US20140127209A1 - Antibody derivatives - Google Patents

Antibody derivatives Download PDF

Info

Publication number
US20140127209A1
US20140127209A1 US13/791,312 US201313791312A US2014127209A1 US 20140127209 A1 US20140127209 A1 US 20140127209A1 US 201313791312 A US201313791312 A US 201313791312A US 2014127209 A1 US2014127209 A1 US 2014127209A1
Authority
US
United States
Prior art keywords
canceled
antibody
amino acid
derivative
seq
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US13/791,312
Other languages
English (en)
Inventor
Kenneth Grabstein
William Brady
Gordon King
Natalie Winblade Nairn
Kurt David SHANEBECK
Paul Heffner SLAGLE
Kenneth Christopher THORNTON
Michael Peter VANBRUNT
Andrea Wang
Hengyu XU
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.)
MedImmune Ltd
Original Assignee
MedImmune Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by MedImmune Ltd filed Critical MedImmune Ltd
Priority to US13/791,312 priority Critical patent/US20140127209A1/en
Assigned to ALLOZYNE, INC. reassignment ALLOZYNE, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: SHANEBECK, KURT DAVID, BRADY, WILLIAM, KING, GORDON, SLAGLE, PAUL HEFFNER, WANG, ANDREA, NAIRN, NATALIE WINBLADE, THORNTON, KENNETH CHRISTOPHER, XU, HENGYU, GRABSTEIN, KENNETH, VANBRUNT, MICHAEL PETER
Publication of US20140127209A1 publication Critical patent/US20140127209A1/en
Priority to US14/886,978 priority patent/US10202447B2/en
Assigned to MEDIMMUNE LIMITED reassignment MEDIMMUNE LIMITED ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ALLOZYNE, INC.
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/24Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against cytokines, lymphokines or interferons
    • C07K16/244Interleukins [IL]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/24Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against cytokines, lymphokines or interferons
    • C07K16/244Interleukins [IL]
    • C07K16/248IL-6
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/46Hybrid immunoglobulins
    • C07K16/468Immunoglobulins having two or more different antigen binding sites, e.g. multifunctional antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • A61K2039/507Comprising a combination of two or more separate antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/31Immunoglobulins specific features characterized by aspects of specificity or valency multispecific
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/33Crossreactivity, e.g. for species or epitope, or lack of said crossreactivity
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/40Immunoglobulins specific features characterized by post-translational modification
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/567Framework region [FR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/622Single chain antibody (scFv)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value

Definitions

  • This disclosure relates to novel antibody derivatives, methods for preparing them, compositions containing them, and their use in therapy.
  • Monospecific antibodies such as the naturally-occurring IgG, have two identical antigen binding paratopes, as they are made of two identical heavy chains and two identical light chains.
  • Bispecific antibodies are engineered immunoglobulin derivatives that have two different binding paratopes, usually directed at different antigens or epitopes.
  • bispecifics have been developed for a variety of different applications with potential use in cancer therapy, inflammatory conditions, and thrombolytic therapy, to name a few.
  • these applications include the retargeting of effector molecules (prodrug-converting enzymes, radio-isotopes, complement components), effector cells (CTLs, NK cells), and the delivery of prodrugs or chemotherapeutic agents.
  • CTLs effector cells
  • chemotherapeutic agents In the context of inflammation, bispecifics have been developed to inhibit two or more cytokines.
  • intracellular bispecific antibodies Intrabodies
  • Kontermann and Müller 1999.
  • an intracellularly expressed diabody was used to inhibit functional expression of two cell surface receptors (Jendreyko et al., 2003).
  • Bispecifics must have strong and selective binding to a disease-related antigen, and are designed to be non-immunogenic by a variety of techniques, such as antibody humanization, the use of transgenic humanized mice, or de-immunization.
  • the widespread development of bispecific antibodies has been hampered by the difficulty of producing materials of sufficient quality and in sufficient quantity for in vivo preclinical and clinical studies.
  • efficient bispecific antibody production has required both a novel structural format that enables the formation of stable homogenous bispecific proteins, and an efficient expression system that leads to high-level production.
