EP2486050A1 - Polypeptides qui se lient à l'il-23r - Google Patents

Polypeptides qui se lient à l'il-23r

Info

Publication number
EP2486050A1
EP2486050A1 EP10709310A EP10709310A EP2486050A1 EP 2486050 A1 EP2486050 A1 EP 2486050A1 EP 10709310 A EP10709310 A EP 10709310A EP 10709310 A EP10709310 A EP 10709310A EP 2486050 A1 EP2486050 A1 EP 2486050A1
Authority
EP
European Patent Office
Prior art keywords
polypeptide
loop
seq
human
amino acid
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP10709310A
Other languages
German (de)
English (en)
Inventor
Anke Kretz-Rommel
Martha Wild
Katherine Bowdish
Elise Chen
Daniela Oltean
Maria Gonzalez
Mili Kapoor
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Anaphore Inc
Original Assignee
Anaphore Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US12/577,067 external-priority patent/US20100105620A1/en
Application filed by Anaphore Inc filed Critical Anaphore Inc
Priority claimed from PCT/US2010/023804 external-priority patent/WO2011043835A1/fr
Publication of EP2486050A1 publication Critical patent/EP2486050A1/fr
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4726Lectins
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/54Interleukins [IL]

Definitions

  • sequence listing is filed in this application in electronic format only and is incorporated by reference herein.
  • sequence listing text file "10- .SequenceListing.txt” was created on , and is bytes in size.
  • the invention relates broadly to the treatment of inflammatory and autoimmune diseases as well as cancer.
  • the invention relates to polypeptides that bind to the IL-23R subunit of the IL-23R heterodimeric receptor and that block interaction of IL-23 with its receptor.
  • IL-23 is an essential cytokine for generation and survival of Thl7 cells.
  • Thl7 cells play a critical role in pathology of many autoimmune diseases, including rheumatoid arthritis, inflammatory bowel disease, psoriasis, systemic lupus erythematosus (SLE) and multiple sclerosis.
  • IL-23R is a key target on Thl7 cells.
  • the IL-23 heterodimeric receptor is composed of two subunits: IL-23R and IL-12R i, with IL-23R being the subunit unique to the IL-23 pathway.
  • IL-12R i is shared with the IL-12 receptor and hence the IL-12 pathway.
  • the IL-23 cytokine is composed of two subunits: pl9 and p40, with the pl9 subunit being unique to IL-23, and p40 shared with IL-12. Binding of IL-23 to the heterodimeric IL- 23 receptor mediates activation of certain T cell subsets, NK cells and myeloid cells.
  • IL-23R genetic variation in IL-23R has been associated with susceptibility to psoriasis and Crohn's disease and also has been implicated in susceptibility to ankylosing spondylitis, Vogt-Koyanagi-Harada disease, Systemic Sclerosis, Beliefs disease (BD), Primary Sjogren's Syndrome, Goodpasture disease. Also, importance of IL-23 in Graft Versus Host disease and chronic ulcers has been suggested, and IL-23 has been implicated in tumorigenesis.
  • Blockade of the IL-23 pathway is efficacious in many preclinical models of autoimmune disease.
  • the nature of shared ligand and receptor subunits between IL- 23 and IL-12 pathways has led to more complex biology than previously appreciated, and separation of IL-23 blockade from IL-12 blockade appears to have important therapeutic implications regarding both efficacy and safety.
  • Blockade of one or the other, or both can be done at the level of the cytokine subunits or the receptor subunits.
  • IL-23 can be detected systemically and is more abundant.
  • cytokine is cell bound and/or not abundant as demonstrated in autoimmune tissues such as synovium from rheumatoid arthritis patients.
  • Targeting receptors will more efficiently block in patients with receptor variants that might be more susceptible to IL-23 signaling (i.e. low threshold variants where very little ligand is required for signaling).
  • the invention is directed to a polypeptide having a trimerizing domain and at least one polypeptide sequence that binds to human IL-23R without activating IL-23 heterodimeric receptor.
  • the polypeptide of the invention does not bind to at least one of human IL-12Rpi or human IL-12RP2, and the polypeptide competes with native human IL-23 for binding to human IL-23R.
  • the trimerizing domain may include a polypeptide of a human tetranectin trimerizing domain (SEQ ID NO: 108) having up to five amino acid substitutions at positions 26, 30, 33, 36, 37, 40, 41, 42, 45, 46, 47, 48, 49, 50 and 51.
  • polypeptides can form a trimeric complex.
  • the polypeptides may trimerize to form a trimeric complex.
  • the polypeptide of the invention includes at least one polypeptide that binds IL-23R and is linked to one of the N-terminus and the C-terminus of the trimerizing domain, and also includes a modulator of inflammation positioned at the other of the N-terminus and the C-terminus.
  • the polypeptide of the invention may also have a polypeptide that binds IL-23 linked to each of the N-terminus and the C-terminus, wherein the polypeptide at the N-terminus is the same or different than the polypeptide at the C- terminus.
  • the polypeptide may also have a therapeutic agent covalently attached to the polypeptide
  • the polypeptide of the invention includes a C-Type Lectin Like Domain (CLTD) and wherein one of loops 1, 2, 3 or 4 of loop segment A or loop segment B of the CTLD comprises a polypeptide sequence that binds IL-23.
  • CLTD C-Type Lectin Like Domain
  • the polypeptide sequence of the CTLD is selected from the group consisting of SEQ ID NO: 133, 134, 135, 167, 137, 138, 139, 140, and 141.
  • the invention is also directed to a method of preventing activation of IL-23R by IL-23 in cells that express IL-23R.
  • the method includes contacting the cell with the trimeric complex of the invention.
  • the invention includes a pharmaceutical composition including the trimeric complex and at least one pharmaceutically acceptable excipient.
  • the composition can be administered to treat an immune disorder or cancer.
  • the composition may also include a modulator of inflation, a chemotherapeutic agent or a cytotoxic agent.
  • the invention is directed to method for preparing the polypeptide of the invention.
  • the method includes selecting a first polypeptide that binds to IL-23R and fusing the first polypeptide with one of the N-terminus or the C-terminus of a multimerizing domain.
  • the method may also include selecting a second polypeptide sequence that is a modulator of inflammation; and fusing the second polypeptide with the other of the N- terminus or the C-terminus of the multimerizing domain.
  • the first polypeptide may be selected so that it does not bind to at least one of IL-12R i or IL-12R 2.
  • the polypeptides can be used to prepare a trimeric complex that prevents activation of IL-23R in a cell expressing IL-23R.
  • the invention is directed to a polypeptide that competes with native human IL-23 for binding to native IL-23R, wherein the polypeptide does not activate human IL-23R and does not bind to at least one of IL-12Rpi or IL-12Rp2.
  • the polypeptide may be a CTLD that has been modified in one of loops 1, 2, 3 or 4 of loop segment A or in loop segment B for binding to IL-23R, and may be selected from one of SEQ ID NO: 133, 134, 135, 167, 137, 138, 139, 140, and 141.
  • Figures 1A and IB show the polypeptide sequence of human IL-23 (SEQ ID NO: 1), human IL-23R (SEQ ID NO: 5), human IL-12Rpi (SEQ ID NO: 6), human IL-12RP2 (SEQ ID NO: 7), human IL-12A (SEQ ID NO: 3), and human IL-12B (SEQ ID NO: 2).
  • Figures 2A, B, C and D show examples of tetranectin trimerizing module variants for use with exemplary polypeptides of the invention.
  • Figure 3 shows alignment of the amino acid sequences of the trimerising structural element of the tetranectin protein family. Amino acid sequences (one letter code)
  • Residues at a and d positions in the heptad repeats are listed in boldface.
  • the listed consensus sequence (SEQ ID NO: 108) of the tetranectin protein family trimerizing structural element comprise the residues present at a and d positions in the heptad repeats shown in the figure in addition to the other conserved residues of the region.
  • "*" denotes an aliphatic hydrophobic residue.
  • Figure 4 shows an alignment of the amino acid sequences of ten CTLDs of known 3D-structure.
  • the sequence locations of main secondary structure elements are indicated above each sequence, labeled in sequential numerical order as " N", denoting a a-helix number N, and " ⁇ ", denoting ⁇ -strand number M.
  • the four cysteine residues involved in the formation of the two conserved disulfide bridges of CTLDs are indicated and enumerated in the Figure as "CI”, “CII”, “CIII” and “CIV” respectively.
  • the two conserved disulfide bridges are CI-CIV and CII-CIII, respectively.
  • the various loops 1-4 and LSB (loop 5) in the human tetranectin sequence are indicated by underlining.
  • the ten C-type lectins are hTN: human tetranectin (SEQ ID NO: 109), MBP: mannose binding protein (SEQ ID NO: 110); SP-D: surfactant protein D (SEQ ID NO: 111); LY49A: NK receptor LY49A (SEQ ID NO: 112); Hl-ASR: HI subunit of the asialoglycoprotein receptor (SEQ ID NO: 113); MMR-4: macrophage mannose receptor domain 4 (SEQ ID NO: 114); IX-A (SEQ ID NO: 115) and IX-B (SEQ ID NO: 116): coagulation factors IX/X-binding protein domain A and B, respectively; Lit: lithostatine (SEQ ID NO: 117); TU14: tunicate C-type lectin (SEQ ID NO: 118). All of these CT
  • Figure 5 depicts an alignment of the amino acid sequences of tetranectins isolated from human (Swissprot P05452) (SEQ ID NO: 119), mouse (Swissprot P43025) (SEQ ID NO: 120), chicken (Swissprot Q9DDD4) (SEQ ID NO: 121), bovine (Swissprot Q2KIS7) (SEQ ID NO: 122), Atlantic salmon (Swissprot B5XCV4) (SEQ ID NO: 123), frog
  • Figure 6 shows the PCR strategy for creating randomized loops in a CTLD.
  • Figure 7 shows the DNA and amino acid sequence of the human tetranectin CTLD modified to contain restriction sites for cloning, indicating the Ca2+ binding sites. Restriction sites are underscored with solid lines. Loops are underlined with dashed lines. Calcium coordinating residues are in bold italics and include Site 1 : Dl 16, E120, G147, E150, 51; Site 2: £>143, D145, E150, D165.
  • the CTLD domain starts at amino acid A45 in bold ⁇ i.e. ALQTVCL). Changes to the native tetranectin (TNCTLD) base sequence are shown in lower case.
  • the restriction sites were created using silent mutations that did not alter the native amino acid sequence.
  • Figure 8 shows a number of sequences of polypeptides of the invention that bind to IL-23R.
  • the sequences were produced according to the method of the invention by selecting polypeptides from a library of polypeptides having the scaffold structure of a human tetranectin CTLD that have been modified in one more loop regions.
  • the CTLD scaffold of these sequences starts at A45 of human tetranectin (SEQ ID NO: 119).
  • SEQ ID NO: 119 human tetranectin
  • Figure 9 depicts an alignment of the nucleotide and amino acid sequences of the coding regions of the mature forms of human (SEQ ID NOS: 143 [nucleotide sequence] and 142 [amino acid sequence]) and murine tetranectin (SEQ ID NOS: 144 [nucleotide sequence] and 145 [amino acid sequence]) starting at their trimerizing domains, with an indication of known secondary structural elements.
  • Figure 10 shows the results of a competition ELISA. Binding of human IL-23 to human IL-23R in the presence or absence of the polypeptides of the invention was evaluated.
  • Figure 11 shows the results of an experiment comparing IL-23 -induced IL-17 production in the presence of ATRIMERTM complex 4G8 of the invention, native human IL- 23, and Ustekinumab.
  • Figure 12 shows the results of an experiment comparing IL-23 -induced IL-17 production in the presence of ATRIMERTM complex 1 A4 of the invention and Ustekinumab.
  • Figure 13 shows the results of an experiment comparing IL-12-induced IFNy production in the presence of the ATRIMERTM complex 4G8 of the invention, native human IL-23, and Ustekinumab.
  • Figure 14 shows the results of an experiment comparing Stat-3 phosphorylation in NKL cell in response to IL-23 and the polypeptides of the invention.
  • Figure 15 is a table showing experimental results associated with several
  • ATRIMERTM polypeptide complexes of the invention are ATRIMERTM polypeptide complexes of the invention.
  • Figure 16 depicts the three dimensional structure (ribbon format) for human tetranectin, depicting the secondary structural features of the protein. The structure was solved in the Ca 2+ -bound form.
  • Figure 17A depicts the three dimensional overlay structures of the CTLDs for human tetranectin (HTN) and several tetranectin homologues, including human mannose binding protein (MBP), rat mannose binding protein-C (MBP-C), human surfactant protein D, rat mannose binding protein-A (MBP-A), and rat surfactant protein A.
  • the CTLD overlay structures were generated using Swiss PDB Viewer Deep View v. 4.0.1 for Macintosh using the three-dimensional structure of human tetranectin as a template.
  • Figure 17B shows the corresponding amino acid sequences of the CTLDS for human tetranectin and the tetranectin homologues depicted in Figure 17A.
  • FIG. 18A depicts the three dimensional overlay structures of the CTLDs for human tetranectin (HTN) and several tetranectin homologues, including human pancreatitis- associated protein, human dendritic cell-specific ICAM-3 -grabbing non-integrin 2 (DC- SIGNR), rat aggrecan, mouse scavenger receptor, and human scavenger receptor.
  • the CTLD overlay structures were generated using Swiss PDB Viewer Deep View v.
  • FIG. 18B shows the corresponding amino acid sequences of the CTLDS for human tetranectin and the tetranectin homologues depicted in Figure 18 A.
  • 1TDQB rat aggrecan
  • 1UV0A human pancreatitis-associated protein
  • 20X8 A human scavenger receptor
  • 20X9A mouse scavenger receptor
  • 1SL6A human DC-SIGNR
  • the invention is directed to polypeptides that bind IL-23R and that include polypeptide sequences of a multimerizing domain and one or more polypeptide sequences that bind to IL-23R.
  • the polypeptides of the invention function as IL-23R antagonists. Two, three, or more of the polypeptides can multimerize to form a multimeric complex including the polypeptides that bind IL-23R.
  • the polypeptide binds IL-23R, but does not bind IL-12R i or ⁇ ,-12 ⁇ 2.
  • the invention provides methods for treating immune mediated disorders, cancer and other diseases in a subject by administering the polypeptide or multimeric complexes of the polypeptide to a patient in need.
  • IL-23 is a cytokine that functions in innate and adaptive immunity and refers to a hetero-dimeric protein complex belonging to the IL-6 superfamily. The heterodimeric complex is secreted by activated dendritic and phagocytic cells and keratinocytes. IL-23 is also expressed by dermal Langerhans cells. IL-23 A, also known as IL-B30, the pl9 subunit, or simply " i 9," associates with IL-12B, the p40 subunit, to form IL-23 (pl9/p40).
  • the amino acid sequences of IL-23 A ( i 9) (SEQ ID NO: 1) and IL-12B (SEQ ID NO: 2) are shown in FIG. 1.
  • IL-23 is up-regulated by a wide array of pathogens and pathogen-products together with self-signals for danger or injury.
  • IL-23 is up-regulated in psoriatic dermal tissues, in dendritic cells of multiple sclerosis patients and it has as well been shown that IL- 23 is active in promoting tumor incidence and growth.
  • IL-23 not only stimulates neutrophil and macrophage infiltration, but also promotes angiogenesis and inflammatory mediators in the tumor microenvironment.
  • IL-23 can result in down-regulation of IL-12 and interferon ⁇ , both of which are essential cytokines for cytotoxic immune responses, and controls the influx and activity of anti-tumor effector lymphocytes.
  • IL-23 inflicts a repurposing of the adaptive cytotoxic effector response away from anti-tumor immunity and towards proinflammatory and proangiogenic effector pathways that nourish the tumor. Consequently, IL-23 enables the persistence of the recognized tumor cells, accompanied by tumor-associated inflammation. This concept can explain tumor growth in the presence of large quantities of tumor-specific T cells.
  • IL-23 heterodimeric receptor refers to the heterodimeric polypeptide complex of IL-23R and IL-12Rpi . This receptor binds IL-23.
  • the polypeptide sequence of IL-23R and IL-12Rpi are shown in FIG. 1.
  • IL-23R refers to a polypeptide that can complex with IL-12Rpi to form the IL-23 heterodimeric receptor.
  • IL-23R is also referred to as the IL-23R subunit.
  • IL-12R i refers to the polypeptide that complexes with IL-23R to form the IL-23 heterotrimeric receptor and separately and independently with IL-12R 2 to form a heterodimeric IL-12 receptor.
  • the polypeptide sequences of IL-12Rpi and IL-12RP2 are shown in FIG. 1.
  • Inhibitors and “antagonists” or “activators” and “agonists” refer to inhibitory or activating molecules, respectively.
  • “Inhibitors” are compounds that decrease, block, prevent, delay activation, inactivate, desensitize, or down regulate biological function or activity associated with, for example, a gene, protein, ligand, receptor, or cell.
  • Activators are compounds that increase, activate, facilitate, enhance activation, sensitize, or up regulate the biological function or activity of, for example, gene, protein, ligand, receptor, or cell.
  • An "agonist” is a compound that interacts with a target to cause or promote an increase in the activation of the target.
  • An “antagonist” is a compound that opposes the actions of an agonist.
  • An antagonist prevents, reduces, inhibits, or neutralizes the activity of an agonist.
  • An antagonist can also prevent, inhibit, or reduce constitutive activity of a target, e.g., a target receptor, even where there is no identified agonist.
  • a "modulator" of a gene, a receptor, a ligand, or a cell is a molecule that alters an activity of the gene, receptor, ligand, or cell, where activity can be activated, inhibited, or altered in its regulatory properties.
  • the modulator may act alone, or it may use a cofactor, for example, a protein, metal ion, or small molecule.
  • IL-23R antagonist refers to any molecule _that binds to IL-23R either alone or in complex with IL-12Rpi and blocks or dampens receptor signaling through a variety of mechanisms which can include blocking the ability of IL-23 to bind, blocking receptor heterodimer formation, or blocking or inducing changes that affect intracellular signaling, including conformational changes or receptor internalization.
  • binding member refers to a member of a pair of molecules which have binding specificity for one another.
  • the members of a binding pair may be naturally derived or wholly or partially synthetically produced.
  • One member of the pair of molecules has an area on its surface, or a cavity, which binds to and is therefore complementary to a particular spatial and polar organization of the other member of the pair of molecules.
  • the members of the pair have the property of binding specifically to each other.
  • binds when referring to a ligand/receptor, antibody/antigen, or other binding pair, indicates a binding reaction which is determinative of the presence of member of a binding pair in a heterogeneous population of another member of the binding pair.
  • a specified ligand binds to a particular receptor and does not bind in a significant amount to other proteins present in the sample.
  • the term "multimerizing domain” means an amino acid sequence that comprises the functionality that can associate with other amino acid sequence(s) having a multimerizing domain to form multimeric complexes.
  • the multimerizing domain is a dimerizing domain, a trimerizing domain, a tetramerizing domain, a pentamerizing domain, etc. These domains are capable of forming polypeptide complexes of two, three, four, five or more polypeptides of the invention.
  • the polypeptide contains an amino acid sequence - a "trimerizing domain”— which forms a trimeric complex with two other trimerizing domains.
  • a trimerizing domain can associate with other trimerizing domains of identical amino acid sequence (a homotrimer), or with trimerizing domains of different amino acid sequence (a heterotrimer). Such an interaction may be caused by covalent bonds between the components of the trimerizing domains as well as by hydrogen bond forces, hydrophobic forces, van der Waals forces and salt bridges.
