 248 D I Q M T Q S P S S L S A S V G D R V T I T C R A S Q D I D E S L M -
1 GACATCCRGA TGACCCAGTC TCCATCCTCC CTGTCTGCAT CTGTAGGAGA CCGTGTCACC ATCACTTGCC GGGCAAGTCA GGATATTGAT GAGTCTTTAA ' W Y Q Q K P G K A P R L L I Y G V S Y L Q S G V P S R F
S G S G S -
101 TGTGGTACCA GCAGARACCA GGGAAAGCCC CTAGGCTCCT GRTCTRTGGG GTGTCCTATT TGCAARGTGG GGTCCCATCA CGTTTCAGTG GCRGTGGRTC
- G T D F T L T I S S L Q P E D F A T Y Y C Q Q R W K A P F T F G Q
201 TGGGACAGAT CTCCTTTTAC
G T K V E I K R - SEQ ID NO. : 363
DOM-88 - SEQ ID NO. : 249
D I Q M T Q S P S S L S A S V G D R V T I T C R A S Q E I V E D L Y -
1 GACATCCAGA TGACCCAGTC TCCATCCTCC CTGTCTGCRT CTGTAGGAGA CCGTGTTACC ATCRCTTGCC GGGCAAGTCA GGAGATTGTG GRGGATTTAT
- W Y Q Q K P G K A A K L L I Y G A S W L Q S G V P S R F S G S G S -
101 ATTGGTATCR GCRGAAACCA GGGAAAGCCG CTARGCTCCT GATCTATGGT GCGTCCTGGT TGCAAAGTGG GGTCCCATCA CGTTTCAGTG GCAGTGGATC . - G T D F T L T I S S L Q P E D F A T Y Y C Q Q T R R R P
Y T F G Q
201 TGGGACAGRT ACGCGTAGGC GTCCTTATAC
G T K V E I K R 301 GGGACCAAGG TGGAAATCAA ACGG — SEQ ID NO. : 364
DOM-89 - SEQ ID NO. : 250
D I Q M T Q S P A S L S A S V G D R V T I T C R A S Q D I D P M L R -
301 GGGACCAAGG TGGAAATCAA ACGG — SEQ ID NO. : 365
DOM-90 - SEQ ID NO. : 251
- W Y Q Q K P G K A P R L L I Y Y G S V L Q S G V P S R F
S G S G S -
V T F G Q
201 TGGGACAGAT CCATCAGCAG TCTGCAACCT GAAGATTTTG CTACGTACTA CTGTCAACAG CGTTTTCAGG AGCCTGTGAC
G T K V E I K R 301 GGGACCAAGG TGGAAATCAAACGG ~ SEQ ID NO. : 366
DOM-91 - SEQ ID NO. : 252
D I Q M T Q P S S L S A S V G D R V T T C R A S Q i s D E L N -
301 GGGACCAAGG TGGAAATCAA ACGG — SEQ ID NO. : 367
DOM-92 - SEQ ID NO.: 253
- G W V R Q A P G K G L E W V S T I S T G G F S T Y Y A D
S V K G R -
C A K A R
201 GTTCACCATC CTGCMATGA ATTACTGTGC
Y Y Y L S Q I K N F D Y W G Q G T L V T V S S 301 TATTATTATC TAAGAATTTT GTCTCGAGC - SEQ ID NO.: 368
DOM-93 - SEQ ID NO.: 254
E V Q L L E S G G G L V Q P G G S L R L S C A A S G F T F D I Y G M 1 GAGGTGCAGC TGTTGGAGTC TGGGGGAGGC TTGGTACAGC CTGGGGGGTC CCTGCGTCTC TCCTGTGCAG CCTCCGGATT CACCTTTGAT
• T W V R Q A P G K G L E W V S S I S P l G L V T Y Y A D P V K G R - 101 TGACTTGGGT GTCTGGAGTG
GCAGACCCCG
F T I S R D N S K H T L Y L Q M N S L R A E D T A V Y Y C A K L K
201 GTTCACCATC TCCCGCGACA AΓTCCAAGAA CACGCTGTAT CTGCAAATGA ACAGCCTGCG TGCCGAGGAC ACCGCGGTAT ATTACTGTGC
E H G D V P F D Y W G Q G T L V T V S S
301 GAGCATGGGG ATGTTCCTTT TGACTACTGG GGTCAGGGAA CCCTGGTCAC CGTCTCGAGC — SEQ XD NO . : 369
DOM-94 - SEQ ID NO . : 255
E V Q L L E S G G G L V Q P G G S L R L S C A A S G F T F E L Y P M -
1 GAGGTGCAGC TGTTGGAGTC TGGGGGAGGC TTGGTACAGC CTGGGGGGTC CCTGCGTCTC TCCTGTGCAG CCTCCGGATT CACCTTTGAG
S W V R Q A P G K G L E W V S T I S P T G L L T Y Y A D S V K G R -
101 TGAGTTGGGT GCAGACTCCG - F T I S R D N S K N T L Y L Q M K S L R A E D T A V Y Y
C A K F K
201 GTTCACCATC TCCCGCGACA ATTCCAAGAA CACGCTGTAT CTGCAAATGA ACAGCCTGCG TGCCGAGGAC ACCGCGGTAT ATTACTGTGC GAAATTTAAG
R S G K T D D T N F D Y W G Q G T L V T V S S 301 AGGAGTGGGA GTCAGGGAAC GTCTCGAGC — SEQ ID
NO.: 370
DOM-95 - SEQ ID NO. : 256
E V Q L L E S G G G L V Q P G G S L R L S C A A S G F T F R E Y D M - 101 TGCTGTGGGT CCGCCAGGCT CCAGGGAAGG GTCTAGAGTG GGTCTCAACG ATTGTGGGGG ATGGTAATGG TACATACTAC
ATTACTGTGC
R Q F D Y W G Q G T L V T V S S
301 CGTCAGTTTGACTACTGGGG TCAGGGAACC CTGGTCACCG TCTCGAGC — SEQ ID NO. : 371
DOM-96 - SEQ ID NO.: 257
- L W V R Q A P G K G L E W V S S I S P S G R W T Y Y A D
S V K G R -
C A K S L
301 TTTGAGGGTA GTTTTGACTA CTGGGGTCAG GGAACCCTGG TCACCGTCTC GAGC — SEQ ID NO. : 372
DOM-97 - SEQ ID NO.: 258
E V Q L L E S G G G L V Q P G G S L R L S C A A
F E E Y G M - 1 GAGGTGCAGC TGTTGGAGTC TGGGGGAGGC TTGGTACRGC CTGGGGGGTC CCTGCGTCTC TCCTGTGCAG CCTCCGGATT CACCTTTGAG
S W V R Q A P G K G L E W V S T I S P I G V T T Y Y A D S V K G R - 101 TGAGTTGGGT GGTCTCAACG
GCAGACTCCG
• F T I S R D N S K N T L Y L Q M H S L R A E D T A V Y Y C A K N A
201 GTTCACCATC ATTACTGTGC
Y D R K S N F D Y W G Q G T L V T V S S
301 TATGATCGGA AGTCTAATTT TGACTACTGG GGTCAGGGAA CCCTGGTCAC CGTCTCGAGC — SEQ ID NO . : 373
DOM-98 - SEQ ID NO.: 259
E V Q L L E S G G G L V Q P G G S L R L S C A A S G F T F D R Y V M -
1 GAGGTGCAGC TGTTGGAGTC TGGGGGAGGC TTGGTACAGC CTGGGGGGTC CCTGCGTCTC TCCTGTGCAG CCTCCGGATT CACCTTTGAT
V W V R Q A P G K D L E W V S G I T P S G R R T Y Y A D S V K G R -
101 TGGTGTGGGT GCAGACTCCG TGAAGGGCCG - F T I S R D N S K D T L Y L Q M N S L R A E D T A V Y Y
C A K V L
201 GTTCACCATC TCCCGCGACA ATTCCAAGGA CACGCTGTAT CTGCAAATGA ACAGCCTGCG TGCCGAGGAC ACCGCGGTAT ATTACTGTGC GAAAGTGTTG
G R H F D P L L P S F D Y W G Q G T L V T V S S 301 GGGCGTCATT GC — SEQ
ID NO. : 374
DOM-99 - SEQ ID NO.: 260
E V Q L L E S G G G L V Q P G G S L R L S C A A S G F T F E D ϊ A W • 1 GAGGTGCAGC TGTTGGAGTC TGGGGGAGGC TTGGTACAGC CTGGGGGGTC CCTGCGTCTC TCCTGTGCAG CCTCCGGATT CACCTTTGAG
• S W V R Q A P G K G L E W V E T I T P G G F W T Y Y A D S V K G R - 101 TGAGTTGGGT
GCAGACTCCG
• F T I S R D K S K N T L Y L Q M N S L R A E D T A V Y Y C A K T S
201 GTTCACCATC ATTACTGTGC
S G E L Q L V E D F D Y W G Q G T L V T V S S 301 AGTGGGGAGT GTCTCGAGC — ΞΞQ ID
NO.: 375
DOM-100 - SEQ ID NO. : 261
D I Q M T Q S P S S L S A S V G D R V T I T C R A S Q N I K H S L R -
1 GACATCCAGA GAATATTAAG
• W Y Q Q K P G K A P R L L I Y H R S Q L Q S G V P S R F S G S G S -
101 GGTGGTACCA CGTTTCAGTG - G T D F T L T I S S L Q P E D F A T Y Y C Q Q V R H R P
Y T F G Q
201 TGGGACAGAT GTCCTTATAC
G T K V E I K R 301 GGGACCAAGG TGGAAATCAAACGG — SΞQ ID NO. : 376
DOM-101 - SEQ ID NO. : 262
D I Q M T Q S P S S L S A S V G D R V T I T C R A S Q A I G H R L R - 101 GTTGGTATCA GCAGAAACCA GGGAAAGCCC CTAAGCTCCT GATCTATCAT CGGTCCAAGT TGCAAAGTGG GGTCCCATCA
301 GGGACCARGG TGGAAATCAA ACGG — SEQ ID NO . : 377
DOM-102 - SEQ ID NO.: 263
- W Y Q Q K P G K A P K L L I Y H R S H L Q S G V P S R F
S G S G S - 101 GGTGGTACCA GCAGAAACCA GGGAAAGCCC CCAAGCTCCT GATCTATCAT AGGTCCCATT TGCAAAGTGG GGTCCCATCA
CGTTTCAGTG
- G T D F T L T I S S L Q P E D S A T Y Y C Q Q W D R P P Y T F G Q
201 TGGGACAGAT TTCACTCTCA CCATCAGCAG TCTGCAACCT GAAGATTCTG CTACGTACTA CTGTCAACAG TGGGATAGGC CGCCTTATAC
G T K V E I K R
301 GGGACCAAGG TGGAAATCAA ACGG — SEQ ID NO . : 378
DOM-I 03 - SEQ ID NO. : 264
D I Q M T Q S L S A S V G D R V T I T A S Q
I G H R L E -
201 TGGGACAGAT TTCACTCTCA CCATCAGCAG ΓCTGCAACCT GAAGATTTTG CTACGTACTA CTGTCAACAG GTGCGGGCTG TGCCTTATAC GTTTGGCCAA
G T K V E I K R
301 GGGACCAAGG TGGAAATTAA ACGG — SEQ ID NO. : 379
DOM-104 - SEQ ID NO. : 265
Y T F G Q
201 TGGGACAGAT TTCACTCTCA CCATCAGCAG TCTGCAACCT GAAGATTTTG CTACGTACTA CTGTCAACAG GTTCGTTTTT CTCCTTATAC
G T K V E I K R
301 GGGACCAAGG TGGAAATCAAACGG — SEQ ID NO. : 380
DOM-105 - SEQ ID NO. : 266
D I Q M T Q S L S A S V G D R V T T C R A S Q G H R L R -
301 GGGACCAAGG TGGAAATCAAACGG - SEQ ID NO. : 381
DOM-106 - SEQ ID NO. : 267
- W Y Q Q K P G K A P K L L I Y H R S R L Q S G V P S R P
S G S G S -
N T F G Q
201 TGGGACAGAT TTCACTCTCA CCATCAGCAG TCTGCAACCT GAGGATTTTG CTACGTACTA CTGTCAACAG TATAAGGTTA GGCCTAATAC
G T K V E I K R 301 GGGACCAAGG TGGAAATCAAACGG ~ SEQ ID NO. : 382
DOM-107 - SEQ ID NO. : 268
D I Q M T Q S L S A S V G D R V T
I G H R L R - 1 GACATCCAGA TGACCCAGTC TCCATCCTCC CTGTCTGCAT CTGTAGGAGA CCGTGTCACC ATCACTTGCC GGGCAAGTCA GGCTATTGGGCATCGTTAC
- W Y Q Q K P G K A P K L L I Y H R S K L Q S G V P S E P S G S G S - 101 GTTGGTATCA GCAGAAACCA GGGAAAGCCC CTAAGCTCCT GATTTATCAT CGGTCCAAGT TGCAAAGTGG GGTCCCATCA
CGTTTCAGTG GCAGTGGATC
• G T D F T L T I S S L Q P E D F A T Y Y C Q Q T Y S S P H T F G Q
201 TGGGACAGAT TTCACTCTCA CCATCAGCAG TCΓGCAACCT GAAGATTTTG CTACGTACTA CTGTCAACAG ACTTATTCGT CTCCTCATAC GTTCGGCCAA
G T K V E I K R
301 GGGACCAAGG TGGAAATCAAACGG — SEQ ID NO. : 383
DOM-108 - SEQ ID NO.: 269
D I Q M T Q S P S S L S A S V G D R V T I T C R A S Q A I G H R L R -
1 GACATCCAGA TGACCCAGTC TCCATCCTCC CTGTCTGCAT CTGTAGGAGA CCGTGTCACC ATCACTTGCC GGGCAAGTCA GGCTATTGGG CATCGGTTAC - W Y Q Q K P G K A P K L L I Y H R S K L Q S G V P S R F
S G S G S -
101 GTTGGTATCA GCAGAAACCA GGGAAAGCCC CTAAGCTCCT GATCTATCAT CGGTCCAAGT TGCAAAGTGG GGTCCCATCA CGTTTCAGTG GCAGTGGATC
• G T D F T L T I S S L Q P E D F A T Y Y C Q Q R A V R P F T F G Q
201 TGGGACAGAT TTCACTCTCA CCATCAGCAG TCTACAACCT GAAGATTTTG CTACGTACTA CTGTCAACAG AGGGCGGTGA GGCCTTTTAC GTTCGGCCAA
G T K V E I K R
301 GGGACCAAAG TGGAAATCAA ACGG — SEQ ID NO . : 384
DOM-109 - SEQ ID NO. : 270
D I Q M T Q S P S L S A S V G D R V T C R A S Q
I G H R L R ' 1 GACATCCAGA TGACCCAGTC TCCATCCΓCC CTGTCTGCAT CΓGTAGGAGA CCGTGTCACC ATCACTTGCC GGGCAAGTCA
301 GGGACCAAGG TGGMATCAAACGG — SEQ ID NO. : 385
DOM-110 - SEQ ID NO. : 271
- W Y Q Q K P G K A P K L L I Y A G S I L Q S G V P S R F
S G S G S -
101 GGTGGTACCA CGTTTCAGTG GCAGTGGATC
- G T D F T L T I S S L Q P E D F A T Y Y C Q Q T S I R P Y T F G Q
201 TGGGACAGAT TTCACTCTCA CCATCAGCAG TCTGCAACCT GAAGATTTTG CTACGTACTA CTGTCAACAG ACTAGTATTA GGCCTTATAC
G T K V E I K R
301 GGGACCAAGG TGGAAATCAA ACGG — SEQ ID NO. : 386
DOM8-111 - SEQ ID NO. : 272
E V Q L L E L V Q P G G S L R L S C A A
F E R Y P M - 1 GAGGTGCAGC TGTTGGAGTC TGGGGGAGGC TTGGTACAGC CTGGGGGGTC CCTGCGTCTC TCCTGTGCAG CCTCCGGATT CACCTTTGAG
• T W V E Q A P G K G L E W V S T I H G S G S A T Y Y A D S V K G R - 101 TGACGTGGGT CCAGGGAftGG GTCTAGAGTG GGTCTCAACG ATTCATGGTT CTGGTAGTGC TACATACTAC
GCAGACTCCG
• F T I S R D N S K N T L Y L Q M N S L R A E D T A V Y Y C A K G E
201 GTTCACCATc TCCCGCGACA ATTCCAAGAA CACGCTGTAT CTGCAAATGA ACAGCCTGCG TGCCGAGGAC ACCGCGGTAT ATTACTGTGC
Y T S R H N S L G H F D Y W G Q G T L V T V S S
301 TATACTAGTC GC — SEQ
ID NO. : 387
DOM-112 - SEQ ID NO.: 273
E V Q l L E S G G G L V Q P G G S L R L S C A A S G F T F M D Y P M -
1 GAGGTGCAGC TGTTGGAGTC TGGGGGAGGC TTGGTACAGC CTGGGGGGTC CCTGCGTCTC TCCTGTGCAG CCTCCGGATT CACCTTTATG
G W V R Q A P G K G L E W V S S I G P V G M S T Y Y A D S V K G R -
101 TGGGGTGGGT GCAGACTCCG TGAAGGGCCG - F T I S R D N S K N T L Y L Q M N S L R A E D T A V Y Y
C A K Y G
201 GTTCACCATC TCCCGCGACA ATTCCAAGAA CACGCTGTAT CTGCAAATGA ACAGCCTGCG TGCCGAGGAT ACCGCGGTAT ATTACTGTGC GAAATATGGG
G T S G R H N T K F D Y W G Q G T L V T V S S 301 GGGACTAGTG GTCTCGAGC — SEQ ID
NO.: 388
DOM-113 - SEQ ID NO.: 274 E V Q L L E S G G G L V Q P G G S L R L S C A A S G F T
F T E Y P M - 1 GAGGTGCAGC TGTTGGAGTC TGGGGGAGGC TTGGTACAGC CTGGGGGGTC CCTGCGTCTC TCCTGTGCAG CCTCCGGATT CACCTTTACT GAGTATCCTA
S W V E Q A P G K G L E W V S V I S P L G F T T Y Y A D S V K G R - 101 TGAGTTGGGT CCGCCAGGCT CCAGGGAAGG GTCTAGAGTG GGTCTCAGTT ATTTCTCCTC TTGGTTTTAC GACATACTAC
GCAGACTCCG TGAAGGGCCG
• F T I S R D N S K N T L Y L Q M N S L R A E D T A V Y Y C A K W T
201 GTTCACCATC TCCCGCGACA ATTCCAAGAR CACGCTGTAT CTGCAAATGA ACAGCCTGCG TGCCGAGGAT ACCGCGGTAT ATTACTGTGC GAAATGGACT
G G S G I L K S S F D Y W G Q G T L V T V S S
301 GGTGGGAGTG GTATTTTGAA TTCTTCTTTT GACTACTGGG GTCAGGGAAC CCTGGTCACC GTCTCGAGC — SEQ ID NO.: 389
DOM-I 14 - SEQ ID NO.: 275
E V Q L L E S G G G L V Q P G G S L R L S C A A S G F R V S N Y D L -
1 GAGGTGCAGC TGTTGGAGTC TGGGGGAGGC TTGGTACAGC CTGGGGGGTC CCTGCGTCTC TCCTGTGCAG CCTCCGGATT TAGGGTTAGC AATTACGATT
T W V R Q A P G K G L E W V S T I S A T N G S T Y Y A D S V K G R -
101 TGACCTGGGT CCGCCAGGCT CCAGGGAAGG GTCTAGAGTG GGTATCAACC ATTAGTGCCA CAAACGGTAG CACATACTAC
GCAGACTCCG TGAAGGGCCG - F T I S R D N S K N T L Y L Q M N S L R A E D T A V Y Y
C A A V T
201 GTTCACCATC TCCCGTGACA ATTCCAAGAA CACGCTGTAT CTGCAAATGA ACAGCCTGCG TGCCGAGGAC ACCGCGGTAT ATTATTGCGC GGCAGTGACG
W W L L R H N D N L G F W G Q G T L V T V S S 301 TGGTGGTTGT TGCGTCATAA CGACAACTTG GGGTTTTGGG GTCAGGGAAC CCTGGTCACC GTCTCGAGC — SEQ ID
NO.: 390
DOM-115 - SEQ ID NO.: 276
E V Q L L E S G G G L V Q P G G S L R L S C A A S G F S I S Y K N M - 1 GAGGTGCAGC TGTTGGAGTC TGGGGGAGGC TTGGTACAGC CTGGGGGGTC CCTGCGTCTC TCCTGTGCAG CCTCCGGATT TAGCATTAGC
• A W V R Q A P G K G L E W V S A I K A A N G S T Y Y A D S V K G R - 101 TGGCCTGGGT
GCAGACTCCG
• F T I S R D N S K N T L Y L Q M N S L R A E D T A V Y Y C A T G S
201 GTTCACCATC ATTATTGCGC
Q K K R T Y T F D F W G Q G T L V T V S S
301 CAGAAGAAGC GGACCTACAC GTTCGACTTT TGGGGTCAGG GAACCCTGGT CACCGTCTCG AGC — SEQ ID NO . : 391
DOM-120 - SEQ ID NO.: 277
CACCTTTAGG
• G W V R Q A P G K G L E W V S S I N P M G Y Q T Y Y A D S V K G R -
101 TGGGTTGGGT CCAGGGAAGG GTCTAGAGTG GGTCTCATCT ATTAATCCTA TGGGTTATCA GACATACTAC GCAGACTCCG TGAAGGGCCG - F T I S R D N S K N T L Y L Q M N S L R A E D T A V Y Y
C A K H G
201 GTTCACCATC TCCCGCGACA ATTACTGTGC
V G K G T K P H N F D Y W G Q G T L V T V S S 301 GTGGGGAAGG GTCTCGAGC — SEQ ID
NO.: 392
DOM-121 - SEQ ID NO.: 278
E V Q L L E S G G G L V Q P G G S L R L S C A A S G F F E L Y R M -
4 1 GAGGTGCAGC TGTTGGAGTC TGGGGGAGGC TTGGTACAGC CTGGGGGGTC CCTGCGTCTC TCCTGTGCAG CCTCCGGRTT CACCTTTGAG CTGTATAGGA
• S W V R Q A P G K G L E W V S E I S G S G F P T Y Y A D S V K G R - ioi TGTCTTGGGT CCGCCAGGCT CCAGGGAAGG GTCTAGAGTG GGTCTCAGAG ATTAGTGGΓA GTGGTTTTCC TACATACTAC
GCAGACTCCG TGAAGGGCCG
• F T I S R D N S K H T L l' L Q M N S L R A E D T A V Y Y C A K S L
201 GTTCACCATC TCCCGCGACA ATTCCAAGAA CACGCTGTAT CTGCAAATGA ACAGCCTGCG TGCCGAGGAC ACCGCGGTAT ATTACTGTGC GAAAAGTCTG
H D K T Q H H Q E F D Y W G Q G T L V T V S S
301 CATGATAAGA CTCAGCATCA TCAGGAGTTT GACTACTGGG GTCAGGGAAC CCTGGTCACC GTCTCGAGC — SΞQ ID NO.: 393
DOM-123 - SΞQ ID NO.: 279
E V Q l L E S G G G L V Q P G G S L R L S C A A S G F T F I E Y P M '
1 GAGGTGCAGC TGTTGGAGTC TGGGGGAGGC TTGGTACAGC CTGGGGGGTC CCTGCGTCTC TCCTGTGCAG CCTCCGGATT CACCTTTATT GAGTATCCTA
• R W V R Q A P G K G L E W V S L I S P S G V F T Y Y A D S V K G R -
101 TGCGGTGGGT CCGCCAGGCT CCAGGGAAGG GTCTAGAGTG GGTCTCACTT ATTTCTCCGT CTGGTGTGTT TACATACTAC GCAGACTCCG TGAAGGGCCG - F T I S R D N S K N T L Y L Q M N S L R A E D T A V Y Y
C A K G D
201 GTTCACCATC TCCCGCGACA ATTCCAAGAA CACGCTGTAT CTGCAAATGA ACAGCCTGCG TGCCGAGGAC ACCGCGGTAT ATTACTGTGC GAAAGGGGAT
E S S T F D Y W G Q G T L V T V S S 301 GAGTCTAGTA CTTTTGACTA CTGGGGTCAG GGAACCCTGG TCACCGTCTC GAGC — SEQ ID NO. : 394
DOM-124 - SEQ ID NO. : 280
E V Q L L E S G G G L V Q P G G S L R L S C A A S G F T F K R Y D M - 1 GAGGTGCAGC TGTTGGAGTC TGGGGGAGGC TTGGTACAGC CTGGGGGGTC CCTGCGTCTC TCCTGTGCAG CCTCCGGATT CACCTTTAAG
• D W V E Q A P G K G L E W V S T I G S S G Y P T Y Y A D S V K G R - 101 TGGATTGGGT
GCAGACTCCG
F T I S R D N S K N T L Y L Q M N S L R A E D T A V Y Y C A E R M
201 GTTCACCATC ATTACTGTGC
P G Y F P G F A R Q F D Y W G Q G T L V T V S S 301 CCTGGTTATT GC — SEQ
ID NO. : 395
DOM-125 - SEQ ID NO.: 281
CACCTTTTGG
• G W V R Q A P G K G L E W V S T I N D E G R E T Y Y A D S V K G R -
101 TGGGGTGGGT GCAGACTCCG TGAAGGGCCG - F T I S R D N S K N T L Y L Q M N S L R A E D T A V Y Y
C A K K R
201 GTTCACCATC TCCCGCGACA ATTACTGTGC GAAAAAGCGG
V S S S V N A P Y E F D Y W G Q G T L V T V S S 301 GTGTCTAGTT GC — SEQ
ID NO. : 396
DOM-126 - SEQ ID NO.: 282
E V Q L L E S G G G L V Q P G G S L R L C A A S G F
F A N Y S M - 1 GAGGTGCAGC TGTTGGAGTC TGGGGGAGGC TTGGTACAGC CTGGGGGGTC CCTGCGTCTC TCCTGTGCAG CCTCCGGATT CACCTTTGCG S W V R Q A P G K G L E W V S S I D R L G T H T Y Y A D S V K G R - 101 TGAGTTGGGT
GCAGACTCCG F T I S R D N S K N T L Y L Q M N S L E A E D T A V Y Y C A K V L
201 GTTCACCATC ATTACTGTGC
A D L I A G H A E F D Y W G Q G T L V T V S S 301 GCTGATCTTA GTCTCGAGC — SEQ ID
NO. : 397
DOM-127 - SEQ ID NO.: 283
CACCTTTCCG A W V R Q A P G K G L E W V S G I S R S G S M T Y Y A D S V K G R -
101 TGGCGTGGGT GCAGACTCCG TGAAGGGCCG - F T I S R D N S K N T L Y L Q M N S L R A E D T A V Y Y
C A K G V
201 GTTCACCATC TCCCGCGACA ATTCCAAGAA CACGCTGTAT TTGCAAATGA ACAGCCTGCG TGCCGAGGAC ACCGCGGTAT ATTACTGTGC GAAAGGTGTT
D A H V Y Y M E P F F D Y W G Q G T L V T V S S 301 GATGCGCATG SC — SEQ
ID NO. : 398
DOM-128 - SEQ ID NO. : 284
E V Q L L E E G G G L V Q P G G S L R L S C A A S G F T F E R Y Q M - 1 GAGGTGCAGC TGTTGGaGTC TGGGGGAGGC TTGGTACAGC CTGGGGGGTC CCTGCGTCTC TCCTGTGCAG CCTCCGGATT CACCTTTGAG
• A W V R Q A P G K G L E W V S T I S S D G G G T Y Y A D S V K G R -
5 101 TGGCGTGGGT
GCAGACTCCG
• F Γ I S E D N S K N T L Y L Q M N S L R A E D T A V Y Y
C A K P G
, n 201 GTTCACCATc ΓCCCGCGACA ATTCCAAGAA CACGCTGTAT CTGCAAATGA ACAGCCTGCG TGCCGAGGAC ACCGCGGTAT
IU ATTACTGTGC GAAACCGGGT
T V F D Y W G Q G T L V T V S S
301 ACTGTTTTTG ACTACTGGGG TCAGGGAACC CTGGTCACCG TCTCGAGC — SEQ ID NO. : 399
15 DOM-129 - SEQ ID NO. : 285
E V Q L L E S G G G L V Q P G G S L R L S C A A S G F T F P K Y E M -
1 GAGGTGCAGC TGTTGGAGTC TGGGGGAGGC TTGGTACAGC CTGGGGGGTC CCTGCGTCTC TCCTGTGCAG CCTCCGGATT CACATTTCCG AAGTATGAGA 0 - A W V R Q A P G K G L E W V S S I D G D G K S T Y Y A D
S V K G R -
101 TGGCGTGGGT CCGCCAGGCT CCAGGGAAGG GTCTAGAGTG GGTCTCATCT ATTGATGGTG ATGGTAAGTC TACATACTAC GCAGACTCCG TGAAGGGCCG
- F T I S R D N S K N T L Y L Q M N S L R A E D T A V Y Y 5 C A K P D
201 GTTCACCATC ATTCCAAGAA CACGCTGTAT CTGCAAATGA ACAGCCTGCG ACCGCGGTAT ATTACTGTGC GAAACCGGAT
Q F F D Y W G Q G T L V T V S S
301 cAGTTTTTTG ACTACTGGGG TCAGGGAACC CTGGTCACCG TCTCGAGC — SΞQ ID NO. : 400 0
DOM-130 - SEQ ID NO. : 286
E V Q L L E S L V Q P G G S L R L S C T A G F
F A G Y Q M - 5 1 GAGGTGCAGC TGTTGGAGTC TGGGGGAGGC TTGGTACAGC CTGGGGGGTC CCTGCGTCTC TCCTGTACAG CCTCCGGATT CACCTTTGCG GGTTATCAGA S K V R Q A P G K G L E W V S S I T N E G V S T Y Y A D S V K G R - 101 TGTCGTGQGT CCGCCAGGCT CCAGGGAAGG GTCTAGAGTG GGTCTCAAGT ATTACTAATG AGGGTGTTTC TACATACTAC
GCAGACTCCG TGAAGGGCCG F T I S R D H S K K T L Y L Q M N S L R A E D T A V Y Y C A K P G
201 GTTCACCATC TCCCGCGACA ATTCCAAGAA CACGCTGTAT CTGCAAATGA ACAGCCTGCG TGCCGAGGAC ACCGCGGTAT ATTACTGTGC GAAACCGGGG
K Y F D Y W G Q G T L V T V S S
301 AAGTATTTTGACTACTGGGG TCAGGGAACC CTGGTCACCG TCTCGAGC — SEQ ID NO. : 401
DOM-131 - SEQ ID NO.: 287
E V Q L L E S G G G L V Q P G G S L R L S C A A S G F T F G E Y E M -
1 GAGGTGCAGC TGTTGGAGTC TGGGGGAGGC TTGGTACAGC CTGGGGGGTC CCTGCGTCTC TCCTGTGCAG CCTCCGGATT CACCTTTGGG GAGTATGAGA - V W V R Q A P G K G L E W V S S I T S D G L S T Y Y A D
S V K G R - ioi TGGTGTGGGT CCGCCAGGCT CCAGGGTiAGG GTCTGGAGΓG GGTCTCATCT ATTACGTCGG ATGGTCTGAG TACATACTAC
GCAGACTCCG TGAAGGGCCG F T I S R D N S K N T L Y L Q M H S L R A E D T A V Y Y C A K P G
201 GTTCACCATC TCCCGCGACA ATTACTGTGC GAAACCGGGT
I R F D Y W G Q G T L V T V S S
301 ATTCGTTTTG ACTACTGGGG TCAGGGAACC CTGGTCACCG TCTCGAGC — SEQ ID NO. : 402
DOM-132 - SEQ ID NO. : 288
E V Q L L E S G G G L V Q P G G S L R L S C A A G F
F A D Y D M - 1 GaGGTGCAGC TGTTGGAGTC TGGGGGAGGC TTGGTACAGC CTGGGGGGTC CCTGCGTCTC TCCTGTGCAG CCTCCGGATT CACCTTTGCT GATTATGATA
■ A W V R Q A P G K G L E W V S G I V D D G L M T Y Y A D S V K G E - 101 TGGCTTGGGT CCGCCAGGCT CCAGGGAAGG GTCTAGAGTG GGTCTCAGGT ATTGTTGATG ATGGTCTTAT GACATACTAC
GCAGACTCCG TGAAGGGCCG
• F T I S R D N S K JJ T L Y L Q M N S L E A E D T A V Y Y C A K P D
201 GTTCACCATC TCCCGCGACA ATTCCAAGAA CACGCTGTAT CTGCAAATGA ACAGCCTGCG TGCCGAGGAC ACCGCGGTAT ATTACTGTGC GAAACCGGAT
V A F D Y W G Q G T L V T V S N
301 GTTGCTTTTG ACTACTGGGG TCAGGGGACC CTGGTCACCG TCTCGAAC — SEQ ID NO. : 403
DOM-133 - SEQ ID NO.: 289
E V Q L L E S G G G L V Q P G G S L E L S C A A S G F T F I G Y A M -
1 GAGGTGCAGC TGTTGGAGTC TGGGGGAGGC TTGGTACAGC CTGGGGGGTC CCTGCGTCTC TCCTGTGCAG CCTCCGGATT CACCTTTATT GGTTATGCTA - A W V R Q A P G K G L E B V S S I G P L G A T T Y Y A D
S V K G R -
101 TGGCGTGGGT CCGCCAGGCT CCAGGGAAGG GTCTAGAGTG GGTCTCAAGT ATTGGTCCTT TGGGTGCGAC TACATACTAC GCAGACTCCG TGAAGGGCCG
F T I S R D N S K N T L Y L Q M N S L R A E D T A V Y Y C A K L P
201 GTTCACCATC TCCCGCGACA ATTCCAAGAA CACGCTGTAT CTGCAAATGA ACAGCCTGCG TGCCGAGGAC ACCGCGGTAT ATTACTGTGC GAAATTGCCT
A G T S S H S V D F D Y W G Q G T L V T V S S
301 GCTGGTACGA GTAGTCATAG TGTGGATTTT GACTACTGGG GTCAGGGAAC CCTGGTCACC GTCTCGAGC — SΞQ ID NO.: 404
DOM-134 - SEQ ID NO. : 290
E V Q L L E S G G G L V Q P G G S L R L S C A A S G F T F A D Y E M -
301 GTTCAGTTTG ACTACTGGGG TCAGGGAACC CTGGTCACCG TCTCGAGC — SEQ ID NO . : 405
DOM-135 - SEQ ID NO. : 291
E V Q L L E S G G G L V Q P G G S L R L S C A A S G F T F R R Y V M -
1 GAGGTGCAGC TGTTGGAGTC TGGGGGAGGC TTGGTACAGC CTGGGGGGTC CCTGCGTCTC TCCTGTGCAG CCTCCGGATT CACCTTTCGT AGGTATGTTA - G W V R Q A P G K G L E W V S W I E A D G R T T Y Y A D
S V K G R -
101 TGGGGTGGGT CCGCCAGGCT CCAGGGAAGG GTCTAGAGTG GGTCTCATGG ATTGAGGCTG ATGGTCGTAC GACATACTAC GCAGACTCCG TGAAGGGCCG
- F T I S R D N S K K T L Y L Q M N S L R A E D T A V Y Y C A K G L
201 GTTCACCATC TCCCGCGACA ATTCCAAGAA CACGCTGTAT CTGCAAATGA ACAGCCTGCG TGCCGAGGAC ACCGCGGTAT ATTACTGTGC GAAAGGGCTT
T D Q H V I E F D Y W G Q G I L V T V S S
301 ACGGATCAGc ATGTTATTGA GTTTGACTAC TGGGGTCAGG GAACCCTGGT CACCGTCTCG AGC — SEQ ID NO . : 406