  • a variety of approaches have been used to generate bispecific antibodies in prokaryotic and/or eukaryotic systems, primarily involving genetic fusion of the antigen binding domains. Some limited efforts using chemical conjugation have also been tested.
  • Stability of the recombinant bispecific antibodies under storage conditions as well as the stability and half life of these molecules in vivo are critical parameters with strong impact for clinical application.
  • the bispecific has to be sufficiently stable to allow the molecules to induce a therapeutic benefit before being degraded.
  • tandem scFv molecules, as well as diabodies were inactivated under physiological conditions, with varying half-lives depending on the antibody construct tested.
  • One approach to improve the stability of antibody molecules is the generation of disulfide-stabilized molecules introducing cysteine bridges between the VH-VL interfaces to inhibit dissociation of the VH and VL domains.
  • a marked reduction in production yield has been reported for these disulfide-stabilized bispecific diabodies in E - coli . As such there is a need for further bispecific constructs having improved stability, half lives and yields that are suitable for therapeutic applications.
  • CD4 + T-helper (T H ) lymphocytes represent a heterogeneous population of cells that play an essential role in adaptive immunity. These cells include effector cells, which are devoted to protection against pathogens, and regulatory T cells (Tregs), which protect against effector responses to autoantigens.
  • T H derived from the observation that these cells are critical for helping B cells to produce antibodies.
  • CD4 + T cells were also found to be responsible for helping CD8 + T cells differentiate into killer effector cells of the so-called cell-mediated immunity.
  • CD4 + T cells may themselves be immune effector cells in immune reactions such as delayed-type hypersensitivity, in which these cells induce inflammatory reactions mainly characterized by the activation of macrophages.
  • T H 1 cells produce IFN ⁇ and their primary role is the protection against intracellular microbes
  • T H 2 cells produce IL-4, IL-5, and IL-13 and are historically associated with atopy and asthma.
  • T H 1 and T H 2 cell development are under the control of certain transcription factors including T box expressed in T cells (Tbet) and signal transducer and activator of transcription (STAT) 4 for T H 1 cells and GATA-binding protein (GATA)-3 and STATE for T H 2 cells.
  • Tbet T box expressed in T cells
  • STAT signal transducer and activator of transcription
  • GATA GATA-binding protein
  • T H 1 differentiation is mainly driven by IL-12 and IFN ⁇ , while IL-4 (in the absence of IL-12) drives T H 2 differentiation.
  • IL-12 signaling along with antigen presentation, is believed to shift cell differentiation toward the T helper (T H ) 1 phenotype, and is associated with robust production of the proinflammatory cytokine, interferon gamma (IFN- ⁇ ).
  • IFN- ⁇ interferon gamma
  • T H 17 cells A recently described third subset of T helper cells, T H 17 cells, is abundant at mucosal interfaces, where they contain infection with pathogenic bacteria and fungi. These cells produce IL-17A (also referred to as IL-17), IL-17F, and IL-22, cytokines involved in neutrophilia, tissue remodeling and repair, and production of antimicrobial proteins.
  • IL-17A also referred to as IL-17
  • IL-17F also referred to as IL-17F
  • IL-22 cytokines involved in neutrophilia
  • tissue remodeling and repair production of antimicrobial proteins.
  • IL-6 and TGF- ⁇ induces the orphan nuclear receptors, retinoid related orphan receptor (ROR) ⁇ t and ROR ⁇ , which are the key transcription factors in determining the differentiation of the T H 17 lineage as well as the IL-23R.
  • ROR retinoid related orphan receptor
  • STAT3 regulates IL-6-induced expression of ROR ⁇ t and ROR ⁇ and IL-17 production.
  • STAT1 activation inhibits the development of T H 17 cells.
  • IL-6 activates both STAT3 and STAT1
  • STAT3 activation is maintained while STAT1 activation is suppressed in T H 17 cells.
  • IL-23 has been implicated in the maintenance and activation of human T H 17 cells.