  • trimerizing domain of a polypeptide of the invention may be derived from tetranectin as described in U.S. Patent Application Publication No. 2007/0154901 ('901 Application), which is incorporated by reference in its entirety.
  • the mature human tetranectin single chain polypeptide sequence is provided herein as SEQ ID NO: 142.
  • Examples of a tetranectin trimerizing domain includes the amino acids 17 to 49, 17 to 50, 17 to 51 and 17-52 of SEQ ID NO: 99, which represent the amino acids encoded by exon 2 of the human tetranectin gene, and optionally the first one, two or three amino acids encoded by exon 3 of the gene.
  • Other examples include amino acids 1 to 49, 1 to 50, 1 to 51 and 1 to 52, which represents all of exons 1 and 2, and optionally the first one, two or three amino acids encoded by exon 3 of the gene.
  • only a part of the amino acid sequence encoded by exon 1 is included in the trimerizing domain.
  • the N-terminus of the trimerizing domain may begin at any of residues 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16 and 17 of SEQ ID NO: 99.
  • the N terminus is 110 or VI 7 and the C-terminus is Q47, T48, V49, C(S)50, L51 or K52 (numbering according to SEQ ID NO: 99).
  • FIGs 2A-2D provide a number of potential truncation variant of the human tetranectin trimerizing domain.
  • the trimerizing domain is a tetranectin trimerizing structural element ("TTSE") having a amino acid sequence of SEQ ID NO: 108 which is a consensus sequence of the tetranectin family trimerizing structural element as more fully described in US 2007/00154901, which is incorporated herein by reference in its entirety.
  • TTSE tetranectin trimerizing structural element
  • the TTSE embraces variants of a naturally occurring member of the tetranectin family of proteins, and in particular variants that have been modified in the amino acid sequence without adversely affecting, to any substantial degree, the ability of the TTSE to form alpha helical coiled coil trimers.
  • the trimeric polypeptide according to the invention includes a TTSE as a trimerizing domain having at least 66% amino acid sequence identity to the consensus sequence of SEQ ID NO: 108; for example at least 73%, at least 80%, at least 86% or at least 92% sequence identity to the consensus sequence of SEQ ID NO: 108 (counting only the defined (not X) residues). In other words, at least one, at least two, at least three, at least four, or at least five of the defined amino acids in SEQ ID NO: 108 may be substituted.
  • the cysteine at position 50 (C50) of SEQ ID NO: 142 can be advantageously be mutagenized to serine, threonine, methionine or to any other amino acid residue in order to avoid formation of an unwanted inter-chain disulphide bridge, which can lead to unwanted multimerization.
  • Other known variants include at least one amino acid residue selected from amino acid residue nos. 6, 21, 22, 24, 25, 27, 28, 31, 32, 35, 39, 41, and 42 (numbering according to SEQ ID NO: 142), which may be substituted by any non-helix breaking amino acid residue. These residues have been shown not to be directly involved in the intermolecular interactions that stabilize the trimeric complex between three TTSEs of native tetranectin monomers.
  • the TTSE has a repeated heptad having the formula a-b-c-d-e-f-g (N to C), wherein residues a and d (i.e., positions 26, 30, 33, 37, 40, 44, 47, and 51 may be any hydrophobic amino acid (numbering according to SEQ ID NO: 99).
  • the TTSE trimerization domain may be modified by the incorporation of polyhistidine sequence and/or a protease cleavage site, e.g., Blood
  • Coagulating Factor Xa or Granzyme B (see US 2005/0199251, which is incorporated herein by reference), and by including a C-terminal KG or KGS sequence. Also, to assist in purification, Proline at position 2 may be substituted with Glycine .
  • FIGs 2A-2D Particular non- limiting examples of TTSE truncations and variants are shown in FIGs 2A-2D.
  • MBP trimerizing domain is described in PCT Application Serial No. US08/76266, published as WO 2009/036349, which is incorporated by reference in its entirety. This trimerizing domain can oligomerize even further and create higher order multimeric complexes.
  • trimerising domain is capable of interacting with other, similar or identical trimerising domains.
  • the interaction is of the type that produces trimeric proteins or polypeptides.
  • Such an interaction may be caused by covalent bonds between the components of the trimerising domains as well as by hydrogen bond forces, hydrophobic forces, van der Waals forces, and salt bridges.
  • the trimerising effect of trimerizing domain is caused by a coiled coil structure that interacts with the coiled coil structure of two other trimerizing domains to form a triple alpha helical coiled coil trimer that is stable even at relatively high temperatures.
  • the complex is stable at least 60 °C, for example in some embodiments at least 70 °C
  • C-type lectin-like protein and “C-type lectin” are used to refer to any protein present in, or encoded in the genomes of, any eukaryotic species, which protein contains one or more CTLDs or one or more domains belonging to a subgroup of CTLDs, the CRDs, which bind carbohydrate ligands.
  • the definition specifically includes membrane attached C-type lectin-like proteins and C-type lectins, "soluble” C-type lectin-like proteins and C-type lectins lacking a functional transmembrane domain and variant C-type lectin-like proteins and C-type lectins in which one or more amino acid residues have been altered in vivo by glycosylation or any other post-synthetic modification, as well as any product that is obtained by chemical modification of C-type lectin-like proteins and C-type lectins.
  • the CTLD consists of roughly 120 amino acid residues and, characteristically, contains two or three intra-chain disulfide bridges. Although the similarity at the amino acid sequence level between CTLDs from different proteins is relatively low, the 3D-structures of a number of CTLDs have been found to be highly conserved, with the structural variability essentially confined to a so-called loop-region, often defined by up to five loops. Several CTLDs contain either one or two binding sites for calcium and most of the side chains which interact with calcium are located in the loop-region.
  • CTLDs for which 3D structural information is available, it has been inferred that the canonical CTLD is structurally characterized by seven main secondary- structure elements (i.e. five ⁇ -strands and two a-helices) sequentially appearing in the order ⁇ , ⁇ , ⁇ 2, ⁇ 2, ⁇ 3, ⁇ 4, and ⁇ 5.
  • Figure 4 illustrates an alignment of the CTLDs of ten known C-type lectins.
  • the ⁇ -strands are arranged in two anti-parallel ⁇ -sheets, one composed of ⁇ and ⁇ 5, the other composed of ⁇ 2, ⁇ 3 and ⁇ 4.
  • FIG. 1 An additional ⁇ -strand, ⁇ , often precedes ⁇ in the sequence and, where present, forms an additional strand integrating with the ⁇ , ⁇ 5 -sheet.
  • two disulfide bridges one connecting l and ⁇ 5 (Q-Civ) and one connecting ⁇ 3 and the polypeptide segment connecting ⁇ 4 and ⁇ 5 (Cn-Cm) are invariantly found in all CTLDs characterized to date.
  • Figure 5 shows an alignment of CTLDs from human tetranectin and eight other tetranectin or tetranectin like polypeptides.
  • these conserved secondary structure elements form a compact scaffold for a number of loops, which in the present context collectively are referred to as the "loop-region", protruding out from the core.
  • these loops are organized in two segments, loop segment A, LSA, and loop segment B, LSB.
  • LSA represents the long polypeptide segment connecting ⁇ 2 and ⁇ 3 that often lacks regular secondary structure and contains up to four loops.
  • LSB represents the polypeptide segment connecting the ⁇ -strands ⁇ 3 and ⁇ 4.
  • Residues in LSA together with single residues in ⁇ 4, have been shown to specify the Ca 2+ - and ligand-binding sites of several CTLDs, including that of tetranectin.
  • mutagenesis studies involving substitution of one or a few residues, have shown that changes in binding specificity, Ca 2+ -sensitivity and/or affinity can be accommodated by CTLD domains.
  • CLTDs are known, including the following non- limiting examples: tetranectin, lithostatin, mouse macrophage galactose lectin, Kupffer cell receptor, chicken neurocan, perlucin, asialoglycoprotein receptor, cartilage proteoglycan core protein, IgE Fc receptor, pancreatitis-associated protein, mouse
  • ATRIMERTM polypeptide complex or “ATRIMERTM complex” refers to a trimeric complex of three trimerizing domains that also include CLTDs (Anaphore, Inc., San Diego, California).
  • an effective amount refers to an amount of a polypeptide of the invention, optionally in conjunction with a therapeutic agent which is effective for preventing, ameliorating or treating the disease or condition in question whether administered simultaneously or sequentially.
  • an effective amount is the amount of the polypeptide of the invention, and a a therapeutic agent, such as a cytotoxic or immunosuppressive agent, in combination sufficient to decrease the effects of IL-23 on IL- 23R expressing cells, affect other pathways on IL-23R expressing cells working
  • IL-23R synergistically with IL-23R, or affecting other immune cells acting in concert with IL-23R expressing cells, decrease the propensity of a cell to proliferate or survive, or to enhance, or otherwise increase the propensity (such as synergistically) of a cell to undergo apoptosis, reduce tumor volume, or prolong survival of a mammal having a cancer or immune related disease.
  • a “therapeutic agent” refers to a cytotoxic agent, a chemotherapeutic agent, an immunosuppressive agent, an anti-inflammatory agent, an immunostimulatory agent, and/or a growth inhibitory agent.
  • immunosuppressive agent and "modulators of inflammation” as used herein for adjunct therapy refers to substances that act to suppress or mask the immune system of the mammal being treated herein. This would include substances that suppress cytokine production, downregulate or suppress self-antigen expression, inhibit migration of immune cells to sites of chronic inflammation, or mask the MHC antigens. Examples of such agents include but are not limited to 2-amino-6-aryl-5 -substituted pyrimidines (see U.S. Pat. No. 4,665,077); nonsteroidal anti-inflammatory drugs (NSAIDs); azathioprine;
  • cyclophosphamide bromocryptine; danazol; dapsone; glutaraldehyde (which masks the MHC antigens, as described in U.S. Pat. No. 4,120,649); anti-idiotypic antibodies for MHC antigens and MHC fragments; cyclosporin A; steroids such as glucocorticosteroids, e.g., prednisone, methylprednisolone, dexamethasone, and hydrocortisone; methotrexate (oral or subcutaneous); hydroxycloroquine; sulfasalazine; leflunomide; cytokine or cytokine receptor antagonists including anti-interferon-gamma (IFN- ⁇ ), - ⁇ , or -a antibodies, anti-tumor necrosis factor-a antibodies (such as e.g.
  • IFN- ⁇ interferon-gamma
  • - ⁇ anti-tumor necrosis factor-a antibodies
  • infliximab, adalimumab or Cimzia anti-TNFa immunoadhesin (etanercept), anti-tumor necrosis factor- ⁇ antibodies, anti-TGF- ⁇ antibodies, anti-interleukin-2 antibodies and anti-IL-2 receptor antibodies; anti-IL-6 antibodies, anti-IL- 6R antibodies, anti-LFA-1 antibodies, including anti-CDl la and anti-CD 18 antibodies; anti- L3T4 antibodies; heterologous anti-lymphocyte globulin; pan-T antibodies, preferably anti- CD3 or anti-CD4/CD4a antibodies; soluble peptide containing a LFA-3 binding domain (WO 90/08187 published Jul.
  • T-cell receptor fragments (Offner et al., Science, 251 : 430-432 (1991); WO 90/11294; Janeway, Nature, 341 : 482 (1989); and WO 91/01133); and T-cell receptor antibodies (EP 340,109) such as T10B9, integrin inhibitors such as Tysabri, CCR9 or CCR6 antagonists, anti-TLlA antibodies or cytokines known to suppress immune responses such as IL-lO or IL-27.
  • cytotoxic agent refers to a substance that inhibits or prevents the function of cells and/or causes destruction of cells.
  • the term is intended to include radioactive isotopes (e.g. At 211 , 1 131 , 1 125 , Y 90 , Re 186 , Re 188 , Sm 153 , Bi 212 , P 32 and radioactive isotopes of Lu), chemotherapeutic agents, and toxins such as small molecule toxins or enzymatically active toxins of bacterial, fungal, plant or animal origin, or fragments thereof.
  • radioactive isotopes e.g. At 211 , 1 131 , 1 125 , Y 90 , Re 186 , Re 188 , Sm 153 , Bi 212 , P 32 and radioactive isotopes of Lu
  • chemotherapeutic agents e.g. At 211 , 1 131 , 1 125 , Y 90 , Re 186 , Re 188
  • a "chemotherapeutic agent” is a chemical compound useful in the treatment of cancer.
  • chemotherapeutic agents include alkylating agents such as thiotepa and CYTOXAN® cyclosphosphamide; alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa;
  • ethylenimines and methylamelamines including altretamine, triethylenemelamine, triethylenephosphoramide, triethylenethiophosphoramide and trimethylolomelamine;
  • acetogenins especially bullatacin and bullatacinone
  • a camptothecin including the synthetic analogue topotecan
  • bryostatin callystatin; CC-1065 (including its adozelesin, carzelesin and bizelesin synthetic analogues); cryptophycins (particularly cryptophycin 1 and cryptophycin 8); dolastatin; duocarmycin (including the synthetic analogues, KW-2189 and CB1-TM1); eleutherobin; pancratistatin; a sarcodictyin; spongistatin; nitrogen mustards such as chlorambucil, chlornaphazine, cholophosphamide, estramustine, ifosfamide,
  • dynemicin including dynemicin A; bisphosphonates, such as clodronate; an esperamicin; as well as neocarzinostatin chromophore and related chromoprotein enediyne antibiotic chromophores), aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycin, carabicin, carminomycin, carzinophilin, chromomycinis, dactinomycin, daunorubicin, detorubicin, 6-diazo-5-oxo-L-norleucine, ADRIAMYCIN® doxorubicin (including morpholino-doxorubicin, cyanomorpholino-doxorubicin, 2-pyrrolino-doxorubicin and deoxydoxorubicin), epirubicin, esorubicin, idarubicin, marcellomycin
  • etoglucid gallium nitrate; hydroxyurea; lentinan; lonidainine; maytansinoids such as maytansine and ansamitocins; mitoguazone; mitoxantrone; mopidanmol; nitraerine;
  • razoxane rhizoxin; sizofiran; spirogermanium; tenuazonic acid; triaziquone; 2,2',22"- trichlorotriethylamine; trichothecenes (especially T-2 toxin, verracurin A, roridin A and anguidine); urethan; vindesine; dacarbazine; mannomustine; mitobronitol; mitolactol;
  • TAXOL® paclitaxel Bristol-Myers Squibb Oncology, Princeton, N.J.
  • ABRAXANETM Cremophor-free albumin-engineered nanoparticle formulation of paclitaxel
  • TAXOTERE® doxetaxel Rhone- Poulenc Rorer, Antony, France
  • chloranbucil GEMZAR® gemcitabine
  • 6-thioguanine mercaptopurine
  • methotrexate platinum analogs such as cisplatin and carboplatin; vinblastine; platinum; etoposide (VP- 16); ifosfamide; mitoxantrone; vincristine; NAVELBINE® vinorelbine; novantrone;
  • proteasome inhibitors such as bortezomib (Velcade), BCL-2 inhibitors, IAP antagonists (e.g. Smac mimics/xIAP and cIAP inhibitors such as certain peptides, pyridine compounds such as (S)- N- ⁇ 6-benzo[ 1 ,3]dioxol-5-yl- 1 -[5-(4-fluoro-benzoyl)-pyridin-3-ylmethyl]-2-oxo- 1 ,2-dihydro- pyridin-3-yl ⁇ -2-methylamino-propionamide, xIAP antisense), HDAC inhibitors (HDACI) and kinase inhibitors (Sorafenib).
  • IAP antagonists e.g. Smac mimics/xIAP and cIAP inhibitors such as certain peptides
  • pyridine compounds such as (S)- N- ⁇ 6-benzo[ 1 ,3]diox
  • anti-hormonal agents that act to regulate or inhibit hormone action on tumors
  • SERMs selective estrogen receptor modulators
  • tamoxifen including NOLVADEX® tamoxifen
  • raloxifene including NOLVADEX® tamoxifen
  • droloxifene 4-hydroxytamoxifen
  • trioxifene keoxifene
  • LY117018 onapristone
  • aromatase inhibitors that inhibit the enzyme aromatase, which regulates estrogen production in the adrenal glands, such as, for example, 4(5)-imidazoles, aminoglutethimide, MEGASE® megestrol acetate, AROMASIN® exemestane, formestanie, fadrozole, RIVISOR® vorozole, FEMARA® letrozole, and ARIMIDEX® anastrozole
  • anti-androgens such as flutamide
  • ANGIOZYME® ribozyme and a HER2 expression inhibitor
  • vaccines such as gene therapy vaccines, for example, ALLOVECTIN® vaccine, LEUVECTIN® vaccine, and VAXID® vaccine
  • PROLEUKIN® rIL-2 PROLEUKIN® rIL-2
  • LURTOTECAN® topoisomerase 1 inhibitor ABARELIX® rmRH
  • pharmaceutically acceptable salts, acids or derivatives of any of the above are examples of any of the above.
  • a “growth inhibitory agent” when used herein refers to a compound or composition which inhibits growth of a cell, either in vitro or in vivo.
  • the growth inhibitory agent is one that significantly reduces the percentage of cells overexpressing such genes in S phase.
  • growth inhibitory agents include agents that block cell cycle progression (at a place other than S phase), such as agents that induce Gl arrest and M-phase arrest.
  • Classical M-phase blockers include the vincas (vincristine and vinblastine), taxol, and topo II inhibitors such as doxorubicin, epirubicin, daunorubicin, etoposide, and bleomycin.
  • DNA alkylating agents such as tamoxifen, prednisone, dacarbazine, mechlorethamine, cisplatin, methotrexate, 5-fluorouracil, and ara-C. Further information can be found in The Molecular Basis of Cancer, Mendelsohn and Israel, eds., Chapter 1, entitled “Cell cycle regulation, oncogenes, and antineoplastic drugs” by Murakami et al. (WB Saunders: Philadelphia, 1995, pg. 13).
  • agents that induce cell stress such as e.g. arginine depleting agents such as arginase.
  • Synergistic activity means that the effect observed when employing a combination of an IL-23R antagonist and a therapeutic agent is (1) greater than the effect achieved when that IL-23R antagonist or therapeutic agent is employed alone (or individually) and (2) greater than the sum added (additive) effect for that IL-23R antagonist or therapeutic agent.
  • synergy or synergistic effect can be determined by way of a variety of means known to those in the art.
  • the synergistic effect of IL-23R antagonist and a therapeutic agent can be observed in in vitro or in vivo assay formats examining reduction in cytokine release from immune cells, number or type of immune cells present, or in the case of cancer, in reduction of tumor cell number or tumor mass.
  • cancer refers to or describe the physiological condition in mammals that is typically characterized by unregulated cell growth.
  • cancer include but are not limited to, carcinoma including
  • adenocarcinoma lymphoma, blastoma, melanoma, sarcoma, and leukemia. More particular examples of such cancers include squamous cell cancer, small-cell lung cancer, non-small cell lung cancer (NSCLC), gastrointestinal cancer, Hodgkin's and non-Hodgkin's lymphoma, pancreatic cancer, glioblastoma, glioma, cervical cancer, ovarian cancer, liver cancer such as hepatic carcinoma and hepatoma, bladder cancer, breast cancer, colon cancer, colorectal cancer, endometrial carcinoma, myeloma (such as multiple myeloma), salivary gland carcinoma, kidney cancer such as renal cell carcinoma and Wilms' tumors, basal cell carcinoma, melanoma, prostate cancer, vulval cancer, thyroid cancer, testicular cancer, esophageal cancer, and various types of head and neck cancer.
  • NSCLC non-small cell lung cancer
  • gastrointestinal cancer Ho
  • immune related disease means a disease or disorder in which a component of the immune system of a mammal causes, mediates or otherwise contributes to morbidity in the mammal. Also included are diseases in which stimulation or intervention of the immune response has an ameliorative effect on progression of the disease. Included within this term are autoimmune diseases, immune-mediated inflammatory diseases.
  • immune -related and inflammatory diseases examples include systemic lupus
  • erythematosis erythematosis, rheumatoid arthritis, juvenile chronic arthritis, spondyloarthropathies, ankylosing spondylitis, systemic sclerosis (scleroderma), idiopathic inflammatory myopathies (dermatomyositis, polymyositis), primary Sjogren's syndrome, systemic vasculitis, sarcoidosis, autoimmune hemolytic anemia (immune pancytopenia, paroxysmal nocturnal hemoglobinuria), autoimmune thrombocytopenia (idiopathic thrombocytopenic purpura, immune-mediated thrombocytopenia), thyroiditis (Grave's disease, Hashimoto's thyroiditis, juvenile lymphocytic thyroiditis, atrophic thyroiditis), diabetes mellitus, immune -mediated renal disease (glomerulonephritis, tubulointerstitial nephritis), demyelinating diseases of the central and