DOM-136 - SEQ ID NO.: 292
E V Q L L E S G G G L V Q P G G S S C A A
F D G Y R M -
201 GTTCACCATC TCCCGCGACA ATTCCASGAA CACGCTGTAT CTGCAAATGA ACAGCCTGCG TGCCGAGGAC ACCGCGGTAT
301 GGGATGCAGT TTGACTACTG GGGTCAGGGAACCCTGGTCA CCGTCTCGAG C — SEQ ID NO. : 407
DOM-137 - SEQ ID NO.: 293
- G W V R Q A P G K G L E W V S S I G P I G F T T Y Y A D
S V K G R -
101 TGGGTTGGGT CCAGGGAAGG GTCTAGAGTG GGTCTCAAGT ATTGGTCCTA TTGGTTTTAC TACATACTAC GCAGACTCCG TGAAGGGCCG
- F T I S R D N S K N T L Y L Q M H S L R A E D T A V Y Y C A E M K
301 TcσCCTTATA AGCCGCAGTT TGACTACTGG GGTCAGGGAA CCCTGGTCAC CGTCTCGAGC — SEQ ID NO . : 408
DOM-138 - SEQ ID NO. : 294
E V Q L L E S G G G L V P G G S L R L S C A A S G F T A Y W M - 1 G&GGTGCAGC TGTTGGAGTC TGGGGGAGGC TTGGTACAGC CTGGGGGGTC CCTGCGTCTC TCCTGTGCAG CCTCCGGATT CACCTTTTTG
• V W V R Q A P G K G L E W V S S I S P S G T H T Y Y A D S V K G R - 101 TGGTTTGGGT
GCAGACTCCG
F T I S R D N S K N T L Y L Q M N S L R V E D T A V Y Y C A K Y T
201 GTTCACCATC ATTACTGTGC
E P G L G S F D Y W G Q G T L V T V S S
301 GAGCCGGGGT TGGGTTCTTT TGACTACTGG GGTCAGGGAA CCCTGGTCAC CGTCTCGAGC — SEQ ID NO . : 409
DOM-139 - SEQ ID NO. : 295
CACCTTTTCG
G W V R Q A P G K G L E W V S V I S E V G S L T Y Y A D S V K G R -
101 TGGGGTGGGT GCAGACTCCG TGAAGGGCCG - F T I S R D N S K N T L Y L Q M N S L R A E D T A V Y Y
C A K P H
301 GATAGTTCGA TTGGGTTTGA CTACTGGGGT CAGGGAACCC TGGTCACCGT CTCGAGC — SEQ ID NO . :
410
DOM- 141 - SEQ ID NO. : 296
D I Q M T Q S P S S L S A S V G D R V T I T C R A S Q W I G D T L T -
301 GGGACCAAGG TGGAAATCAA ACGG — SEQ ID NO. : 411
DOM-142 - SEQ ID NO.: 297
- W Y Q Q K P G K A P K L L I Y F S S I L Q S G V P S R F
S G S G S -
T T F G R
201 TGGGACAGAT TTCACTCTCA CCATCAGCAG TCTGCAACCT GAAGATTTTG CTACGTACTA CTGTCAACAG TATCATACTT CGCCTACTAC
G T K V K I K R 301 GGGACCAAGG TGAAAATCAA ACGG — SEQ ID NO. : 412
DOM-143 - SEQ ID NO. : 298
D I Q M Q S P S S L S A S V G R V T I T C R A S Q
I E T N L E •
301 GGGACCAAGG TGGAAATCAA ACGG — SEQ ID NO. : 413
DOM-144 - SEQ ID NO. : 299
- W Y Q Q K P G K A P K L L I Y N A S W L Q S G V P S R F
S G S G S -
I T F G Q
201 TGGGACAGAT ATCCTATTAC
G T K V E I K R 301 GGGACCAAGG TGGAAATCAA ACGG — SEQ ID NO. : 414
DOM-145 - SEQ ID NO. : 300
D I Q M L S A S V G D R V T I T C R A S Q
I Y T S L S - 1 GACATCCAGA TGACCCAGTC TCCATCCTCC CTGTCTGCAT CTGTAGGAGA CCGTGTCACC ATCACTTGCC GGGCAAGTCA GACGATTTAT ACTTCGTTAA W Y Q Q K P G K A P K L L I H Y G S V L Q S G V P S R F S G S G S - 101 GTTGGTATCA GCAGAAACCA GGGAAAGCCC CTAAGCTCCT GATCCATTAT GGTTCCGTGT TGCAAAGTGG GGTCCCATCA
CGTTTCAGTG GCAGTGGATC
• G T D F T L T I S S L Q P E D S A T Y Y C Q Q V H Q A P T T F G Q
201 TGGGACAGAT TTCACTCTCA CCATCAGCAG TCTGCAACCT GAAGATTCTG CTACGTACTA CTGTCAACAG GTTCATCAGG CTCCTACGAC GTTCGGCCAA
G T K V E I K R
301 GGGACCAAGG TGGAAATCAA ACGG — SEQ ID NO . : 415
DOM-146 - SEQ ID NO.: 301
D I R M T Q S P S S L S A S V G D R V T I T C R A S Q W I G D S L A -
1 GACATCCGGA TGACCCAGTC TCCATCCTCC CTGTCTGCAT CTGTAGGAGA CCGTGTCACC ATCACTTGCC GGGCAAGTCA GTGGATTGGG GATTCTTTAG - W Y Q Q K P G K A P K L L I Y G I S E L Q S G V P S R F
S G S G S -
101 CGTGGTACCA GCAGAAGCCA GGGAAAGCCC CTAAGCTCCT GATCTATGGT ATTTCCGAGT TGCAAAGTGG GGTCCCATCA. CGTTTCAGTG GCAGTGGATC
G T D F T L T I S S L Q P E D S A T Y Y C Q L S S S M P H T F G Q
201 TGGGACAGAT TTCACTCTCA CCATCAGCAG TCTGCAACCT GAAGATTCTG CTACGTACTA CTGTCAACTG TCTAGTAGTA TGCCTCATAC GTTCGGCCAA
G T K V E I K R 301 GGGACCAAGG TGGAAATCAA ACGG — SEQ ID NO. : 416
DOM-147 - SEQ ID NO. : 302
D I Q M T Q P S S L S A S V G D R V T I T C R A S Q
I E T N L E 1 GACATCCAGA TGACCCAGTC TCCATCCTCC CTGTCTGCAT CTGTAGGAGA CCGTGTCACC ATCACTTGCC GGGCAAGTCA GGAGATTGAG ACGAATTTAG
• W Y Q Q K P G K A P K L I, I Y D S S H L Q S G V P S R F S G S G S - 101 AGTGGTACCA GCAGAAACCA GGGAAAGCCC CTAAGCTCCT GATCTATGAT TCGTCCCATT TGCAAAGTGG GGTCCCATCA
CGTTTCAGTG GCAGTGGATC
• G T D F T L T I S S L Q P E D F A T Y Y C Q Q Y H Q H P P T F G Q
201 TGGGACAGAT TTCACTCTCA CCATCAGCAG TCTGCAACCT GAAGATTTTG CTACGTACTA CTGTCAACAG TATCATCAGA ATCCTCCGAC GTTCGGCCAA
G T K V E I K R
301 GGAACCAAGG TGGAAATCAAACGG — ΞΞQ ID NO. : 417
DOM-149 - SEQ ID NO.: 303
D I Q M T Q S P S S L S A S V G D R V T I T C R A S Q W I G R Q L V -
1 GACATCCAGA TGACCCAGTC TCCATCCTCC CTGTCTGCAT CTGTAGGAGA CCGTGTCACC ATCACTTGCC GGGCAAGTCA GTGGATTGGG AGGCAGTTAG - W Y Q Q K P G K A P K L L I Y G A T E L Q S G V P S R F
S G S G S -
101 TTTGGTACCA GCAGAAACCA GGGAAAGCCC CTAAGCTCCT GATCTATGGG GCGACCGAGT TGCAAAGTGG GGTCCCATCA CGTTTTAGTG GCAGTGGATC
• G T D F T L T I S S L Q P E D F A T Y Y C Q Q Q S K G P L T F G H
201 TGGGACAGAT TTCACTCTCA CCATCAGCAG TCTGCAACCT GAAGATTTTG CTACGTACTA CTGTCAACAG CAGTCGAAGG GTCCTCTTAC
G T K V E I K R
301 GGGACCAAGG TGGAAATCAAACGG — SEQ ID NO. : 418
DOM-150 - SEQ ID NO. : 304
D I Q M T Q S P S S L S A V G D R V T I T C R A S Q T D L N - 1 GACATCCAGA TGACCCAGTC TCCATCCTCC CTGTCTGCAT CTGTAGGAGA CCGTGTCACC ATCACTTGCC GGGCAAGTCA GGGGATTGGT ACTGATTTAa
• B Y Q Q K P G K A P K L L I Y M G S Y L Q S G V P S R F S G S G S - 101 ATTGGTATCA
CGTTTCAGTG
• G T D F T L T I S S L Q P E D F A T Y Y C Q Q I Y S F P I T F G Q
201 TGGGACAGAT TTCCTATTAC
G T K V E I K R
301 GGGACCAAGG TGGAAATCAAACGG — SEQ ID NO. : 419
DOM-154 - SEQ ID NO. : 305
- W Y Q Q K P G K A P K L L I Y F G S L L Q S G V P S R F
S G S R S -
101 ATTGGTATCA GCAGAAACCA GGGAAAGCCC CTAAGCTCCT GATCTATTTT GGTTCCCTGT TGCAAAGTGG GGTCCCATCA CGTTTCAGTG GCAGTAGATC
G T D F T L T I S S L Q P E D F A T Y Y C Q Q H H T R P Y T F G Q
201 TGGGACAGAT TTCACTCTCA CCATCAGCAG TCTGCAACCT GAAGATTTTG CTACGTACTA CTGTCAACAG CATCATACTC GTCCTTATAC
G T K V E I K R 301 GGGACCAAGG TGGAAATCAAACGG — SEQ ID NO. 1 420
DOM-155 - SEQ ID NO.: 306
D I Q M T Q S L S A S V G R V T T C R
M D L E - 1 GACATCCAGA TGACCCAGTC TCCATCCTCC CTGTCTGCAT CTGTAGGAGA CCGTGTCACC ATCACTTGCC GGGCAAGTCA
301 GGGACCAAGG TGGARATCAA ACGG — SΞQ ID NO. : 421
DOM-156 - SEQ ID NO. : 307
- W Y Q Q K P G K A P K L L I Y A A S W L Q S G V P S R F
S G S G S -
V T F G Q
201 TGGGACAGAT TTCACTCTCA CTGTCAACAG TGCCTGTGAC
G T K V E I K R 301 GGGACCAAGG TGGAAATCAA ACGG — SEQ ID NO . I 422
DOM-157 - SEQ ID NO. : 308
D I Q M T Q S P S S L S A S R V T C R A S Q
I G E D L E - 1 GACATCCAGA TGACCCAGTC TCCATCCTCC CTGTCTGCAT CTGTAGGAGA CCGTGTCACC ATCACTTGCC GAGCAAGTCA GAATATTGGG GAGGATTTAG
• W Y Q Q K P G N A p K i L I Y S A S H L Q S G V P S R F S G S G S - 101 AGTGGTACCA GCAGAAACCA GGGAATGCCC CTAAGCTCCT GATCTATAGT GCGTCCCATT TGCAAAGTGG GGTCCCATCA
CGTTTCAGTG GCAGTGGATC
G T D F T L T I S S L Q P E D F A T Y Y C Q Q Y S S Y P V T F G Q
201 TGGGACAGAT TTCACTCTCA CCATCAGCAG TCTGCAACCT GAAGATTTTG CTACGTACTA CTGTCAACAG TATTCTAGTT ATCCTGTTAC GTTCGGCCAA
G T K V E I K R
301 GGGACCAAGG TGGAAATCAA ACGG — SEQ ID NO . : 423
DOM-158 - SEQ ID NO. : 309
D I Q M T Q S P S S L S A S V G D R V T I T C R A S Q P I D E D L E -
1 GACATCCAGA TGACCCAGTC TCCATCCTCC CTGTCTGCAT CCGTAGGAGA CCGTGTCACC ATCACTTGCC GGGCAAGTCA GCCGATTGAT GAGGATTTAG - W Y Q Q K P G N A P K L L l Y S A S Y L Q S G V P S R F
S G S G S -
101 AGTGGTACCA GCAGAAACCA GGGAATGCCC CTAAGCTCCT GATCTATAGT GCGTCCTATT TGCAAAGTGG GGTCCCATCA CGTTTCAGTG GCAGTGGATC
G T D F T L T I S R L Q P E D F A T Y Y C Q Q Y H L L P A T F G Q
201 TGGGACAGAT TTCACTCTCA CCATCAGCAG ACTGCAACCT GAAGATTTTG CTACGTACTA CTGTCAACAG TATCATCTTC TGCCTGCTAC
G T K V E I K R 301 GGGACCAAGG TGGAAATCAA ACGG — SEQ ID NO. : 424
DOM-159 - SEQ ID NO. : 310
D I Q M I Q S P S S L S A S V G D R V T T C R A S
I N E D L E -
301 GGGACCAAGG TGGAAATCAA ACGG — SEQ ID NO. : 425
DOM-160 - SEQ ID NO.: 311
- W Y Q Q K P G K A P K L L I Y H S S E L Q S G V P S R F
S G S G S -
V T F G Q
201 TGGGACAGAT TTCACTCTCA CCATCAGCAG TCTGCAACCT GAAGATTTTG CTACGTACTA CTGTCAACAG TATCATATGT CGCCTGTGAC
G T K V E I K R 301 GGGACCAAGG TGGAAATCAAACGG — SEQ ID NO. Z 426
DOM-I 61 - SEQ ID NO. : 312
D I Q M T Q S S A R V T C R A S Q S D L E -
201 TGGGACAGAT TTCACTCTCA CCATCAGCAG TCTGCAACCT GAAGATTTTG CTACGTACTA CTGTCAACAG TATCATAGTC TGCCTGTTAC
G T K V E I K R
301 GGGACCAAGG TGGAAATCAA ACGG — SEQ ID NO. : 427
DOM-162 - SEQ ID NO. : 313
- W Y Q Q K P G K A P K L L I Y N S S F L Q S G V P S R F
S G S G S -
V T F G Q
201 TGGGGCAGAT CCATCAGCAG TCTGCAACCT GAAGATTTTG CTACGTACTA CTGTCAACAG TGCCTGTTAC GTTCGGCCAA
G T K V E I K R
301 GGGACCAAGG TGGAAATCAA ACGG — SEQ ID NO. : 428
DOM-163 - SEQ ID NO. : 314
D I Q M T Q S P S L S A S V G D R V T I T C R A S Q
I E G N 1 GACATCCAGa TGACCCAGTC GGATATTGAG
• W Y Q Q K P G K A P K L L I Y D S S Q L Q S G V P S R F S G S G S - 101 AGTGGTACCA
CGTTTCAGTG
• G T D F T L T I S S L Q P E D F A T Y Y C Q Q Y H H L P T T F G Q
201 TGGGACAGAT TTCCTACGAC
G T K V E I K R
301 GGGACCAAGG TGGSAATCAA ACGG — SEQ ID NO. : 429
DOM-164 - SEQ ID NO.: 315
D I Q M T Q S P S S L S A S V G D R V T I T C R A S Q S I D T D L E -
1 GACATCCAGA GAGTATTGAT - W Y Q Q K P G K A P K L L I Y D G S W L Q S G V P S R F
S G S G S -
101 AGTGGTATCA CGTTTCAGTG GCAGTGGATC
- G T D F T L T I S S L Q P E D F A T Y Y C Q Q Y R W I P V T F G Q
201 TGGGACAGAT TTTACTCTCA CCATCAGCAG TCTGCAACCT GAAGATTTTG CTACGTACTA CTGTCAACAG TATCGGTGGA TTCCTGTTAC GTTCGGCCAA
G T K V E I K R 301 GGGACCAAGG TGGAAATCAA ACGG — SEQ ID NO. : 430
DOM-165 - SEQ ID NO.: 316
D I Q M T Q S P S S L S A S V G D R V T I T A S T D L E . 1 GACATCCAGA TGACCCAGTC TCCATCCTCC CTGTCTGCAT CTGTAGGAGA CCGTGTCACC ATCACTTGCC GGGCAAGTCA GAGTATTAGT ACTGATTTAG
W Y Q Q K L G K A P K L L I Y D ft S L L Q S G V P S R P S G S G S - 101 AGTGGTACCA GCAGAAACTA GGGAAAGCCC CTAAGCTCCT GATCTATGAT GCTTCCCTTT TGCAAAGTGG GGTCCCATCA
CGTTTCAGTG GCAGTGGATC
- G T D F T L T I S S L Q P E D F A T Y Y C Q Q Y S S L P V T F G Q
201 TGGGACAGAT TTCACTCΓCA CCATCAGCAG TCTGCAACCT GAAGATTTTG CTACGTACTA CTGTCAACAG TATTCGAGTC TGCCTGTTAC GTTCGGCCM
G T K V E I K R
301 GGGACCAAGG TGGAAATCAA ACGG — SEQ ID NO. : 431
DOM-166 - SEQ ID NO. : 317
D I Q M T Q S P S S L S A S V G D R V T I T C R A S Q P I T T S L E -
1 GACATCCAGA TGACCCAGTC TCCATCCTCC CTGTCTGCAT CTGTAGGAGA CCGTGTCACC ATCACTTGCC GGGCAAGTCA GCCTATTACG ACGTCTTTAG - W Y Q Q K P G K A P K L L I Y D A S M L Q S G V P S R F
S G S G S -
101 AGTGGTACCA GCAGAAACCA GGGAAAGCCC CTAAGCTCCT GATCTATGAT GCGTCCATGT TGCAAAGTGG GGTCCCATCA CGTTTCAGTG GCAGTGGATC
- G T D F T L T I S S L Q P E D F A T Y Y C Q Q Y W V T P V T F G Q
201 TGGGACAGAT TTCACTCTCA CCATCAGCAG TCTGCAACCT GAAGATTTTG CTACGTACTA CTGTCAACAG TATTGGGTTA CGCCTGTTAC GTTCGGCCAA
G T K V E I K R 301 GGGACCAAGG TGGAAATCAA ACGG — SEQ ID NO . : 432
DOM-167 - SEQ ID NO. : 318
D I Q M Q S P S S L S A S V G D R V T T C R A S Q
I H T N L E •
301 GGGACCAAGG TGGGAATCAA ACGG — SEQ ID NO. : 433
DOM-168 - SEQ ID NO. : 319
- W Y Q Q K P G K A P K L L I Y D G S M L Q S G V P S R F
S G S G S -
V T F G Q
201 TGGGACAGAT TTCACTCTCA CCATCAGCAG TCTGCAACCT GAAGATTTTG CTACGTACTA CTGTCAACAG TATAGTGTGT CGCCTGTTAC
G T K V E I K R 301 GGGACCAAGG TGGAAATCAA ACGG — SEQ ID NO. '. A3 A
DOM-I 69 - SEQ ID NO. : 320
D I Q M T Q S P S L S A S V G D R V T I T C R A S Q
I D N N L E - 1 GACATCCAGA TGACCCAGTC TCCATCCTCC CTGTCTGCAT CTGTAGGAGA CCGTGTCACC ATCACTTGCC GGGCAAGTCA GAGTATTGAT AATAATTTAG W Y Q Q K P G E A P K L L I Y D G S L L Q S G V P S R F S G S G S - 101 AGTGGTACCA GCAGAAACCA GGGGAAGCCC CTAAGCTCCT GATCTATGAT GGGTCCCTTT TGCAAAGTGG GGTCCCATCA
CGTTTCAGTG GCAGTGGATC
- G T D F T L T I S S L Q P E D F A T Y Y C Q Q Y H L H P V T F G Q
201 TGGGACAGAT TTCACTCTTA CCATCAGCAG TCTGCAACCT GAAGATTTTG CTACGTACTA CTGTCAACAG TATCATCTTC ATCCTGTTAC GTTCGGCCAA
G T K V E I K R
301 GGGACCAAGG TGGAAATCAA ACGG — SΞQ ID NO. : 435
DOM-170 - SEQ ID NO.: 321
D I Q M T Q S P S S L S A S V G D R V T I T C R A S Q D I D T N L E -
1 GACATCCAGA TGACCCAGTC TCCATCCTCC CTGTCTGCAT CTGTAGGAGA CCGTGTCACC ATCACTTGCC GGGCAAGTCA GGATATTGAT ACGAATTTAG - W Y Q Q K P G E A P K L L I Y D R S T L Q S G V P S R F
S G S G S -
101 AGTGGTATCA GCAGAAACCA GGGGAAGCCC CTAAGCTCCT GATCTATGAT CGTTCCACGT TGCAAAGTGG GGTCCCATCA CGTTTCAGTG GCAGTGGATC
- G T D F T L T I S S L Q P E D F A T Y Y C Q Q Y D S Y P V T F G Q
201 TGGGACAGAT TTCACTCTCA CCATCAGCAG TCTGCAACCT GAAGATTTTG CTACGTACTA CTGTCAACAG TATGATTCTT ATCCTGTGAC
G T K V E I K R 301 GGGACCAAGG TGGAAATCAA ACGG — SΞQ ID NO. . 436
DOM-171 - SEQ ID NO. : 322
D I Q M T Q S P S L S A S V G D R V T T C R A S Q S N L E -
301 GGGACCAAGG TGGMATCAA ACGG — SEQ ID NO. : 437
DOM-172 - SEQ ID NO.: 323
- W Y Q Q K P G K A P K L L I Y L S S R L Q S G V P S R F
S G S G S -
Y T F G Q
201 TGGGACAGAT TTTACTCTCA CCATCAGCAG TCTGCAACCT GAAGATTTTG CTACGTACTA CTGTCAACAG CTGAAGAAGC CTCCTTATAC
G T K V E I K R 301 GGGACCAAGG TGGAAATCAA ACGG — SEQ ID NO. I 438
DOM-173 - SEQ ID NO.: 324
D I Q M Q S P S S L S A S V G D R V T I T C R A S Q
I K N R L A - 1 GACATCCAGA TGACCCAGTC TCCATCCTCC CTGTCTGCAT CTGTAGGAGA CCGTGTCACC ATCACTTGCC GGGCAAGTCA GAAGATTAaG AATCGGTTAG
• W Y Q Q K P G K A P K L L I Y E V S H L Q S G V P S R F S G S G S - 101 CGTGGTACCA GCAGAAACCA GGGAAAGCCC CTAAGCTCCT GATCTATGAG GTTTCCCATT TGCAAAGTGG GGTCCCATCA
CGTTTCAGTG GCAGTGGATC
G T D F T L T I G S L Q P E D F A T Y Y C Q Q R R Q S P Y T F G Q
201 TGGGACAGAT TTCACTCTCA CCATCGGCAG TCTGCAACCT GAAGATTTTG CTACGTACTA CTGTCAACAG AGGAGGCAGT CGCCTTATAC GTTCGGCCAA
G T K V E I K R
301 GGGACCAAGG TGGARATCAAACGG — SEQ ID NO. : 439
DOM-174 - SEQ ID NO.: 325
D I Q M T Q S P S S L S A S V G D R V T I T C R A S E D I G E E L F -
1 GACATCCAGA TGACCCAGTC TCCATCCTCC CTGTCTGCAT CTGTAGGAGA CCGTGTCACC ATCACTTGCC GGGCAAGTGA GGATATTGGG GAGGAGTTAT - W Y Q Q E P G K A P K L L I Y S A S T L Q S E V P S R F
E G S G S -
101 TTTGGTATCA GCAGAAACCA GGGAAAGCCC CTAAGCTCCT GATCTATTCG GCGTCCACGT TGCAAAGTGA GGTCCCATCA CGTTTCAGTG GCAGTGGATC
G T D F T L T I S S L Q H E D F A T Y Y C Q Q V Y E W P Y T F G Q
201 TGGGACAGAT TTCACTCTCA CCATCAGCAG TCTGCAACAT GAAGATTTTG CTACGTACTA CTGTCARCAG GTTTATGAGT GGCCTTATAC GTTCGGCCAA
G T K V E I K R 301 GGGACCAAGG TGGAAATCAA ACGG — SEQ ID NO. : 440
DOM-175 - SEQ ID NO. : 326
D I Q M T Q S P S S L S A S V G D R V T I T C R A S Q P I S G G L R -
1 GACATCCAGA TGACCCAGTC TCCATCCTCC CTGTCTGCAT CTGTAGGAGA CCGTGTCACC ATCACTTGCC GGGCAAGTCA GCCTATTTCT GGGGGTTTAA
301 GGGACCAAGG TGGAAATCAA ACGG — SEQ ID NO. : 441
DOM-176 - SEQ ID NO.: 327
E V Q L L E S G G G L V Q P G G S L R L S C A A S G F T F D A Y E M -
101 TGGGGTGGGT
GCAGACTCCG
- F T I S R D N S K N T L Y L Q M H S L R A E D T A V Y Y C A K P G
201 GTTCACCATC ATTACTGTGC
D N V G I F D Y W G Q G T L V T V S S
301 GATAATGTTG GTATTTTTGA CTACTGGGGT CAGGGAACCC TGGTCACCGT CTCGAGC — SEQ ID NO . : 442
DOM-177 - SEQ ID NO.: 328
CACCTTTAGT • V W V R Q A P G K G L E W V S A I D E W G F A T Y Y A D S V K G R -
101 TGGTGTGGGT CCGCCAGGCT CCAGGGAAGG GTCTAGAGTG GGTCTCAGCG ATTGATGAGT GGGGTTTTGC GACATACTAC GCAGACTCCG TGAaGGGCCG - F T I S R D N S K N T L Y L Q M H S L R A E D T A V Y Y
C A K H W
201 GTTCACCATC TCCCGCGACA ATTCCAAGAA CACGCTGTAT CTGCAAATGA ACAGCCTGCG TGCCGAGGAC ACCGCGGTAT ATTACTGTGC GAAACATTGG
E F T S D T S R F D Y W G Q G T L V T V S S 301 GAGTTTACGT CTGATACGTC GCGTTTTGAC TACTGGGGTC AGGGAACCCT GGTCACCGTC TCGAGC — SEQ ID
NO.: 443
DOM-178 - SEQ ID NO. : 329 E V Q L L E S G G G L V Q P G G S L R L S C A A S G F T
F E D F D M -
1 GAGGTGCAGC TGTTGGAGTC TGGGGGAGGC TTGGTACAGC CTGGGGGGTC CCTGCGTCTC TCCTGTGCAG CCTCCGGATT CACCTTTGAG GATTTTGATA
- A W V R Q A P G K G L E W V S S I N D Q G S L T Y Y A D S V K G R -
101 TGGCTTGGGT CCGCCAGGCT CCAGGGAAGG GTCTAGAGTG GGTCTCATCT ATTAATGATC AGGGTTCTCT GACATACTAC GCAGACTCCG TGAAGGGCCG
- F T I S R D N S K N T L Y L Q M N S L R A E D T A V Y Y C A K P D 201 GTTCACCATC TCCCGCGACA ATTCCAAGAA CACGCTGTAT CTGCAAATGA ACAGCCTGCG TGCCGAGGAC ACCGCGGTAT
ATTACTGTGC GAAACCGGAT
Q F F D Y W G Q G T L V T V S S
301 CAGTTTTTTG ACTACTGGGG TCAGGGAACC CTGGTCACCG TCΓCGAGC — SEQ ID NO. : 444
DOM-179 - SEQ ID NO.: 330
E V Q L L E S G G G L V Q P G G S L R L S C A A S G F T F S A Y D M -
1 GAGGTGCAGC TGTTGGAGTC TGGGGGAGGC TTGGTACAGC CTGGGGGGTC CCTGCGTCTC TCCTGTGCAG CCTCCGGATT CACCTTTAGT GCTTATGATA M W V R Q A P G K G L E W V S R I S P Q G Q R T Y Y A D S V K G R -
101 TGATGTGGGT CCGCCAGGCT CCAGGGAAGG GTCTAGAGTG GGTCTCACGG ATTAGTCCTC AGGGTCAGCG GACATACTAC GCAGACTCCG TGAAGGGCCG - F T I S R D N S K H T L Y L Q M N S L R A E D T A V Y Y
C A K I R
201 GTTCACCATC TCCCGCGACA ATTCCAAGAA CACGCTGTAT CTGCAAATGA ACAGCCTGCG TGCCGAGGAC ACCGCGGTAT ATTACTGTGC GAAAATTCGT
G Q S R I P M R F D Y W G Q G T L V 301 GGGCAGTCGc GGATTCCTAT GAGGTTTGAC TACTGGGGTC AGGGAACCCT GGTC — SEQ ID NO. : 445
DOM-180 - SEQ ID NO.: 331
E V Q L L E S G G G L V Q P G G S L R L S C A A S G F T F T D Y E M -
1 GAGGTGCAGC TGTTGGAGTC TGGGGGAGGC TTGGTACAGC CTGGGGGGTC CCTGCGTCTC TCCTGTGCAG CCTCCGGATT CACCTTTACG GATTATGAGA
• G W V R Q A P G K G L E W V S T I T S L G E S T Y Y A D S V K G R - 101 TGGGGTGGGT CCGCCAGGCT CCAGGGAAGG GTCTAGAGTG GGTCTCAACG ATTACTAGTT TGGGTGAGAG TACATACTAC
GCAGACTCCG TGAAGGGCCG
- F T I S R D N S K N T L Y L Q M N S L R A E D T A V Y Y C A K P G
201 GTTCACCATC TCCCGCGACA ATTCCAAGAA CACGCTGTAT CTGCAAATGA ACAGCCTGCG TGCCGAGGAC ACCGCGGTAT ATTACTGTGC GAAACCTGGT
R I F D Y W G Q G T L V T V S S
301 CGTATTTTTG ACTACTGGGG TCAGGGAACC CTGGTCACCG TCTCGAGC — SΞQ ID NO. : 446
DOM- 181 - SEQ ID NO. : 332
E V Q L L E S G G G L V Q P G G S L R L S C A A S G F T F A F Y P M -
1 GAGGTGCAGC TGTTGGAGTC TGGGGGAGGC TTGGTACAGC CTGGGGGGTC CCTGCGTCTC TCCTGTGCAG CCTCCGGATT CACCTTTGCT TTTTATCCTA • M W V R Q A P G K G L E W V S W I D A T G T K T Y Y A D S V K G R -
101 TGATGTGGGT CCGCCAGGCT CCAGGGAAGG GTCTAGAGTG GGTCTCATGG ATTGATGCTA CGGGTACGAG GACATACTAC GCAGACTCCG TGAAGGGCCG - F T I S R D K S K N T L Y L Q M N S L R A E D T A V Y Y
C A E G N
201 GTTCACCATC TCCCGCGACA ATTCCAAGAA CACGCTGTAT CTGCAAATGA ACAGCCTGCG TGCCGAGGAC ACCGCGGTAT ATTACTGTGC GGAAGGTAAT
Y G S S Y T M G V F D Y W G Q G T L V T V S S 301 TATGGGAGTT CGTATACTAT GGGGGTTTTT GACTACTGGG GTCAGGGAAC CCTGGTCACC GTCTCGAGC ~ SEQ XD
NO.: 447
DOM-182 - SEQ ID NO.: 333 E V Q L L E S G G G L V Q P G G S L R L S C A A S G F T
F D E Y P M -
1 GAGGTGCAGC TGTTGGAGTC TGGGGGAGGC TTGGTACAGC CTGGGGGGTC CCTGCGTCTC TCCTGTGCAG CCTCCGGATT CACCTTTGAT GAGTATCCGA
- Y W V R Q A P G K G L E W V S S I G P S G P N T Y Y A D S V K G R -
101 TGTATTGGGT CCGCCAGGCT CCAGGGAAGG GTCTAGAGTG GGTCTCATCG ATTGGTCCTT CTGGTCCGAA TACATACTAC GCAGACTCCG TGAAGGGCCG
- F T I S R D N S K N T L Y L Q M N S L R A E D T A V Y Y C A K S P 201 GTTCACCATC TCCCGCGACA ATTCCAAGAA CACGCTGTAT CTGCAAATGA ACAGCCTGCG TGCCGAGGAC ACCGCGGTAT
ATTACTGTGC GAAATCTCCG
Y F D V I P S Y F D Y W G Q G T L V T V S S
301 TATTTTGATG TTATTCCTAG TTATTTTGAC TACTGGGGTC AGGGAACCCT GGTCACCGTC TCGAGC~ SΞQ ID NO.: 448
DOM-183 - SEQ ID NO.: 334
E V Q L L E S G G G L V Q P G G S L R L S C A A S G F T F A D Y G M - 1 GAGGTGCAGC TGTTGGAGTC TGGGGGAGGC TTGGTACAGC CTGGGGGGTC CCTGCGTCTC TCCTGTGCAG CCTCCGGATT
CACCTTTGCG GATTACGGTA • G W V R Q A P G K G L E W V S S I Q S S G L R T Y Y A D S V K G R -
101 TGGGTTGGGT CCGCCAGGCT CCAGGGAftGG GTCTAGAGTG GGTCTCATCT ATTCAGTCGT CGGGTTTGCG GACATACTAC GCAGACTCCG TGARGGGCCG - F T I S R D N S K N T L Y L Q M N S L R A E D T A V Y Y
C A K R A
201 GTTCACCATC TCCCGCGACA ATTCCAAGAa CACGCTGTAT CTGCAAATGA ACAGCCTGCG TGCCGAGGAT ACCGCGGTAT ATTACTGTGC GAAACGGGCT
N S R R G F D Y W G Q G T L V T V S S 301 AATTCTCGTA GGGGTTTTGA CTACTGGGGT CAGGGAACCC TGGTCACCGT CTCGAGC — SEQ ID NO . :
449
DOM-184 - SEQ ID NO. : 335 E V Q L L E S G G G L V Q P G G S L R L S C A A S G F T
F S D Y E M -
1 GAGGTGCAGC TGTTGGAGTC TGGGGGAGGC TTGGTACAGC CTGGGGGGTC CCTGCGTCTC TCCTGTGCAG CCTCCGGATT CACCTTTTCT GATTATGAGA
- M W V R Q A P G K G L E W V S S I T S H G G S T Y Y A D S V K G R -
101 TGATGTGGGT CCGCCAGGCT CCAGGGA&GG GTCTAGAGTG GGTCTCATCT ATTACTAGTC ATGGTGGGTC TACATACTAC GCAGACTCCG TGAAGGGCCG
- F T I S R D N S K K T L Y L Q M N S L R A E D T A V Y Y C A K P D 201 GTTCACCATC TCCCGCGACA ATTCCAAGAA CACGCTGTAT CTGCAAATGA ACAGCCTGCG TGCCGAGGAT ACCGCGGTAT
ATTACTGTGC GAAACCTGAT
K D F D Y W G Q G T L V T V S S
301 AAGGATTTTG ACTACTGGGG TCAGGGA&CC CTGGTCACCG TCTCGAGC — SEQ ID NO. : 450
DOM-185 - SEQ ID NO. : 336
E V Q L L E S G G G L V Q P G G S L R L S C A A S G F T F A H Y P M -
1 GAGGTGCAGC TGTTGGAGTC TGGGGGAGGC TTGGTACAGC CTGGGGGGTC CCTGCGTCTC TCCTGTGCAG CCTCCGGATT CACCTTTGCG CATTATCCGA S B V R Q A P G K G L E W V S S I G R L G N R T Y Y A D S V K G R -
101 TGTCGTGGGT CCGCCAGGCT CCMGGMGG GTCTAGAGTG GGTCTCATCG ATTGGTAGGC TGGGTAATCG TACATACTAC GCAGACTCCG TGAAGGGCCG - F T I S R D N S K N T L Y L Q M N S L R A E D T A V Y Y
C A K R A
201 GTTCACCATC TCCCGCGACA ATTCCAAGAA CACGCTGTAT CTGCAAATGA ACAGCCTGCG TGCCGAGGAC ACCGCGGTAT ATTACTGTGC GAAACGTGCT