  • IL-22 was originally described in mice and humans as a cytokine characteristic of fully differentiated T H 17 cells. Recently, however, a distinct subset of human skin-homing memory T cells has been shown to produce IL-22, but neither IL-17 nor IFN ⁇ . Differentiation of IL-22 producing T cells, now named T H 22 cells, could be promoted by stimulation of naive T cells in the presence of IL-6 and TNF or by the presence of plasmacytoid dendritic cells, and appears to be independent of RORC. The human T H 22 cell population coexpresses the chemokine receptor CCR6 and the skin-homing receptors CCR4 and CCR10, which led to hypotheses that these cells may be important in skin homeostasis and pathology.
  • T H 1 cells were long considered to be the major effectors in multiple autoimmune diseases, while T H 2 cells have been known to be involved in atopy and asthma. More recently, T H 17 cells have been implicated as culprits in a plethora of autoimmune and other inflammatory diseases in mice and humans. Many of the disease states previously associated with T H 1 cells, e.g., experimental autoimmune encephalomyelitis (EAE, a model for multiple sclerosis), collagen-induced arthritis, and some forms of colitis, were shown to be caused by IL-23-dependent T H 17 cells or other IL-17-producing lymphoid cell types. An imbalance between T H 17 and Treg cell function may be central in some of these diseases.
  • EAE experimental autoimmune encephalomyelitis
  • T H 17 cells Although many studies have analyzed the role of T H 17 cells in animal models of intestinal inflammation and autoimmunity, there are only a few studies investigating the role of T H 17 cells in patients with Crohn's disease. An increased number of T cells are found expressing retinoid related orphan receptor-ct (ROR ⁇ t), the master transcription factor for T H 17 cells, in the lamina intestinal of patients with Crohn's disease. Two independent studies showed that T H 17 cells in human peripheral blood and in the gut from healthy individuals and patients with Crohn's disease (Acosta-Rodriguez et al., 2007; Annunziato et al., 2007). These two studies showed that these cells are characterized by the expression of ROR ⁇ t, IL23R and CCR6, whereas they lack CXCR3, a chemokine receptor that is characteristic for T H 1 cells.
  • ROR ⁇ t retinoid related orphan receptor-ct
  • T H 17/T H 1 T cell populations which also expressed both IL17A and IFN ⁇ , which they named “T H 17/T H 1” cells.
  • Acosta-Rodriguez et al. (2007) identified T H 17 cells that can be characterized by CCR6 + CCR4 + expression, while CCR6 + CXCR3 + -expressing T H 1 cells also included a subset which produced both IL17A and IFN ⁇ .
  • very recent findings implicate CD161 as a novel surface marker for human T H 17 cells and demonstrate the exclusive origin of these cells from a CD161 + CD4 + T cell progenitor.
  • the interactions between T H 1 and T H 17 cells and the role of IFN ⁇ on T H 17 cells may be more complex than previously assumed and require further analysis to delineate the specific contributions of these cell lineages to Crohn's disease and other autoimmune diseases.
  • IL-6 a protein encoded by the IL6 gene, is an interleukin that acts both as a pro-inflammatory and an anti-inflammatory cytokine. It is secreted by T cells and macrophages to stimulate immune response, e.g. during infection and after trauma, IL-6's role as an anti-inflammatory cytokine is mediated through its inhibitory effects on TNF-alpha and IL-1, and activation of IL-1ra and IL-10.
  • IL-23 is a heterodimeric cytokine consisting of two subunits, one called p40, which is shared with another cytokine, IL-12, and another called p19 (the IL-23 alpha subunit which is encoded by the IL-23A gene) (see FIG. 10A ).
  • the two subunits of IL-23 are linked by a disulfide bridge.
  • IL-23 is an important part of the inflammatory response against infection. It promotes upregulation of the matrix metalloprotease MMP9, increases angiogenesis and reduces CD8+T-cell infiltration.