  • hypersensitivity pneumonitis transplantation associated diseases including graft rejection and graft-versus-host-disease, immune-mediated or autoimmune eye diseases such as uveitis, dry eye, Beliefs disease (BD) .
  • Infectious diseases include AIDS (HIV infection), hepatitis A, B, C, D, and E, bacterial infections, fungal infections, protozoal infections and parasitic infections.
  • a "B-cell malignancy” is a malignancy involving B cells.
  • Hodgkin's disease including lymphocyte predominant Hodgkin's disease (LPHD); non- Hodgkin's lymphoma (NHL); follicular center cell (FCC) lymphoma; acute lymphocytic leukemia (ALL); chronic lymphocytic leukemia (CLL); hairy cell leukemia; plasmacytoid lymphocytic lymphoma; mantle cell lymphoma; AIDS or HIV-related lymphoma; multiple myeloma; central nervous system (CNS) lymphoma; post-transplant lymphoproliferative disorder (PTLD); Waldenstrom's macroglobulinemia (lymphoplasmacytic lymphoma);
  • LPHD lymphocyte predominant Hodgkin's disease
  • NHL non- Hodgkin's lymphoma
  • FCC follicular center cell
  • ALL acute lymphocytic leukemia
  • CLL
  • MALT lymphoid tissue lymphoma mucosa-associated lymphoid tissue lymphoma
  • lymphoma lymphoma/leukemia.
  • NCL Non-Hodgkin's lymphoma
  • grade/follicular NHL grade/follicular NHL, relapsed or refractory NHL, front line low grade NHL, Stage III/IV NHL, chemotherapy resistant NHL, small lymphocytic (SL) NHL, intermediate
  • grade/follicular NHL intermediate grade diffuse NHL, diffuse large cell lymphoma, aggressive NHL (including aggressive front-line NHL and aggressive relapsed NHL), NHL relapsing after or refractory to autologous stem cell transplantation, high grade
  • immunoblastic NHL high grade lymphoblastic NHL, high grade small non-cleaved cell NHL, bulky disease NHL, etc.
  • TAA tumor-associated antigens
  • TSA tumor-specific antigens
  • TAAs or TSAs exibiting on the surface of tumor cells include but are not limited to alfafetoprotein, carcinoembryonic antigen (CEA), CA-125, MUC-1, glypican-3, tumor associated glycoprotein-72 (TAG-72), epithelial tumor antigen, tyrosinase, melanoma associated antigen, MART-1, gplOO, TRP-1, TRP-2, MSH-1, MAGE-1, -2, -3, -12, RAGE-1, GAGE 1-, -2, BAGE, NY-ESO-1, beta-catenin, CDCP-1, CDC-27, SART-1, EpCAM, CD20, CD23, CD33, EGFR, HER-2, breast tumor-associated antigens BTA-1 and BTA-2, RCASl (receptor-binding cancer antigen expressed on SiSo cells), PLACenta-specific 1 ( PLAC-1), syndecan, MN (gp250), idiotype, among others.
  • Tumor associated antigens also include the blood group antigens, for example, Le a , Le b , LeX, LeY, H-2, B-l, B-2 antigens. (See Table 19 at the end of the specification). Ideally, for the purposes of this invention, TAA or TSA targets do not get internalized upon binding.
  • non-natural amino acid refers to an amino acid that is not one of the 20 common amino acids including, for example, amino acids that occur by modification (e.g. post-translational modifications) of a naturally encoded amino acid (including but not limited to, the 20 common amino acids or pyrolysine and selenocysteine) but are not themselves naturally incorporated into a growing polypeptide chain by the translation complex.
  • non-naturally-occurring amino acids include, but are not limited to, N-acetylglucosaminyl-L-serine, N-acetylglucosaminyl-L- threonine, and O-phosphotyrosine.
  • Constantly modified variants applies to both amino acid and nucleic acid sequences. With respect to particular nucleic acid sequences, conservatively modified variants refers to those nucleic acids which encode identical or essentially identical amino acid sequences or, where the nucleic acid does not encode an amino acid sequence, to essentially identical nucleic acid sequences. Because of the degeneracy of the genetic code, a large number of functionally identical nucleic acids may encode any given protein.
  • amino acid sequences one of skill will recognize that an individual substitution to a nucleic acid, peptide, polypeptide, or protein sequence which substitutes an amino acid or a particular percentage of amino acids in the encoded sequence for a conserved amino acid is a "conservatively modified variant.” Conservative substitution tables providing functionally similar amino acids are well known in the art.
  • samples or assays comprising a given, e.g., protein, gene, cell, or organism, are treated with a potential activator or inhibitor and are compared to control samples without the inhibitor.
  • Control samples i.e., not treated with antagonist, are assigned a relative activity value of 100%.
  • Inhibition is achieved when the activity value relative to the control is about 90% or less, typically 85% or less, more typically 80%> or less, most typically 75% or less, generally 70%> or less, more generally 65%> or less, most generally 60%> or less, typically 55% or less, usually 50%> or less, more usually 45%) or less, most usually 40%> or less, preferably 35% or less, more preferably 30% or less, still more preferably 25% or less, and most preferably less than 25%.
  • Activation is achieved when the activity value relative to the control is about 110%, generally at least 120%, more generally at least 140%, more generally at least 160%, often at least 180%, more often at least 2-fold, most often at least 2.5-fold, usually at least 5-fold, more usually at least 10-fold, preferably at least 20-fold, more preferably at least 40-fold, and most preferably over 40-fold higher.
  • Endpoints in activation or inhibition can be monitored as follows. Activation, inhibition, and response to treatment, e.g., of a cell, physiological fluid, tissue, organ, and animal or human subject, can be monitored by an endpoint.
  • the endpoint may comprise a predetermined quantity or percentage of, e.g., an indicator of inflammation, oncogenicity, or cell degranulation or secretion, such as the release of a cytokine, toxic oxygen, or a protease.
  • the endpoint may comprise, e.g., a predetermined quantity of ion flux or transport; cell migration; cell adhesion; cell proliferation; potential for metastasis; cell differentiation; and change in phenotype, e.g., change in expression of gene relating to inflammation, apoptosis, transformation, cell cycle, or metastasis (see, e.g., Knight (2000) Ann. Clin. Lab. Sci. 30: 145- 158; Hood and Cheresh (2002) Nature Rev. Cancer 2:91-100; Timme, et al. (2003) Curr. Drug Targets 4:251-261; Robbins and Itzkowitz (2002) Med. Clin. North Am. 86: 1467-1495; Grady and Markowitz (2002) Annu. Rev. Genomics Hum. Genet. 3: 101-128; Bauer, et al. (2001) Glia 36:235-243; Stanimirovic and Satoh (2000) Brain Pathol. 10: 113-126).
  • An endpoint of inhibition is generally 75% of the control or less, preferably 50% of the control or less, more preferably 25% of the control or less, and most preferably 10% of the control or less.
  • an endpoint of activation is at least 150% the control, preferably at least two times the control, more preferably at least four times the control, and most preferably at least 10 times the control.
  • composition that is "labeled” is detectable, either directly or indirectly, by spectroscopic, photochemical, biochemical, immunochemical, isotopic, or chemical methods.
  • useful labels include P, P, S, C, H, I, stable isotopes, fluorescent dyes, electron-dense reagents, substrates, epitope tags, or enzymes, e.g., as used in enzyme-linked immunoassays, or fluorettes (see, e.g., Rozinov and Nolan (1998) Chem. Biol. 5:713-728).
  • amino acid modification(s) and “modification(s)” refer to amino acid substitutions, deletions or insertions or any combinations thereof in an amino acid sequence relative to another amino acid sequence, for example a native amino acid sequence.
  • substitutional variants herein are those that have at least one amino acid residue in a native CTLD sequence removed and a different amino acid inserted in its place at the same position.
  • the substitutions may be single, where only one amino acid in the molecule has been substituted, or they may be multiple, where two or more amino acids have been substituted in the same molecule.
  • Specific reference to more than one amino acid substitution in a CTLD refers to multiple substitutions in which each individual amino acid substitution can occur at any amino acid position within the CTLD, including consecutive and non-consecutive amino acid positions.
  • specific reference to more than one amino acid insertion or deletion in a CTLD refers to multiple insertions or deletions in which each individual amino acid insertion or deletion can occur at any amino acid position within the CTLD, including consecutive and non-consecutive amino acid positions.
  • nucleic acid molecule encoding refers to the order or sequence of deoxyribonucleotides along a strand of deoxyribonucleic acid. The order of these deoxyribonucleotides determines the order of amino acids along the polypeptide chain. The DNA sequence thus encodes the amino acid sequence.
  • Randomize refers to ensembles of polypeptide or nucleic acid sequences or segments, in which the amino acid residue or nucleotide at one or more sequence positions may differ between different members of the ensemble of polypeptides or nucleic acids, such that the amino acid residue or nucleotide occurring at each such sequence position may belong to a set of amino acid residues or nucleotides that may include all possible amino acid residues or nucleotides or any restricted subset thereof.
  • the invention is directed to a polypeptide having a multimerizing domain and at least one polypeptide binding member that binds to IL-23R.
  • the binding member may either be linked to the multimerizing domain, for example at the N- or the C-terminus.
  • the polypeptides of the invention are non-natural polypeptides, for example, fusion proteins of a multimerizing domain and a polypeptide sequence that binds an IL-23R.
  • the non-natural polypeptides may also be natural polypeptides wherein the naturally occurring amino acid sequence has been altered by the addition, deletion, or substitution of amino acids.
  • polypeptides having a C-type Lectin Like Domain examples include polypeptides having a C-type Lectin Like Domain (CTLD) wherein one or more of the loop regions of the domains have been modified as described herein.
  • CTLD C-type Lectin Like Domain
  • the polypeptide that binds to IL-23R is a fragment or variant of a natural polypeptide that binds to the receptor, wherein when the naturually occurring polypeptide, variant or fragment is fused to a multimerizing domain, the fusion protein is no longer a naturally occurring polypeptide. Accordingly, the invention does not exclude naturally occurring polypeptide, fragments or variants thereof from being a part of fusion protein of the invention.
  • the polypeptide is an IL-23R antagonist that binds to IL-23R and prevents signaling through the IL-23 pathway.
  • the polypeptide binds IL23-R (SEQ ID NO: 5) or variants thereof.
  • the polypeptides of the invention bind to one or more sites on IL-23R that prevents binding of the native IL-23 ligand and thereby prevent activation of the receptor by the IL-23 ligand.
  • the polypeptides of the invention do not have agonist activity and do not activate the IL-23 heterdimeric receptor.
  • the polypeptide does not specifically bind to IL- 12R i or IL-12R 2. Accordingly, use of the polypeptide of the invention in therapeutic compositions can avoid the consequences of the unwanted blocking the activity of IL-12 for certain therapies.
  • a monomeric polypeptide of the invention includes at least two segments: a multimerizing domain that is capable of forming a multimeric complex with other multimerizing domains, and a polypeptide sequence that binds to IL-23R.
  • the sequence that binds to IL-23R may be fused with the multimerizing domain at the N- terminus, at the C-terminus, or at both the N- and C-termini of the domain.
  • the polypeptide that binds to IL-23R at the N-terminus is different than the polypeptide that binds IL-23R at the C terminus of the trimerizing domain.
  • a first polypeptide that binds IL-23R is fused at one of the N- terminus and the C-terminus of a trimerizing domain
  • a second polypeptide that is a modulator of inflammation is fused at the other of the N-terminus or the C-terminus of the trimerizing domain.
  • Modulators that are not polypeptides can be linked to the trimerizing domain, either covalently or non-covalently, as would be understood by one of skill in the art.
  • other polypeptide and non-polypeptide therapeutic agents can be linked to the trimerizing module.
  • another aspect of the invention includes a multimerizing domain having a polypeptide that binds to IL-23R on one end of the domain (one of either of the N-terminus or C-terminus), and a polypeptide that binds to tumor-associated (TAA) or tumor-specific antigens (TSA) on the other end (the other of the N-terminus and the C- terminus).
  • TAA tumor-associated
  • TSA tumor-specific antigens
  • the domain that binds to TAA's or TSA's may be peptides, such as for example CTLDs, single chain antibodies, or any type of domain that specifically binds to the desired target.
  • the activity of death receptor agonists can be enhanced by designing a molecule with binding activity mediated through an IL-23R binding polypeptide one end of a trimerizing domain that drives the drug to sites of inflammation in the setting of cancer and that allows clustering of the death receptor specific polypeptide on the second end of the trimerizing domain.
  • the polypeptide binds to a death receptors at lower affinity than to IL-23R. More specifically, the polypeptide that binds to IL-23R may bind with least 2 times greater affinity, for example, 2, 2.5, 3, 3.5, 4, 4.5 5, 10, 15, 20, 50 and 100 times greater, than the polypeptide binds the death receptor.
  • Indications for trimeric complexes having both IL-23R-binding polypeptide(s) and TAA or TSA targeting agent(s) include non-small cell lung cancer (NSCLC), colorectal cancer, ovarian cancer, renal cancer, pancreatic cancer, sarcomas, non-hodgkins lymphoma (NHL), multiple myeloma, breast cancer, prostate cancer, melanoma, glioblastoma, neuroblastoma.
  • NSCLC non-small cell lung cancer
  • NDL non-hodgkins lymphoma
  • multiple myeloma breast cancer
  • prostate cancer melanoma
  • glioblastoma neuroblastoma.
  • a polypeptide that specifically binds to an IL-23 receptor is contained in the loop region of a CTLD.
  • the polypeptide may be a portion of the IL-23 polypeptide, or may be sequence that is identified as provided here.
  • the sequence is contained in a loop region of a CLTD, and the CTLD is fused to a trimerizing domain at the N-terminus or C-terminus of the domain either directly or through the appropriate linker.
  • the polypeptide of the invention may include a second CLTD domain, fused at the other of the N-terminus and C-terminus, wherein the sequence of the CTLDs and/or their affinity for IL-23R may be the same or different.
  • the polypeptide includes a polypeptide that binds to an IL-23R at one of the termini of the trimerizing domain and a CLTD at the other of the termini.
  • One, two or three of the polypeptides can be part of a trimeric complex containing up to six specific binding members for IL-23R.
  • the polypeptide sequences that bind IL-23R can have a binding affinity for IL- 23R that is about equal to the binding affinity that native IL-23 has for IL-23R. In certain embodiments, the polypeptides of the invention have a binding affinity for the IL-23R that is greater or less than the binding affinity that native IL-23 has for the same IL-23R.
  • the polypeptides of the invention can include one or more amino acid mutations in a native IL-23 (pi 9) sequence, or a random sequence, that has selective binding affinity for IL-23R, but not IL-12R i or IL-12R 2.
  • a native IL-23 (pi 9) sequence or a random sequence, that has selective binding affinity for IL-23R, but not IL-12R i or IL-12R 2.
  • binding affinity of such binding members to the IL-23R is approximately equal (unchanged) or greater than (increased) as compared to native IL-23, and the binding affinity of the binding member to IL-12R i or IL- 12R 2 is less than or nearly eliminated as compared to native sequence IL-23
  • the binding affinity of the binding member, for purposes herein is considered "selective" for IL-23R.
  • the affinity of the binding member for IL-23R is less than the affinity of IL- 23 for the receptor, but the binding member is still selective for the receptor if it has greater affinity for IL-23R than its affinity for IL-12R l or IL-12R 2.
  • Preferred IL-23R selective antagonists of the invention will have at least 5-fold, preferably at least a 10-fold greater binding affinity to IL-23R as compared to IL-12R i or IL-12RP2, and even more preferably, will have at least 100-fold greater binding affinity to IL-23R as compared to a IL-12Rpi or IL-12RP2.
  • the respective binding affinity of the antagonists can be determined and compared to the binding properties of native IL-23, or a portion thereof, by ELISA, RIA, and/or BIAcore assays, known in the art.
  • Preferred IL-23R selective antagonists of the invention will not inhibit IL-12 signaling in at least one type of mammalian cell, and such signal inhibition can be determined by known art methods such as ELISA.
  • IL-23R antagonist comprises an antibody or an antibody fragment.
  • antibody is used to describe an immunoglobulin whether natural or partly or wholly synthetically produced.
  • antibody should be construed as covering any specific binding member or substance having a binding domain with the required receptor specificity.
  • this term covers antibody fragments, derivatives, functional equivalents and homologues of antibodies, including any polypeptide comprising an immunoglobulin binding domain, whether natural or wholly or partially synthetic. Chimeric molecules comprising an immunoglobulin binding domain, or equivalent, fused to another polypeptide are therefore included.
  • the term also covers any polypeptide or protein having a binding domain which is, or is homologous to, an antibody binding domain, e.g. antibody mimics. These can be derived from natural sources, or they may be partly or wholly synthetically produced. Examples of antibodies are the immunoglobulin isotypes and their isotypic subclasses; fragments which comprise an antigen binding domain such as Fab, Fab', F(ab') 2 , scFv, Fv, dAb, Fd; and diabodies.
  • the invention in another aspect relates to a multimeric complex of three polypeptides, each of the polypeptides comprising a multimerizing domain and at least one polypeptide that binds to IL-23R.
  • the multimeric complex comprises a polypeptide having a multimerizing domain selected from a polypeptide having substantial homology to a human tetranectin trimerizing structural element, or other human trimerizing polyeptides including mannose binding protein (MBP) trimerizing domain, a collectin neck region polypeptide, and others.
  • MBP mannose binding protein
  • the multimeric complex can be comprised of any of the polypeptides of the invention wherein the polypeptides of the multimeric complex comprise multimerizing domains that are able to associate with each other to form a multimer.
  • the multimeric complex is a homomultimeric complex comprised of polypeptides having the same amino acid sequences.
  • the multimeric complex is a heteromultimeric complex comprised of polypeptides having different amino acid sequences such as, for example, different multimerizing domains, and/or different polypeptides that bind to an IL-23R.
  • the heteromultimeric complexes can include a therapeutic agent and IL-23R antagonists.
  • the invention relates to a method for preparing a polypeptide that prevents activation of IL-23R in a cell expressing IL-23R.
  • the method includes the steps of: (a) selecting a first polypeptide(s) that specifically binds IL-23R; (b) grafting the first polypeptide(s) into one or two loop regions of tetranectin CTLD to form a first binding determinant or directly fusing the polypeptide to the tetranectin trimerizing domain, and (c) fusing the first CTLD with one of the N-terminus or the C-terminus of a tetranectin trimerizing domain.
  • the polypeptide that binds IL-23R does not bind IL-12R l or IL-12R 2.