T P V P I K G L F D Y W G Q G T L V T V S S 301 ACGCCTGTGC CGATTAAGGG TTTGTTTGAC TACTGGGGTC AGGGAACCCT GGTCACCGTC TCGAGC — SEQ ID
NO.: 451
DOM-186 - SEQ ID NO. : 337 E V Q L L E S G G G L V Q P G G S L R L S C A A S G L T
F G R Y E M -
1 GAGGTGCAGC TGTTGGAGTC TGGGGGAGGC TTGGTACAGC CTGGGGGGTC CCTGCGTCTC TCCTGTGCAG CCTCCGGACT CACCTTTGGG AGGTATGAGA
- A W V R Q A P G K G L E W V S S I D S D G W V T Y Y A D S V K G R -
101 TGGCGTGGGT CCGCCAGGCT CCAGGGAAGG GTCTAGAGTG GGTCTCAAGT ATTGATTCGG ATGGTTGGGT GACATACTAC GCAGACTCCG TGAAGGGCCG
- F T I S R D N S K N T L Y L Q M N S L R A E D T A V Y Y C A Q P D 201 GTTCACCATC TCCCGCGACA ATTCCAAGAA CACGCTGTAT CTGCAAATGA ACAGCCTGCG TGCCGAGGAC ACCGCGGTAT
ATTACTGTGC GCAACCGGAT
S L F D Y W G Q G T L V T V S S
301 TCGTTGTTTG ACTACTGGGG TCAGGGAACC CTGGTCACCG TCTCGAGC — SΞQ ID NO . : 452
DOM-187 - SEQ ID NO. : 338
E V Q L L E S G G G L V Q P G G S L R L S C A A S G F T F S S Y S M -
1 GAGGTGCAGC TGTTGGAGTC TGGGGGAGGC TTGGTACAGC CTGGGGGGTC CCTGCGTCTC TCCTGTGCAG CCTCCGGATT CACCTTΓTCT AGTTATTCTA • V W V R Q A P G K G L E W V S G I N R G G T R T Y Y A D S V K G R -
101 TGGTGTGGGT CCGCCAGGCT CCAGGGAAGG GTCTAGAGTG GGTCTCAGGT ATTAATCGGG GTGGTACTCG TACATACTAC GCAGACTCCG TGAAGGGCCG - F T I S R D N S K M T L Y L Q M N S L R A E D T A V Y Y
C A K G W
201 GTTCACCATC TCCCGCGACA ATTCCAAGAA CACGCTGTAT CTGCAAATGA ACAGCCTGCG TGCCGAGGAC ACCGCGGTAT ATTACTGTGC GAAAGGTTGG
R R G F D Y W G Q G T L V T V S S 301 AGGAGGGGGT TTGACTACTG GGGTCAGGGA ACCCTGGTCA CCGTCTCGAG C — SEQ ID NO. : 453
DOM-188 - SEQ ID NO. : 339
E V Q L L E S G G G L V Q P G G S L R L S C A A S G F T F T R Y R M -
1 GAGGTGCAGC TGTTGGAGTC TGGGGGAGGC TTGGTACAGC CTGGGGGGTC CCTGCGTCTC TCCTGTGCAG CCTCCGGATT CACCTTTACG CGTTATAGGA
S W V R Q A P G K G L E W V S G I S R D G Y R T Y Y A D S V K G R - 101 TGTCTTGGGT CCGCCAGGCT CCAGGGAAGG GTCTGGAGTG GGTCTCAGGG ATTTCGAGGG STGGTTATCG GACATACTAC
GCAGACTCCG TGAAGGGCCG
F T I S R D N S K N T L Y L Q M N S L R A E D T A V Y Y C A K G M
201 GTTCACCATC TCCCGCGACA ATTCCAAGAA CACGCTGTAT CTGCAAATGA ACAGCCTGCG TGCCGAGGAC ACCGCGGTAT ATTACTGTGC GAAAGGTATG
T A S F D Y W G Q G T L V T V S S
301 ACTGCGTCGT TTGACTACTG GGGTCAGGGA ACCCTGGTCA CCGTCTCGAG C — SEQ ID NO. : 454
DOM-189 - SEQ ID NO.: 340
E V Q L L E S G G G L V Q P G G S L R L S C A A S G F T F Q M Y P M -
1 GAGGTGCAGC TGTTGGAGTC TGGGGGAGGC TTGGTACAGC CTGGGGGGTC CCTGCGTCTC TCCTGTGCAG CCTCCGGATT CACCTTTCAG ATGTATCCGA G W V R Q A P G K G L E W V S M I E P A G D L T Y Y A D S V K G R -
101 TGGGGTGGGT CCGCCAGGCT CCAGGGAAGG GTCTAGAGTG GGTCTCAATG ATTGAGCCGG CTGGTGATCT TACATACTAC GCAGACTCCG TGAAGGGCCG - F T I S R D N S K N T L Y L Q M N S L R A E D T A V Y Y
C A K Y Q
201 GTTCACCATC TCCCGCGACA ATTCCAAGAA CACGCTGTAT CTGCAAATGA ACAGCCTGCG TGCCGAGGAC ACCGCGGTAT ATTACTGTGC GASATATCAG
E O P W F D Y W G Q G T L V T V S S 301 GAGCAGCCTT GGTTTGACTA CTGGGGTCAG GGAACCCTGG TCACCGTCTC GAGC — SEQ ID NO. : 455
DOM-190 - SEQ ID NO.: 341
E V Q L L E S G G G L V Q P G G S L R L S C A A S G F T F S M Y D M -
1 GAGGTGCAGC TGTTGGAGTC TGGGGGAGGC TTGGTACAGC CTGGGGGGTC CCTGCGTCTC TCCTGTGCAG CCTCCGGATT CACCTTTAGT ATGTATGATA
H W V R Q A P G K G L E W V S T I L S D G T D T Y Y A D S V K G R - 101 TGCATTGGGT CCGCCAGGCT CCAGGGAAGG GTCTAGAGTG GGTCTCAACG ATTTTGTCTG ATGGTACGGA TACATACTAC
GCAGACTCCG TGAAGGGCCG
- F T I S R D N S K N T L Y L Q M N S L R A E D T A V Y Y C A K Y G
201 GTTCACCATC TCCCGCGACA ATTCCAAGAA CACGCTGTAT CTGCAAATGA ACAGCCTGCG TGCCGAGGAC ACCGCGGTAT ATTACTGTGC GAAATATGGG
A M F D Y W G Q G T L V T V S S
301 GCTATGTTTG ACTACTGGGG TCAGGGAACC CTGGTCACCG TCTCGAGC — SEQ ID NO. : 456
DOM-191 - SEQ ID NO.: 342
E V Q L L E S G G G L V Q P G G S L R L S C A A S G F T F K L Y P M -
1 GAGGTGCAGC TGTTGGAGTC TGGGGGAGGC TTGGTACAGC CTGGGGGGTC CCTGCGTCTC TCCTGTGCAG CCTCCGGATT CACCTTTAAG TTGTATCCGA
301 TGGGATTATc TGTTTGACTA CTGGGGTCAG GGAACCCTGG TCACCGTCTC GAGC — SEQ ID NO. : 457
DOM-I 92 - SEQ ID NO. : 343
E V Q L L E S G G G L V Q P G G S L R L S C A A S G F T F S R Y P M -
101 TGAGTTGGGT
GCAGACTCCG TGAAGGGCCG
- F T I S R D N S K N T L Y L Q M N S L R A E D T A V Y Y C A E G H
201 GTTCACCATC TCCCGCGACA ATTCCAAGAA CACGCTGTAT CTGCAAATGA ACAGCCTGCG TGCCGAGGAC ACCGCGGTAT ATTACTGTGC GGAAGGGCAT
Q A P F D Y W G Q G T L V T V S S
301 CAGGCGCCGT TTGACTACTG GGGTCAGGGAACCCTGGTCA CCGTCTCGAG C — SEQ ID NO. : 458
DOM-193 - SEQ ID NO.: 344
• S W V R Q A P G K G L E W V S T I N A N G I R T Y Y A D S V K G R -
101 TGTCTTGGGT CCGCCAGGCT CCAGGGAAGG GTCTAGAGTG GGTCTCAACT ATTAATGCGA ATGGTATTCG GACATACTAC GCCGACTCCG TGAAGGGCCG - F T I S R D N S K N T L Y L Q M H G L R A E D T A V Y Y
C A K G G
201 GTTCACCATC TCCCGCGACA ATTCCAAGAA CACGCTGTAT CTGCAAATGA ACGGCCTGCG TGCCGAGGAC ACCGCGGTAT ATTACTGTGC GAAAGGGGGG
V W R W G T G H K F D Y W G Q G T L V T V S S 301 GTTTGGAGGT GGGGGACTGG GCATAAGTTT GACTACTGGG GTCAGGGAAC CCTGGTCACC GTCTCGAGC — SEQ ID
NO.: 459
DOM-194 - SEQ ID NO. : 345 E V Q L L E S G G G L V Q P G G S L R L S C A A S G F T
F K Q Y D M -
1 GAGGTGCAGC TGTTGGAGTC TGGGGGAGGC TTGGTACAGC CTGGGGGGTC CCTGCGTCTC TCCTGTGCAG CCTCCGGATT CACCTTTAAG CAGTATGATA
- R W V R Q A P G K G L E W V S T I S Q N G T K T Y Y A D S V K G R -
101 TGCGGTGGGT CCGCCAGGCT CCAGGGAAGG GTCTAGAGTG GGTCTCAACG ATTTCGCAGA ATGGTACTAA GACATACTAC GCAGACTCCG TGAAGGGCCG
- F T I S E D N S K N T L Y L Q M N S L R A E D T A V Y Y C A K S R 201 GTTCACCATC TCCCGCGACA ATTCCAAGAA CACGCTGTAT CTGCAAATGA ACAGCCTGCG TGCCGAGGAC ACCGCGGTAT
ATTACTGTGC GARATCGAGG
T G R Y F D Y W G Q G T L V T V S S
301 ACTGGTAGGT ATTTTGACTA CTGGGGTCAG GGAACCCTGG TCACCGTCTC GAGC — SEQ ID NO. : 460
DOM-195 - SEQ ID NO. : 346
E V Q L L E S G G G L V Q P G G S L R L S C A A S G F T F G T Y D M -
1 GAGGTGCAGC TGTTGGAGTC TGGGGGAGGC TTGGTACAGC CTGGGGGGTC CCTGCGTCTC TCCTGTGCAG CCTCCGGATT CACCTTTGGG ACTTATGATA • G W V R Q A P G K G L E W V S R I N W Q G D R T Y Y A D S V K G R -
101 TGGGGTGGGT CCGCCAGGCT CCAGGGAAGG GTCTAGAGTG GGTCTCAAGG ATTAATTGGC AGGGTGATCG TACATACTAC GCAGACTCCG TGAAGGGCCG - F T I S R D N S K N T L Y L Q M N S L R A E D T A V Y Y
C A K A G
201 GTTCACCATC TCCCGCGACA ATTCCAAGAR CACGCTGTAT CTGCAAATGA ACAGCCTGCG TGCCGAGGAC ACCGCGGTAT ATTACTGTGC GAAAGCGGGG
F G H Y V D G L G F D Y W G Q G T L V T V S S 301 TTTGGTCATT ATGTTGATGG TCTTGGGTTT GACTACTGGG GTCAGGGAAC CCTGGTCACC GTCTCGAGC — SEQ ZD
NO. : 461
DOM-196 - SEQ ID NO. : 347 E V Q L L E S G G G L V Q P G G S L R L S C A A S G F T
F S G Y E M -
1 GAGGTGCAGC TGTTGGAGTC TGGGGGAGGC TTGGTACAGC CTGGGGGGTC CCTGCGTCTC TCCTGTGCAG CCTCCGGATT CACCTTTAGT GGGTATGAGA
- A W V R Q A P G K G L E W V S S I T D M G D S T Y . Y A D S V K G R -
101 TGGCGTGGGT CCGCCAGGCT CCAGGGAAGG GTCTAGAGTG GGTCTCATCT ATTACTGATA TGGGTGATTC TACATACTAC GCAGACTCCG TGAAGGGCCG
- F T I S R D N S K N T L Y L Q M N S L R A E D T A V Y Y C A K P G 201 GTTCACCATC TCCCGCGACA ATTCCAAGAA CACGCTGTAT CTGCAAATGA ACAGCCTGCG TGCCGAGGAC ACCGCGGTAT
ATTACTGTGC GAAACCTGGG
T A F D Y W G P G T L V T V S S
301 ACTGCGTTTGACTACTGGGG TCCGGGAACC CTGGTCACCG TCTCGAGC — SEQ ID NO. : 462
DOM-197 - SEQ ID NO.: 348
E V Q L L E S G G G L V Q P G G S L R l S C A A S G F T F A K Y K M -
1 GAGGTGCAGC TGTTGGAGTC TGGGGGAGGC TTGGTACAGC CTGGGGGGTC CCTGCGTCTC TCCTGTGCAG CCTCCGGATT CACCTTTGCG AAGTATAAGA W W V R Q A P G K G L E W V S S I T P K G H S T Y Y A D S V K G R -
101 TGTGGTGGGT CCGCCftGGCT CCAGGGAAGG GTCTAGAGTO GGTCTCAAGT ATTACTCCGA AGGGTCATTC TACATACTAC GCAGACTCCG TGAAGGGCCG - F T - I S R D N E K N T L Y L Q M N S L R A E D T A V Y Y
C A K R P
201 GTTCACCATC TCCCGCGACA ATTCCAAGAA CACGCTGTAT CTGCAAATGA ACAGCCTGCG TGCCGAGGAT ACCGCGGTAT ATTACTGTGC GARAAGGCCG
M T P F D Y W G Q G T L V T V S S 301 ATGACTCCGT TTGACTACTG GGGTCAGGGA ACCCTGGTCA CCGTCTCGAG C — SEQ ID NO. : 463
DOM-198 - SEQ ID NO.: 349
E V Q L L E S G G G L V Q P G G S L R L S C A A S G F T F E R Y N M -
1 GAGGTGCAGC TGTTGGAGTC TGGGGGAGGC TTGGTACAGC CTGGGGGGTC CCTGCGTCTC TCCTGTGCAG CCTCCGGATT CACCTTTGAG CGGTATAATA
S W V R Q A P G K G L E VJ V S S I R P R G G K T Y Y A D S V K G R - 101 TGTCTTGGGT CCGCCAGGCT CCAGGGAAGG GTCTAGAGTG GGTCTCATCT ATTCGGCCGC GGGGTGGGAA GACATACTAC
GCAGACTCCG TGAAGGGCCG
• F T I S R D N S K N T L Y L Q M N S L R A E D T A V Y Y C A K W R
201 GTTCACCATC TCCCGCGACA ATTCCAAGAA CACGCTGTAT CTGCAAATGA ACAGCCTGCG TGCCGAGGAC ACCGCGGTAT ATTACTGTGC GAAATGGCGG
R E G Y T G S K F D Y W G Q G T' L V T V S S
301 CGGGAGGGGT ATACTGGTTC TAAGTTTGAC TACTGGGGTC AGGGAACCCT GGTCACCGTC TCGAGC — SEQ ID NO.: 464
DOM-199 - SEQ ID NO. : 350
E V Q L L E S G G G L V Q P G G S L R L S C A A S G F T F E R Y G M -
1 GAGGTGCAGC TGTTGGAGTC TGGGGGAGGC TTGGTACAGC CTGGGGGGTC CCTGCGTCTC TCCTGTGCAG CCTCCGGATT CACCTTTGAG AGGTATGGTA • T W V R Q A P G K G L E W V S S I W P R G Q K T Y Y A D S V K G R -
101 TGACTTGGGT CCGCCAGGCT CCAGGGAAGG GTCTGGAGTG GGTCTCAAGT ATTTGGCCGA GGGGTCAGAA GACATACTAC GCAGACTCCG TGAAGGGCCG F T I S R D N S K N T L Y L Q M H S L R A E D T A V Y Y
C A K G N
201 GTTCACCATC TCCCGCGACA ATTCCAAGAA CACGCTGTAT CTGCAAATGA ACAGCCTGCG TGCCGAGGAC ACCGCGGTAT ATTACTGTGC GAAAGGGAAT
S R Y V F D Y W G Q G T L V T V S S 301 AGTCGGTATG TTTTTGACTA CTGGGGTCAG GGAACCCTGG TCACCGTCTC GAGC — SEQ XD NO. : 465
DOM-200 - SEQ ID NO. : 351
E V Q L L E S G G G L V Q P G G S L R L S C A A S G F T F T N Y S M -
1 GAGGTGCAGC TGTTGGAGTC TGGGGGAGGC TTGGTACAGC CTGGGGGGTC CCTGCGTCTC TCCTGTGCAG CCTCCGGATT CACCTTTACT AATTATAGTA G W V R Q A P G K G L E W V S T I R P N G T K T Y Y A D S V K G R - 101 TGGGTTGGGT CCGCCAGGCT CCAGGGAAGG GTCTAGAGTG GGTCTCAACT ATTCGTCCTA ATGGTACTAA GACATACTAC GCAGACTCCG TGAAGGGCCG
- F T I S R D N S K N T L Y L Q M N S L R A E D T A V Y Y C A K R S
201 GTTCACCATC TCCCGCGACA ATTCCAAGAA CACGCTGTAT CTGCAAATGA ACAGCCTGCG TGCCGAGGAT ACCGCGGTAT ATTACTGTGC GAAACGGTCG
S A H L Q R F D Y W G Q G T L V T V S S
301 TCTGCGCATC TTCAGAGGTT TGACTACTGG GGTCAGGGAA CCCTGGTCAC CGTCTCGAGC — SEQ ID NO . : 466
DOM-201 - SEQ ID NO.: 352
E V Q L L E S G G G L V Q P G G S L R L S C A A E G F T F G N Y S M -
1 GAGGTGCAGC TGTTGGAGTC TGGGGGAGGC TTGGTACAGC CTGGGGGGTC CCTGCGTCTC TCCTGTGCAG CCTCCGGATT CACCTTTGGT AATTATTCGA • G W V R Q A P G K G L E W V S S I G R H G G R T Y Y A D S V K G R -
101 TGGGTTGGGT CCGCCAGGCT CCAGGGAAGG GTCTAGAGTG GGTCTCATCG ATTGGTCGTC ATGGTGGGCG TACATACTAC
GCAGACTCCG TGAAGGGCCG - F T I S R D N S K K T i Y L Q M N S L R A E D T A V Y Y
C A K K G
201 GTTCACCATC TCCCGCGACA ATTCCAAGAA CACGCTGTAT CTGCAAATGA ACAGCCTGCG TGCCGAGGAC ACCGCGGTAT ATTACTGTGC GAAAAAGGGG
S T Y P R F D Y W G Q G T L V T V S S 301 AGTACTTATc CTAGGTTTGA CTACTGGGGT CAGGGAACCC TGGTCACCGT CTCGAGC — SEIQ ID NO. :
467
DOM-202 - SEQ ID NO.:. 353 E V Q L L E Ξ G G G L V Q P G G S L R L S C T A S G F T
F S H Y E M -
1 GAGGTGCAGC TGTTGGAGTC TGGGGGAGGC TTGGTACAGC CTGGGGGGTC CCTGCGTCTC TCCTGTACAG CCTCCGGATT CACCTTTTCG CATTATGAGA
- G W V R Q A P G K G L E W V S S I E P F G G G T Y Y A D S V K G R -
101 TGGGGTGGGT CCGCCAGGCT CCAGGGAAGG GTCTAGAGTG GGTCTCAAGT ATTGAGCCTT TTGGTGGTGG GACATACTAC
GCAGACTCCG TGAAGGGCCG
- F T I S R D N S K N T L Y L Q M N S L R A E D T A V Y Y C A K V Y 201 GTTCACCATC TCCCGCGACA ATTCCAAGAA CACGCTGTAT CTGCA&ATGA ACAGCCTGCG TGCCGAGGAT ACCGCGGTAT
ATTACTGTGC GAAAGTGTAT
P Q G S F D Y W G Q G T L V T V S S
301 CCTCAGGGTT CTTTTGACTA CTGGGGTCAG GGAACCCTGG TCACCGTCTC GAGC — SEQ ID NO. : 468
DOM-203 - SEQ ID NO. : 354
E V Q L L E S G G G L V Q P G G S L R L S C A A S G F T F S N Y T M
1 GAGGTGCAGC TGTTGGAGTC TGGGGGAGGC TTGGTACAGC CTGGGGGGTC CCTGCGTCTC TCCTGTGCAG CCTCCGGATT CACCTTTAGT AATTATACTA • G W V R Q A P G K G L E B V S S I R P D G K I T Y Y A D S V K G R -
101 TGGGGTGGGT CCGTCAGGCT CCAGGGAAGG GTCTAGAGTG GGTCTCATCT ATTCGGCCTG ATGGTAAGAT TACATACTAC GCAGACTCCG TGARGGGCCG - F T I E R D N S K N T L Y L Q M N S L R A E D T A V Y Y
C A E V Y
201 GTTCACCATC TCCCGCGACA ATTCCAAGSA CACGCTGTAT CTGCAAATGA ACAGCCTGCG TGCCGAGGAC ACCGCGGTAT ATTACTGTGC GGAAGTTTAT
S S C A M C T P L L F D Y W G Q G T L V T V S S 301 TCTTCGTGTG CGATGTGTAC TCCGCTTTTG TTTGACTACT GGGGTCAGGG AACCCTGGTC ACCGTCTCGA GC — SΞQ
ID NO. : 469
DOM-204 - SEQ ID NO. : 355 E V Q l L E S G G G L V Q P G G S L R L S C A Λ S G F T
F K R Y S M -
1 GAGGTGCAGC TGTTGGAGTC TGGGGGGGGC TTGGTACAGC CTGGGGGGTC CCTGCGTCTC TCCTGTGCAG CCTCCGGATT CACCTTTAAG CGGTATTCGA
- A W V R Q A P G K G L E W V S D I G P B G F S T Y Y A D S V K G R -
101 TGGCGTGGGT CCGCCAGGCT CCAGGGAAGG GTCTAGAGTG GGTCTCAGAT ATTGGGCCG& GGGGTTTTTC GACATACTAC GCAGACTCCG TGAAGGGCCG
- F T I S R D N S K N T L Y L Q M H S L R A E D T A V Y Y C A K V G 201 GTTCACCATC TCCCGCGACA ATTCCAAGAA CACGCTGTAT CTGCAAATGA ACAGCCTGCG TGCCGAGGAC ACCGCGGTAT
ATTACTGTGC GAAAGTGGGT
R G Q R D T S Q P F D Y W G Q G T L V T V S S
301 cGTGGTCAGc GTGATACTAG TCAGCCGTTT GACTACTGGG GTCAGGGAAC CCTGGTCACC GTCTCGAGC — SΞQ ID NO. : 470
DOM-205 - SEQ ID NO.: 356
E V Q L L E S G G G L V Q P G G S L R L S C A A S G F T F A S Y Q M - 1 GAGGTGCAGC TGTTGGAGTC TGGGGGAGGC TTGGTACAGC CTGGGGGGTC CCTGCGTCTC TCCTGTGCAG CCTCCGGATT
CACCTTTGCT TCTTATCAGA • A W V R Q A P G K G L E W V S G I T S G G L S T Y Y A D S V K G R -
101 TGGCTTGGGT CCGCCAGGCT CCAGGGAaGG GTCTAGAGTG GGTCTCAGGG ATTACTTCGG GTGGTCTTAG TACGTACTAC GCAGACTCCG TGAAGGGCCG - F T I S R D M S K N T L Y L Q M N S L R A E D T A V Y Y
C A K P G
201 GTTCACCATC ATTACTGTGC
R G F D Y W G Q G T L V T V S S 301 AGGGGTTTTG ACTACTGGGG TCAGGGAACC CTGGTCACCG TCTCGAGC — SEQ ID NO. : 471
DOM-206 - SEQ ID NO.: 357
E V Q L L E S G G G L V Q P G G S L R L S C A A S G F T F A S Y E M -
101 TGACTTGGGT GGTCTCAGGG ATTTCTTCTG
GCAGACTCCG TGAAGGGCCG
F T I S R D N S K N T L Y L Q M N S L R A E D T A V Y Y C A K P G
201 GTTCACCATC ATTACTGTGC GAAACCGGGG
V L F D Y W G Q G T L V T V S S
301 GTGTTGTTTG ACTACTGGGG TCAGGGAACC CTGGTCACCG TCTCGAGC — SEQ ID NO. : 472
DOM-207 - SEQ ID NO.: 358
CACCTTTGAT AAGTATTTGA S W V R Q A P G K G L E W V S G I E P L G D V T Y Y A D S V K G R .
101 TGTCGTGGGT CCGCCAGGCT CCAGGGAAGG GTCTGGAGTG GGTCTCAGGT ATTGAGCCTC TGGGTGATGT TACATACTAC GCAGACTCCG TGAAGGGCCG - F T I S R D N S K N T L Y L Q M N S L R A E D T A V Y Y
C A K E A
201 GTTCACCATC TCCCGCGACA ATTCCAAGAA CACGCTGTAT CTGCAAATGA ACAGCCTGCG TGCCGAGGAC ACCGCGGTAT ATTACTGTGC GAAAGAGGCT
S G D F D Y W G Q G T L V T V S S 301 TCGGGGGATT TTGACTACTG GGGTCAGGGA ACCCTGGTCA CCGTCTCGAG C — SEQ ID NO. : 473
DOM-208 - SEQ ID NO.: 359
E V Q L L E S G G G L V Q P G G S L R L S C A A S G F T F T E Y E M -
1 GAGGTGCAGC TGTTGGAGTC TGGGGGAGGC TTGGTACAGC CTGGGGGGTC CCTGCGTCTC TCCTGTGCAG CCTCCGGGTT CACCTTTACT GAGTATGAGA
S W V R Q A P G K G L E W V S S I D N V G S S T Y Y A D S V K G R - 101 TGTCTTGGGT CCGCCAGGCT CCAGGGAAGG GTCTAGAGTG GGTCTCAAGT ATTGATAATG TGGGTAGTAG TACATACTAC
GCAGACTCCG TGAAGGGCCG
F T I S R D N S K N T L Y L Q M N S L R A E D T A V Y Y C A K P G
201 GTTCACCATC TCCCGCGACA ATTCCAAGAA CACGCTGTAT CTGCAAATGA ACAGCCTGCG TGCCGAGGAT ACCGCGGTAT ATTACTGTGC GAAACCTGGG
K L F D Y W G Q G T L V T V S S
301 AAGCTTTTTGACTACTGGGG TCAGGGAACC CTGGTCACCG TCTCGAGC — SEQ ID NO. : 474
DOM-209 - SEQ ID NO. : 360
E V Q L L E S G G G L V Q P G G S L R L S C A A S G F T F A D Y E M -
1 GAGGTGCAGC TGTTGGAGTC TGGGGGAGGC TTGGTACAGC CTGGGGGGTC CCTGCGTCTC TCCTGTGCAG CCTCCGGATT CACCTTTGCT GATTATGAGA • W W V R Q A P G K G L E W V S A I S R Q G F A T Y Y A D S V K G R -
101 TGTGGTGGGT CCGCCAGGCT CCAGGGAAGG GTCTAGAGTG GGTCTCAGCG ATTTCTAGGC AGGGTTTTGC TACATACTAC GCAGACTCCG TGAAAGGCCG - F T I S R D H S K N T L Y L Q M N S L R A E D T A V Y Y
C A K D L
201 GTTCACCATC TCCCGCGACA ATTCCAAGAA CACGCTGTAT CTGCAAATGA ACAGCCTGCG TGCCGAGGAC ACCGCGGTAT
ATTACTGTGC GAAAGATCTG
E R D D F D Y W G Q G T L V T V S S 301 GAGCGGGATG ATTTTGACTA CTGGGGTCAG GGAACCCTGG TCACCGTCTC GAGC — SEQ ID NO. : 475
Among the DOM clones described, a number of observations have been made regarding homology among clones, and properties of amino acids predicted at particular positions in the anti-CD40L dAbs. These are shown in Tables 9 and 10 below:
Table 9. Predictions for anti-VH CD40L clones: (using Standard single letter amino acid code)
Figure imgf000260_0001
* If amino acid at position 99 (dummy) is P then G or D is frequently found at position 100. Table 10. Predictions for anti-VK CD40L clones:
Figure imgf000260_0002
257
Figure imgf000261_0001
*If amino acid at position 28 (dummy) is D then E is frequently found at position 34. **If amino acid at position 34 (dummy) is E then Y is frequently found at position 91 and H or D at position 92.
Example 8: PEGylation of DOM8-24
Site specific maleimide-PEGylation of DOM8-24 requires a solvent accessible cysteine to be provided on the surface of the protein, in this example, at the C-terminus. The cysteine residue, once reduced to give the free thiol, can then be used to specifically couple the protein to PEG via a wide range of chemistries such as maleimide or another thiol to give a disulphide. A wide range of chemical modified PEGs of different sizes and branched formats are available from Nektar (formally known as Shearwater Corp). This allows the basic dAb-cys monomer to be formatted in a variety of ways for example as a PEGylated monomer, dimer, trimer, tetramer etc. The size of the PEGs is given in kDa but can also be referred to as K (i.e. "4OK PEG" = 40 kDaPEG).
PCR construction of DOM8-24cys
The site of attachment for the PEG may be placed elsewhere on the surface of the dAb as . long as the targeted amino acid is solvent accessible and the resultant PEGylated protein still maintains antigen binding. Thus it is also possible to engineer the cys into any one of frameworks 1-4 of the dAb for PEGylation and still maintain some antigen binding. The following oligonucleotides were used to specifically PCR DOM8-24 with a SaR and BamYU. sites for cloning and also to introduce a C-terminal cysteine residue.
Sail
Ala Ser Thr GIu VaI GIn Leu Leu GIu Ser GIy GIy GIy Leu VaI
258 GCG TCG ACG GAG GTG CAG CTG TTG GAG TCT GGG GGA GGC TTG GTA
CGC AGC TGC CTC CAC GTC GAC AAC CTC AGA CCC CCT CCG AAC CAT
GIn Pro GIy GIy Ξer Leu Arg Leu Ser Cys Ala Ala Ser GIy Phe CAG CCT GGG GGG TCC CTG CGT CTC TCC TGT GCA GCC TCC GGA TTC
GTC GGA CCC CCC AGG GAC GCA GAG AGG ACA CGT CGG AGG CCT AAG
Thr Phe Ser Asn Tyr Gin Met Ala Trp VaI Arg GIn Ala Pro GIy ACC TTT AGT AAT TAT CAG ATG GCG TGG GTC CGC CAG GCT CCA GGG
TGG AAA TCA TTA ATA GTC TAC CGC ACC CAG GCG GTC CGA GGT CCC
Lys GIy Leu GIu Trp VaI Ser Ser lie Thr Ser GIu GIy GIy Ser AAG GGT CTA GAG TGG GTC TCA AGT ATT ACT AGT GAG GGT GGT TCG
TTC CCA GAT CTC ACC CAG AGT TCA TAA TGA TCA CTC CCA CCA AGC
Thr Tyr Tyr Ala Asp Ser VaI Lys GIy Arg Phe Thr lie Ser Arg ACA TAC TAC GCA GAC TCC GTG AAG GGC CGG TTC ACC ATC TCC CGC
TGT ATG ATG CGT CTG AGG CAC TTC CCG GCC AAG TGG TAG AGG GCG
Asp Asn Ser Lys Asn Thr Leu Tyr Leu GIn Met Asn Ser Leu Arg GAC AAT TCC AAG AAC ACA CTG TAT CTG CAA ATG AAC AGC CTG CGT
CTG TTA AGG TTC TTG TGT GAC ATA GAC GTT TAC TTG TCG GAC GCA
Ala GIu Asp Thr Ala VaI Tyr Tyr Cys Ala Lys Pro GIy Lys Asn GCC GAG GAC ACC GCG GTA TAT TAC TGT GCG AAA CCG GGT AAG AAT
CGG CTC CTG TGG CGC CAT ATA ATG ACA CGC TTT GGC CCA TTC TTA
259 Phe Asp Tyr Trp GIy Gin GIy Thr Leu VaI Thr VaI Ser Cys ***
316 TTT GAC TAC TGG GGT CAG GGA ACC CTG GTC ACC GTC TCG TGC TAA
AAA CTG ATG ACC CCA GTC CCT TGG GAC CAG TGG CAG AGC ACG ATT
BamHI
*** GIy Ser (SEQ ID NI: 85]
361 TAA GGA TCC (SEQ ID NO: 83)
ATT CCT AGG (SEQ ID NO: 84 J
The DNA sequence of the PCR primers used to amplify the engineered dAb are shown 5 below.
Forward primer
5'- AGTGCGTCGACGGAGGTGCAGCTGTTGGAGTCT-S' (SEQ ID NO: 86)
Reverse primer
5'- AAAGGATCCTTATTAGCACGAGACGGTGACCAGGGTTCCCTG-S' (SEQ ID NO: 87)
:0 The PCR reaction (50μL volume) was set up as follows: 200μM dNTP's, 0.4μM of each primer, 5 μL of 1Ox P/wTurbo buffer (Stratagene), 100 ng of template plasmid (DOM8-24), lμL of P/wTurbo enzyme (Stratagene) and the volume adjusted to 50μL using sterile water. The following PCR conditions were used: initial denaturing step 94 0C for 2 mins, then 25 cycles of 94 0C for 30 sees, 64 0C for 30 sec and 72 °C for 30 sec. A final extension step was also included
:5 of 72 0C for 5 mins. The PCR product was purified and digested with SaR and BamYLl and ligated into the vector (pDOM5) which had also been cut with the same restriction enzymes. Correct clones were verified by DNA sequencing.
Expression and purification of DOM8-24
DOM8-24 vector was transformed into HB2151 electro-competent cells. Cells carrying
-0 the dAb plasmid were selected for using lOOμg/mL carbenicillin. Cultures were set up in 2L baffled flasks containing 500 mL of terrific broth (Sigma-Aldrich) and lOOμg/mL carbenicillin.