  • Crohn's disease and ulcerative colitis are the two main disease entities of inflammatory bowel diseases (IBDs). Crohn's disease has an average annual incidence rate of 6.3 per 100,000 people in the US. Although their exact aetiology is still not completely understood, it has been proposed that their pathogenesis is characterized by an exaggerated immune response in genetically susceptible individuals. For many years it has been assumed that Crohn's disease is mainly mediated by T H 1 cells, while ulcerative colitis is a T H 2-like type of inflammation. This has been supported by increased levels of T H 1 cytokines such as IFN ⁇ and interleukin 12 (IL-12) in Crohn's disease and an increased expression of certain T H 2 cytokines such as IL-13 in ulcerative colitis.
  • T H 1 cytokines such as IFN ⁇ and interleukin 12 (IL-12) in Crohn's disease
  • IL-12 interleukin 12
  • Ustekinumab (CNTO 1275; StelaraTM; Centocor, Inc., Malvern, Pa.) is a human, immunoglobulin G1 kappa (IgG1 ⁇ ) monoclonal antibody that specifically binds the shared p40 subunit of IL-12 and IL-23 and inhibits the interactions of IL-12 and IL-23 with the cell surface IL-12R ⁇ 1 receptor, thus preventing IL-12- or IL-23-mediated signaling cascades.
  • IgG1 ⁇ immunoglobulin G1 kappa
  • Tocilizumab (Actemra) is a humanized recombinant IgG1k monoclonal antibody against the IL-6 receptor. Tocilizumab was approved by the FDA on Jan. 8, 2010 for the treatment of rheumatoid arthritis.
  • bispecific constructs e.g. one that is specific for IL-6 and IL-23 (and optionally IL-12 as well) represents a significant challenge.
  • bivalent bispecific constructs need not only specific antigen binding and neutralizing domains, but also to be stable, have a long mean residence time and efficacy in vivo. While many efforts have been made to create bispecific antibodies, all efforts to date have failed to create such stable molecules with long in vivo residence times. Moreover, the many efforts to create bispecifics through genetic fusion methods have not succeeded in creating readily manufactured molecules that are stable and high affinity. The bispecific constructs described herein solve these problems for the first time.
  • Bispecific antibodies targeting T H 17 cells have been developed by targeting IL23 and IL17A, (Mabry R. et al., 2009).
  • the inventors aim to provide useful bispecific antibodies.
  • the inventors' novel approach uses highly efficient production of scFv in prokaryotic systems, and a site-specific chemical conjugation method to generate large quantities of bispecific proteins, avoiding many of the problems that plague alternative methods of bispecific antibody generation.
  • a key step is the use of a flexible linker that is chemically attached to the polypeptide chains, prior to refolding, that allow each scFv to refold independently of the other to lead to a functional bispecific construct.
  • the inventors method involves use of in vivo site specific incorporation of non natural aminoacids functioning as reactive sites for covalent and site specific binding of a linker, such as PEG, to the target protein (see WO 2007/130453, the entire contents of which are herein incorporated by reference).
  • An advantage of the method is that the chemistry used to conjugate scFvs to the linker is orthogonal to the 20 natural amino acids.
  • single-chain variable fragments are readily produced in large quantities and can be easily purified.
  • the antibodies are subsequently humanized and converted to scFv.
  • the Inventors have identified a number of humanized monoclonal antibodies for specific targets, namely human IL-6, human IL-23 and human IL-12.
  • the present invention provides bivalent, bispecific constructs comprising an anti-IL-6 antibody, or derivative thereof, and an anti-IL-23 antibody, or derivative thereof, methods of making such constructs, and use of such constructs in therapy.
  • the antibodies of the bivalent, bispecific constructs of the present invention may be isolated monoclonal antibodies, preferably, they are isolated human monoclonal antibodies.
  • the antibodies of the bivalent, bispecific constructs of the present invention may be chimeric antibodies.
  • the framework regions of the antibodies, or derivatives thereof, of the present invention have been humanized.
  • the antibody derivatives of the present invention may include the entire variable region, the heavy chain of the variable region (VH), the light chain of the variable region (VL), a Fab, a Fab′, a F(ab′)2, a Fv, a scFv, a dAb or a complementarity determining region (CDR).
  • the antibody derivatives of the present invention entirely retain, or substantially retain, the antigen binding activity of the antibodies from which they are derived.