  • the tetranectin CTLD has up to five loop regions into which binding members for IL-23R may be inserted or identified by selection from a randomized library as described here. Accordingly, when a polypeptide of the invention includes a CTLD, the polypeptide may have up to five binding members for IL-23R attached to the trimerizing domain through the CTLD. Each of the binding members may be the same or different.
  • a receptor antagonist can be bound to one terminus of a trimerizing domain and one or more therapeutic agents may be bound to the second terminus.
  • the agent may be bound directly or through an appropriate linker as understood to those of skill in the art. Such agents may act in the same pathway as the antagonist, or may act in a different pathway for immune disorders, cancers and other conditions.
  • the agent may be covalently linked to the trimerizing domain via a peptide bond to a side chain in the trimerizing domain or via a bond to a cysteine residue.
  • Other ways of covalently coupling the agent to the module can also be used as shown in, for example, US 6,190,886, which is incorporated by reference herein.
  • a specific binding member for IL-23R can be obtained from a random library of polypeptides by selection of members of the library that specifically bind to the receptor.
  • a number of systems for displaying phenotypes with putative ligand binding sites are known. These include: phage display (e.g. the filamentous phage fd [Dunn (1996), Griffiths and Duncan (1998), Marks et al. (1992)], phage lambda [Mikawa et al. (1996)]), display on eukaryotic virus (e.g. baculovirus [Ernst et al. (2000)]), cell display (e.g. display on bacterial cells [Benhar et al. (2000)], yeast cells [Boder and Wittrup (1997)], and mammalian cells [Whitehorn et al. (1995)], ribosome linked display [Schaffitzel et al.
  • US2007/0275393 which is incorporated herein by reference in its entirety, specifically describes a procedure for accomplishing a display system for the generation of CLTD libraries.
  • the general procedure includes (1) identification of the location of the loop- region, by referring to the 3D structure of the CTLD of choice, if such information is available, or, if not, identification of the sequence locations of the ⁇ 2, ⁇ 3 and ⁇ 4 strands by sequence alignment with known sequences, as aided by the further corroboration by identification of sequence elements corresponding to the ⁇ 2 and ⁇ 3 consensus sequence elements and ⁇ 4-8 ⁇ 3 ⁇ 4 ⁇ characteristics, also disclosed above; (2) subcloning of a nucleic acid fragment encoding the CTLD of choice in a protein display vector system with or without prior insertion of endonuclease restriction sites close to the sequences encoding ⁇ 2, ⁇ 3 and ⁇ 4; and (3) substituting the nucleic acid fragment encoding some or all of the loop-region of the CTLD of choice with randomly selected members
  • a complex may be formed that functions as a homo-trimeric protein that blocks natural IL-23 from binding and activating IL-23R.
  • peptides with IL-23R binding activity must be identified first. To accomplish this, peptides with known binding activity can be used or additional new peptides identified by screening from display libraries. A number of different display systems are available, such as but not limited to phage, ribosome and yeast display.
  • libraries can be constructed and initially screened for binding to IL-23R, either as single monomeric CTLD domains, or individual peptides displayed on the surface of phage. Once sequences with IL-23R binding activity have been identified these sequences would subsequently be grafted on to the trimerization domain of human tetranectin to create potential protein therapeutics capable of binding IL-23R.
  • the first strategy would be to construct and/or use random peptide phage display libraries. Random linear peptides and/or random peptides constructed as disulfide constrained loops would be individually displayed on the surface of phage particles and selected for binding to the desired IL-23R through phage display
  • a second strategy for construction of phage display libraries and trimerization domain constructs would include obtaining CTLD derived binders. Libraries can be constructed by randomizing the amino acids in one or more of the five different loops within the CTLD scaffold of human tetranectin displayed on the surface of phage. Binding to the IL- 23R can be selected for through phage display panning. After obtaining CTLD clones with peptide loops demonstrating IL-23R binding activity, these CTLD clones can then be grafted on to the trimerization domain of human tetranectin and screened for antagonist activity.
  • a third strategy for construction of phage display libraries and trimerization domain constructs would include taking known sequences with binding capabilities to IL- 23R and graft these directly on to the trimerization domain of human tetranectin and screen for binding activity.
  • a fourth strategy includes using peptide sequences with known binding capabilities to the IL-23R and first improve their binding by creating new libraries with randomized amino acids flanking the peptide or/and randomized selected internal amino acids within the peptide, followed by selection for improved binding through phage display. After obtaining binders with improved affinity, the binders of these peptides can be grafted on to the trimerization domain of human tetranectin and screening for antagonist activity.
  • initial libraries can be constructed as either free peptides displayed on the surface of phage particles, as in the first strategy (above), or as constrained loops within the CTLD scaffold as in the second strategy also discussed above. After obtaining binders with improved affinity, grafting of these peptides on to the trimerization domain of human tetranectin and screening for antagonist activity would occur.
  • Peptide display library kits such as, but not limited to, the New England Biolabs Ph.D. Phage display Peptide Library Kits are sold commercially and can be purchased for use in selection of new and novel peptides with IL-23R binding activity. Three forms of the New England Biolabs kit are available: the Ph.D. -7 Peptide Library Kit containing linear random peptides 7 amino acids in length, with a library size of 2.8x10 9 independent clones, the Ph.D.- C7C Disulfide Constrained Peptide Library Kit containing peptides constructed as disulfide constrained loops with random peptides 7 amino acids in length and a library size of 1.2x10 9 independent clones, and the Ph.D.
  • ribosome display libraries might be beneficial where larger/longer random peptides are involved.
  • NNK or NNS random nucleotide strategy is used for disulfide constrained libraries. However, these random positions are flanked by cysteine amino acid residues, to allow for disulfide bridge formation. The N terminal cysteine is often preceded by an additional amino acid such as alanine.
  • a flexible linker made up to but not limited to several glycine residues may act as a spacer between the peptides and the gene III protein for any of the above random peptide libraries.
  • the human tetranectin CTLD shown in FIGs. 4 and 5 contains five loops (four loops in LSA and one loop comprising LSB), which can be altered to confer binding of the CTLD to different protein targets. Random amino acid sequences can be placed in one or more of these loops to create libraries from which CTLD domains with the desired binding properties can be selected. Construction of these libraries containing random peptides constrained within any or all of the five loops of the human tetranectin CTLD can be accomplished (but is not limited to) using either a NNK or NNS as described above in strategy 1.
  • a single example of a method by which seven random peptides can be inserted into loop 1 of the TN CTLD is as follows.
  • PCR can be accomplished using primers IX for (SEQ ID NO: ) and IX rev2 (SEQ ID NO: ) in a PCR reaction without template to generate fragment A, and primers BstXlfor (SEQ ID NO: ) and PstBssRevC (SEQ ID NO: ) can be used in a separate PCR reaction without template to generate fragment B.
  • PCR can be performed using a high fidelity polymerase or taq blend and standard PCR thermocycling conditions. These two overlapping fragments can then be purified and used together, along with the outer primers Bglforl2 (SEQ ID NO: 5) and PstRev (SEQ ID NO: 6), to generate the desired DNA fragment by PCR.
  • This purified fragment can then be ligated into a similarly digested phage display vector such as pPHCPAB (SEQ ID NO: 150) or pANA27 (SEQ ID NO: 164) containing the restriction modified CTLD fused to Gene III, (See Fig. 6).
  • Modification of other loops by replacement with randomized amino acids can be similarly performed as shown above.
  • the replacement of defined amino acids within a loop with randomized amino acids is not restricted to any specific loop, nor is it restricted to the original size of the loops. Likewise, total replacement of the loop is not required, partial replacement is possible for any of the loops. In some cases retention of some of the original amino acids within the loop, such as the calcium coordinating amino acids shown in Figure 7 may be desirable. In these cases, replacement with randomized amino acids may occur for either fewer of the amino acids within the loop to retain the calcium coordinating amino acids, or additional randomized amino acids may be added to the loop to increase the overall size of the loop yet still retain these calcium coordinating amino acids.
  • Very large peptides can be accommodated and tested by combining loop regions such as loops 1 and 2 or loops 3 and 4 into one larger replacement loop.
  • CTLDs such as but not limited to the MBL CTLD, can be used instead of the CTLD of tetranectin. Grafting of peptides into these CTLDs can occur using methods similar to those described above.
  • the polypeptides that bind to an IL- 23R can be identified using a combinatorial peptide library, and a library of nucleic acid sequences encoding the polypeptides of the library, based upon a CTLD backbone, wherein the CTLDs of the polypeptides have been modified according to a number of exemplary schemes, which have been labeled for the purposes of identification only as Schemes (a)-(h): [0133]
  • the invention provides a combinatorial peptide library, and a library of nucleic acid sequences encoding the polypeptides of the library, wherein the CTLDs of the polypeptides have been modified according to a number of schemes, which have been labeled for the purposes of identification only as Schemes (a)- j). While each scheme is more particularly described herein, the modifications are at least as follows:
  • amino acid modifications in at least one of four loops in loop segment A (LSA) of the CTLD wherein the amino acid modifications comprise random substitution of at least five amino acids within Loop 1 and random substitution of at least three amino acids within Loop 2;
  • amino acid modifications in at least one of four loops in loop segment A (LSA) of the CTLD wherein the amino acid modifications comprise at least one amino acid insertion in Loop 3 and random substitution of at least three amino acids within Loop 3;
  • amino acid modifications in at least one of four loops in loop segment A (LSA) of the CTLD wherein the amino acid modifications comprise a modification that combines two loops into a single loop, wherein the two combined loops are Loop 3 and Loop 4;
  • amino acid modifications in at least one of the five loops in loop segment A (LSA) and loop segment B (LSB) of the CTLD wherein the amino acid modifications comprise random substitution of at least five amino acid residues in Loop 3 and random substitution of at least three amino acids within Loop 5
  • amino acid modifications in at least one of the four loops in loop segment A (LSA) of the CTLD wherein the amino acid modifications comprise random substitution of at least one amino acid and insertion of at least six amino acids in Loop 3;
  • amino acid modifications in at least one of the four loops in the loop segment A (LSA) of the CTLD wherein the amino acid modifications comprise a mixture of (1) random substitution of at least six amino acids in Loop 3 and (2) random substitution of at least six amino acids and at least one amino acid insertion in Loop 3; and
  • amino acid modifications in at least one of the four loops in the loop segment A (LSA) of the CTLD wherein the amino acid modifications comprise at least four or more amino acid insertions in at least one of the four loops in the loop segment A (LSA) or loop 5 in loop segment B (LSB) of the CTLD.
  • the invention provides a combinatorial polypeptide library comprising polypeptide members having a randomized C-type lectin domain (CTLD), wherein the randomized CTLD includes amino acid modifications in at least one of the four loops in LSA or in the loop in LSB of the CTLD, wherein the amino acid modifications comprise at least one amino acid insertion in Loop 1 and random substitution of at least five amino acids within Loop 1.
  • CTLD C-type lectin domain
  • the CTLD when the CTLD is from human tetranectin, the CTLD also has a random substitution of Arginine-130.
  • this peptide is located immediately adjacent to the C-terminal peptide of Loop 2 in the C-terminal direction.
  • this peptide is Gly-130.
  • the CTLD when the CTLD is from human or mouse tetranectin, the CTLD includes a substitution of Lysine-148 to Alanine in Loop 4.
  • the amino acid modifications when the combinatorial library has the modified CTLD of Scheme (a), the amino acid modifications comprise two amino acid insertions in Loop 1 and random substitution of at least five amino acids within Loop 1. In other embodiments, when the combinatorial library has the modified CTLD of scheme (a) and the CTLD is from human tetranectin, the amino acid modifications comprise at least one amino acid insertion in Loop 1 , random substitution of at least five amino acids within Loop 1 , and include a random substitution of Arginine 130.
  • the amino acid modifications when the combinatorial library has the modified CTLD of scheme (a) and the CTLD is from human tetranectin, the amino acid modifications comprise two amino acid insertions in Loop 1 , random substitution of five amino acids within Loop 1, and a random substitution of Arginine 130.
  • the amino acid modifications when the combinatorial library has the modified CTLD of scheme (a) and the CTLD is from mouse tetranectin, the amino acid modifications comprise two amino acid insertions in Loop 1 , random substitution of five amino acids within Loop 1 , and a random substitution of Leucine 130.
  • the amino acid modifications can further comprise a substitution of Lysine- 148 to Alanine.
  • the CTLD comprises two amino acid insertions in Loop 1 , random substitution of at least five amino acids within Loop 1, random substitution of Arginine- 130 or other amino acid located outside and adjacent to loop 2 in the C-terminal direction, and a substitution of lysine-148 to alanine in Loop 4.
  • the invention provides a combinatorial polypeptide library comprising polypeptide members having a randomized C-type lectin domain (CTLD), wherein the randomized CTLD comprises amino acid modifications in at least one of the four loops in the LSA of the CTLD, wherein the amino acid modifications comprise random substitution of at least five amino acids within Loop 1 and random substitution of at least three amino acids within Loop 2.
  • CTLD C-type lectin domain
  • the amino acid modifications comprise random substitution of at least five amino acids within Loop 1 , random substitution of at least three amino acids within Loop 2, and random substitution of Arginine-130, or other amino acid located outside and adjacent to loop 2 in the C-terminal direction.
  • the amino acid modifications include random substitutions of at least five amino acids in Loop 1, random substitution of at least three amino acids in Loop 2, and include a random substitution of Arginine 130.
  • the amino acid modifications include random substitutions of five amino acids in Loop 1 , random substitution of three amino acids in Loop 2, and a random substitution of Arginine 130.
  • the amino acid modifications include random substitutions of at least five amino acids in Loop 1, random substitution of at least three amino acids in Loop 2, and include a random substitution of Leucine 130.
  • the amino acid modifications include random substitutions of five amino acids in Loop 1, random substitution of three amino acids in Loop 2, and a random substitution of Leucine 130.
  • the amino acid modifications can further comprise a substitution of Lysine- 148 to Alanine.
  • the amino acid modifications comprise random substitution of at least five amino acids within Loop 1 , random substitution of at least three amino acids within Loop 2, and random substitution of Arginine-130, or other amino acid located outside and adjacent to loop 2 in the C-terminal direction and a substitution of Lysine-148 to Alanine in Loop 4.
  • the invention provides a combinatorial polypeptide library comprising polypeptide members that have a randomized C-type lectin domain (CTLD), wherein the randomized CTLD comprises amino acid modifications in at least one of the four loops in loop segment A (LSA) of the CTLD, wherein the amino acid
  • modifications comprise random substitution of at least seven amino acids within Loop 1 and at least one amino acid insertion in Loop 4.
  • the polypeptide members of the combinatorial library further comprise random substitution of at least two amino acids within Loop 4.
  • the amino acid modifications comprise three amino acid insertions within Loop 4 and optionally further comprise random substitution of at least two amino acids.
  • the amino acid modifications comprise random substitution of at least seven amino acids within Loop 1 , at least three amino acid insertions in Loop 4, and random substitution of at least two amino acids within Loop 4.
  • the amino acid modifications comprise random substitution of seven amino acids within Loop 1 , three amino acid insertions in Loop 4, and random substitution of two amino acids within Loop 4.
  • the invention provides a combinatorial polypeptide library comprising polypeptide members that have a randomized C-type lectin domain (CTLD), wherein the randomized CTLD comprises amino acid modifications in at least one of the four loops in the loop segment A (LSA) of the CTLD, wherein the amino acid modifications comprise at least one amino acid insertion in loop 3 and random substitution of at least three amino acids within Loop 3.
  • CTLD C-type lectin domain
  • the amino acid modifications when the combinatorial library has the modified CTLD of Scheme (d), can further comprise at least one amino acid insertion in Loop 4, and can further comprise random substitution of at least three amino acids within Loop 4.
  • the amino acid modifications can comprise three amino acid insertions in Loop 3.
  • the amino acid modifications can comprise three amino acid insertions in Loop 4.
  • the amino acid modifications comprise random substitution of at least three amino acids within Loop 3, random substitution of at least three amino acids within Loop 4, at least one amino acid insertion in Loop 3 and at least one amino acid insertion in Loop 4.
  • the amino acid modifications comprise random substitution of at least three amino acids within Loop 3, random
  • the amino acid modifications comprise random substitution of three amino acids within Loop 3, random substitution of three amino acids within Loop 4, three amino acid insertions in Loop 3, and three amino acid insertions in Loop 4.
  • the amino acid modifications can further compr random substitution of Lysine- 148 to Alanine or in Loop 4.
  • the invention provides a combinatorial polypeptide library comprising polypeptide members that have a randomized C-type lectin domain (CTLD), wherein the randomized CTLD comprises amino acid modifications in at least one of the four loops in the loop segment A (LSA) of the CTLD, wherein the amino acid modifications comprise a modification that combines two Loops into a single Loop, wherein the two combined Loops are Loop 3 and Loop 4.
  • CTLD C-type lectin domain
  • the amino acid modifications comprise random substitution of at least six amino acids within Loop 3 and random substitution of at least four amino acids within Loop 4.
  • the amino acid modifications comprise random substitution of six amino acids within Loop 3 and random substitution of four amino acids within Loop 4.
  • the amino acid modifications can further comprise random substitution of Proline- 144.