260 The cultures were grown at 30 0C at 200rpm to an O.D.600 of 1-1.5 and then induced with ImM IPTG (isopropyl-beta-D-thiogalactopyranoside, from Melford Laboratories). The expression of the dAb was allowed to continue for 12-16 hrs at 30 0C. It was found that most of the dAb was present in the culture media. Therefore, the cells were separated from the media by centrifugation (8,000Xg for 30 mins), and the supernatant used to purify the dAb. Per litre of supernatant, 10 mL of Streamline Protein A (Amersham Biosciences) was added and the dAb allowed to batch bind with stirring for 3 hours at room temperature, or overnight at 4 0C. The resin was then allowed to settle under gravity for an hour before the supernatant removed. The agarose was then packed into a XK 16 column (Amersham Pharmacia) and was washed with 10 column volumes of 2xPBS. The bound dAb was eluted with 100 mM glycine pH 3.0 and protein containing fractions were then neutralized by the addition of 1/5 volume of 1 M Tris pH 8.0.
PEGylation of DOM8-24cvs using MAL activated PEG
Monomer PEGylation
The cysteine residue which had been engineered onto the surface of the VH dAb was specifically modified with a single linear or branched PEG-MAL to give monomeric modified protein. mPEG-MAL formats which may be used to PEGylate a monomeric VH or Vk dAb. The PEGs may be of MW from 500 to 60,000 (eg, from 2,000 to 40,000) in size.
Figure imgf000264_0001
mPEG-MAL mPEG2-MAL
2.5 ml of 500 μM DOM8-24cys was reduced with 5 mM dithiothreitol and left at room temperature for 20 minutes. The sample was then buffer exchanged using a PD-10 column
(Amersham Pharmacia). The column had been pre-equilibrated with 5 mM EDTA, 20 mM sodium phosphate pH 6.5, 10% glycerol, and the sample applied and eluted following the
261 manufactures guidelines. The eluted sample (3.5 ml of -360 μM dAb) was placed on ice until required. A four fold molar excess of 3OK PEG-MAL or 4OK PEG2-MAL (Nektar) was weighed out and added directly to the reduced dAb solution and gently mixed until the polymer had dissolved. The reaction was left to proceed at room temperature for 3 hours.
Purification of 3OK and 4OK PEGylated DOM8-24cvs monomer
The PEGylated dAb was purified using cation exchange chromatography as the isoelectric point (pi) of the protein is ~8. 40 μL of 40% glacial acetic acid was added per mL of the 30K or 4OK PEG DOM8-24cys reaction to reduce the pH to ~4. The sample was then applied to a ImL Resource S cation exchange column (Amersham Pharmacia), which had been pre- equilibrated with 50 mM sodium acetate pH 4.0. The PEGylated material was separated from the unmodified dAb by running a linear sodium chloride gradient from 0 to 500 mM over 20 column volumes. Fractions containing PEGylated dAb only were identified using SDS-PAGE and then pooled and the pH increased to 8 by the addition of 1/5 volume of IM Tris pH 8.0
In vitro functional binding assay: CD40 ligand receptor assay (CD40L RBA)
The CD40L assay was used to measure the binding of CD40L to CD40 and the ability of binding entities (including monovalent antibody fragments such a dAbs) to block this interaction, as shown Figure 7.
A 96 well Nunc Maxisorp assay plate was coated overnight at 40C with 100 μl per well of recombinant human CD40/Fc (R&D Systems) at 0.5 μg/ml in carbonate buffer. The plate was washed 3 times with 300 μl of 0.05% Tween/PBS and 3 times with 300 μl of PBS using a Tecan plate washer. The wells were blocked using 200 μl of PBS containing 2% (w/v) BSA and incubated for a minimum of 1 h at room temperature. The wells were washed as above, then 50 μl of purified dAb protein was added to each well. To each well 50 μl of CD40L, at 6 ng/ml in diluent (for a final concentration of 3 ng/ml), was also added and the plate incubated for 1 hr at room temperature. The plate was washed as described previously and 100 μl biotinylated anti- CD40L antibody, 0.5 μg/ml in diluent, was added and incubated for 1 hr at room temperature. The plate was washed as described above, then 100 μl HRP conjugated anti-biotin antibody (1:5000 dilution in diluent) added to each well and the plate incubated for 1 hr at room
262 temperature. The plate was washed again as described above using a Tecan plate washer and the assay developed using 100 μl of SureBlue 1 -Component TMB Micro Well Peroxidase solution (the plate was left at room temperature for up to 20 min). The reaction was stopped by the addition of 100 μl I M hydrochloric acid. The OD450nm of the plate was assayed within 30 minutes of acid addition. The OD450nm is proportional to the amount of bound streptavidin- HRP conjugate, therefore the greater the degree of dAb inhibition the lower the OD45onm of the resulting signal. The following controls were included; 0 ng/ml CD40L (diluent only), 3 ng/ml CD40L and 3 ng/ml CD40L with 1 μg/ml anti-CD40L antibody.
The results of the CD40L RBA are shown in Fig. 9. It can be seen that PEGylated DOM8-24cys has a similar affinity for CD40L as the unmodified protein. The size of the polymer attached (either 30 or 40K) does not seem to significantly affect the IC 50 of~l nM.
Example 9: Generation and Expression of a Dual Specific dAb Dimer (T)OM7h-26 — DOM-24)
A dual specific dAb dimer was constructed essentially as described in WO2004/003019 and as described below.
A PCR fragment introducing 5' Notl and 3' EcoRI restriction sites into the DOM-24
DNA sequence was generated using the primers VH-5'-NotI (5'- GTATGTCTGGCGGCCGCAGAGGTGCAGCTGTTGGAGTCTGGGGGAGGCTTG-S ' ; SEQ ID NO: 90) and VH-NO-XhoI (5'-TAGAATTCTTATTAGCTGGAGACGGTGACCAGGGT- 3'; SEQ ID NO: 91). The PCR product was electrophoresed on a 1% agarose gel and the appropriate band was excised and gel cleaned using the Qiagen Gel Extraction Kit. The purified DNA was digested with Notl and EcoRI for 1 hour at 370C and then the digested DNA fragment purified using the Qiagen PCR Cleanup Kit. The digested PCR product was ligated using T4 DNA ligase into the vector pCH3, derived from pPICz-alpha (Invitrogen Ltd, Paisley, UK), which had been previously been digested with Notl and EcoRI, thereby enabling the ligation of the DOM-24 DNA sequence 3' relative to the anti-human serum albumin dAb DNA sequence (DOM7h-26). One micro-litre of the ligation mix was used to electroporate 50 μl of E. coli TOPlOF' (0.2 mm diameter cuvettes; 25 μFD; 200Ω; 2.50O kV) using a Biorad Genepulser II with pulse controller module. The electroporated cells were immediately resuspended in 1 ml low salt LB medium (10 g/1 tryptone; 5 g/1 yeast extract; 5 g/1 NaCl; pH to 7.5 with 4 M NaOH;
263 then autoclaved at 1210C for 15 min) and 100 μl of the cell suspension plated onto low salt LB agar plates (10 g/1 tryptone; 5 g/1 yeast extract; 5 g/1 NaCl; pH to 7.5 with 4 M NaOH; 15 g/1 agar; then autoclaved at 1210C for 15 min) supplemented with 25 μg/ml zeocin (Invitrogen Ltd, Paisley, UK) and grown at 37°C overnight.
5 Several individual clones from the overnight growth plate were analysed by DNA sequencing using the alpha-factor sequencing primer (S'-TACTATTGCCAGCATTGCTGC-S'; SEQ ID NO: 92) and the reverse 3' AOXl primer (5'-GCAAATGGCATTCTGACATCC-S'; SEQ ID NO: 93). A plasmid prep derived from a single clone containing the desired DNA sequence was made by inoculating 100 ml of low salt LB medium supplemented with 25 μg/ml
0 zeocin and grown overnight at 37°C with shaking at 250 rpm, followed by purification of the plasmid DNA using a Qiagen Plasmid Midi Prep Kit (Qiagen).
The concentration and purity of the DNA was determined by measuring the absorbance at
280 nm and 260 nm. Between 10 and 50 μg of DNA was digested with Pmel, Sad or BstXI restriction enzymes and the DNA digestion confirmed by analysis on a 0.8% agarose gel. The
5 digested DNA was purified using the Qiagen Qiaex II Kit (Qiagen) and the DNA eluted into
10 μl purified water per 20 μl of Qiaex II resin.
Competent yeast cells were made by streaking the wild type Pichia pastoris strain XSS onto a YPD agar plate (for 1 litre of media: 20 g peptone from meat type 1; 10 g yeast extract; 2O g agar dissolved in 900 ml water and sterilise by autoclaving at 1210C for 15 minutes;
:0 followed by the aseptic addition of 100ml of 20% destrose and the desired amount of zeocin once the media had cooled below 5O0C) and grown at 3O0C for 36-48 h until colonies appeared. A single colony was inoculated into 5 ml of YPD medium (as for YPD agar, without the addition of 20 g agar) and grown overnight in a 50 ml baffled flask at 300C with shaking at 250 rpm. Five hundred millilitres of YPD medium in a 21 baffled flask was inoculated with 0.1-0.5 ml of
:5 the overnight culture and grown at 300C with shaking at 250 rpm overnight to an OD600 of approximately 1.3-1.5. The culture was cooled on ice then the Pichia harvested by centrifugation at 1500 g for 5 min at 40C. The supernatant was discarded and the pellet resuspended in 500 ml ice cold ultrapure water. The Pichia were recovered again by centrifugation at 150O g for 5 min at 40C. The pellet was resuspended and centrifuged for a further three times, the first time in
264 250 ml ice cold ultrapure water, then twice in 20 ml ice cold IM sorbitol. The competent Pichia were kept on ice for up to 6 hours prior to use.
To transform the Pichia by electroporation, between 10 to 20 μg of linearised vector in 10 μl of water was mixed with 80 μl of competent Pichia in a precooled microfuge tube and 5 incubate on ice for 5 min. The Pichia mixture was then transferred to a precooled 0.2 cm gap electroporation cuvette, and electroporated: 25 μF, resistance set to infinity, 0.54 kV in a Biorad Genepulser II with pulse controller module. Immediately 1 ml ice cold 1 M sorbitol was added to the electroporation cuvette and the electroporated Pichia transferred into a 15 ml polypropylene tube. This culture was incubated at 3O0C without shaking for 2 hours to allow the
10 cells to recover. The cells were then plated onto YPDS agar plates (YPD agar supplemented with I M sorbitol) and zeocin added to final concentrations of 100 μg/ml, 500 μg/ml, 1000 μg/ml, and 2000 μg/ml. The plates were incubated at 30°C in the dark for 2-10 days until colonies appeared. Several clones were picked from each plate and restreaked onto YPD plates supplemented with the same amount of zeocin as they were originally selected against. For
[5 small to medium scale shaking flask expression the Mut status of each X33 clone was not determined.
Individual restreaked colonies were used to inoculate 50 ml of BMGY medium (for 1 litre of medium: 20 g peptone from meat type 1; 10 g yeast extract were dissolved in 700 ml with water and sterilise by autoclaving at 1210C for 15 minutes; followed by the aseptic addition of
JO 100 ml 1 M KPO4 pH 6.0; 100 ml 10% glycerol; and 100 ml 1OxYNB (Yeast Nitrogen Base); 2 ml of 500x biotin) in a 250 ml flask and grown at 3O0C with shaking at 250 rpm until an OD600 of 2 to 6 was attained. The Pichia was recovered by centrifugation at 1500 g for 5 min at room temperature. The resulting pellet was resuspended in 20 ml BMMH medium (as for BMGY medium except the 100 ml 10% glycerol was replaced with 100 ml of 5% methanol), and
>5 returned to a fresh 250 ml baffled flask and incubated at 300C with shaking at 250 rpm for up to 5 days post induction. At 24 h intervals, 0.5 ml samples were recovered and assessed by analysis of the supernatant by SDS-PAGE.
Dimer dAb was purified from the culture supernatant using Streamline protein-A matrix (Amersham Biosciences). Following binding of the dimer dAb, the matrix was transferred to an
265 empty 20 ml chromatography column containing a frit and the supernatant allowed to flow through the column, retaining the matrix. The matrix was washed with 10 times the matrix volume of high salt buffer (10 mM Phosphate buffer, 2.7 mM KCl, 487 mM NaCl, pH 7.4). The bound protein was eluted with 0.1 M glycine pH 3 supplemented with 0.15 M NaCl and 0.8 ml 5 fractions were collected which were immediately neutralised with the addition of 0.2 ml of 1 M Tris-HCl pH 8.0.
Simultaneous Antigen Binding of the Dual Specific dAb Dimer
To demonstrate that the dual specific dimeric dAb was functional and could bind both antigens simultaneously, an antigen binding ELISA was performed. Fifty microlitres per well of
10 human serium albumin at 100 μg/ml in PBS was coated on to a Maxisorb ELISA plate (Nunc) overnight at 40C. The plate was washed 3 times with 250 μl/well of PBS supplemented with 0.1% (v/v) Tween 20 (Sigma). The plate was then blocked for 1 h at room temperature with 200 μl per well of PBSM (PBS supplemented with 2% (w/v) low fat milk powder). The plate was washed as before, then 50 μl per well of doubling dilutions of the dimer dAb in PBSM were
L 5 added and incubated for 1 hour at room temperature. The plate was washed as before then 50 μl per well of a 5 nM solution of biotinylated CD40L in PBSM added and incubated for 1 hour at room temperature. The plate was washed as before then 50 μl per well of streptavidin-HRP (Amersham Biosciences) diluted 1 in 4000 in PBSM was added and incubated for 1 hour at room temperature. The plate was washed 4 times with 250 μl/well of PBS supplemented with 0.1%
10 (v/v) Tween 20, followed by two washes with 250 μl/well of PBS. The ELISA was developed by adding 50 μl per well of SureBlue 1-Component TMB Micro Well Peroxidase solution (KPL, Gaithersberg, MD) and incubated at room temperature for several minutes until the desired colour intensity was obtained. The reaction was stopped by the addition of 100 μl of I M hydrochloric acid and read at 450 nm (Figure 10).
25 Generation and Expression of a Dual Specific Fab (DOM7h-2:CK DOM-24:CH)
The produce a dual specific Fab the method used was essentially the same method as described for the dual specific dAb dimer (DOM7h-26— DOM-24) except that the DNA for the DOM7h-2 and DOM-24 dAbs were cloned into separate pPICz-alpha based vectors containing the human CK or human CHl domain respectively. Each of the two vectors were designed to
266 enable the expression of a single polypeptide consisting of the CHl or CK domain fused in frame to the 3' end of the appropriate dAb. To obtain expression of the complete Fab fragment, competent Pichia pastoris strain X33 were cotransformed with two linearised vectors. Purification was as described for the dimer Fab.
5 Simultaneous Antigen Binding of the Dual Specific Fab
The simultaneous antigen binding of the dual specific Fab was carried out using the same assay as described for the dual specific dAb dimmer above. The results are shown in Figure 11.
Inhibition of CD40 mediated CD54 upregulation disruption of CD40 / CD40L interaction in L3055 cells.
[0 Monomeric dAb molecules were assayed for their ability to disrupt the binding of cellularly associated CD40L to CD40. This was determined by measuring the level inhibition of CD40 mediated CD54 upregulation on group 1 Burkitt lymphoma cell line L3055 by FACS. CD40 displayed on the L3055 cells was stimulated by CD40L expressed on the surface of fibroblasts (typically mouse L-cells) transfected with the CD40L gene as previously described by
5 Garrone et al., 1995 (Garrone P., Neidhardt E. V., Garcia E., Galibert L., van Kooten C5 Banchereau J. (1995) J Exp Med 182, 1265-1273.)
Samples containing the dAbs were preincubated with the L-cells cells for 1 hour prior to the addition of the L3055 cells. Both cell lines were co-cultured for 22 hours following which the L-cells were stained for FACS analysis, as described by Baker et al., 1998 (Baker, M.P., 10 Eliopoulos, A.G., Young, L.S., Armitage, RJ., Gregory, CD., Gordon, J. (1998) Blood, 92 (8), 2830-2843) and by Challa et al., 2002 (Challa, A., Eliopoulos, A.G., Holder, MJ., Burguete, A.S., Pound, J.D., Chamba, A., Grafton, G., Armitage, RJ., Gregory, CD., Martinez- Valdez, H., Young, L. and Gordon, J. (2002) Blood, 99 (9), 3411-3418.
The FACS analysis of DOM-24 (monomeric form) at a concentration of 12μm showed [5 inhibition.
FACS analysis of the Vk DOM-116 at a concentration of 9μM showed inhibition compared with a control blank dAb and the stimulated cell trace (Figure 13).
267 Effect of anti-CD40L therapy on primary immune response to KLH immunisation.
To show the effect of anti-CD40L therapy on the primary immune response to KLH immunization a similar study to that described by Gobburu, J.V.S. et al., 1998 can be carried out (Gobburu, J.V.S., Tenhoor, C, Rogge, M.C., Frazier, D.E., Thomas, D., Benjamin, C, Hess, 5 D.M., & Jusko, WJ. (1998) .1PET, 286, 925-930.
A study example could be as follows: Cynomolgus monkeys are injected with an anti- CD40L antibody or dAb format at a dosing of 10mg/kg at time Oh, 168h and 336h. At 24h the animals receive a single intradermal injection of 100 μg highly purified keyhole limpet hemocyanin (KLH). The serum is tested for a KLH response prior to the start and following 10 completion of the study. The animals are allowed to recover for a suitable time to ensure that the dAb molecule has cleared the circulation, then injected with KLH (without dAb) to determine if the animal can raise an immune response against KLH. Alternative antigens can be used for the study if an earlier response to KLH is detected prior to the start of the study.
The immune response to KLH will be measured by a suitable ELISA, RIA or antibody [ 5 titer assay. It is anticipated that the results would show that that the PEglyated monomeric DOM- 24 would suppress the animals immune response to KLH. This would be shown in the reduction of the antibody titer compared to a positive control. The results could be normalized such that the positive control was deemed to be 100 and the reduction in immune response after the administration of the dAbs would be in the range of 10 to 90% ie 90 to 10 units.
>0 Sequence of the DOM7h-26— DOM-24
LEICREVQLLESGGGL VQPGGSLRLSCTASGFTFDEYNMS WVRQAPGKGLEWVSTILPH GDRTYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAKQDPLYRFDYWGQG TLVTVSSAAAEVQLLESGGGLVQPGGSLRLSCAASGFTFSNYQMAWVRQAPGKGLEW VSSITSEGGSTYYADSVKGRFTISRDNSKNTL YLQMNSLRAEDTA VYYCAKPGKNFDY 15 WGQGTLVTVSS (SEQ ID NO: 88)
Sequence of the DQM7h-26
268 DIQMTQSPSSLSASVGDRVTITCRASQKIATYLNWYQQKPGKAPKLLIYRSSSLQSA VPS RFSGSGSGTVFTLTISSLQPEDFATYYCQQTYAVPPTFGQGTKVEIKR (SEQ ID NO: 89)
AU patents, patent applications, and published references cited herein are hereby incorporated by reference in their entirety. While this invention has been particularly shown and described with references to preferred embodiments thereof, it will be understood by those skilled in the art that various changes in form and details may be made therein without departing from the scope of the invention encompassed by the appended claims.
269 Annex 1; polypeptides which enhance half-life in vivo.
Alpha- 1 Glycoprotein (Orosomucoid) (AAG) Alpha- 1 Antichyromotrypsin (ACT) Alpha- 1 Antitrypsin (AAT)
Alpha- 1 Microglobulin (Protein HC) (AIM)
Alpha-2 Macroglobulin (A2M)
Antithrombin III (AT III)
Apolipoprotein A-I (Apo A-1)
Apoliprotein B (Apo B)
Beta-2-microglobuIin (B2M)
Ceruloplasmin (Cp)
Complement Component (C3)
Complement Component (C4)
Cl Esterase Inhibitor (Cl INH)
C-Reactive Protein (CRP)
Cystatin C (Cys C)
Ferritin (FER)
Fibrinogen (FIB)
Fibronectin (FN)
Haptoglobin (Hp) Hemopexin (HPX)
Immunoglobulin A (IgA)
Immunoglobulin D (IgD)
Immunoglobulin E (IgE)
Immunoglobulin G (IgG)
Immunoglobulin M (IgM)
Immunoglobulin Light Chains (kapa/lambda)
Lipoprotein(a) [Lp(a)J
Mannose-bindign protein (MBP)
Myoglobin (My o)
Neonatal Fc (FcRn)
Plasminogen (PSM)
Prealbumin (Transthyretin) (PAL)
Retinol-binding protein (RBP)
Rheomatoid Factor (RF)
Serum Amyloid A (SAA)
Soluble Tranferrin Receptor (sTfR)
Transferrin (Tf)
271 Annex 2
Figure imgf000275_0001
272
Figure imgf000276_0001
273
Figure imgf000277_0001
274
Figure imgf000278_0001
TGF-β/IL-1 • Prostaglndin synthesis by osteoblasts
• IL-6 production by intestinal epithelial cells (inflammation model)
• Stimulates IL-Il and IL-6 in lung fibroblasts (inflammation model)
275
Figure imgf000279_0001
76
Figure imgf000280_0001
277
Figure imgf000281_0001
278
Figure imgf000282_0001
279
Figure imgf000283_0001
280
Figure imgf000284_0001
Figure imgf000285_0001
282
Figure imgf000286_0001
283
Figure imgf000287_0001
Figure imgf000288_0001
285 Annex 3: Oncology combinations
Figure imgf000289_0001
Figure imgf000290_0001
Figure imgf000291_0001