  • the amino acid modifications comprise random substitution of six amino acids within Loop 3, random substitution of four amino acids within Loop 4, and a random substitution of proline 144, resulting in a combined Loop 3 and Loop 4 amino acid sequence, comprising, for example, NWEXXXXXXX XGGXXXN (SEQ ID NO: ), wherein X is any amino acid and wherein the amino acid sequence of SEQ ID NO: forms a single Loop region.
  • combinatorial library comprise the sequence NWEXXXXXXX XGGXXXN (SEQ ID NO: ), wherein X is any amino acid and wherein the amino acid sequence of SEQ ID NO: forms a single loop from combined and modified Loop 3 and Loop 4.
  • the invention provides a combinatorial polypeptide library comprising polypeptide members that have a randomized C-type lectin domain (CTLD), wherein the randomized CTLD comprises amino acid modifications in at least one of the four loops in the loop segment A (LSA) of the CTLD, wherein the amino acid modifications comprise at least one amino acid insertion in Loop 4 and random substitution of at least three amino acids within Loop 4.
  • the amino acid modifications comprise four amino acid insertions in Loop 4.
  • the amino acid modifications comprise at least four amino acid insertions in Loop 4 and random substitution of at least three amino acids within Loop 4.
  • the amino acid substitutions comprise four amino acid insertions in Loop 4 and random substitution of three amino acids within Loop 4.
  • the polypeptide members of the combinatorial library comprise a modified Loop 3 and a modified Loop 5, wherein the modified Loop 3 comprises randomization of five amino acid residues and the modified Loop 5 comprises randomization of three amino acid residues.
  • the polypeptide members of the combinatorial library comprise a modified Loop 3, a modified Loop 5, and a modified Loop 4, wherein the modification to Loop 4 abrogates plasminogen binding.
  • the amino acid modifications can further comprise one or more amino acid modifications in Loop 4 that modulates plasminogen binding affinity of the CTLD, for example, the substitution of Lysine 148 to Alanine.
  • the amino acid modifications when the CTLD is from human tetranectin, comprise random substitution of at least five amino acid residues in Loop 3, random substitution of at least three amino acid residues in Loop 5, and substitution of Lysine 148 to Alanine in Loop 4.
  • the amino acid modifications comprises random substitution of five amino acid residues in Loop 3 and random substitution of three amino acid residues in Loop 5, and, in another specific embodiment, when the CTLD is from human tetranectin, the amino acid modifications further comprise substitution of Lysine 148 to Alanine in Loop 4.
  • the invention provides a combinatorial polypeptide library comprising polypeptide members that have a randomized C-type lectin domain (CTLD), wherein the randomized CTLD comprises amino acid modifications in at least one of the four loops in the loop segment A (LSA) of the CTLD, wherein the amino acid modifications comprise random substitution of at least one amino acid and at least six amino acid insertions.
  • CTLD C-type lectin domain
  • the amino acid modifications can further comprise one or more amino acid modifications in Loop 4 that modulates plasminogen binding affinity of the CTLD, for example, the substitution of lysine 148 to Alanine.
  • the members of the combinatorial library when the CTLD is from human tertranectin, have random substitution of at least one amino acid and insertion of at least six amino acids in Loop 3, and substitution of Lysine 148 to Alanine in Loop 4.
  • the amino acid modifications comprise random substitution of one amino acid and insertion of six amino acids in Loop 3.
  • the members of the combinatorial library when the CTLD is from human tertranectin, have random substitution of one amino acid and insertion of six amino acids in Loop 3, and substitution of lysine 148 to alanine in Loop 4.
  • one of the substitutions is the substitution of Isoleucine 140.
  • the invention provides a combinatorial polypeptide library comprising polypeptide members that have a randomized C-type lectin domain (CTLD), wherein the randomized CTLD comprises amino acid modifications in at least one of the four loops in the loop segment A (LSA) of the CTLD, wherein the amino acid modifications comprise a mixture of random substitution of six amino acids in Loop 3 and random substitution of six amino acids and one amino acid insertion in Loop 3.
  • the mixture further comprises random substitution of six amino acids and two amino acid insertions in Loop 3.
  • the amino acid modifications comprises a mixture of random substitution of six amino acids in Loop 3, random substitution of six amino acids and one amino acid insertion in Loop 3, and random substitution of six amino acids and two amino acid insertions in Loop 3.
  • the amino acid modifications when the CTLD is from human tetranectin, the amino acid modifications further comprise a substitution of Lysine 148 to Alanine in Loop 4.
  • the invention provides a combinatorial polypeptide library comprising polypeptide members that have a randomized C-type lectin domain (CTLD), wherein the randomized CTLD comprises amino acid modifications in at least one of the four loops in the loop segment A (LSA) of the CTLD, wherein the amino acid modifications in at least one of the four loops in the loop segment A (LSA) of the CTLD, wherein the amino acid modifications comprise at least four or more amino acid insertions in at least one of the four loops in the loop segment A (LSA) or loop 5 in loop segment B (LSB) of the CTLD.
  • CTLD randomized C-type lectin domain
  • the combinatorial library comprises one or more amino acid modifications to the Loop 4 region (alone or in combination with modifications to other regions of the CTLD)
  • certain of the modification(s) are designed to maintain, modulate, or abrogate the metal ion-binding affinity of the CTLD.
  • Such modifications affect the plasminogen-binding activity of the CTLD (see, e.g., Nielbo, et al, Biochemistry, 2004, 43 (27), pp 8636-8643; or Graversen 1998).
  • polypeptide members of the libraries can comprise one or more amino acid modifications (e.g., by insertion, substitution, extension, or randomization) in any
  • the randomized CTLD can comprise one or more amino acid modifications in the loop of the LSB loop region (Loop 5), either alone, or in combination with one or more amino acid modifications in any one, two, three, or four loops of the LSA loop region (Loops 1-4).
  • the invention provides a combinatorial polypeptide library comprising polypeptide members that have a randomized C-type lectin domain (CTLD), wherein the randomized CTLD comprises one or more amino acid modifications in at least one of the four loops in loop segment A (LSA) and one or more amino acid modifications in the loop in loop segment B (LSB) (Loop 5) of the CTLD, wherein the one or more amino acid modifications comprises randomization of the LSB amino acid residues.
  • C-type lectin domain C-type lectin domain
  • the polypeptide members of the combinatorial libraries can have one or more amino acid modifications in any two, three, four, or five loops in the loop region (LSA and LSB) of the CTLD (e.g., any random combination of random amino acid modifications to two loops, to three loops, to four loops, or to all five loops).
  • the polypeptide members of the combinatorial libraries can further comprise additional amino acid modifications to regions of the CTLD outside of the loop region (LSA and LSB), such as in the a-helices or ⁇ -strands (see, e.g., FIG. 1).
  • CTLD loop regions can be extended beyond the exemplary constructs detailed in the non-limiting Examples below.
  • the invention also provides a library of nucleic acid molecules encoding polypeptides of the combinatorial polypeptide library according to any one of the above-described aspects and embodiments.
  • the invention provides a library of nucleic acid sequences encoding the polypeptides of the library, wherein the CTLDs of the polypeptides have been modified according to Schemes (a)-(j).
  • polypeptides having preferred binding characteristics have been identified by one or more of modification schemes (a)-(h), including for example, SEQ ID NOS: 133-141 as set forth in Figure 8.
  • known polypeptides that bind to IL-23R can be cloned directly on to either the N or C terminal end trimerization domain as free linear peptides or as disulfide constrained loops using cysteines.
  • Single chain antibodies or domain antibodies capable of binding IL-23R can also be cloned on to either end of the trimerization domain.
  • peptides with known binding properties can be cloned directly into any one of the loop regions of the TN CTLD.
  • Peptides selected for as disulfide constrained loops or as complementary determining regions of antibodies might be quite amenable to relocation into the loop regions of the CTLD of human tetranectin. For all of these constructs, binding as a monomer, as well as binding and blocking activation as a trimer, when fused with the trimerization domain can then be tested for.
  • Another aspect the invention relates to a method preventing activation of IL-23R in a cell expressing IL-23R.
  • the method includes contacting the cell with an IL-23R binding polypeptide of the invention that includes a trimerizing domain and at least one polypeptide that specifically binds to the IL-23R.
  • the method comprises contacting the cell with a trimeric complex of the invention.
  • the IL-23R binding polypeptide may be an antagonist of IL-23R (or the heterodimeric receptor), or may bind to IL-23R to allow the local delivery of a therapeutic agent associated with the trimerizing domain, as described above, to a tumor, to a site of inflamation or other desired location presenting IL-23R.
  • the invention in another aspect relates to a method of treating a subject having a an immune disorder or a tumor by administering to the subject a therapeutically effective amount of IL-23R antagonist including polypeptide having a trimerizing domain and at least one polypeptide that specifically binds to the IL-23R.
  • the method comprises administering to the subject a trimeric complex of the invention.
  • Another aspect of the invention is directed to a combination therapy.
  • Formulations comprising IL-23R antagonists and therapeutic agents are also provided by the present invention. It is believed that such formulations will be particularly suitable for storage as well as for therapeutic administration.
  • the formulations may be prepared by known techniques. For instance, the formulations may be prepared by buffer exchange on a gel filtration column.
  • IL-23R antagonists and therapeutic agents described herein can be employed in a variety of therapeutic applications. Among these applications are methods of treating various cancers. IL-23R antagonists and therapeutic agents can be administered in accord with known methods, such as intravenous administration as a bolus or by continuous infusion over a period of time, by intramuscular, intraperitoneal, intracerobrospinal, subcutaneous, intraarticular, intrasynovial, intrathecal, oral, topical, or inhalation routes.
  • intravenous administration as a bolus or by continuous infusion over a period of time
  • intramuscular, intraperitoneal, intracerobrospinal subcutaneous, intraarticular, intrasynovial, intrathecal, oral, topical, or inhalation routes.
  • administration may be performed through mini-pump infusion using various commercially available devices.
  • Effective dosages and schedules for administering the IL-23R antagonists may be determined empirically, and making such determinations is within the skill in the art. Single or multiple dosages may be employed. It is presently believed that an effective dosage or amount of the antagonist used alone may range from about 1 ⁇ g/kg to about 100 mg/kg of body weight or more per day. Interspecies scaling of dosages can be performed in a manner known in the art, e.g., as disclosed in Mordenti et al., Pharmaceut. Res., 8: 1351 (1991).
  • IL-23R antagonist When in vivo administration of IL-23R antagonist is employed, normal dosage amounts may vary from about 10 ng/kg to up to 100 mg/kg of mammal body weight or more per day, preferably about 1 ⁇ g/kg/day to 10 mg/kg/day, depending upon the route of administration.
  • Guidance as to particular dosages and methods of delivery is provided in the literature [see, for example, U.S. Pat. Nos. 4,657,760; 5,206,344; or 5,225,212].
  • administration targeting one organ or tissue for example, may necessitate delivery in a manner different from that to another organ or tissue.
  • the dosage of IL-23R antagonist that must be administered will vary depending on, for example, the mammal which will receive IL-23R antagonist, the route of administration, and other drugs or therapies being administered to the mammal.
  • the one or more other therapies may include but are not limited to, administration of radiation therapy, cytokine(s), growth inhibitory agent(s), chemotherapeutic agent(s), cytotoxic agent(s), tyrosine kinase inhibitors, ras farnesyl transferase inhibitors, angiogenesis inhibitors, and cyclin-dependent kinase inhibitors or any other agent that enhances susceptibility of cancer cells to killing by IL-23R antagonists which are known in the art.
  • Preparation and dosing schedules for chemotherapeutic agents may be used according to manufacturers' instructions or as determined empirically by the skilled practitioner. Preparation and dosing schedules for such chemotherapy are also described in Chemotherapy Service Ed., M. C. Perry, Williams & Wilkins, Baltimore, Md. (1992). The chemotherapeutic agent may precede, or follow administration of the Apo2L variant, or may be given simultaneously therewith.
  • polypeptides of in the invention and therapeutic agents may be administered concurrently (simultaneously) or sequentially.
  • a non natural polypeptide of the invention, or multimeric (e.g., trimeric) complex thereof, and a therapeutic agent are administered concurrently.
  • a polypeptide or trimeric complex is administered prior to administration of a therapeutic agent.
  • a therapeutic agent is administered prior to a polypeptide or trimeric complex.
  • treated cells in vitro can be analyzed. Where there has been in vivo treatment, a treated mammal can be monitored in various ways well known to the skilled practitioner. For instance, tumor tissues can be examined pathologically to assay for cell death or serum can be analyzed for immune system responses.
  • the invention relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a therapeutically effective amount of the polypeptide of the invention along with a pharmaceutically acceptable carrier or excipient.
  • pharmaceutically acceptable carrier or “pharmaceutically acceptable excipient” includes any and all solvents, dispersion media, coating, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like that are physiologically compatible. Examples of
  • pharmaceutically acceptable carriers or excipients include one or more of water, saline, phosphate buffered saline, dextrose, glycerol, ethanol and the like as well as combinations thereof.
  • isotonic agents for example, sugars, polyalcohols such as mannitol, sorbitol, or sodium chloride in the composition.
  • Pharmaceutically acceptable substances such as wetting or minor amounts of auxiliary substances such as wetting or emulsifying agents, preservatives or buffers, which enhance the shelf life or effectiveness of the of the antibody or antibody portion also may be included.
  • disintegrating agents can be included, such as cross-linked polyvinyl pyrrolidone, agar, alginic acid or a salt thereof, such as sodium alginate and the like.
  • the pharmaceutical composition can include one or more of the following, carrier proteins such as serum albumin, buffers, binding agents, sweeteners and other flavoring agents; coloring agents and polyethylene glycol.
  • compositions can be in a variety of forms including, 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.
  • liquid solutions e.g. injectable and infusible solutions
  • dispersions or suspensions tablets, pills, powders, liposomes and suppositories.
  • the preferred form will depend on the intended route of administration and therapeutic application.
  • the compositions are in the form of injectable or infusible solutions, such as compositions similar to those used for passive immunization of humans with antibodies.
  • the mode of administration is parenteral (e.g., intravenous, subcutaneous, intraperitoneal, intramuscular).
  • the polypeptide (or trimeric complex) is administered by intravenous infusion or injection.
  • the polypeptide or trimeric complex is administered by intramuscular or subcutaneous injection.
  • compositions include, but are not limited to, rectal, transdermal, vaginal, transmucosal or intestinal administration.
  • compositions are typically 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 (i.e. polypeptide or trimeric complex) in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered 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.
  • 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.
  • An article of manufacture such as a kit containing IL-23R antagonists and therapeutic agents useful in the treatment of the disorders described herein comprises at least a container and a label.
  • Suitable containers include, for example, bottles, vials, syringes, and test tubes.
  • the containers may be formed from a variety of materials such as glass or plastic.
  • the label on or associated with the container indicates that the formulation is used for treating the condition of choice.
  • the article of manufacture may further comprise a container comprising a pharmaceutically-acceptable buffer, such as phosphate-buffered saline, Ringer's solution, and dextrose solution.
  • the article of manufacture may also comprise a container with another active agent as described above.
  • an appropriate amount of a pharmaceutically-acceptable salt is used in the formulation to render the formulation isotonic.
  • pharmaceutically-acceptable carriers include saline, Ringer's solution and dextrose solution.
  • the pH of the formulation is preferably from about 6 to about 9, and more preferably from about 7 to about 7.5. It will be apparent to those persons skilled in the art that certain carriers may be more preferable depending upon, for instance, the route of administration and concentrations of IL-23R antagonist and therapeutic agent.
  • compositions can be prepared by mixing the desired molecules having the appropriate degree of purity with optional pharmaceutically acceptable carriers, excipients, or stabilizers (Remington's Pharmaceutical Sciences, 16th edition, Osol, A. ed. (1980)), in the form of lyophilized formulations, aqueous solutions or aqueous suspensions.
  • Acceptable carriers, excipients, or stabilizers are preferably nontoxic to recipients at the dosages and concentrations employed, and include buffers such as Tris, HEPES, PIPES, phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol;
  • buffers such as Tris, HEPES, PIPES, phosphate, citrate, and other organic acids
  • antioxidants including ascorbic acid and methionine
  • preservatives such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzal
  • polypeptides such as serum albumin, gelatin, or immunoglobulins
  • proteins such as serum albumin, gelatin, or immunoglobulins
  • hydrophilic polymers such as polyvinylpyrrolidone
  • amino acids such as glycine, glutamine, asparagine, histidine, arginine, or lysine
  • sugars such as sucrose, mannitol, trehalose or sorbitol
  • salt-forming counter-ions such as sodium
  • non-ionic surfactants such as TWEENTM, PLURONICSTM or polyethylene glycol (PEG).
  • Such carriers include ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts, or electrolytes such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, and cellulose-based substances.
  • ion exchangers alumina, aluminum stearate, lecithin
  • serum proteins such as human serum albumin
  • buffer substances such as glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts, or electrolytes
  • protamine sulfate disodium hydrogen phosphate
  • potassium hydrogen phosphate sodium chloride
  • colloidal silica magnesium trisilicate
  • Carriers for topical or gel-based forms include polysaccharides such as sodium carboxymethylcellulose or methylcellulose, polyvinylpyrrolidone, polyacrylates, polyoxyethylene-polyoxypropylene-block polymers, polyethylene glycol, and wood wax alcohols.
  • conventional depot forms are suitably used.
  • Such forms include, for example, microcapsules, nano-capsules, liposomes, plasters, inhalation forms, nose sprays, sublingual tablets, and sustained-release preparations.
  • Formulations to be used for in vivo administration should be sterile. This is readily accomplished by filtration through sterile filtration membranes, prior to or following lyophilization and reconstitution.
  • the formulation may be stored in lyophilized form or in solution if administered systemically. If in lyophilized form, it is typically formulated in combination with other ingredients for reconstitution with an appropriate diluent at the time for use.
  • An example of a liquid formulation is a sterile, clear, colorless unpreserved solution filled in a single-dose vial for subcutaneous injection.
  • Therapeutic formulations generally are placed into a container having a sterile access port, for example, an intravenous solution bag or vial having a stopper pierceable by a hypodermic injection needle.
  • the formulations are preferably administered as repeated intravenous (i.v.), subcutaneous (s.c), intramuscular (i.m.) injections or infusions, or as aerosol formulations suitable for intranasal or intrapulmonary delivery (for intrapulmonary delivery see, e.g., EP 257,956).
  • sustained-release preparations include
  • sustained-release matrices include polyesters, hydrogels (e.g., poly(2-hydroxyethyl- methacrylate) as described by Langer et al., J. Biomed. Mater. Res., 15: 167-277 (1981) and Langer, Chem. Tech., 12: 98-105 (1982) or poly(vinylalcohol)), polylactides (U.S. Pat. No.
  • the polypeptide of the invention can be expressed in any suitable standard protein expression system by culturing a host transformed with a vector encoding the polypeptide under such conditions that the polypeptide is expressed.
  • the expression system is a system from which the desired protein may readily be isolated.
  • prokaryotic expression systems are available since high yields of protein can be obtained and efficient purification and refolding strategies.
  • selection of appropriate expression systems is within the knowledge of one skilled in the art.
  • the isolated polynucleotide encodes a polypeptide that specifically binds IL-23R and a trimerizing domain.
  • the isolated polynucleotide encodes a first polypeptide that specifically binds IL-23R, and a trimerizing domain.
  • the polypeptide that specifically binds IL-23R and the trimerizing domain are encoded in a single contiguous polynucleotide sequence (a genetic fusion).
  • polypeptide that specifically binds IL-23R and the trimerizing domain are encoded by non-contiguous polynucleotide sequences. Accordingly, in some embodiments the at least one polypeptide that specifically binds IL-23R and the trimerizing domain are expressed, isolated, and purified as separate polypeptides and fused together to form the polypeptide of the invention.
  • these recombinant DNA constructs may be inserted in- frame into any of a number of expression vectors appropriate to the chosen host.
  • the expression vector comprises a strong promoter that controls expression of the recombinant polypeptide constructs.
  • the resulting polypeptide can be isolated and purified using suitable standard procedures well known in the art, and optionally subjected to further processing such as e.g. lyophilization.
  • Standard techniques may be used for recombinant DNA molecule, protein, and polypeptide production, as well as for tissue culture and cell transformation. See, e.g., Sambrook, et al. (below) or Current Protocols in Molecular Biology (Ausubel et al., eds., Green Publishers Inc. and Wiley and Sons 1994). Purification techniques are typically performed according to the manufacturer's specifications or as commonly accomplished in the art using conventional procedures such as those set forth in Sambrook et al. (Molecular Cloning: A Laboratory Manual. Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY (1989), or as described herein. Unless specific definitions are provided, the
  • a flexible molecular linker optionally may be interposed between, and covalently join, the specific binding member and the trimerizing domain.
  • the linker is a polypeptide sequence of about 1-20 amino acid residues.
  • the linker may be less than 10 amino acids, most preferably, 5, 4, 3, 2, or 1. It may be in certain cases that 9, 8, 7 or 6 amino acids are suitable.
  • the linker is essentially non-immunogenic, not prone to proteolytic cleavage and does not comprise amino acid residues which are known to interact with other residues (e.g. cysteine residues).
  • conjugates are covalently attached (hereinafter "conjugated") to one or more chemical groups.
  • Chemical groups suitable for use in such conjugates are preferably not significantly toxic or immunogenic.
  • the chemical group is optionally selected to produce a conjugate that can be stored and used under conditions suitable for storage.
  • a variety of exemplary chemical groups that can be conjugated to polypeptides are known in the art and include for example carbohydrates, such as those carbohydrates that occur naturally on glycoproteins, polyglutamate, and non-proteinaceous polymers, such as polyols (see, e.g., U.S. Pat. No. 6,245,901).
  • a polyol for example, can be conjugated to polypeptides of the invention at one or more amino acid residues, including lysine residues, as is disclosed in WO 93/00109, supra.
  • the polyol employed can be any water-soluble poly(alkylene oxide) polymer and can have a linear or branched chain. Suitable polyols include those substituted at one or more hydroxyl positions with a chemical group, such as an alkyl group having between one and four carbons.
  • the polyol is a poly(alkylene glycol), such as poly(ethylene glycol) (PEG), and thus, for ease of description, the remainder of the discussion relates to an exemplary embodiment wherein the polyol employed is PEG and the process of conjugating the polyol to a polypeptide is termed "pegylation.”
  • PEG poly(ethylene glycol)
  • pegylation the process of conjugating the polyol to a polypeptide
  • other polyols such as, for example, poly(propylene glycol) and polyethylene- polypropylene glycol copolymers, can be employed using the techniques for conjugation described herein for PEG.
  • the average molecular weight of the PEG employed in the pegylation of the Apo- 2L can vary, and typically may range from about 500 to about 30,000 daltons (D) .
  • the average molecular weight of the PEG is from about 1,000 to about 25,000 D, and more preferably from about 1,000 to about 5,000 D.
  • pegylation is carried out with PEG having an average molecular weight of about 1,000 D.
  • the PEG homopolymer is unsubstituted, but it may also be substituted at one end with an alkyl group.
  • the alkyl group is a C1-C4 alkyl group, and most preferably a methyl group.
  • PEG preparations are commercially available, and typically, those PEG preparations suitable for use in the present invention are nonhomogeneous preparations sold according to average molecular weight.
  • polypeptide of the invention can be further modified using techniques known in the art, such as, conjugated to a small molecule compounds (e.g., a chemotherapeutic); conjugated to a signal molecule (e.g., a fluorophore); conjugated to a molecule of a specific binding pair (e.g,. biotin/streptavidin, antibody/antigen); or stabilized by glycosylation, PEGylation, or further fusions to a stabilizing domain (e.g., Fc domains).
  • a small molecule compounds e.g., a chemotherapeutic
  • a signal molecule e.g., a fluorophore
  • conjugated to a molecule of a specific binding pair e.g,. biotin/streptavidin, antibody/antigen
  • a stabilizing domain e.g., Fc domains
  • a variety of methods for pegylating proteins are known in the art. Specific methods of producing proteins conjugated to PEG include the methods described in U.S. Pat. Nos. 4,179,337, 4,935,465 and 5,849,535.
  • the protein is covalently bonded via one or more of the amino acid residues of the protein to a terminal reactive group on the polymer, depending mainly on the reaction conditions, the molecular weight of the polymer, etc.
  • the polymer with the reactive group(s) is designated herein as activated polymer.
  • the reactive group selectively reacts with free amino or other reactive groups on the protein.
  • the PEG polymer can be coupled to the amino or other reactive group on the protein in either a random or a site specific manner.
  • the type and amount of the reactive group chosen, as well as the type of polymer employed, to obtain optimum results will depend on the particular protein or protein variant employed to avoid having the reactive group react with too many particularly active groups on the protein. As this may not be possible to avoid completely, it is recommended that generally from about 0.1 to 1000 moles, preferably 2 to 200 moles, of activated polymer per mole of protein, depending on protein concentration, is employed. The final amount of activated polymer per mole of protein is a balance to maintain optimum activity, while at the same time optimizing, if possible, the circulatory half-life of the protein.
  • polyol when used herein refers broadly to polyhydric alcohol compounds.
  • Polyols can be any water-soluble poly(alkylene oxide) polymer for example, and can have a linear or branched chain.
  • Preferred polyols include those substituted at one or more hydroxyl positions with a chemical group, such as an alkyl group having between one and four carbons.
  • the polyol is a poly(alkylene glycol), preferably poly(ethylene glycol) (PEG).
  • PEG poly(ethylene glycol)
  • PEG poly(ethylene glycol)
  • polyols of the invention include those well known in the art and those publicly available, such as from commercially available sources.
  • half-life extending molecules can be attached to the N-or C- terminus of the trimerization domain including serum albumin-binding peptides, IgG-binding peptides or peptides binding to FcRn.
  • the vectors discussed in the following Examples are derived from vectors that have been previously described [See US 2007/0275393]. Certain vector sequences are provided in the Sequence Listing and one of skill will be able to derive vectors given the description provided herein.
  • the pPhCPAB phage display vector (SEQ ID NO: 150) has the gill signal peptide coding region has been fused with a linker to the hTN sequence encoding ALQT (etc.). The C-terminal end of the CTLD region is fused via a linker to the remaining gill coding region.
  • the murine TN CTLD phage display vectors are similarly designed. Another embodiment of these vectors is pANA27 (SEQ ID NO: 164) in which the gene III C-terminal region has been truncated and the suppressible stop codon at the end of the hTN coding sequence has been altered to encode glutamine.
  • the murine vector pANA28 (SEQ ID NO: 165) was constructed in a similar fashion.
  • Loop 1 The nucleotide and amino acid sequences of human tetranectin, and the positions of loops 1, 2, 3, 4, and 5 (LSB) are shown in FIG. 9.
  • the coding sequences for Loop 1 were modified to encode the sequences shown in Table 2, where the five amino acids AAEGT (SEQ ID NO: ) were substituted with seven random amino acids encoded by the nucleotides NNK NNK NNK NNK NNK NNK NNK (SEQ ID NO: ); N denotes A, C, G, or T; K denotes G or T.
  • the amino acid arginine immediately following Loop 2 was also fully randomized by using the nucleotides NNK in the coding strand. This amino acid was randomized because the arginine contacts amino acids in Loop 1 , and might constrain the configurations attainable by Loop 1 randomization.
  • the coding sequence for Loop 4 was altered to encode an alanine (A) instead of the Lysine 148 (K) in order to abrogate plasminogen binding, which has been shown to be dependent on the Loop 4 lysine (Graversen et al, 1998).
  • the human Loop 1 extended library was generated using overlap PCR in the following manner (primer sequences are shown in Table 3).
  • Primers lXfor (SEQ ID NO: ) and IXrev (SEQ ID NO: ) were mixed and extended by PCR, and primers BstXlfor (SEQ ID NO: ) and PstBssRevC (SEQ ID NO: ) were mixed and extended by PCR.
  • the resulting fragments were purified from gels, and mixed and extended by PCR in the presence of the outer primers Bglforl2 (SEQ ID NO: ) and PstRev (SEQ ID NO: ).
  • the resulting fragment was gel purified and cut with Bgl II and Pst I and cloned into a phage display vector pPhCPAB or pANA27.
  • the phage display vector pPhCPAB was derived from pCANTAB (Pharmacia), and contained a portion of the human tetranectin CTLD fused to the Ml 3 gene III protein.
  • the CTLD region was modified to include BgUl and Pstl restriction enzyme sites flanking Loops 1-4, and the 1-4 region was altered to include stop codons, such that no functional gene III protein could be produced from the vector without ligation of an in- frame insert.
  • pANA27 was derived from pPhCPAB by replacing the BamHl to Clal regions with the BamHl to Clal sequence of SEQ ID NO: 164 (pANA27). This replaces the amber suppressible stop codon with a glutamine codon and truncates the amino terminal region of gene III.
  • Ligated material was transformed into electrocompetent XL 1 -Blue E. coli (Stratagene) and four to eight liters of cells were grown overnight and DNA isolated to generate a master library DNA stock for panning. A library size of 1.5 x 10 8 was obtained, and clones examined showed diversified sequence in the targeted regions.
  • Loop 1-2 libraries of human tetranectin C-type lectin binding domains (“Human 1-2")
  • the coding sequences for Loop 1 were modified to encode the sequences shown in Table 2, where the five amino acids AAEGT (SEQ ID NO: ; human) were replaced with five random amino acids encoded by the nucleotides NNK NNK NNK NNK NNK ((SEQ ID NO: ); N denotes A, C, G, or T; K denotes G or T).
  • Loop 2 including the neighboring arginine
  • the four amino acids TGAR in human were replaced with four random amino acids encoded by the nucleotides NNK NNK NNK NNK (SEQ ID NO: ).
  • Loop 4 was altered to encode an alanine (A) instead of the lysine (K) in the loop, in order to abrogate plasminogen binding, which has been shown to be dependent on the Loop 4 lysine (Graversen et al, 1998).
  • the human 1-2 library was generated using overlap PCR in the following manner (primer sequences are shown in Table 3). Primers 1-2 for (SEQ ID NO: ) and 1-2 rev (SEQ ID NO: ) were mixed and extended by PCR. The resulting fragment was purified from gels, mixed and extended by PCR in the presence of the outer primers Bglforl2 (SEQ ID NO: ) and PstRevl2 (SEQ ID NO: ). The resulting fragment was gel purified and cut with Bgl II and Pst I and cloned into similarly digested phage display vector pPhCPAB or pANA27, as described above. A library size of 4.86 x 10 8 was obtained, and clones examined showed diversified sequence in the targeted regions.
  • Loop 1-4 library of human C-type lectin binding domains (“Human 1-4")
  • the coding sequences for Loop 1 were modified to encode the sequences shown in Table 2, where the seven amino acids DMAAEGT (SEQ ID NO: ) for human were substituted with seven random amino acids encoded by the nucleotides NNS NNS NNS NNS NNS NNS NNS NNS (SEQ ID NO: ) (N denotes A, C, G, or T; S denotes G or C).
  • Loop 4 was modified and extended to encode the sequences shown in Table 1 , where two amino acids of Loop 4, KT for human, were replaced with five random amino acids encoded by the nucleotides NNS NNS NNS NNS (SEQ ID NO: ) for human.
  • the human 1-4 library was generated using overlap PCR in the following manner (primer sequences are shown in Table 3).
  • Primers BglBssfor (SEQ ID NO: ) and BssBglrev (SEQ ID NO: ) were mixed and extended by PCR, and primers BssPstfor (SEQ ID NO: ) and PstBssRev (SEQ ID NO: ) were mixed and extended by PCR.
  • the resulting fragments were purified from gels, mixed and extended by PCR in the presence of the outer primers Bglfor (SEQ ID NO: ) and PstRev (SEQ ID NO: ).
  • the resulting fragment was gel purified and cut with Bgl II and Pst I restriction enzymes, and cloned into similarly digested phage display vector pPhCPAB or pANA27, as described above.
  • a library size of 2 x 10 9 was obtained, andl2 clones examined prior to panning showed diversified sequence in the targeted regions.
  • Loop 3-4 extended libraries of human C-type lectin binding domains (“Human 3-4X")
  • the coding sequences for Loop 3 were modified to encode the sequences shown in Table 2, where the three amino acids EIT of human tetranectin were replaced with six random amino acids encoded by the nucleotides NNK NNK NNK NNK NNK NNK (SEQ ID NO: ) in the coding strand (N denotes A, C, G, or T; K denotes G or T).
  • the three amino acids KTE in human were replaced with six random amino acids encoded by the nucleotides NNK NNK NNK NNK NNK NNK (SEQ ID NO: ).
  • the human 3-4 extended library was generated using overlap PCR in the following manner (primer sequences are shown in Table 3).
  • Primers H Loop 1-2-F (SEQ ID NO: ) and H Loop 3-4 Ext-R (SEQ ID NO: ) were mixed and extended by PCR, and primers H Loop 3-4 Ext-F (SEQ ID NO: and H Loop 5-R (SEQ ID NO: ) were mixed and extended by PCR.
  • the resulting fragments were purified from gels, and mixed and extended by PCR in the presence of additional H Loop 1-2-F (SEQ ID NO: ) and H Loop 5-R (SEQ ID NO: ).
  • the resulting fragment was gel purified and cut with Bgl II and Pst I restriction enzymes, and cloned into similarly digested phage display vector pPhCPAB or pANA27, as described above.
  • a library size of 7.9 x 10 8 was obtained, and clones examined showed diversified sequence in the targeted regions.
  • the human 3-4 combo library was generated using overlap PCR in the following manner (primer sequences are shown in Table 3).
  • Primers H Loop 1-2-F (SEQ ID NO: ) and H Loop 3-4 Combo-R (SEQ ID NO: ) were mixed and extended by PCR and the resulting fragment was purified from gels and mixed and extended by PCR in the presence of additional H Loop 1-2-F (SEQ ID NO: ) and H loop 5-R (SEQ ID NO: ).
  • the resulting fragment was gel purified and cut with Bgl II and Pst I restriction enzymes, and cloned into similarly digested phage display vector pPhCPAB or pANA27, as described above.
  • a library size of 4.95 x 10 9 was obtained, and clones examined showed diversified sequence in the targeted regions.
  • Loop 4 extended libraries of human tetranectin C-type lectin binding domains ("Human 4")
  • the coding sequences for Loop 4 were modified to encode the sequences shown in Table 2, where the three amino acids KTE of human tetranectin were replaced with seven random amino acids encoded by the nucleotides NNK NNK NNK NNK NNK NNK NNK ((SEQ ID NO: ); N denotes A, C, G, or T; K denotes G or T).
  • the human 4 extended library was generated using overlap PCR in the following manner (primer sequences are shown in Table 3). Primers H Loop 1-2-F (SEQ ID NO: ) and H Loop 3-R (SEQ ID NO: ) were mixed and extended by PCR, and primers H Loop 4 Ext-F (SEQ ID NO: ) and H Loop 5-R (SEQ ID NO: ) were mixed and extended by PCR. The resulting fragments were purified from gels, and mixed and extended by PCR in the presence of additional H Loop 1-2-F (SEQ ID NO: ) and H Loop 5-R (SEQ ID NO: ).
  • the resulting fragment gel purified and was cut with Bgl II and Pst I restriction enzymes, and cloned into similarly digested phage display vector pPhCPAB or pANA27, as described above.
  • a library size of 2.7 x 10 9 was obtained, and clones examined showed diversified sequence in the targeted regions.
  • Loop 3 altered libraries of human tetranectin C-type lectin binding domains
  • the coding sequences for Loop 3 were modified to encode the sequences shown in Table 2, where the six amino acids ETEITA (SEQ ID NO: ) of human were replaced with six, seven, or eight random amino acids encoded by the nucleotides NNK NNK NNK NNK NNK NNK NNK (SEQ ID NO: ), NNK NNK NNK NNK NNK NNK NNK (SEQ ID NO: ), and NNK NNK NNK NNK NNK NNK NNK NNK NNK (SEQ ID NO: ); N denotes A, C, G, or T; and K denotes G or T.
  • ETEITA SEQ ID NO:
  • Loop 4 the three amino acids KTE in human were replaced with six random amino acids encoded by the nucleotides NNK NNK NNK NNK NNK NNK NNK (SEQ ID NO: ).