Claims

1. A method of antagonizing an activity of CD40 or CD40L in an individual, the method comprising administering an antibody polypeptide that is monovalent for binding to CD40L (gp39) to said individual wherein said polypeptide antagonizes an activity of CD40 or CD40L or both.
2. A method of antagonizing the binding of CD40 to CD40L in an individual, the method comprising administering an antibody polypeptide that is monovalent for binding to CD40L (gp39) to said individual, wherein said polypeptide antagonizes the binding of CD40 to CD40L in said individual.
3. A method of treating or preventing a symptom of autoimmune disease in an individual, the method comprising administering a monovalent anti-CD40L antibody polypeptide to said individual in an amount effective to treat or prevent a symptom of autoimmune disease.
4. A method of treating or preventing an autoimmune disease in an individual, the method comprising administering a monovalent anti-CD40L antibody polypeptide to said individual in an amount effective to treat or prevent a symptom of autoimmune disease.
5. The method of claim 3, wherein said autoimmune disease is a disease selected from the group , consisting of systemic lupus erythematosis, multiple sclerosis, rheumatoid arthritis, Diabetes, allograft rejection, xenograft transplant rejection, and graft vs. host disease.
6. The method of claim 3, wherein said antibody polypeptide inhibits the binding of CD40L to CD40.
7. The method of claim 3, wherein said antibody polypeptide comprises an amino acid sequence selected from the group consisting of SEQ ID NOs 7-82, SEQ ID NOs 246-360, and DOM8-24.
8. The method of claim 3, wherein said antibody polypeptide has an amino acid sequence at least 85% identical to a sequence selected from the group consisting of SEQ ID NOs 7-82 and 246-360, which antibody polypeptide specifically and monovalently binds CD40L.
9. The method of claim 6, wherein said antibody polypeptide inhibits binding of CD40L to CD40 with an IC50 in the range of 20 pM to 1.5 μM, inclusive.
10. The method of claim 3, wherein said antibody polypeptide is PEG-linked.
11. The method of claim 10, wherein said PEG-linked antibody polypeptide has a hydrodynamic size of at least 24 kD.
12. The method of claim 10 wherein said PEG is linked to said antibody at a cysteine or lysine residue.
13. The method of claim 10 wherein the total PEG size is from 20 to 60 kD, inclusive.
14. The method of claim 10 wherein said PEG-linked antibody polypeptide has a hydrodynamic size of at least 20O kD.
15. The method of claim 10 wherein said PEG-linked antibody polypeptide has an increased in vivo half-life relative to the same polypeptide composition lacking linked polyethylene glycol.
16. The method of claim 15 wherein the tα-half life of the polypeptide composition is increased by 10% or more.
17. The method of claim 15 wherein the tα-half life of the polypeptide composition is in the range of 0.25 minutes to 12 hours.
18. The method of claim 15 wherein the tβ-half life of the polypeptide composition is increased by 10% or more.
19. The method of claim 15 wherein the tβ-half life is in the range of 12 to 48 hours.
20. The method of claim 1 or 2 wherein said antibody polypeptide is a camelid or nurse shark VHH domain.
21. An antibody polypeptide that is monovalent for binding to CD40L (gp39), wherein binding of said antibody polypeptide to CD40L does not substantially agonize CD40 and/or CD40L activity.
22. The antibody of claim 21 wherein said antibody polypeptide inhibits the binding of CD40 to CD40L, and does not substantially agonize signaling by CD40.
23. The antibody polypeptide of claim 21 wherein binding of said antibody polypeptide to CD40L does not substantially induce JNK phosphorylation in Jurkat T-cells.
24. The antibody polypeptide of claim 21 wherein binding of said antibody polypeptide to CD40L does not substantially induce IFN-γ secretion by Jurkat T-cells co-stimulated with anti- CD3 antibody.
25. The antibody polypeptide of claim 21 wherein the presence of said antibody polypeptide in a standard platelet aggregation assay does not result in aggregation of more than 25% over the aggregation observed in a negative control assay.
26. The antibody polypeptide of claim 21 in which said antibody polypeptide is selected from the group consisting of an antibody single variable domain polypeptide, dAb, FAb, an scFv, an Fv, or a disulfide-bonded Fv.
27. An antibody polypeptide that is monovalent for binding to CD40L (gp39), wherein the presence of said antibody polypeptide in a standard platelet aggregation assay does not result in aggregation of more than 25% over the aggregation observed in a negative control assay.
28. A PEG-linked antibody polypeptide of claim 21.
29. The PEG-linked antibody polypeptide of claim 28 which has a hydrodynamic size of at least 24 kD.
30. The PEG-linked antibody polypeptide of claim 28 wherein said PEG is linked to said antibody at a cysteine or lysine residue.
31. The PEG-linked antibody polypeptide of claim 28 wherein the total PEG size is from 20 to 60 kD, inclusive.
32. The PEG-linked antibody polypeptide of claim 28 which has a hydrodynamic size of at least . 20O kD.
33. The PEG-linked antibody polypeptide of claim 28 which has an increased in vivo half-life relative to the same polypeptide composition lacking linked polyethylene glycol.
34. The PEG-linked antibody polypeptide of claim 33 wherein the tα-half life of the polypeptide composition is increased by 10% or more.
35. The PEG-linked antibody polypeptide of claim 33 wherein the tα-half life of the polypeptide composition is in the range of 0.25 minutes to 12 hours.
36. The PEG-linked antibody polypeptide of claim 33 wherein the tβ-half life of the polypeptide composition is increased by 10% or more.
37. The PEG-linked antibody polypeptide of claim 33 wherein the tβ-half life is in the range of 12 to 48 hours.
38. The antibody of claim 21 which comprises an amino acid sequence selected from the group consisting of SEQ ID NOs 7-82 and 246-360.
39. The antibody polypeptide of claim 21 which is free of an Fc domain.
40. The antibody polypeptide of claim 39 wherein the presence of said antibody polypeptide in a standard platelet aggregation assay does not result in aggregation of more than 25% over the aggregation observed in a negative control assay.
41. The antibody of claim 21 which inhibits binding of CD40L to CD40 with an IC50 in the range of 20 pM to 1.5 μM, inclusive.
42. A composition comprising the antibody polypeptide of claim 21 and a second antibody polypeptide which binds a ligand other than CD40L, wherein said antibody polypeptide which binds a ligand other than CD40L binds a ligand selected from the group consisting of HSA, TNFα, IL-I, IL-2, IL-4, IL-6, IL-8, IL- 12, IL-18, IFN-γ, CD2, CD4, CD8, LFAl, LFA3, VLA4, CD80, B7-1, CD28, CD86, B7-2, CTLA-4 CD28, Inducible costimulatory molecule (ICOS), CD27, CD30, OX40, CD45, CD69, CD3, CD70, Inducible costimulatory molecule ligand (ICOSL), OX40L, HVEM (Herpes Virus Entry Mediator), LIGHT.
43. An antibody polypeptide which has an amino acid sequence at least 85% identical to a sequence selected from the group consisting of SEQ ID NOs 7-82, SEQ ID NOs 246-360, and DOM8-24, which antibody polypeptide specifically and monovalent!}' binds CD40L.
44. An antigen-binding polypeptide, said polypeptide comprising a immunoglobulin single variable domain which specifically and monovalently binds CD40L, wherein said polypeptide substantially inhibits the binding of CD40L to CD40.
45. The antigen-binding polypeptide of claim 44 which inhibits the binding of CD40 to CD40L with an IC50 in the range of 20 pM to 1.5 μM, inclusive.
46. The antigen-binding polypeptide of claim 44 wherein binding of said polypeptide to CD40L does not substantially agonize CD40 and/or CD40L activity.
47. The antigen-binding polypeptide of claim 44 wherein binding of said polypeptide to CD40L does not substantially induce JNK phosphorylation in Jurkat T-cells.
45. The antigen-binding polypeptide of claim 44 wherein binding of said polypeptide to CD40L does not substantially induce IFN-γ secretion by Jurkat T-cells co-stimulated with anti-CD3 antibody.
46. The antigen-binding polypeptide of claim 44 wherein the presence of said antibody polypeptide in a standard platelet aggregation assay does not result in aggregation of more than 25% over the aggregation observed in a negative control assay.
47. The antigen-binding polypeptide of claim 44 in which said antigen-binding polypeptide is selected from the group consisting of an antibody single variable domain polypeptide, dAb, FAb, an scFv, an Fv, or a disulfide-bonded Fv.
48. A polypeptide according to claim 21 or 44, wherein the anti-CD40L variable domain or dAb 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 sequence of one or more of said framework regions collectively comprises 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.
49. A polypeptide according to claim 21 or 44, wherein the amino acid sequences of FWl, FW2, FW3 and FW4 of the anti-CD40L variable domain or dAb 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.
50. The polypeptide according to claim 49, wherein the amino acid sequences of said FWl, FW2 and FW3 of the anti-CD40L variable domain or dAb are the same as the amino acid sequences of corresponding framework regions encoded by human germline antibody gene segments.
51. The polypeptide according to claim 49, wherein said human germline antibody gene segment is selected from the group consisting of DP47, DP45, DP48 and DPK9.
52. A PEG-linked antigen-binding polypeptide comprising a single immunoglobulin variable domain which specifically and monovalently binds CD40L.
53. The PEG-linked polypeptide of claim 52 which has a hydrodynamic size of at least 24 kD.
54. The PEG-linked polypeptide of claim 52 wherein said PEG is linked to said antibody at a cysteine or lysine residue.
55. The PEG-linked polypeptide of claim 52 wherein the total PEG size is from 20 to 60 IcD, inclusive.
56. The PEG-linked polypeptide of claim 52 which has a hydrodynamic size of at least 200 kD.
57. The PEG-linked polypeptide of claim 52 which has an increased in vivo half-life relative to the same polypeptide composition lacking linked polyethylene glycol.
58. The PEG-linked polypeptide of claim 52 wherein the tα-half life of the polypeptide composition is increased by 10% or more.
59. The PEG-linked polypeptide of claim 52 wherein the tα-half life of the polypeptide composition is in the range of 0.25 minutes to 12 hours.
60. The PEG-linked polypeptide of claim 52 wherein the tβ-half life of the polypeptide composition is increased by 10% or more.
61. The PEG-linked polypeptide of claim 60 wherein the tβ-half life is in the range of 12 to 48 hours.
62. An immunoglobulin variable domain polypeptide, said polypeptide comprising a single immunoglobulin variable domain which specifically and monovalently binds CD40L, and which comprises an amino acid sequence selected from the group consisting of SEQ ID NOs 7-82, SEQ ID NOs 246-360, and DOM8-24.
63. An immunoglobulin variable domain polypeptide which has an amino acid sequence at least 85% identical to a sequence selected from the group consisting of SEQ ID NOs 7-82, SEQ ID NOs 246-360, and DOM8-24, which polypeptide specifically and monovalently binds CD40L.
64. The immunoglobulin variable domain polypeptide of claim 63 which comprises a single immunoglobulin variable domain which specifically and monovalently binds CD40L.
65. The immunoglobulin variable domain polypeptide of claim 63 which antagonizes the binding of CD40L.
66. The immunoglobulin variable domain polypeptide of claim 63 which inhibits the binding of CD40 to CD40L and which has an IC50 in the range of 20 pM to 1.5 μM, inclusive.
67. The immunoglobulin variable domain polypeptide of claim 63 wherein said antigen-binding polypeptide inhibits the interaction ' of CD40 with CD40L, but does not substantially agonize signaling by CD40.
68. A composition comprising the immunoglobulin variable domain polypeptide of claim 63 and a second immunoglobulin variable domain polypeptide which binds a ligand other than CD40L.
69. The composition of claim 68 wherein said antibody polypeptide which binds a ligand other than CD40L binds a ligand selected from the group consisting of HSA, TNFα, IL-I, IL-2, IL-4, IL-6, IL-8, IL-12, IL-18, IFN-γ, CD2, CD4, CDS, LFAl, LFA3, VLA4, CD80, B7-1, CD28, CD86, B7-2, CTLA-4 CD28, Inducible costimulatory molecule (ICOS), CD27, CD30, OX40, CD45, CD69, CD3, CD70, Inducible costimulatory molecule ligand (ICOSL), OX40L, HVEM (Herpes Virus Entry Mediator), LIGHT.
70. An extended release pharmaceutical formulation comprising a monovalent anti-CD40L antibody polypeptide.
71. The extended release pharmaceutical formulation of claim 70, wherein said antibody polypeptide is selected from the group consisting of a dAb, Fab, scFv, Fv, or a disulfide bonded Fv.
72. The formulation of claim 70 wherein said monovalent anti-CD40L antibody polypeptide comprises a single immunoglobulin variable domain that binds CD40L.
73. An antibody single variable domain polypeptide that binds to CD40L, said polypeptide comprising the sequence ofDOM8-24.
74. An antibody single variable domain polypeptide that binds to CD40L, wherein the polypeptide has an amino acid sequence that is identical to the amino acid sequence of D0M8- 24, or differs from the amino acid sequence of DOM8-24 at no more than 25 amino acid positions and has a sequence that is at least 80% homologous to the sequence of DOM8-24.
75. An antibody single variable domain polypeptide that binds to CD40L, wherein the polypeptide has an amino acid sequence that is identical to the amino acid sequence of D0M8- 24, or differs from the amino acid sequence of DOM8-24 at no more than 25 amino acid positions and has a CDRl sequence that is at least 50% homologous to the CDRl sequence of DOM8-24.
76. An antibody single variable domain polypeptide that binds to CD40L, wherein the polypeptide has an amino acid sequence that is identical to the amino acid sequence of D0M8- 24, or differs from the amino acid sequence of DOM8-24 at no more than 25 amino acid positions and has a CDR2 sequence that is at least 50% homologous to the CDR2 sequence of DOM8-24.
77. An antibody single variable domain polypeptide that binds CD40L, wherein the polypeptide has an amino acid sequence that is identical to the amino acid sequence of DOM8-24, or differs from the amino acid sequence of DOM8-24 at no more than 25 amino acid positions and has a CDR3 sequence that is at least 50% homologous to the CDR3 sequence of DOM8-24.
78. An antibody single variable domain polypeptide that binds CD40L, wherein the dAb has an amino acid sequence that is identical to the amino acid sequence of DOM8-24, or differs from the amino acid sequence of DOM8-24 at no more than 25 amino acid positions and has a CDRl sequence that is at least 50% homologous to the CDRl sequence of DOM8-24 and has a CDR2 sequence that is at least 50% homologous to the CDR2 sequence of DOM8-24 .
79. An antibody single variable domain polypeptide that binds CD40L, wherein the dAb has an amino acid sequence that is identical to the amino acid sequence of DOM8-24, or differs from the amino acid sequence of DOM8-24 at no more than 25 amino acid positions and has a CDR2 sequence that is at least 50% homologous to the CDR2 sequence of DOM8-24 and has a CDR3 sequence that is at least 50% homologous to the CDR3 sequence of DOM8-24 .
80. An antibody single variable domain polypeptide that binds CD40L, wherein the dAb has an amino acid sequence that is identical to the amino acid sequence of DOM8-24, or differs from the amino acid sequence of DOM8-24 at no more than 25 amino acid positions and has a CDRl sequence that is at least 50% homologous to the CDRl sequence of DOM8-24 and has a CDR3 sequence that is at least 50% homologous to the CDR3 sequence of DOM8-24 .
81. An antibody single variable domain polypeptide that binds CD40L, wherein the dAb has an amino acid sequence that is identical to the amino acid sequence of DOM8-24, or differs from the amino acid sequence of DOM8-24 at no more than 25 amino acid positions and has a CDRl
' sequence that is at least 50% homologous to the CDRl sequence of DOM8-24 and has a CDR2 sequence that is at least 50% homologous to the CDR2 sequence of DOM8-24 and has a CDR3 sequence that is at least 50% homologous to the CDR3 sequence of DOM8-24.
82. A CD40L antagonist having a CDRl sequence that is at least 50% homologous to the CDRl sequence of DOM8-24.
83. A CD40L antagonist having a CDR2 sequence that is at least 50% homologous to the CDR2 sequence of DOM8-24.
84. A CD40L antagonist having a CD.R3 sequence that is at least 50% homologous to the CDR3 sequence of DOM8-24.
85. A CD40L antagonist having a CDRl sequence that is at least 50% homologous to the CDRl sequence of DOM8-24 and a CDR2 sequence that is at least 50% homologous to the CDR2 sequence of DOM8-24.
86. A CD40L antagonist having a CDR2 sequence that is at least 50% homologous to the CDR2 sequence of DOM8-24 and a CDR3 sequence that is at least 50% homologous to the CDR3 sequence of DOM8-24.
87. A CD40L antagonist having a CDRl sequence that is at least 50% homologous to the CDRl sequence of DOM8-24 and a CDR3 sequence that is at least 50% homologous to the CDR3 sequence of DOM8-24.
88. A CD40L antagonist having a CDRl sequence that is at least 50% homologous to the CDRl sequence of DOM8-24 and a CDR2 sequence that is at least 50% homologous to the CDR2 sequence of DOM8-24 and a CDR3 sequence that is at least 50% homologous to the CDR3 sequence of DOM8-24. -
89. The CD40L antagonist of claim 88, wherein said antagonist is an antibody single variable domain polypeptide selected from the group consisting of an antibody single variable domain polypeptide, dAb, FAb, an scFv, an Fv, or a disulfide-bonded Fv.
90. An antibody polypeptide that inhibits the binding of DOM8-24 to CD40L.
91. An antibody polypeptide that binds to the same epitope of CD40L bound by DOM8-24.
92. A method of treating or preventing a symptom of autoimmune disease in an individual, the method comprising administering the antibody polypeptide of claim 90 or 91 to said individual in an amount effective to treat or prevent a symptom of autoimmune disease.
93. An antibody polypeptide that binds to the same epitope of CD40L bound by an antibody single variable domain polypeptide comprising a sequence selected from the group consisting of SEQ ED Nos: 7-82 and 246-360.
94. An antibody single variable domain polypeptide comprising the sequence of CDRl , CDR2, and/or CDR3 of DOM8-24.
95. An antibody single variable domain polypeptide comprising the sequence of CDRl, CDR2, and/or CDR3 of an antibody single variable domain polypeptide comprising a sequence selected from the group consisting of SEQ ID Nos. 7-82 and 246-360.
96. The antibody single variable domain polypeptide of claim 94, wherein said polypeptide comprises the sequence of two or more of CDRl, CDR2, or CDR3 of DOM8-24.
97. The antibody single variable domain polypeptide of claim 95, wherein said polypeptide comprises the sequence of two or more of CDRl, CDR2, or CDR3 of an antibody single variable domain polypeptide comprising a sequence selected from the group consisting of SEQ ID Nos. 7-82 and 246-360.
98. A dual specific ligand comprising a first immunoglobulin single variable domain having a binding specificity to a first antigen and a second single variable domain having a binding activity to a second antigen, wherein the first antigen is CD40L and the second single variable domain is an Antigen Presenting Cell surface antigen or a T cell surface antigen.
99. A ligand according to claim 98 wherein the Antigen Presenting Cell surface antigen is selected from one of the group consisting of, activated macrophage surface antigens, activated B cell surface antigens, co-stimulatory signal pathway surface antigens, and MHC.
100. A ligand according to claim 99 wherein the MHC is class II.
299
101. A ligand according to claim 100 wherein the MHC class II is alpha.
102. A ligand according to claim 100 wherein the MHC class II is beta.
103. A ligand according to claim 99 wherein the Antigen Presenting Cell surface antigen is selected from the group consisting of CD28, Inducible costimulatory molecule (ICOS)5 CD27, CD30, OX40, CD45, CD69, CDS, CD70, Inducible costimulatory molecule ligand (ICOSL), OX40L, CD80, CD86, HVEM (Herpes Virus Entry Mediator), and LIGHT.
104. A ligand according to claim 99 wherein the Antigen Presenting Cell surface antigen is selected from the group consisting of CD28, Inducible costimulatory molecule (ICOS), CD27, CD30, OX40, CD45, CD69, and CD3.
105. A ligand according to claim 99 wherein the surface antigen is a B7 gene surface antigen.
106. A ligand according to claim 105 wherein the B7 gene surface antigen is B7- 2, or B7-1.
300
PCT/GB2005/003562 2004-09-17 2005-09-16 Compositions monovalent for cd40l binding and methods of use WO2006030220A1 (en)

Priority Applications (15)

Application Number Priority Date Filing Date Title
EP05784064.7A EP1797126B1 (en) 2004-09-17 2005-09-16 Compositions monovalent for cd40l binding and methods of use
US11/663,247 US8524236B2 (en) 2004-09-17 2005-09-16 Methods of antagonizing the binding of CD40 to CD40L with CD40L-specific monovalent polypeptides in autoimmune individuals
CN200580039317.5A CN101061140B (en) 2004-09-17 2005-09-16 Compositions monovalent for CD40L binding and methods of use
CA2581017A CA2581017C (en) 2004-09-17 2005-09-16 Compositions monovalent for cd40l binding and methods of use
AU2005283932A AU2005283932B8 (en) 2004-09-17 2005-09-16 Compositions monovalent for CD40L binding and methods of use
ES05784064.7T ES2442455T3 (en) 2004-09-17 2005-09-16 Monovalent compositions for binding to CD40L and use procedures
JP2007531827A JP5808880B2 (en) 2004-09-17 2005-09-16 Monovalent composition for CD40L binding and methods of use
KR1020077008642A KR101360140B1 (en) 2004-09-17 2005-09-16 Compositions monovalent for CD40L binding and methods of use
MX2007003187A MX2007003187A (en) 2004-09-17 2005-09-16 Compositions monovalent for cd40l binding and methods of use.
KR1020137005252A KR20130039341A (en) 2004-09-17 2005-09-16 Compositions monovalent for cd40l binding and methods of use
BRPI0515404-9A BRPI0515404A (en) 2004-09-17 2005-09-16 use of an antibody polypeptide, antibody polypeptide, cd40l antagonist, composition, extended release pharmaceutical formulation, and, specific double ligand
NZ553847A NZ553847A (en) 2004-09-17 2005-09-16 Single variable domain monovalent for binding to CD40L (gp39)
IL181724A IL181724A (en) 2004-09-17 2007-03-05 Isolated antibody polypeptides which monovalently bind to cd40l derivatives and compositions thereof
NO20071312A NO20071312L (en) 2004-09-17 2007-03-12 Bonds that are monovalent for CD40L binding and method of use
HK07108149.6A HK1101182A1 (en) 2004-09-17 2007-07-26 Compositions monovalent for cd40l binding and methods of use

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US61081904P 2004-09-17 2004-09-17
US60/610,819 2004-09-17
US11/105,512 2005-04-08
US11/102,512 US7563443B2 (en) 2004-09-17 2005-04-08 Monovalent anti-CD40L antibody polypeptides and compositions thereof

Publications (2)

Publication Number Publication Date
WO2006030220A1 WO2006030220A1 (en) 2006-03-23
WO2006030220A9 true WO2006030220A9 (en) 2006-10-19

Family

ID=36074265

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2005/003562 WO2006030220A1 (en) 2004-09-17 2005-09-16 Compositions monovalent for cd40l binding and methods of use

Country Status (16)

Country Link
US (5) US7563443B2 (en)
EP (2) EP2371862A3 (en)
JP (1) JP5808880B2 (en)
KR (2) KR101360140B1 (en)
AU (1) AU2005283932B8 (en)
BR (1) BRPI0515404A (en)
CA (1) CA2581017C (en)
ES (1) ES2442455T3 (en)
GE (1) GEP20125471B (en)
HK (1) HK1101182A1 (en)
IL (1) IL181724A (en)
MX (1) MX2007003187A (en)
NO (1) NO20071312L (en)
NZ (1) NZ553847A (en)
RU (1) RU2364420C2 (en)
WO (1) WO2006030220A1 (en)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9096651B2 (en) 2007-09-26 2015-08-04 Chugai Seiyaku Kabushiki Kaisha Method of modifying isoelectric point of antibody via amino acid substitution in CDR
US9228017B2 (en) 2009-03-19 2016-01-05 Chugai Seiyaku Kabushiki Kaisha Antibody constant region variant
US9334331B2 (en) 2010-11-17 2016-05-10 Chugai Seiyaku Kabushiki Kaisha Bispecific antibodies
US9340615B2 (en) 2009-05-15 2016-05-17 Chugai Seiyaku Kabushiki Kaisha Anti-AXL antibody
US9399680B2 (en) 2007-12-05 2016-07-26 Chugai Seiyaku Kabushiki Kaisha Nucleic acids encoding anti-NR10 antibodies

Families Citing this family (344)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2366718A3 (en) * 2002-06-28 2012-05-02 Domantis Limited Ligand
US9453251B2 (en) 2002-10-08 2016-09-27 Pfenex Inc. Expression of mammalian proteins in Pseudomonas fluorescens
PT1639011E (en) * 2003-06-30 2009-01-20 Domantis Ltd Pegylated single domain antibodies (dab)
KR20070047327A (en) 2004-07-26 2007-05-04 비오겐 아이덱 엠에이 아이엔씨. Anti-cd154 antibodies
EP1774017B1 (en) 2004-07-26 2013-05-15 Pfenex Inc. Process for improved protein expression by strain engineering
US7563443B2 (en) 2004-09-17 2009-07-21 Domantis Limited Monovalent anti-CD40L antibody polypeptides and compositions thereof
GB0521621D0 (en) 2005-10-24 2005-11-30 Domantis Ltd Tumor necrosis factor receptor 1 antagonists for treating respiratory diseases
JP5185624B2 (en) * 2004-12-02 2013-04-17 ドマンティス リミテッド Bispecific antibodies targeting serum albumin and GLP-1 or PYY
JP2008533115A (en) * 2005-03-18 2008-08-21 ドマンティス リミテッド Antibodies against Candida antigen
WO2006106905A1 (en) 2005-03-31 2006-10-12 Chugai Seiyaku Kabushiki Kaisha Process for production of polypeptide by regulation of assembly
MX2007012222A (en) 2005-04-06 2007-12-06 Bristol Myers Squibb Co Methods for treating immune disorders associated with graft transplantation with soluble ctla4 mutant molecules.
BRPI0609224B1 (en) 2005-05-18 2021-08-17 Ablynx N.V IMPROVED NANOBODIES AGAINST TUMOR NECROSIS ALPHA FACTOR
EP2532679B1 (en) * 2005-10-21 2017-04-12 Novartis AG Human antibodies against il13 and therapeutic uses
UA96139C2 (en) * 2005-11-08 2011-10-10 Дженентек, Інк. Anti-neuropilin-1 (nrp1) antibody
EP3345616A1 (en) 2006-03-31 2018-07-11 Chugai Seiyaku Kabushiki Kaisha Antibody modification method for purifying bispecific antibody
CN101479381B (en) 2006-03-31 2015-04-29 中外制药株式会社 Method for control of blood kinetics of antibody
US7993648B2 (en) 2006-05-03 2011-08-09 The Regents of the Universitry of Colorado Immunostimulatory regimen comprising administering type 1 interferon and agonistic anti-CD40 antibody
TW200812616A (en) * 2006-05-09 2008-03-16 Genentech Inc Binding polypeptides with optimized scaffolds
EP1854810A1 (en) * 2006-05-09 2007-11-14 PanGenetics B.V. Deimmunized antagonistic anti-human CD40 monoclonal antibody from the ch5D12 antibody
EP2826788B1 (en) 2006-06-21 2017-12-13 MUSC Foundation for Research Development Targeting complement factor h for treatment of diseases by the use of cr2-fh molecules
WO2008074840A2 (en) 2006-12-19 2008-06-26 Ablynx N.V. Amino acid sequences directed against a metalloproteinase from the adam family and polypeptides comprising the same for the treatment of adam-related diseases and disorders
CA2673331A1 (en) 2006-12-19 2008-06-26 Ablynx N.V. Amino acid sequences directed against gpcrs and polypeptides comprising the same for the treatment of gpcr-related diseases and disorders
US9512236B2 (en) 2006-12-19 2016-12-06 Ablynx N.V. Amino acid sequences directed against GPCRS and polypeptides comprising the same for the treatment of GPCR-related diseases and disorders
SI2125894T1 (en) * 2007-03-22 2019-05-31 Biogen Ma Inc. Binding proteins, including antibodies, antibody derivatives and antibody fragments, that specifically bind cd154 and uses thereof
US9580719B2 (en) 2007-04-27 2017-02-28 Pfenex, Inc. Method for rapidly screening microbial hosts to identify certain strains with improved yield and/or quality in the expression of heterologous proteins
CA2685326A1 (en) 2007-04-27 2008-11-06 Dow Global Technologies Inc. Method for rapidly screening microbial hosts to identify certain strains with improved yield and/or quality in the expression of heterologous proteins
JP5240870B2 (en) 2007-07-03 2013-07-17 アブリンクス エン.ヴェー. Methods for providing improved immunoglobulin sequences
TWI464262B (en) 2007-09-26 2014-12-11 中外製藥股份有限公司 Antibody constant region mutant
AU2008328781A1 (en) 2007-11-27 2009-06-04 Ablynx N.V. Amino acid sequences directed against heterodimeric cytokines and/or their receptors and polypeptides comprising the same
US20110091462A1 (en) 2008-03-05 2011-04-21 Ablynx N.V. Novel antigen binding dimer-complexes, methods of making and uses thereof
GB0809069D0 (en) 2008-05-19 2008-06-25 Univ Leuven Kath Gene signatures
CA2720763A1 (en) 2008-04-07 2009-10-15 Ablynx Nv Amino acid sequences directed against the notch pathways and uses thereof
WO2009127691A1 (en) 2008-04-17 2009-10-22 Ablynx N.V. Peptides capable of binding to serum proteins and compounds, constructs and polypeptides comprising the same
CN102099378B (en) 2008-05-16 2016-01-20 埃博灵克斯股份有限公司 For CXCR4 and other GPCRs aminoacid sequence and comprise the polypeptide of described aminoacid sequence
US9109026B2 (en) * 2008-06-03 2015-08-18 Abbvie, Inc. Dual variable domain immunoglobulins and uses thereof
US9193780B2 (en) 2008-06-05 2015-11-24 Ablynx N.V. Amino acid sequences directed against envelope proteins of a virus and polypeptides comprising the same for the treatment of viral diseases
US8802634B2 (en) 2008-07-11 2014-08-12 Phylogica Limited CD40-L Inhibitory Peptides
US20110097339A1 (en) * 2008-07-18 2011-04-28 Domantis Limited Compositions monovalent for CD28 binding and methods of use
JP2012507260A (en) * 2008-07-18 2012-03-29 ドマンティス リミテッド Monovalent compositions for CD28 binding and methods of use
US20100136584A1 (en) * 2008-09-22 2010-06-03 Icb International, Inc. Methods for using antibodies and analogs thereof
TWI440469B (en) 2008-09-26 2014-06-11 Chugai Pharmaceutical Co Ltd Improved antibody molecules
WO2010043650A2 (en) 2008-10-14 2010-04-22 Ablynx Nv Amino acid sequences directed against cellular receptors for viruses and bacteria
JP2012511014A (en) * 2008-12-05 2012-05-17 エイエルエス・セラピー・デベロップメント・インスティテュート Methods for treating neurodegenerative diseases
US9044459B2 (en) 2008-12-05 2015-06-02 Als Therapy Development Institute Method for the treatment of neurodegenerative diseases
JP5823871B2 (en) 2008-12-10 2015-11-25 アブリンクス エン.ヴェー. Amino acid sequences directed against the Angiopoietin / Tie system for the treatment of diseases and disorders associated with angiogenesis and polypeptides comprising the same
BRPI0923806A2 (en) * 2008-12-30 2015-07-14 Centocor Ortho Biotech Inc Serum markers for predicting clinical response to anti-tnfa antibodies in patients with ankylosing spondylitis.
EP2210902A1 (en) * 2009-01-14 2010-07-28 TcL Pharma Recombinant monovalent antibodies
WO2010100135A1 (en) 2009-03-05 2010-09-10 Ablynx N.V. Novel antigen binding dimer-complexes, methods of making/avoiding and uses thereof
EP3674317A1 (en) 2009-03-19 2020-07-01 Chugai Seiyaku Kabushiki Kaisha Antibody constant region variant
EP2417163B1 (en) 2009-04-10 2019-02-27 Ablynx N.V. Improved amino acid sequences directed against il-6r and polypeptides comprising the same for the treatment of il-6r related diseases and disorders
AU2010243551B2 (en) 2009-04-30 2015-03-26 Ablynx Nv Method for the production of domain antibodies
PT2437785E (en) * 2009-06-04 2015-04-20 Novartis Ag Methods for identification of sites for igg conjugation
SI2438087T1 (en) 2009-06-05 2017-10-30 Ablynx N.V. Trivalent anti human respiratory syncytial virus (hrsv) nanobody constructs for the prevention and/or treatment of respiratory tract infections
RS20120461A1 (en) 2009-07-02 2013-06-28 Musc Foundation For Research Development Methods of stimulating liver regeneration
EP2451839B1 (en) 2009-07-10 2020-04-22 Ablynx N.V. Method for the production of variable domains
CA2768462A1 (en) * 2009-07-16 2011-01-20 Glaxo Group Ltd. Improved anti-serum albumin binding single variable domains
CA2768981A1 (en) * 2009-07-29 2011-02-03 Rudolf Maria De Wildt Ligands that bind tgf-beta receptor rii
SI2805731T1 (en) 2009-09-03 2019-02-28 Ablynx N.V. Stable formulations of polypeptides and uses thereof
WO2011037158A1 (en) 2009-09-24 2011-03-31 中外製薬株式会社 Modified antibody constant regions
DK2993231T3 (en) 2009-09-24 2018-10-22 Ucb Biopharma Sprl Bacterial strain for recombinant protein expression with protease-low DEGP-retaining chaperone activity and deactivated Tsp and ptr genes
WO2011057158A1 (en) 2009-11-05 2011-05-12 Taligen Therapeutics, Inc. Treatment of paroxysmal nocturnal hemoglobinuria, hemolytic anemias and disease states involving intravascular and extravascular hemolysis
US20120269814A1 (en) * 2009-11-10 2012-10-25 Amgen Inc. Anti-c mpl antibodies
US9644022B2 (en) 2009-11-30 2017-05-09 Ablynx N.V. Amino acid sequences directed against human respiratory syncytial virus (HRSV) and polypeptides comprising the same for the prevention and/or treatment of respiratory tract infections
WO2011073180A1 (en) 2009-12-14 2011-06-23 Ablynx N.V. Single variable domain antibodies against ox40l, constructs and therapeutic use
WO2011083141A2 (en) 2010-01-08 2011-07-14 Ablynx Nv Method for generation of immunoglobulin sequences by using lipoprotein particles
GB201000587D0 (en) 2010-01-14 2010-03-03 Ucb Pharma Sa Bacterial hoist strain
GB201000590D0 (en) 2010-01-14 2010-03-03 Ucb Pharma Sa Bacterial host strain
GB201000591D0 (en) 2010-01-14 2010-03-03 Ucb Pharma Sa Bacterial hoist strain
AU2011212442A1 (en) 2010-02-05 2012-08-09 Ablynx Nv Peptides capable of binding to serum albumin and compounds, constructs and polypeptides comprising the same
US9120855B2 (en) 2010-02-10 2015-09-01 Novartis Ag Biologic compounds directed against death receptor 5
CN105380904A (en) 2010-02-11 2016-03-09 埃博灵克斯股份有限公司 Methods and compositions for the preparation of aerosols
WO2011108714A1 (en) 2010-03-04 2011-09-09 中外製薬株式会社 Antibody constant region variant
WO2011117423A1 (en) 2010-03-26 2011-09-29 Ablynx N.V. Immunoglobulin single variable domains directed against cxcr7
EP2927242A1 (en) 2010-04-09 2015-10-07 Amgen, Inc Btnl9 proteins, nucleic acids, and antibodies and uses thereof
EP2569332B1 (en) 2010-05-14 2019-01-09 The Regents of the University of Colorado, A Body Corporate Improved complement receptor 2 (cr2) targeting groups
US9932403B2 (en) 2010-05-20 2018-04-03 Ablynx Nv Biological materials related to HER3
USRE49339E1 (en) 2010-06-22 2022-12-20 The Regents Of The University Of Colorado, A Body Corporate Antibodies to the C3D fragment of complement component 3
WO2011161263A1 (en) 2010-06-25 2011-12-29 Ablynx Nv Pharmaceutical compositions for cutaneous administration
KR20130120439A (en) 2010-07-09 2013-11-04 제넨테크, 인크. Anti-neuropilin antibodies and methods of use
GB201012599D0 (en) 2010-07-27 2010-09-08 Ucb Pharma Sa Process for purifying proteins
WO2012042026A1 (en) 2010-09-30 2012-04-05 Ablynx Nv Biological materials related to c-met
KR101853405B1 (en) * 2010-10-20 2018-05-02 주식회사 티움바이오 Fusion protein having Factor IX activity
PT2632946T (en) 2010-10-29 2018-03-01 Ablynx Nv Method for the production of immunoglobulin single variable domains
WO2012058768A1 (en) 2010-11-05 2012-05-10 Zymeworks Inc. Stable heterodimeric antibody design with mutations in the fc domain
EP3575321B1 (en) 2010-11-08 2023-07-26 Ablynx N.V. Cxcr2 binding polypeptides
WO2012072731A2 (en) * 2010-12-01 2012-06-07 Glaxo Group Limited Improved anti-serum albumin binding single variable domains
CN103492565B (en) 2011-02-25 2021-01-29 中外制药株式会社 Fc gamma RIIb specific Fc antibodies
EP2691415B1 (en) 2011-03-28 2018-07-11 Ablynx N.V. Method for producing solid formulations comprising immunoglobulin single variable domains
EP2691418A1 (en) 2011-03-28 2014-02-05 Ablynx N.V. Bispecific anti-cxcr7 immunoglobulin single variable domains
SI2699601T1 (en) * 2011-04-21 2018-04-30 Bristol-Myers Squibb Company Antibody polypeptides that antagonize cd40
UA117218C2 (en) 2011-05-05 2018-07-10 Мерк Патент Гмбх Amino acid sequences directed against il-17a, il-17f and/or il17-a/f and polypeptides comprising the same
WO2012152823A1 (en) 2011-05-09 2012-11-15 Ablynx Nv Method for the production of immunoglobulin single variable domains
KR102072250B1 (en) 2011-05-27 2020-03-02 아블린쓰 엔.브이. Inhibition of bone resorption with rankl binding peptides
AU2012273928A1 (en) 2011-06-23 2014-02-06 Ablynx Nv Immunoglobulin single variable domains directed against IgE
SI2731973T1 (en) 2011-07-13 2018-04-30 Ucb Biopharma Aprl Bacterial host strain expressing recombinant dsbc
CA2842099A1 (en) 2011-07-27 2013-01-31 Glaxo Group Limited Antigen binding constructs
CN108178800B (en) * 2011-08-17 2022-06-17 葛兰素集团有限公司 Modified single variable domain antibodies with reduced binding to anti-drug antibodies
EP3311837A1 (en) 2011-09-23 2018-04-25 Ablynx NV Prolonged inhibition of interleukin-6 mediated signaling
EP2762493B1 (en) 2011-09-30 2021-06-09 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule promoting disappearance of antigens having plurality of biological activities
TW201326209A (en) 2011-09-30 2013-07-01 Chugai Pharmaceutical Co Ltd Therapeutic antigen-binding molecule with a FcRn-binding domain that promotes antigen clearance
EP2747783B1 (en) 2011-09-30 2017-06-14 Ablynx N.V. Biological materials related to c-met
CN104245732B (en) * 2011-10-13 2020-04-07 百时美施贵宝公司 Antibody polypeptides antagonizing CD40L
CN109897103A (en) 2011-11-04 2019-06-18 酵活有限公司 There is the antibody design of the stabilization heterodimeric of mutation in Fc structural domain
CA2854921A1 (en) 2011-11-23 2013-05-30 Amgen Inc. Methods of treatment using an antibody against interferon gamma
EP2847230B1 (en) 2012-05-10 2020-08-12 Zymeworks Inc. Heteromultimer constructs of immunoglobulin heavy chains with mutations in the fc domain
DK3207938T3 (en) * 2012-05-11 2020-03-16 Medimmune Ltd CTLA-4 VARIANTS
GB201208367D0 (en) 2012-05-14 2012-06-27 Ucb Pharma Sa Biological product
WO2014004586A1 (en) 2012-06-25 2014-01-03 Zymeworks Inc. Process and methods for efficient manufacturing of highly pure asymmetric antibodies in mammalian cells
CA2878400A1 (en) 2012-07-19 2014-01-23 Amgen Inc. Btnl3 proteins, nucleic acids, and antibodies and uses thereof
US10413620B2 (en) 2012-08-17 2019-09-17 The Regents Of The University Of Colorado, A Body Corporate Light-emitting versions of the monoclonal antibody to C3D (MAB 3D29) for imaging
AU2013302441B2 (en) 2012-08-17 2018-05-10 The Regents Of The University Of Colorado, A Body Corporate Compositions and methods for detecting complement activation
EP2906683B1 (en) 2012-10-15 2017-05-31 Bristol-Myers Squibb Company Mammalian cell culture processes for protein production
US9914785B2 (en) 2012-11-28 2018-03-13 Zymeworks Inc. Engineered immunoglobulin heavy chain-light chain pairs and uses thereof
CA2910632A1 (en) 2013-04-29 2014-12-04 Agrosavfe N.V. Compositions comprising polypeptides that bind glucosylceramide of a fungal pest
WO2016071438A2 (en) 2014-11-05 2016-05-12 Agrosavfe Nv Transgenic plant comprising a polynucleotide encoding a variable domain of heavy-chain antibody
PT3050896T (en) 2013-09-27 2021-08-24 Chugai Pharmaceutical Co Ltd Method for producing polypeptide heteromultimer
EP2883883A1 (en) 2013-12-16 2015-06-17 Cardio3 Biosciences S.A. Therapeutic targets and agents useful in treating ischemia reperfusion injury
GB201403775D0 (en) 2014-03-04 2014-04-16 Kymab Ltd Antibodies, uses & methods
US10435475B2 (en) 2014-03-07 2019-10-08 Bristol-Myers Squibb Company Method of using antibody polypeptides that antagonize CD40 to treat IBD
NL2013007B1 (en) 2014-06-16 2016-07-05 Ablynx Nv Methods of treating TTP with immunoglobulin single variable domains and uses thereof.
NL2013661B1 (en) 2014-10-21 2016-10-05 Ablynx Nv KV1.3 Binding immunoglobulins.
MA40764A (en) 2014-09-26 2017-08-01 Chugai Pharmaceutical Co Ltd THERAPEUTIC AGENT INDUCING CYTOTOXICITY
CA2963037A1 (en) 2014-10-10 2016-04-14 Ablynx N.V. Treatment of rsv infection
TR201909589T4 (en) 2014-10-10 2019-07-22 Ablynx Nv Inhalation device for use in aerosol therapy of respiratory diseases.
JP6830437B2 (en) 2014-12-10 2021-02-17 リージェンツ オブ ザ ユニバーシティ オブ ミネソタ Genetically modified cells, tissues and organs to treat the disease
MA41294A (en) 2014-12-19 2017-11-08 Chugai Pharmaceutical Co Ltd ANTI-MYOSTATIN ANTIBODIES, POLYPEPTIDES CONTAINING FC REGION VARIANTS, AND METHODS OF USE
EP3233921B1 (en) 2014-12-19 2021-09-29 Chugai Seiyaku Kabushiki Kaisha Anti-c5 antibodies and methods of use
PT3233910T (en) 2014-12-19 2020-03-17 Ablynx Nv Cysteine linked nanobody dimers
GB201501613D0 (en) 2015-01-30 2015-03-18 Ucb Biopharma Sprl Treatment of autoimmune disorders with CD154 antibodies
MA41459A (en) 2015-02-03 2017-12-12 Als Therapy Development Inst ANTI-CD40L ANTIBODIES AND METHODS FOR TREATING CD40L ILLNESSES OR DISORDERS
WO2016124568A1 (en) 2015-02-03 2016-08-11 INSERM (Institut National de la Santé et de la Recherche Médicale) Anti-rho gtpase conformational single domain antibodies and uses thereof
KR102605798B1 (en) 2015-02-05 2023-11-23 추가이 세이야쿠 가부시키가이샤 Antibodies comprising an ion concentration dependent antigen-binding domain, fc region variants, il-8-binding antibodies, and uses therof
WO2016135041A1 (en) 2015-02-26 2016-09-01 INSERM (Institut National de la Santé et de la Recherche Médicale) Fusion proteins and antibodies comprising thereof for promoting apoptosis
TWI759261B (en) 2015-02-27 2022-04-01 日商中外製藥股份有限公司 Use of IL-6 receptor antibody for preparing pharmaceutical composition
US9139653B1 (en) 2015-04-30 2015-09-22 Kymab Limited Anti-human OX40L antibodies and methods of treatment
US9512229B2 (en) 2015-03-03 2016-12-06 Kymab Limited Synergistic combinations of OX40L antibodies for the treatment of GVHD
US9434785B1 (en) 2015-04-30 2016-09-06 Kymab Limited Anti-human OX40L antibodies and methods of treating graft versus host disease with the same
US11142587B2 (en) 2015-04-01 2021-10-12 Chugai Seiyaku Kabushiki Kaisha Method for producing polypeptide hetero-oligomer
US10259882B2 (en) 2015-05-07 2019-04-16 Agenus Inc. Anti-OX40 antibodies
US10927186B2 (en) 2015-05-13 2021-02-23 Ablynx N.V. T cell recruiting polypeptides based on TCR alpha/beta reactivity
CA2985700C (en) 2015-05-13 2022-06-28 Ablynx N.V. T cell recruiting polypeptides based on cd3 reactivity
EP3331563B1 (en) 2015-08-05 2023-04-19 Janssen Biotech, Inc. Anti-cd154 antibodies and methods of using them
CN105384825B (en) 2015-08-11 2018-06-01 南京传奇生物科技有限公司 A kind of bispecific chimeric antigen receptor and its application based on single domain antibody
US20170059561A1 (en) * 2015-08-28 2017-03-02 The Florida International University Board Of Trustees Thermally Stable Electrochemical Sensor With Long Shelf-Life
US11078251B2 (en) 2015-09-18 2021-08-03 INSERM (Institut National de la Santé et de la Recherche Médicale) T cell receptors (TCR) and uses thereof for the diagnosis and treatment of diabetes
WO2017055484A1 (en) 2015-09-29 2017-04-06 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for determining the metabolic status of lymphomas
EP3362088B1 (en) 2015-10-12 2020-11-25 Institut National de la Sante et de la Recherche Medicale (INSERM) An agent capable of depleting cd8 t cells for the treatment of myocardial infarction or acute myocardial infarction
WO2017081190A1 (en) 2015-11-13 2017-05-18 INSERM (Institut National de la Santé et de la Recherche Médicale) Anti- nkg2d single domain antibodies and uses thereof
JP7256011B2 (en) 2015-11-27 2023-04-11 アブリンクス エン.ヴェー. Polypeptides that inhibit CD40L
EP3383430A4 (en) 2015-12-02 2019-12-18 Agenus Inc. Antibodies and methods of use thereof
JP7141336B2 (en) 2015-12-25 2022-09-22 中外製薬株式会社 Anti-myostatin antibodies and methods of use
CN108368166B (en) 2015-12-28 2023-03-28 中外制药株式会社 Method for improving purification efficiency of polypeptide containing FC region
US20200264165A1 (en) 2016-01-04 2020-08-20 Inserm (Institut National De La Sante Et De Larecherche Medicale) Use of pd-1 and tim-3 as a measure for cd8+ cells in predicting and treating renal cell carcinoma
WO2017129558A1 (en) 2016-01-25 2017-08-03 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for predicting or treating myelopoiesis-driven cardiometabolic diseases and sepsis
ES2924741T3 (en) 2016-01-28 2022-10-10 Inst Nat Sante Rech Med Methods to Increase the Potency of Immune Checkpoint Inhibitors
WO2017129763A1 (en) 2016-01-28 2017-08-03 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and pharmaceutical compositions for the treatment of signet ring cell gastric cancer
WO2017129790A1 (en) 2016-01-28 2017-08-03 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and pharmaceutical composition for the treatment of cancer
AU2017233658B2 (en) 2016-03-14 2023-09-21 Chugai Seiyaku Kabushiki Kaisha Cell injury inducing therapeutic drug for use in cancer therapy
WO2017174681A1 (en) 2016-04-06 2017-10-12 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and pharmaceutical compositions for the treatment of age-related cardiometabolic diseases
WO2017182609A1 (en) 2016-04-22 2017-10-26 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and pharmaceutical composition for the treatment of inflammatory skin diseases associated with desmoglein-1 deficiency
EP3452505A1 (en) 2016-05-02 2019-03-13 Ablynx NV Treatment of rsv infection
JP7166512B2 (en) 2016-05-03 2022-11-08 インスティチュート ナショナル デ ラ サンテ エ デ ラ ルシェルシュ メディカル (インセルム) CD31shed as a molecular beacon for imaging inflammation
WO2017191300A1 (en) 2016-05-06 2017-11-09 INSERM (Institut National de la Santé et de la Recherche Médicale) Pharmaceutical compositions for the treatment of chemoresistant acute myeloid leukemia (aml)
EP3454863A1 (en) 2016-05-10 2019-03-20 INSERM (Institut National de la Santé et de la Recherche Médicale) Combinations therapies for the treatment of cancer
WO2017202962A1 (en) 2016-05-24 2017-11-30 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and pharmaceutical compositions for the treatment of non small cell lung cancer (nsclc) that coexists with chronic obstructive pulmonary disease (copd)
WO2017202890A1 (en) 2016-05-27 2017-11-30 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and compositions for predicting and treating myeloma
WO2018007442A1 (en) 2016-07-06 2018-01-11 Ablynx N.V. Treatment of il-6r related diseases
WO2018014260A1 (en) 2016-07-20 2018-01-25 Nanjing Legend Biotech Co., Ltd. Multispecific antigen binding proteins and methods of use thereof
US11053308B2 (en) 2016-08-05 2021-07-06 Chugai Seiyaku Kabushiki Kaisha Method for treating IL-8-related diseases
WO2018029182A1 (en) 2016-08-08 2018-02-15 Ablynx N.V. Il-6r single variable domain antibodies for treatment of il-6r related diseases
US11246905B2 (en) 2016-08-15 2022-02-15 President And Fellows Of Harvard College Treating infections using IdsD from Proteus mirabilis
WO2018041989A1 (en) 2016-09-02 2018-03-08 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for diagnosing and treating refractory celiac disease type 2
CN110248674B (en) * 2016-09-07 2021-10-26 萨辛生命科学有限公司 Synthetic antibodies against VEGF and uses thereof
EP3510407A1 (en) 2016-09-08 2019-07-17 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for diagnosing and treating nephrotic syndrome
WO2018050833A1 (en) 2016-09-15 2018-03-22 Ablynx Nv Immunoglobulin single variable domains directed against macrophage migration inhibitory factor
WO2018068201A1 (en) 2016-10-11 2018-04-19 Nanjing Legend Biotech Co., Ltd. Single-domain antibodies and variants thereof against ctla-4
WO2018083248A1 (en) 2016-11-03 2018-05-11 Kymab Limited Antibodies, combinations comprising antibodies, biomarkers, uses & methods
WO2018089628A1 (en) 2016-11-09 2018-05-17 Agenus Inc. Anti-ox40 antibodies, anti-gitr antibodies, and methods of use thereof
WO2018087391A1 (en) 2016-11-14 2018-05-17 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and pharmaceutical compositions for modulating stem cells proliferation or differentiation
KR20190080934A (en) 2016-11-16 2019-07-08 아블린쓰 엔.브이. CD123 and T cell mobilization polypeptides capable of binding to TCR alpha / beta
WO2018091621A1 (en) 2016-11-17 2018-05-24 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and pharmaceutical compositions for increasing endogenous protein level
WO2018099968A1 (en) 2016-11-29 2018-06-07 Ablynx N.V. Treatment of infection by respiratory syncytial virus (rsv)