  • the coding sequence for loop 4 was altered to encode an alanine (A) instead of the lysine (K) in the loop, in order to abrogate plasminogen binding, which has been shown to be dependent on the loop 4 lysine (Graversen et al, 1998).
  • the human Loop 3 altered library was generated using overlap PCR in the following manner.
  • Primers HLoop3F6, HLoop3F7, and HLoop3F8 (SEQ ID NOS: , respectively) were individually mixed with HLoop4R (SEQ ID NO: ) and extended by PCR.
  • the resulting fragments were purified from gels, and mixed and extended by PCR in the presence of oligos H Loop 1-2F (SEQ ID NO: ), HuBglfor (SEQ ID NO: ) and PstRev (SEQ ID NO: ).
  • the resulting fragments were gel purified, digested with Bgll and Pstl restriction enzymes, and cloned into similarly digested phage display vector pPhCPAB or pANA27, as above. After library generation, the three libraries were pooled for panning.
  • the human loop 3 loop library is generated using overlap PCR in the following manner.
  • Primers Loop3AF2 (SEQ ID NO: ) and Loop3AR2 (SEQ ID NO: ) are mixed and extended by PCR, and primers Loop3BF (SEQ ID NO: ) and Loop3BR (SEQ ID NO: ) are mixed and extended by PCR.
  • the resulting fragments are purified from gels, mixed, and subjected to PCR in the presence of primers Bgl for (SEQ ID NO: ) and Loop30R (SEQ ID NO: ).
  • loop 3 and 5 altered libraries of human C-type lectin binding domains
  • the coding sequences for loops 3 and 5 were modified to encode the sequences shown in Table 2, where the five amino acids TEITA of human were replaced with five amino acids encoded by the nucleotides NNK NNK NNK NNK NNK (SEQ ID NO: ), and the three amino acids AAN of human were replaced with three amino acids encoded by the nucleotides NNK NNK NNK.
  • the coding sequence for loop 4 was altered to encode an alanine (A) instead of the lysine (K) in the loop, in order to abrogate plasminogen binding, which has been shown to be dependent on the loop 4 lysine (Graversen et al, 1998).
  • the human loop 3 and 5 altered library was generated using overlap PCR in the following manner.
  • Primers h3-5AF (SEQ ID NO: ) and h3-5AR (SEQ ID NO: ) were mixed and extended by PCR, and primers h3-5BF (SEQ ID NO: ) and h3-5 BR (SEQ ID NO: ) were mixed and extended by PCR.
  • the resulting fragments were purified from gels, and mixed and extended by PCR in the presence of h3-5 OF (SEQ ID NO: ) and PstRev (SEQ ID NO: ).
  • the resulting fragment was gel purified, digested with Bgl I and Pst I restriction enzymes, and cloned into similarly digested phage display vector pPhCPAB or pANA27 as above.
  • Phage generated from human library 1-4 were panned on recombinant human IL- 23R/Fc chimera (R&D Systems). Screening of these binding panels after three, four, and/or five rounds of panning using an ELISA plate assay identified receptor-specific binders in all cases.
  • the master library DNA was transformed by electroporation into bacterial strain TGI (Stratagene). Cells were allowed to recover for one hour with shaking at 37°C in SOC (Super-Optimal broth with Catabolite repression) medium prior to increasing the volume 10-fold by adding super broth (SB) to a final concentration of 20% glucose and 20 ⁇ g/mL carbenicillin. After shaking at 37°C for one hour, the
  • carbenicillin concentration was increased to 50 ⁇ g/mL for another hour, after which 400 mL of SB with 2% glucose and 50 ⁇ g/mL carbenicillin were added, along with helper phage M13K07 to a final concentration of 5xl0 9 pfu/mL. Incubation was continued at 37°C without shaking for 30 minutes, and then with shaking at 100-150 rpm for another 30 min. Cells were centrifuged at 3200g at 4°C for 20 minutes, then resuspended in 500 mL SB medium containing 50 ⁇ g/mL carbenicillin and 50 ⁇ g/mL kanamycin. Cells were grown overnight at room temperature (RT) with shaking at 150 rpm.
  • RT room temperature
  • Phage were isolated by pelleting the bacterial cells by centrifugation at 15,000 g and 4°C for 20 min. The supernatant was incubated with one-fourth volume (usually 250 mL of supernatant/bottle + 62.5 mL PEG solution) of 20% PEG/2.5 M NaCl on ice for 30 min. The phage is pelleted by centrifugation at 15,000 g and 4°C for 20 min. The phage pellet was resuspended in 1% bovine serum albumin (BSA) in phosphate buffered saline (PBS) containing 0.1% sodium azide
  • BSA bovine serum albumin
  • PBS phosphate buffered saline
  • phage was resuspended in Buffer D, containing 0.05% boiled cassein, 0.025% Tween-20, and protease inhibitors. Material was filter-sterilized using Whatman Puradisc 25 mm diameter, 0.2 ⁇ pore size filters.
  • Phage generated from human library 1-4 were panned on recombinant human IL- 23R/Fc chimera (R&D Systems cat #1686-MR). Library panning was performed either using a plate or a bead format. For the plate format, six to eight wells of a 96-well Immulon HB2 ELISA plate were coated with 250 -1000 ng/well of carrier- free human IL-23R/Fc in Dulbecco's PBS.
  • bound phage were washed once with either IX PBS/0.05%> Tween or with Buffer D, and were eluted using glycine buffer, pH 2.2, containing 1 mg/mL BSA. After neutralization with 2 M Tris base (pH 11.5) the eluted phage were incubated for 15 minutes at room temperature with two to four milliliters of TGI (Stratagene), XLl-Blue (Stratagene), ER2738 (Lucigen or NEB), or SS320 (Lucigen) cells at an optical density of approximately 0.9 measured at 600 nm (OD 60 o) in yeast extract-tryptone (YT) medium .
  • Phage were prepared from this infection using the protocol above, but scaled down by about 20%> (volume). Phage prepared from eluted phage were subjected to additional rounds of panning. At each round, titers of input and output phage were determined by plating on agar with appropriate antibiotics, and colonies from these plates were used later for screening for binders by ELISA.
  • human IL-23R was biotinylated and purified using a Sulfo-NHS micro biotinylation kit (Thermo-Scientific) according to the manufacturer's instructions. Phage were generated for panning from the master library as per the protocol above, except that the phage pellet was resuspended in a casein buffer containing 0.5% boiled casein, 0.025% Tween 20 in PBS with added EDTA-free protease inhibitors (Roche).
  • streptavidin magnetic beads (2 tubes with 50 or 0.5 mg each of Myone Tl Dynabeads (Invitrogen)) were washed several times in 0.5%> boiled casein, 1% Tween 20 to remove preservatives.
  • a 150 aliquot of the phage prep was preincubated with one tube of beads for 30 min at 37°C to remove streptavidin binders.
  • the phage prep was then removed from the beads and 1 ⁇ g of biotinylated IL-23R was added along with 10 ⁇ ⁇ of human Fc at 100 ⁇ g/mL and incubated for 2 h at 37°C with rotation.
  • ELISA plates were prepared by incubating overnight at 4°C with 50-100 ⁇ ⁇ of PBS containing 75-100 ng/well of recombinant human IL-23R/Fc. A duplicate plate coated with human IgG Fc (R&D Systems) was used as a control. Plates were washed 3 times with PBS, blocked for 1 h at 37°C with 3% milk in IX PBS, and incubated for 1 hour with 100 uL/well of each milk-treated phage mixture.
  • Binders to human IL-23R were identified from the third and fourth rounds of panning. Examples of the sequences from the randomized regions of Loops 1 and 4 from phage-displayed CTLD binders to human IL-23R/Fc chimera are given in Table 4.
  • Loop 4 the second and fifth amino acids were always glycine, the fourth amino acid was always one of the cyclic amino acids tryptophan or phenylalanine, the first amino acid was hydrophobic, and usually a cyclic amino acid, such as phenylalanine, tyrosine, or tryptophan, and the third amino acid was hydrophobic, and was usually valine.
  • the Loop 1 region had less of a consensus, though glycine and serine appeared
  • ELISA assays indicated that these binders did not cross-react with either human IgGl Fc or with recombinant mouse IL-23R.
  • ELISA and Biacore binding assays indicated that purified monomeric CTLD or full-length trimers from candidate clones 001-69.4G8 and other competed with IL-23 for binding to the human IL-23R.
  • Competitive candidates have been identified that have nanomolar affinities.
  • Loop 4 region of the human IL-23R appeared to be a relevant motif, a shuffling approach was developed preserving the diversity of Loop 4 regions already obtained by panning, but resorting them with all possible Loop 1 regions from the original naive library.
  • DNA from the round 4 panning of human IL-23R was digested with EcoRI and BssHll restriction enzymes, which cut between the Loop 1 and Loop 4 regions, and a fragment of about 1.4 kb, containing the Loop 4 region, was isolated.
  • the original human 1-4 library DNA was digested with the same enzymes, and a fragment of about 3.5 kb, containing the Loop 1 region, was isolated. These fragments were ligated together and a new hi -4 shuffle library was generated as described above.
  • the library was panned using the bead protocol (supra), except that at each round of panning the amount of biotinylated recombinant human IL-23R/Fc was decreased about 10-fold, from 200 ng, (to 20 ng, to 2 ng,) to 0.1 ng. Phage supernatants from colonies were screened by ELISA as described above and binders were identified and sequenced. Loop 1 and 4 sequences of the affinity-matured binders appear in Table 5, SEQ ID NOS: ).
  • Loop 1 and 4 sequences from affinity-matured human Loop 1-4 binders to human IL-23R
  • a separate affinity maturation library was generated in which the diversity of the Loop 1 regions obtained in the initial panning round 4 was maintained, a limited selection of Loop 4 options was utilized, and Loop 3 was randomized in six positions. This was achieved by generating primers to amplify the Loop 1 region using DNA from the original panning round 4 of the human Loop 1-4 library as template, along with primers Bglfor (SEQ ID NO: ) and H1-3-4R (SEQ ID NO: ). This primer encodes the following amino acid sequence for loops 3 and 4:
  • Affinity matured IL-23R binding sequences obtained from these libraries are provided in Table 6. Some of the binders obtained were altered by swapping more favorable loop 4 or loop 1 sequences for others to obtain additional affinity-matured binders, and these are included in Table 6.
  • Table 7 shows some additional clones that were made with a primer similar to Hl- 3-4R (SEQ ID NO: ), but having coding sequences resulting in the selection of the following loop modications.
  • Another affinity maturation library was generated by limiting loop 4 to five amino acid sequences: FGVFG (SEQ ID NO: ), WGVFG, FGYFG, WGYFG, and WGVWG (SEQ ID NOS: , respectively), while maintaining the GlySer found at the beginning of loop 1 in IL- 23R binders, and varying the subsequent five amino acids in loop 1 using an NNK strategy.
  • Primers GSXX (SEQ ID NO: ) and 090827 BssBglrev (SEQ ID NO: ) were mixed and extended using PCR, and primers FGVFGfor, FGYFGfor, WGVFGfor, WGYFGfor, and
  • WGVWGfor SEQ ID NOS: to ) were mixed individually with primer Pst Loop 4 rev
  • H4E amino acids diverges from the TN sequence numbering in the last four candidates listed, because of the introduction in loop 4 of three additional amino acids.
  • H4E corresponds to El 50 in the human TN sequence [7, SEQ ID NO: 131], for example.
  • pANAl SEQ ID NO: 151
  • pANA3 SEQ ID NO: 153
  • pANA3 is the C-terminal HA-His-tagged version of pANAl .
  • pANA12 (SEQ ID NO: 162) is the C-terminal HA-StrepII-tagged version of pANAl .
  • the loop regions can be sub-cloned into ATRIMERTM polypeptide copmlexexpression vectors pANA4 or p ANA 10 to produce secreted ATRIMERTM polypeptide complexes in E. coli.
  • pANA4 (SEQ ID NO: 154) is a pBAD based expression vector containing C-terminal His/Myc-tagged full length human TN with an ompA signal peptide to direct the proteins to periplasm or growth medium
  • p ANA 10 (SEQ ID NO: 160) is the C-terminal HA-StrepII-tagged version of pANA4.
  • the cultures were incubated at 37°C on a shaker at 200 rpm to an OD 6 oo of 0.5, then cooled to room temperature.
  • IPTG was added to a final concentration of 0.05 mM for pANAl and pANA12, while arabinosis was added to a final concentration of 0.002- 0.02% for pANA4 and pANAlO.
  • the induction was performed overnight at room temperature
  • periplasmic proteins were extracted by osmotic shock or gentle sonication.
  • the 6xHis-tagged proteins were purified using Ni -NTA affinity chromatography. Briefly, periplasmic proteins were reconstituted in a His-binding buffer (100 mM HEPES, pH 8.0, 500 mM NaCl, 10 mM imidazole) and loaded onto a Ni -NTA column pre-equilibrated with His-binding buffer. The column was washed with lOx volume of binding buffer. The bound proteins were eluted with an elution buffer (100 mM HEPES, pH 8.0, 500 mM NaCl, 500 mM imidazole). The purified proteins were dialyzed into IX PBS buffer and bacterial endotoxin was removed by anion exchange.
  • a His-binding buffer 100 mM HEPES, pH 8.0, 500 mM NaCl, 10 mM imidazole
  • the strep II-tagged monomeric CTLDs and ATRIMERTM polypeptide complexes were purified by Strep-Tactin affinity chromatography. Briefly, periplasmic proteins were reconstituted in IX PBS buffer and loaded onto a Strep-Tactin column pre-equivalent with IX PBS buffer. The column was washed with 10X volume of PBS buffer. The proteins were eluted with elution buffer (IX PBS with 2.5 mM desthiobiotin). The purified proteins were dialyzed into IX PBS buffer and bacterial endotoxin was removed by anion exchange. [0267] For some cell assays, ATRIMERTM polypeptide complexes were produced by mammalian cells.
  • DNA fragments encoding loop regions were sub-cloned into the mammalian expression vector pANA2 or pANAl 1 to produce ATRIMERTM polypeptide complexes in the HEK293 transient expression system.
  • pANA2 (SEQ ID NO: 152) is a modified pCEP4 vector containing a C-terminal His tag.
  • pANAl 1 (SEQ ID NO: 161) is the C-terminal HA-StrepII-tagged version of pANA2.
  • the DNA fragments encoding loop region were obtained by double digestion with Bglll and Mfel and ligated into the expression vectors pANA2 and pANAl 1 pre-digested with Bglll and Mfel.
  • the expression plasmids were purified from bacteria using a Qiagen HiSpeed Plasmid Maxi Kit (Qiagene).
  • Qiagene Qiagen HiSpeed Plasmid Maxi Kit
  • transient transfection was performed using Qiagen SuperFect Reagent according to the manufacturer's protocol. The day after transfection, the medium was removed and changed to 293 Isopro serum-free medium (Irvine Scientific). Two days later, glucose in 0.5 M HEPES buffer was added into the media to a final concentration of 1%. The tissue culture supernatant was collected 4-7 days after transfection for purification.
  • the transient transfection was performed by Invitrogen's 293Fectin according to the manufacturer's protocol. The next day, IX volume of fresh medium was added into the culture. The tissue culture supernatant was collected 4-7 days after transfection for purification.
  • ATRIMERTM polypeptide complex purification from mammalian tissue culture supernatant was performed as described for E. coli produced ATRIMERTM polypeptide complexes.
  • ELISA assays performed as described in Example 9, demonstrated that none of the phage-displayed binders cross-reacted with either human IgGl Fc or with recombinant mouse IL-23R/FC (R&D Systems).
  • IL-23R positive human IL- 23R
  • Assays were performed generally as follows. Individual wells in Immulon HB2 plates were incubated overnight at 4°C with 100 PBS containing 100 ng of an anti-human IgG Fc (R&D MAB 110 clone 97924). Plates were washed five times with PBS/0.05% Tween 20, and wells were incubated for 1.5 h at RT with 100 each of PBS containing 50 ng of recombinant human IL-23R/Fc.
  • IL-23 -containing solutions were prepared as follows. Human IL-23 (eBioscience) was added at a concentration of 100 ng/mL. Competitor was included at a final concentration of 1 ⁇ g/mL.
  • FIG. 10 An example of the results of the competition assay (inhibiting IL-23/IL-23R interaction) using the ATRIMERTM polypeptide complexes from the initial panning is presented in Figure 10.
  • ATRIMERTM polypeptide complexes having the CTLD from clones 59-3B5, 61-p4G3, 78-2E6 and 056-53.H4E from the affinity-matured panning procedure were used in a competition assay with IL-23 for binding to IL-23R.
  • ATRIMERTM polypeptide complexes were tested in competition ELISA more extensively to determine IC50 values. As shown in Table 11, ATRIMERTM polypeptide complexes displayed low to subnanomolar IC50s.
  • ATRIMERTM polypeptide complex 056-53 The ATRIMERTM polypeptide complex 056-53. H4E was chosen as a standard for comparison, and additional competition assays were performed with affinity-matured ATRIMERTM polypeptide complexes. Table 12 provides the ratio of the IC50 of tested ATRIMERTM polypeptide complexes to that of 056-53. H4E performed in the same assay, in order to better compare competition results among assays.
  • CM5 chip GE Healthcare
  • CM5 chip GE Healthcare
  • a low-density receptor surface less than 200 RU, was used for all of the analyses.
  • ATRIMERTM polypeptide complex dilutions (1-500 nM) were injected over the IL-23R surface at 30 ⁇ /min and kinetic constants were derived from the sensorgram data using the Biaevaluation software (version 3.1, GE Healthcare). Data collection was 3 minutes for the association and 5 minutes for dissociation.
  • the anti-human IgG surface was regenerated with a 30s pulse of 3M magnesium chloride. All sensorgrams were double- referenced against an activated and blocked flow-cell as well as buffer injections.
  • ATRIMERTM complexes binding to IL-23R do not recognize IL-12Rpi or IL- 12RP2
  • a Biacore 3000 biosensor (GE Healthcare) was used to evaluate the interaction of human IL-12R l/Fc or IL-12R 2/Fc with IL-23R binding ATRIMERTM complexes.
  • IL-23R binding ATRIMERTM polypeptide complexes were amine-coupled to CM5 chips (GE Healthcare) then IL-23R (IL-23R) was injected over the chip surface.
  • A5F competes with binding of IL-23 to the IL-23R
  • Example 16 Testing activity of selected ATRIMERTM polypeptide complex in cell based assay
  • PBMC peripheral blood mononuclear cells
  • Table 18 shows the results for affinity-matured ATRIMERTM polypeptide complexes tested in the PBMC assay.
  • the ability of the ATRIMERTM polypeptide complexes to block IL-23-induced IL-17, IL-17F, and IL-22 production was measured for ATRIMERTM polypeptide complexes as indicated.
  • the results are shown as a ratio with the numerator being the IC50 for the ATRIMERTM polypeptide complexes compared to the IC50 for ustekinumab. Results of more than one assay are shown for some ATRIMERTM polypeptide complexes.
  • ATRIMERTM complexes to the natural killer cell line NKL expressing the heterodimeric IL- 23 receptor was determined.
  • ATRIMERTM complexes at a concentration of 150 ⁇ g/mL or IL- 23 at 50 ng/mL as positive control were incubated at 37°C with 140,000 NKL cells/well in a 96-well plate. After 10 min, cells were centrifuged at 1200 rpm for 5 min, and washed with PBS twice. Then, cells were lysed and treated according to the protocol provided in the Stat3 phosphorylation kit that was obtained from Cell Signaling Technology (PATH SCAN ® Phospho Stat3 Sandwich ELISA kit, Cat #7300, Cell Signaling Technology, Inc., Danvers, MA).
  • Stat-3 phopshorylation was measured by adsorbance at 450 nM using a Molecular Devices ELISA plate reader.
  • Figure 14 exemplary for complexes of 056-53. H4E and H4EP1E9 , no activation of IL-23R receptor by the ATRIMERTM complexes was observed, while IL-23 resulted in STAT-3 phosphorylation as expected. Similar results were obtained for all other atrimers tested such as 101-51-1 A4, 101-51-1 A7, 105-08- 1A8, 101-54- 4B6, H4E E137A, 101-113-6C 108 and 101-54-4B10 as summarized in FIGs. 15A and 15B.
  • any numerical values recited herein include all values from the lower value to the upper value in increments of one unit provided that there is a separation of at least two units between any lower value and any higher value.
  • concentration of a component or value of a process variable such as, for example, size, angle size, pressure, time and the like, is, for example, from 1 to 90, specifically from 20 to 80, more specifically from 30 to 70, it is intended that values such as 15 to 85, 22 to 68, 43 to 51, 30 to 32, etc. are expressly enumerated in this specification.
  • one unit is considered to be 0.0001, 0.001, 0.01 or 0.1 as appropriate.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Zoology (AREA)
  • Genetics & Genomics (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Toxicology (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