EP3548894B1 (en) 2016-12-02 2021-10-20 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and compositions for diagnosing renal cell carcinoma
WO2018134389A1 (en) 2017-01-23 2018-07-26 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and compositions for treating infections
JP7341060B2 (en) 2017-02-10 2023-09-08 アンスティチュ ナショナル ドゥ ラ サンテ エ ドゥ ラ ルシェルシュ メディカル Methods and pharmaceutical compositions for the treatment of cancer associated with MAPK pathway activation
JP7186401B2 (en) 2017-02-28 2022-12-09 フエー・イー・ベー・フエー・ゼツト・ウエー Means and methods for oral delivery of proteins
US11440951B2 (en) 2017-03-13 2022-09-13 The Government Of The United States, As Represented By The Secretary Of The Army Therapeutic antibodies to Marburg virus
EP3600427A1 (en) 2017-03-24 2020-02-05 INSERM - Institut National de la Santé et de la Recherche Médicale Methods and compositions for treating melanoma
WO2018178029A1 (en) 2017-03-27 2018-10-04 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and compositions for treating degenerative muscular and/or neurological conditions or diseases
WO2018178030A1 (en) 2017-03-27 2018-10-04 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and compositions for treating degenerative muscular and/or neurological conditions or diseases
WO2018178078A1 (en) 2017-03-28 2018-10-04 INSERM (Institut National de la Santé et de la Recherche Médicale) New tau species
WO2018178237A1 (en) 2017-03-30 2018-10-04 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for the treatment of mitochondrial genetic diseases
WO2018203545A1 (en) 2017-05-02 2018-11-08 国立研究開発法人国立精神・神経医療研究センター Method for predicting and evaluating therapeutic effect in diseases related to il-6 and neutrophils
US11891451B2 (en) 2017-05-11 2024-02-06 Vib Vzw Glycosylation of variable immunoglobulin domains
KR20200013683A (en) 2017-05-17 2020-02-07 인쎄름 (엥스띠뛰 나씨오날 드 라 쌍떼 에 드 라 흐쉐르슈 메디깔) FLT3 inhibitors for improving pain treatment by opioids
KR20200010294A (en) 2017-05-24 2020-01-30 에이엘에스 테라피 디벨럽먼트 인스티튜트 Therapeutic Anti-CD40 Ligand Antibodies
JP7320457B2 (en) 2017-06-02 2023-08-03 アブリンクス エン.ヴェー. aggrecan-binding immunoglobulin
TWI826376B (en) 2017-06-02 2023-12-21 德商麥克專利有限公司 Polypeptides binding adamts5, mmp13 and aggrecan
MX2019014448A (en) 2017-06-02 2020-02-10 Merck Patent Gmbh Mmp13 binding immunoglobulins.
EP3630847A1 (en) 2017-06-02 2020-04-08 Merck Patent GmbH Adamts binding immunoglobulins
WO2018224615A1 (en) 2017-06-08 2018-12-13 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and compositions for treating hyperpigmentation disorders
JP7265487B2 (en) 2017-06-08 2023-04-26 アンスティチュ ナショナル ドゥ ラ サンテ エ ドゥ ラ ルシェルシュ メディカル Chimeric receptors for use in whole-cell sensors for detecting analytes of interest
WO2018234843A1 (en) 2017-06-22 2018-12-27 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and pharmaceutical compositions for the treatment of fibrosis with agents capable of inhibiting the activation of mucosal-associated invariant t (mait) cells
WO2019000223A1 (en) 2017-06-27 2019-01-03 Nanjing Legend Biotech Co., Ltd. Chimeric antibody immune effctor cell engagers and methods of use thereof
WO2019002548A1 (en) 2017-06-29 2019-01-03 INSERM (Institut National de la Santé et de la Recherche Médicale) Treating migraine by agonising trek1, trek2 or heteromers including them
US11155607B2 (en) 2017-07-19 2021-10-26 Vib Vzw Serum albumin binding agents
WO2019016310A1 (en) 2017-07-20 2019-01-24 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and compositions for treating cancers
EP3658581A1 (en) 2017-07-24 2020-06-03 INSERM (Institut National de la Santé et de la Recherche Médicale) Antibodies and peptides to treat hcmv related diseases
WO2019020593A1 (en) 2017-07-25 2019-01-31 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and pharmaceutical compositions for modulating monocytopoiesis
WO2019043026A1 (en) 2017-08-30 2019-03-07 INSERM (Institut National de la Santé et de la Recherche Médicale) Anti-mesothelin radiolabelled single domain antibodies suitable for the imaging and treatment of cancers
EP3457139A1 (en) 2017-09-19 2019-03-20 Promise Advanced Proteomics Antibody-like peptides for quantifying therapeutic antibodies
KR20200063139A (en) * 2017-09-19 2020-06-04 엠에이비 디스커버리 게엠베하 Efficacy CD40 antibody
EP3684471A1 (en) 2017-09-20 2020-07-29 Institut National de la Sante et de la Recherche Medicale (INSERM) Methods and pharmaceutical compositions for modulating autophagy
US11873347B2 (en) 2017-10-31 2024-01-16 Vib Vzw Antigen-binding chimeric proteins and methods and uses thereof
MA50893A (en) 2017-11-15 2020-09-23 Novo Nordisk As X-FACTOR BINDERS REINFORCING FX ACTIVATION
WO2019101995A1 (en) 2017-11-27 2019-05-31 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and pharmaceutical compositions for cardiac regeneration
JP7369127B2 (en) 2017-12-28 2023-10-25 ナンジン レジェンド バイオテック カンパニー,リミテッド Single domain antibodies against TIGIT and variants thereof
US20210072244A1 (en) 2018-01-04 2021-03-11 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and compositions for treating melanoma resistant
WO2019137541A1 (en) 2018-01-15 2019-07-18 Nanjing Legend Biotech Co., Ltd. Single-domain antibodies and variants thereof against pd-1
EP3743096A1 (en) 2018-01-25 2020-12-02 INSERM (Institut National de la Santé et de la Recherche Médicale) Antagonists of il-33 for use in methods for preventing ischemia reperfusion injury in an organ
JP2021512947A (en) 2018-02-06 2021-05-20 アブリンクス・エヌ・フェー How to treat the first episode of TTP with an immunoglobulin single variable domain
EP3752134A1 (en) 2018-02-16 2020-12-23 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and compositions for treating vitiligo
US11858960B2 (en) 2018-03-01 2024-01-02 Vrije Universiteit Brussel Human PD-L1-binding immunoglobulins
WO2019180204A1 (en) 2018-03-23 2019-09-26 Université Libre de Bruxelles Wnt signaling agonist molecules
WO2019193375A1 (en) 2018-04-04 2019-10-10 INSERM (Institut National de la Santé et de la Recherche Médicale) Use of fzd7 inhibitors for the treatment of retinal neovascularization
WO2019207030A1 (en) 2018-04-26 2019-10-31 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for predicting a response with an immune checkpoint inhibitor in a patient suffering from a lung cancer
CN112654397A (en) 2018-09-05 2021-04-13 国家医疗保健研究所 Methods and compositions for treating asthma and allergic diseases
US20220048995A1 (en) 2018-09-10 2022-02-17 INSERM (Institut National de la Santé et de la Recherche Médicale) Use of an inhibitor of ntsr1 activation or expression for preventing weight loss, muscle loss, and protein blood level decrease in subjects in need thereof
US20220047567A1 (en) 2018-09-10 2022-02-17 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for the treatment of neurofibromatosis
WO2020058201A1 (en) 2018-09-17 2020-03-26 INSERM (Institut National de la Santé et de la Recherche Médicale) Use of inhibitors of phosphatase activity of soluble epoxide for the treatment of cardiometabolic diseases
JP2022511337A (en) 2018-09-19 2022-01-31 インサーム (インスティテュート ナショナル デ ラ サンテ エ デ ラ ルシェルシェ メディカル) Methods and Pharmaceutical Compositions for the Treatment of Cancers Resistant to Immune Checkpoint Treatment
EP3856772A1 (en) 2018-09-25 2021-08-04 Institut National de la Santé et de la Recherche Médicale (INSERM) Use of antagonists of th17 cytokines for the treatment of bronchial remodeling in patients suffering from allergic asthma
WO2020070062A1 (en) 2018-10-01 2020-04-09 INSERM (Institut National de la Santé et de la Recherche Médicale) Use of tim-3 inhibitors for the treatment of exacerbations in patients suffering from severe asthma
JP2022512626A (en) 2018-10-04 2022-02-07 アンスティチュ ナショナル ドゥ ラ サンテ エ ドゥ ラ ルシェルシュ メディカル Methods and pharmaceutical compositions for the treatment of mucosal inflammatory diseases
EP3860653A1 (en) 2018-10-05 2021-08-11 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and systems for controlling the agonistic properties of antibody variable domains by light
EP3636657A1 (en) 2018-10-08 2020-04-15 Ablynx N.V. Chromatography-free antibody purification method
US20210347898A1 (en) 2018-10-09 2021-11-11 INSERM (Institut National de la Santé et de la Recherche Médicale) Use of alpha-v-integrin (cd51) inhibitors for the treatment of cardiac fibrosis
WO2020079162A1 (en) 2018-10-18 2020-04-23 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for inducing full ablation of hematopoiesis
WO2020104479A1 (en) 2018-11-20 2020-05-28 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and compositions for treating cancers and resistant cancers with anti transferrin receptor 1 antibodies
WO2020115261A1 (en) 2018-12-07 2020-06-11 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and compositions for treating melanoma
WO2020120592A1 (en) 2018-12-12 2020-06-18 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and compositions for predicting and treating melanoma
WO2020120644A1 (en) 2018-12-13 2020-06-18 INSERM (Institut National de la Santé et de la Recherche Médicale) New anti tau svqivykpv epitope single domain antibody
WO2020127885A1 (en) 2018-12-21 2020-06-25 INSERM (Institut National de la Santé et de la Recherche Médicale) Compositions for treating cancers and resistant cancers
EP4059569A1 (en) 2019-01-03 2022-09-21 Institut National De La Sante Et De La Recherche Medicale (Inserm) Methods and pharmaceutical compositions for enhancing cd8+ t cell-dependent immune responses in subjects suffering from cancer
EP3921031A1 (en) 2019-02-04 2021-12-15 Institut National de la Santé et de la Recherche Médicale (INSERM) Methods and compositions for modulating blood-brain barrier
WO2020169707A1 (en) 2019-02-21 2020-08-27 INSERM (Institut National de la Santé et de la Recherche Médicale) Foxo1 inhibitor for use in the treatment of latent virus infection
WO2020178313A1 (en) 2019-03-05 2020-09-10 INSERM (Institut National de la Santé et de la Recherche Médicale) New biomarkers and biotargets in renal cell carcinoma
JP2022524768A (en) 2019-03-08 2022-05-10 リンクシス ベスローテン フェンノートシャップ Internalized binding molecule that targets receptors involved in cell proliferation or cell differentiation
JP2022526334A (en) 2019-03-25 2022-05-24 アンスティチュ ナショナル ドゥ ラ サンテ エ ドゥ ラ ルシェルシュ メディカル Methods of Treatment of Tauopathy Disorders by Targeting New Tau Species
EP3946330A1 (en) 2019-03-29 2022-02-09 Institut National de la Santé et de la Recherche Médicale (INSERM) Methods for the treatment of keloid, hypertrophic scars and/or hyperpigmentation disorders
EP3947737A2 (en) 2019-04-02 2022-02-09 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods of predicting and preventing cancer in patients having premalignant lesions
US20220175815A1 (en) 2019-04-03 2022-06-09 Orega Biotech Combination therapies based on pd1 and il-17b inhibitors
US20220220565A1 (en) 2019-04-30 2022-07-14 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and compositions for treating melanoma
US11434291B2 (en) 2019-05-14 2022-09-06 Provention Bio, Inc. Methods and compositions for preventing type 1 diabetes
WO2020239934A1 (en) 2019-05-28 2020-12-03 Vib Vzw Cd8+ t-cells lacking plexins and their application in cancer treatment
EP3976650A1 (en) 2019-05-28 2022-04-06 Vib Vzw Cancer treatment by targeting plexins in the immune compartment
CN114206938A (en) 2019-06-20 2022-03-18 国家医疗保健研究所 Anti-protease NEXIN-1 conformational single domain antibodies and uses thereof
CN114401743A (en) 2019-08-02 2022-04-26 法国国家健康和医学研究院 Use of neutralizing granzyme B for providing cardioprotection in a subject who has experienced a myocardial infarction
WO2021048292A1 (en) 2019-09-11 2021-03-18 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and compositions for treating melanoma
WO2021063968A1 (en) 2019-09-30 2021-04-08 INSERM (Institut National de la Santé et de la Recherche Médicale) Method and composition for diagnosing chronic obstructive pulmonary disease
US20220354811A1 (en) 2019-10-03 2022-11-10 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and compositions for modulating macrophages polarization
WO2021064184A1 (en) 2019-10-04 2021-04-08 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and pharmaceutical composition for the treatment of ovarian cancer, breast cancer or pancreatic cancer
WO2021078359A1 (en) 2019-10-21 2021-04-29 INSERM (Institut National de la Santé et de la Recherche Médicale) Use of inhibitors of cubilin for the treatment of chronic kidney diseases
WO2021094308A1 (en) 2019-11-12 2021-05-20 INSERM (Institut National de la Santé et de la Recherche Médicale) New serological marker for the latent form of toxoplasmosis
GB201918279D0 (en) 2019-12-12 2020-01-29 Vib Vzw Glycosylated single chain immunoglobulin domains
WO2021144426A1 (en) 2020-01-17 2021-07-22 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and compositions for treating melanoma
EP4110810A1 (en) 2020-02-28 2023-01-04 Orega Biotech Combination therapies based on ctla4 and il-17b inhibitors
MX2022012376A (en) 2020-03-31 2023-02-15 Biotalys NV Anti-fungal polypeptides.
WO2021198511A1 (en) 2020-04-03 2021-10-07 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and compositions for treatment of sars-cov-2 infection
CN113527488A (en) 2020-04-22 2021-10-22 迈威(上海)生物科技股份有限公司 Single variable domain antibody targeting human programmed death ligand 1(PD-L1) and derivative thereof
WO2021224401A1 (en) 2020-05-07 2021-11-11 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and compositions for determining a reference range of β-galactose exposure platelet
EP4149558A1 (en) 2020-05-12 2023-03-22 INSERM (Institut National de la Santé et de la Recherche Médicale) New method to treat cutaneous t-cell lymphomas and tfh derived lymphomas
WO2021229104A1 (en) 2020-05-15 2021-11-18 Université de Liège Anti-cd38 single-domain antibodies in disease monitoring and treatment
US20230302050A1 (en) 2020-05-20 2023-09-28 Institut Curie Single Domain Antibodies and Their Use in Cancer Therapies
EP4153627A1 (en) 2020-05-20 2023-03-29 Institut Curie Synthetic single domain library
EP4161583A1 (en) 2020-06-05 2023-04-12 Institut National de la Santé et de la Recherche Médicale (INSERM) Methods and pharmaceutical compositions for treating ocular diseases
KR20230030644A (en) 2020-06-29 2023-03-06 인쎄름 (엥스띠뛰 나씨오날 드 라 쌍떼 에 드 라 흐쉐르슈 메디깔) Anti-protein single-domain antibodies and polypeptides comprising them
WO2022008597A1 (en) 2020-07-08 2022-01-13 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and pharmaceutical composition for the treatment of infectious diseases
EP4189395A1 (en) 2020-07-28 2023-06-07 Institut National de la Santé et de la Recherche Médicale (INSERM) Methods and compositions for preventing and treating a cancer
US20230323299A1 (en) 2020-08-03 2023-10-12 Inserm (Institut National De La Santé Et De La Recherch Médicale) Population of treg cells functionally committed to exert a regulatory activity and their use for adoptive therapy
EP4214235A1 (en) 2020-09-16 2023-07-26 LinXis B.V. Internalizing binding molecules
WO2022063957A1 (en) 2020-09-24 2022-03-31 Vib Vzw Biomarker for anti-tumor therapy
CA3196737A1 (en) 2020-09-24 2022-03-31 Massimiliano Mazzone Combination of p2y6 inhibitors and immune checkpoint inhibitors
WO2022064049A1 (en) 2020-09-28 2022-03-31 INSERM (Institut National de la Santé et de la Recherche Médicale) Method for diagnosing brucella infection
EP4229090A1 (en) 2020-10-16 2023-08-23 Université d'Aix-Marseille Anti-gpc4 single domain antibodies
WO2022084300A1 (en) 2020-10-20 2022-04-28 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for diagnosis and monitoring form of coronavirus infection
WO2022084531A1 (en) 2020-10-23 2022-04-28 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and compositions for treating glioma
EP4240761A1 (en) 2020-11-05 2023-09-13 Institut National de la Santé et de la Recherche Médicale (INSERM) Use of il-6 inhibitors for the treatment of acute chest syndrome in patients suffering from sickle cell disease
WO2022101481A1 (en) 2020-11-16 2022-05-19 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and compositions for predicting and treating uveal melanoma
WO2022120388A2 (en) 2020-12-04 2022-06-09 Tidal Therapeutics, Inc. Ionizable cationic lipids and lipid nanoparticles, and methods of synthesis and use thereof
TW202222840A (en) 2020-12-11 2022-06-16 大陸商上海華奧泰生物藥業股份有限公司 Cd73 antigen binding protein and the application thereof
WO2022148480A1 (en) 2021-01-11 2022-07-14 星济生物(苏州)有限公司 ANTIGEN-BINDING PROTEIN TARGETING STAPHYLOCOCCUS AUREUS α-HEMOLYSIS AND APPLICATION THEREOF
JP2024502758A (en) 2021-01-13 2024-01-23 上海華奥泰生物藥業股▲フン▼有限公司 CD73 binding protein and its use
WO2022152862A1 (en) 2021-01-14 2022-07-21 Institut Curie Her2 single domain antibodies variants and cars thereof
EP4282413A1 (en) 2021-01-21 2023-11-29 Natural Medicine Institute Of Zhejiang Yangshengtang Co., Ltd. Composition and method for treating tumors
WO2022169825A1 (en) 2021-02-03 2022-08-11 Mozart Therapeutics, Inc. Binding agents and methods of using the same
WO2022175392A1 (en) 2021-02-17 2022-08-25 Vib Vzw Inhibition of slc4a4 in the treatment of cancer
AU2022222994A1 (en) 2021-02-19 2023-09-28 Seoul National University R&Db Foundation Single domain antibody against cd47 and use thereof
AU2022222423A1 (en) 2021-02-19 2023-09-28 Seoul National University R&Db Foundation Single domain antibody against pd-l1 and use thereof
CN116917322A (en) 2021-02-19 2023-10-20 沙裴隆有限公司 Bispecific single domain antibodies against PD-L1 and CD47 and uses thereof
CN117321076A (en) 2021-02-19 2023-12-29 美国卫生及公众服务部代表 Single domain antibodies neutralizing SARS-CoV-2
WO2022194908A1 (en) 2021-03-17 2022-09-22 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and compositions for treating melanoma
KR20230162797A (en) 2021-03-22 2023-11-28 스타맵 바이오로직스 (쑤저우) 씨오., 엘티디 Antigen-binding protein targeting pneumolysin and uses thereof
KR20240012352A (en) 2021-03-23 2024-01-29 인쎄름 (엥스띠뛰 나씨오날 드 라 쌍떼 에 드 라 흐쉐르슈 메디깔) How to Diagnose and Treat T-Cell Lymphoma
KR20230166096A (en) 2021-04-02 2023-12-06 오리셀 테라퓨틱스 컴퍼니 리미티드 CLDN18.2 antigen binding protein and its applications
WO2022214681A1 (en) 2021-04-09 2022-10-13 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for the treatment of anaplastic large cell lymphoma
EP4326871A1 (en) 2021-04-19 2024-02-28 INSERM (Institut National de la Santé et de la Recherche Médicale) Use of splice switching oligonucleotides for exon skipping-mediated knockdown of pim2
EP4326903A1 (en) 2021-04-23 2024-02-28 Inserm (Institut National De La Sante Et De La Recherche Medicale) Methods and compositions for treating cell senescence accumulation related disease
WO2022242892A1 (en) 2021-05-17 2022-11-24 Université de Liège Anti-cd38 single-domain antibodies in disease monitoring and treatment
US20230174651A1 (en) 2021-06-23 2023-06-08 Janssen Biotech, Inc. Materials and methods for hinge regions in functional exogenous receptors
IL309559A (en) 2021-07-09 2024-02-01 Luxembourg Inst Of Health Lih Dimeric protein complexes and uses thereof
WO2023285362A1 (en) 2021-07-12 2023-01-19 INSERM (Institut National de la Santé et de la Recherche Médicale) Use of il-36 inhibitors for the treatment of netherton syndrome
WO2023006975A2 (en) 2021-07-30 2023-02-02 INSERM (Institut National de la Santé et de la Recherche Médicale) Chimeric proteins and methods of immunotherapy
WO2023057601A1 (en) 2021-10-06 2023-04-13 Biotalys NV Anti-fungal polypeptides
WO2023076876A1 (en) 2021-10-26 2023-05-04 Mozart Therapeutics, Inc. Modulation of immune responses to viral vectors
WO2023078900A1 (en) 2021-11-03 2023-05-11 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and compositions for treating triple negative breast cancer (tnbc)
WO2023110937A1 (en) 2021-12-14 2023-06-22 INSERM (Institut National de la Santé et de la Recherche Médicale) Depletion of nk cells for the treatment of adverse post-ischemic cardiac remodeling
WO2023118165A1 (en) 2021-12-21 2023-06-29 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and compositions for treating melanoma
EP4209508A1 (en) 2022-01-11 2023-07-12 Centre national de la recherche scientifique Nanobodies for the deneddylating enzyme nedp1
WO2023144235A1 (en) 2022-01-27 2023-08-03 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for monitoring and treating warburg effect in patients with pi3k-related disorders
WO2023144303A1 (en) 2022-01-31 2023-08-03 INSERM (Institut National de la Santé et de la Recherche Médicale) Cd38 as a biomarker and biotarget in t-cell lymphomas
EP4231017A1 (en) 2022-02-17 2023-08-23 Promise Proteomics Detection and quantification of anti-drug antibodies and anti-self antibodies
EP4257609A1 (en) 2022-04-08 2023-10-11 iOmx Therapeutics AG Combination therapies based on pd-1 inhibitors and sik3 inhibitors
WO2023198648A1 (en) 2022-04-11 2023-10-19 Institut National de la Santé et de la Recherche Médicale Methods for the diagnosis and treatment of t-cell malignancies
WO2023198851A1 (en) 2022-04-14 2023-10-19 Institut National de la Santé et de la Recherche Médicale Methods for controlling the tumor cell killing by light
WO2023198874A1 (en) 2022-04-15 2023-10-19 Institut National de la Santé et de la Recherche Médicale Methods for the diagnosis and treatment of t cell-lymphomas
WO2023217904A1 (en) 2022-05-10 2023-11-16 Institut National de la Santé et de la Recherche Médicale Syncitin-1 fusion proteins and uses thereof for cargo delivery into target cells
US20240002331A1 (en) 2022-06-08 2024-01-04 Tidal Therapeutics, Inc. Ionizable cationic lipids and lipid nanoparticles, and methods of synthesis and use thereof
WO2023237661A1 (en) 2022-06-09 2023-12-14 Institut National de la Santé et de la Recherche Médicale Use of endothelin receptor type b agonists for the treatment of aortic valve stenosis
EP4299124A1 (en) 2022-06-30 2024-01-03 Universite De Montpellier Anti-mglur2 nanobodies for use as biomolecule transporter
WO2024003310A1 (en) 2022-06-30 2024-01-04 Institut National de la Santé et de la Recherche Médicale Methods for the diagnosis and treatment of acute lymphoblastic leukemia
WO2024008274A1 (en) 2022-07-04 2024-01-11 Universiteit Antwerpen T regulatory cell modification
WO2024008755A1 (en) 2022-07-04 2024-01-11 Vib Vzw Blood-cerebrospinal fluid barrier crossing antibodies
WO2024008799A1 (en) 2022-07-06 2024-01-11 Institut National de la Santé et de la Recherche Médicale Methods for the treatment of proliferative glomerulonephritis
WO2024013234A1 (en) 2022-07-13 2024-01-18 Institut National de la Santé et de la Recherche Médicale Methods for diagnosis, prognosis, stratification and treating of myocarditis
WO2024018003A1 (en) 2022-07-21 2024-01-25 Institut National de la Santé et de la Recherche Médicale Extracellular vesicles functionalized with an erv syncitin and uses thereof for cargo delivery
WO2024018426A1 (en) 2022-07-22 2024-01-25 Janssen Biotech, Inc. Enhanced transfer of genetic instructions to effector immune cells
WO2024018046A1 (en) 2022-07-22 2024-01-25 Institut National de la Santé et de la Recherche Médicale Garp as a biomarker and biotarget in t-cell malignancies
WO2024023283A1 (en) 2022-07-29 2024-02-01 Institut National de la Santé et de la Recherche Médicale Lrrc33 as a biomarker and biotarget in cutaneous t-cell lymphomas
WO2024028433A1 (en) 2022-08-04 2024-02-08 Institut National de la Santé et de la Recherche Médicale Methods for the treatment of lymphoproliferative disorders
WO2024033399A1 (en) 2022-08-10 2024-02-15 Institut National de la Santé et de la Recherche Médicale Sigmar1 ligand for the treatment of pancreatic cancer
WO2024033400A1 (en) 2022-08-10 2024-02-15 Institut National de la Santé et de la Recherche Médicale Sk2 inhibitor for the treatment of pancreatic cancer
WO2024038112A1 (en) 2022-08-17 2024-02-22 Institut National de la Santé et de la Recherche Médicale Improved anti-albumin nanobodies and their uses
WO2024047110A1 (en) 2022-08-31 2024-03-07 Institut National de la Santé et de la Recherche Médicale Method to generate more efficient car-t cells

Family Cites Families (63)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SE307996B (en) 1965-11-30 1969-01-27 Asea Ab
US4839295A (en) 1984-06-08 1989-06-13 Pierce Chemical Company Measurement of protein using bicinchoninic acid
EP0368684B2 (en) 1988-11-11 2004-09-29 Medical Research Council Cloning immunoglobulin variable domain sequences.
CA2016842A1 (en) 1989-05-16 1990-11-16 Richard A. Lerner Method for tapping the immunological repertoire
SE509359C2 (en) 1989-08-01 1999-01-18 Cemu Bioteknik Ab Use of stabilized protein or peptide conjugates for the preparation of a drug
US5977307A (en) 1989-09-07 1999-11-02 Alkermes, Inc. Transferrin receptor specific ligand-neuropharmaceutical agent fusion proteins
GB9015198D0 (en) 1990-07-10 1990-08-29 Brien Caroline J O Binding substance
JPH06508511A (en) 1990-07-10 1994-09-29 ケンブリッジ アンティボディー テクノロジー リミティド Method for producing specific binding pair members
CA2046830C (en) 1990-07-19 1999-12-14 Patrick P. Deluca Drug delivery system involving inter-action between protein or polypeptide and hydrophobic biodegradable polymer
US5843440A (en) 1990-10-03 1998-12-01 Redcell Canada, Inc. Cellular and serum protein anchors for modulating pharmacokinetics
AU666330B2 (en) 1991-09-03 1996-02-08 Catsclaw Limited Apparatus for arresting the progress of vehicles
US5474771A (en) 1991-11-15 1995-12-12 The Trustees Of Columbia University In The City Of New York Murine monoclonal antibody (5c8) recognizes a human glycoprotein on the surface of T-lymphocytes, compositions containing same
DK1024191T3 (en) * 1991-12-02 2008-12-08 Medical Res Council Preparation of autoantibodies displayed on phage surfaces from antibody segment libraries
US6005079A (en) 1992-08-21 1999-12-21 Vrije Universiteit Brussels Immunoglobulins devoid of light chains
EP1498427B1 (en) 1992-08-21 2009-12-16 Vrije Universiteit Brussel Immunoglobulins devoid of light chains
WO1995006481A1 (en) 1993-09-02 1995-03-09 Trustees Of Dartmouth College Methods for inducing antigen-specific t cell tolerance
NZ273208A (en) 1993-09-02 2000-12-22 Dartmouth College Suppressing a humoral immune response against a thymus dependant (TD) antigen
US5869049A (en) 1993-09-02 1999-02-09 Trustees Of Dartmouth College Methods of inducing T cell unresponsiveness to bone marrow with gp39 antagonists
US5922545A (en) 1993-10-29 1999-07-13 Affymax Technologies N.V. In vitro peptide and antibody display libraries
SE9400088D0 (en) 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
US5683693A (en) 1994-04-25 1997-11-04 Trustees Of Dartmouth College Method for inducing T cell unresponsiveness to a tissue or organ graft with anti-CD40 ligand antibody or soluble CD40
GB9416721D0 (en) 1994-08-18 1994-10-12 Short Brothers Plc A bias yarn assembly forming device
US5876950A (en) 1995-01-26 1999-03-02 Bristol-Myers Squibb Company Monoclonal antibodies specific for different epitopes of human GP39 and methods for their use in diagnosis and therapy
ES2159726T3 (en) 1995-03-10 2001-10-16 Roche Diagnostics Gmbh FORMS OF PHARMACEUTICAL ADMINISTRATION CONTAINING POLYPEPTIDES, IN THE FORM OF MICROPARTICLES, AND PROCEDURE FOR MANUFACTURING.
US5833987A (en) 1995-06-07 1998-11-10 Trustees Of Dartmouth College Treatment of T cell mediated autoimmune disorders
GB2302024B (en) 1995-06-09 1998-10-07 Malix Polymer Engineering Limi Obstetric vacuum extractor
DE69621940T2 (en) 1995-08-18 2003-01-16 Morphosys Ag PROTEIN - / (POLY) PEPTIDE LIBRARIES
US6440418B1 (en) * 1995-11-07 2002-08-27 Idec Pharmaceuticals Corporation Methods of treating autoimmune diseases with gp39-specific antibodies
US6340459B1 (en) 1995-12-01 2002-01-22 The Trustees Of Columbia University In The City Of New York Therapeutic applications for the anti-T-BAM (CD40-L) monoclonal antibody 5C8 in the treatment of reperfusion injury in non-transplant recipients
US20040248201A1 (en) * 1996-06-27 2004-12-09 Serge Muyldermans Recognition molecules interacting specifically with the active site or cleft of a target molecule
PL188408B1 (en) * 1996-07-08 2005-01-31 Biogen Therapeutic application of bam (cd40l) technology in treating diseases related to smooth muscles
EP0988321A2 (en) 1997-06-20 2000-03-29 Innogenetics N.V. B7-binding molecules for treating immune diseases
DE69803392D1 (en) 1997-06-20 2002-02-28 Tanox Pharma B V ANTI-CD40L IMMUNOTOXINE FOR THE TREATMENT OF DISEASES
WO1999000143A1 (en) 1997-06-27 1999-01-07 Biogen, Inc. Cd154 blockade therapy for autoimmune diseases
GB9722131D0 (en) 1997-10-20 1997-12-17 Medical Res Council Method
CN1203178C (en) 1997-10-27 2005-05-25 尤尼利弗公司 Multivalent antigen-binding proteins
CA2325983C (en) 1998-04-20 2009-02-17 Genzyme Corporation Drug delivery of proteins from polymeric blends
IL127127A0 (en) 1998-11-18 1999-09-22 Peptor Ltd Small functional units of antibody heavy chain variable regions
AU4770300A (en) 1999-05-14 2000-12-05 Medical Research Council Protein scaffold and its use to multimerise monomeric polypeptides
FI991456A (en) 1999-06-24 2000-12-25 Nokia Networks Oy EMI gasket
US6482411B1 (en) * 1999-08-27 2002-11-19 Board Of Regents, The University Of Texas System Methods of reducing bone loss with CD40 ligand
GB0006398D0 (en) * 2000-03-16 2000-05-03 Novartis Ag Organic compounds
AU2001264946A1 (en) 2000-05-24 2001-12-03 Imclone Systems Incorporated Bispecific immunoglobulin-like antigen binding proteins and method of production
US20030012781A1 (en) 2000-06-06 2003-01-16 Anderson Darrell Non-agonistic antibodies to human gp39, compositions containing, and therapeutic use thereof
EP1320590A2 (en) 2000-07-13 2003-06-25 Millennium Pharmaceuticals, Inc. 47885, a novel human ubiquitin-activating enzyme and uses therefor
AU8867501A (en) 2000-09-01 2002-03-13 Biogen Inc Co-crystal structure of monoclonal antibody 5c8 and cd154, and use thereof in drug design
ES2368623T3 (en) 2000-12-13 2011-11-18 Bac Ip B.V. PROTEIN MATRICES OF VARIABLE DOMAINS OF CAMILIDAE HEAVY CHAIN IMMUNOGLOBULIN.
US20050089932A1 (en) 2001-04-26 2005-04-28 Avidia Research Institute Novel proteins with targeted binding
US20030157561A1 (en) 2001-11-19 2003-08-21 Kolkman Joost A. Combinatorial libraries of monomer domains
WO2003002609A2 (en) 2001-06-28 2003-01-09 Domantis Limited Dual-specific ligand and its use
EP1419179B1 (en) 2001-08-10 2010-03-03 Aberdeen University Antigen binding domains from fish
US20050130124A1 (en) 2001-10-05 2005-06-16 Wiersma Erik J. Phagemid display system
JP2005289809A (en) 2001-10-24 2005-10-20 Vlaams Interuniversitair Inst Voor Biotechnologie Vzw (Vib Vzw) Mutant heavy-chain antibody
AR039067A1 (en) * 2001-11-09 2005-02-09 Pfizer Prod Inc ANTIBODIES FOR CD40
EP2366718A3 (en) * 2002-06-28 2012-05-02 Domantis Limited Ligand
US8129330B2 (en) * 2002-09-30 2012-03-06 Mountain View Pharmaceuticals, Inc. Polymer conjugates with decreased antigenicity, methods of preparation and uses thereof
EP2390268B1 (en) 2002-11-08 2017-11-01 Ablynx N.V. Single domain antibodies directed against tumour necrosis factor-alpha and uses therefor
CA2511910A1 (en) 2002-12-27 2004-07-15 Domantis Limited Dual specific single domain antibodies specific for a ligand and for the receptor of the ligand
GB0230201D0 (en) 2002-12-27 2003-02-05 Domantis Ltd Retargeting
PT1639011E (en) 2003-06-30 2009-01-20 Domantis Ltd Pegylated single domain antibodies (dab)
EP1675878A2 (en) 2003-10-24 2006-07-05 Avidia, Inc. Ldl receptor class a and egf domain monomers and multimers
US7563443B2 (en) * 2004-09-17 2009-07-21 Domantis Limited Monovalent anti-CD40L antibody polypeptides and compositions thereof

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9096651B2 (en) 2007-09-26 2015-08-04 Chugai Seiyaku Kabushiki Kaisha Method of modifying isoelectric point of antibody via amino acid substitution in CDR
US9399680B2 (en) 2007-12-05 2016-07-26 Chugai Seiyaku Kabushiki Kaisha Nucleic acids encoding anti-NR10 antibodies
US9228017B2 (en) 2009-03-19 2016-01-05 Chugai Seiyaku Kabushiki Kaisha Antibody constant region variant
US9340615B2 (en) 2009-05-15 2016-05-17 Chugai Seiyaku Kabushiki Kaisha Anti-AXL antibody
US9334331B2 (en) 2010-11-17 2016-05-10 Chugai Seiyaku Kabushiki Kaisha Bispecific antibodies

Also Published As

Publication number Publication date
AU2005283932A1 (en) 2006-03-23
JP5808880B2 (en) 2015-11-10
KR20070057953A (en) 2007-06-07
NO20071312L (en) 2007-06-08
GEP20125471B (en) 2012-04-25
JP2008513425A (en) 2008-05-01
AU2005283932B8 (en) 2012-01-19
US7829096B2 (en) 2010-11-09
WO2006030220A1 (en) 2006-03-23
CA2581017A1 (en) 2006-03-23
US20100092483A1 (en) 2010-04-15
MX2007003187A (en) 2008-01-16
US7927596B2 (en) 2011-04-19
HK1101182A1 (en) 2007-10-12
EP1797126A1 (en) 2007-06-20
BRPI0515404A (en) 2007-11-27
KR101360140B1 (en) 2014-02-20
KR20130039341A (en) 2013-04-19
US20060062784A1 (en) 2006-03-23
IL181724A0 (en) 2007-07-04
IL181724A (en) 2012-08-30
RU2364420C2 (en) 2009-08-20
ES2442455T3 (en) 2014-02-11
RU2007114266A (en) 2008-10-27
EP2371862A3 (en) 2012-10-24
CA2581017C (en) 2016-10-11
US8524236B2 (en) 2013-09-03
US20090297524A1 (en) 2009-12-03
US20100092482A1 (en) 2010-04-15
US7563443B2 (en) 2009-07-21
AU2005283932B2 (en) 2012-01-12
EP1797126B1 (en) 2013-10-23
NZ553847A (en) 2010-01-29
US20110172400A1 (en) 2011-07-14
EP2371862A2 (en) 2011-10-05

Similar Documents

Publication Publication Date Title
KR101360140B1 (en) Compositions monovalent for CD40L binding and methods of use
US20210047414A1 (en) Compositions monovalent for cd28 binding and methods of use
US20130230540A1 (en) Compositions monovalent for cd28 binding and methods of use
EP2315778A2 (en) Compositions monovalent for cd28 binding and methods of use
AU2017202454A1 (en) Compositions monovalent for cd28 binding and methods of use
WO2014120916A1 (en) Pegylated domain antibodies monovalent for cd28 binding and methods of use

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KM KP KR KZ LC LK LR LS LT LU LV LY MA MD MG MK MN MW MX MZ NA NG NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SM SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): BW GH GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LT LU LV MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 181724

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 12007500541

Country of ref document: PH

WWE Wipo information: entry into national phase

Ref document number: 553847

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 2581017

Country of ref document: CA

Ref document number: 2007531827

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: MX/a/2007/003187

Country of ref document: MX

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2005283932

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 07032752

Country of ref document: CO

WWE Wipo information: entry into national phase

Ref document number: 2720/DELNP/2007

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 9989

Country of ref document: GE

Ref document number: 2005784064

Country of ref document: EP

Ref document number: 1020077008642

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: 2007114266

Country of ref document: RU

ENP Entry into the national phase

Ref document number: 2005283932

Country of ref document: AU

Date of ref document: 20050916

Kind code of ref document: A

WWP Wipo information: published in national office

Ref document number: 2005283932

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 200580039317.5

Country of ref document: CN

WWP Wipo information: published in national office

Ref document number: 2005784064

Country of ref document: EP

ENP Entry into the national phase

Ref document number: PI0515404

Country of ref document: BR

WWE Wipo information: entry into national phase

Ref document number: 11663247

Country of ref document: US

WWE Wipo information: entry into national phase

Ref document number: 1020137005252

Country of ref document: KR