La présente invention a pour objet des polypeptides qui se lient à l'IL-23R comprenant des polypeptides ayant un domaine de multimérisation, par exemple de trimérisation, et une séquence polypeptidique qui se lie à l'IL-23R. Le domaine de multimérisation peut être dérivé de la tétranectine humaine. Les peptides se liant à l'IL-23R inhibent l'activation de l'IL-23R par l'IL-23 native et peuvent être utilisés en tant qu'agents thérapeutiques pour une série troubles immunitaires et de cancers. La présente invention concerne aussi des procédés permettant de sélectionner des polypeptides et de préparer des complexes multimères.
EP10709310A 2009-10-09 2010-02-10 Polypeptides qui se lient à l'il-23r Withdrawn EP2486050A1 (fr)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US2009006027 2009-10-09
US12/577,067 US20100105620A1 (en) 2008-10-10 2009-10-09 Polypeptides that bind Trail-R1 and Trail-R2
PCT/US2010/023804 WO2011043835A1 (fr) 2009-10-09 2010-02-10 Polypeptides qui se lient à l'il-23r
US12/703,752 US20110086770A1 (en) 2009-10-09 2010-02-10 Combinatorial Libraries Based on C-type Lectin-like Domain

Publications (1)

Publication Number Publication Date
EP2486050A1 true EP2486050A1 (fr) 2012-08-15

Family

ID=42153789

Family Applications (1)

Application Number Title Priority Date Filing Date
EP10709310A Withdrawn EP2486050A1 (fr) 2009-10-09 2010-02-10 Polypeptides qui se lient à l'il-23r

Country Status (1)

Country Link
EP (1) EP2486050A1 (fr)

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO2011043835A1 *

Similar Documents

Publication Publication Date Title
WO2011043835A1 (fr) Polypeptides qui se lient à l'il-23r
US20120135938A1 (en) Polypeptides that Bind TRAIL-R1 and TRAIL-R2
US11384131B2 (en) Superagonists, partial agonists and antagonists of interleukin-2
JP6694409B2 (ja) Tnfsf一本鎖分子
US20110028403A1 (en) HSP70-Based Treatment for Autoimmune Diseases and Cancer
AU2005265150B2 (en) C-MET kinase binding proteins
JP2013507123A (ja) C型レクチンドメインをベースとするコンビナトリアルライブラリー
US20090053252A1 (en) Bimer or an oligomer of a dimer, trimer, quatromer or pentamer of recombinant fusion proteins
US20120021995A1 (en) Polypeptides that Bind TRAIL-R1 and TRAIL-R2
KR20040018316A (ko) 스캐폴드 구조의 c-타입 렉틴 유사 도메인을 가지는단백질의 조합 라이브러리
JP5462623B2 (ja) Elr−cxcケモカインの高親和性アンタゴニスト
JPH1072492A (ja) ペプチド化合物
CN114075298B (zh) 一种索烃化的var2csa重组蛋白及其制备方法和应用
US7192738B2 (en) IGF binding proteins
US20110086806A1 (en) Polypeptides that Bind IL-23R
EP2486050A1 (fr) Polypeptides qui se lient à l'il-23r
JP5832032B2 (ja) 血球成熟促進活性を有する物質のスクリーニング方法
JPH09289892A (ja) c−MPLリガンドアゴニスト抗体
WO2022078518A1 (fr) Molécule d'il-2 modifiée et utilisation associée
JP2839837B2 (ja) 顆粒球コロニー刺激因子受容体のリガンド結合領域蛋白質をコードしているdna
WO2023070038A2 (fr) Variants p40 d'il-12 humaine et leurs utilisations
AU2010303879A1 (en) Combinatorial libraries based on C-type lectin domain
Class et al. Patent application title: HSP70-Based Treatment for Autoimmune Diseases and Cancer Inventors: Isabelle Caroline Le Poole (Downers Grove, IL, US) Josephus Dirk Nieland (Arhus C, DK) Thor Las Holtet (Ronde, DK) Thor Las Holtet (Ronde, DK)
JPH03277284A (ja) 遺伝子及びその製造法

Legal Events

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

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20120509

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO SE SI SK SM TR

AX Request for extension of the european patent

Extension state: AL BA RS

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

Free format text: STATUS: THE APPLICATION HAS BEEN WITHDRAWN

18W Application withdrawn

Effective date: 20121102