  • the antibody derivatives are scFv.
  • the present invention further provides an anti-IL-6 antibody, or derivative thereof, and an anti-IL-23 antibody, or derivative thereof, methods of making such antibodies, or derivatives thereof and use of such such antibodies, or derivatives thereof alone or in combination in therapy
  • the antibodies and antibody derivatives of the present invention may be modified to incorporate one or more non-natural amino acids.
  • the anti-IL-6 antibody, or derivative thereof may comprise particular motifs from the CDR regions of the 13A8 antibody.
  • the present invention provides an anti-IL-6 antibody, or derivative thereof, which comprises:
  • a CDR2 region comprising the amino acid sequence YIYTDX 1 STX 2 YANWAKG, wherein X 1 is selected from the group consisting of glycine, asparagine, glutamine, cysteine, serine, threonine, tyrosine; and X 2 is selected from the group consisting of phenylalanine, tryptophan, and tyrosine; and preferably X 1 is serine or threonine and X2 is tryptophan or tyrosine; (SEQ ID NO. 335) and/or a CDR5 region comprising the amino acid sequence RX 1 STLX 2 S, wherein X 1 and X 2 are independently alanine or threonine. (SEQ ID NO. 336)
  • the anti-IL-6 antibody, or derivative thereof may comprise at least one, at least two, at least three, at least four, at least five or six CDR regions whose amino acid sequence is selected from the group consisting of SEQ ID NOs. 10-15.
  • the CDR region may be selected from the CDRs of the heavy chain of the variable region (VH) (i.e. SEQ ID NOs. 10-12) and/or from the CDRs of the light chain of the variable region (VL) (i.e. SEQ ID NOs 13-15).
  • VH variable region
  • the anti-IL-6 antibody, or derivative thereof may comprise the entire VH and/or VL of an anti-IL-6 antibody, or derivative thereof.
  • the anti-IL-6 antibody, or derivative thereof may the comprise the VH of an anti-IL-6 antibody, the VH having the sequence of SEQ ID NO. 259 and/or the VL of an anti-IL-6 antibody, the VL having the sequence of SEQ ID NO. 261.
  • the bivalent, bispecific construct comprises an anti-IL-6 antibody, or derivative thereof, which is a scFv comprising a heavy chain comprising at least one, at least two or three CDR regions the amino acid sequence of SEQ ID NO. 10-12 and a light chain comprising at least one, at least two or three CDR regions the amino acid sequence of SEQ ID NOs. 13-15.
  • the scFv may comprise the amino acid sequence of SEQ ID NO. 259 and a light chain comprising the amino acid sequence of SEQ ID NO. 261.
  • the invention also provides an anti-IL-6 antibody, or derivative thereof, or a bivalent bispecific construct comprises an anti-IL-6 antibody, or derivative thereof according to the above clauses based on antibodies 28D2, 18D4, 8C8, 9H4 and 9C8 in which reference to SEQ ID NOs 10-15 is replaced by reference, respectively, to SEQ ID NOs 20-25, 30-35, 40-45, 50-55 and 60-65.
  • reference to SEQ ID NOs. 259 and 261 may be replaced by references to SEQ ID NOs. 263 and 265.
  • the present invention also encompasses bivalent, bispecific constructs having anti-IL-6 antibodies, or derivatives thereof, which comprise at least one CDR region whose amino acid sequence has at least 90%, at least 95%, at least 98%, or at least 99% identity to an amino acid sequence selected from the group consisting of SEQ ID NO.s 10-15.
  • the present invention also encompasses bivalent, bispecific constructs having anti-IL-6 antibodies, or derivatives thereof, which comprise at least one CDR region whose amino acid sequence comprises one or more amino acid additions, deletions or substitutions to an amino acid sequence selected from the group consisting of SEQ ID NO.s 10-15.
  • the CDR region comprises at least one conservative amino acid substitution to an amino acid sequence selected from the group consisting of SEQ ID NO.s 10-15.
  • the present invention also provides bivalent, bispecific construct comprising an anti-IL-6 antibody, or derivative thereof, which comprises at least one CDR region that binds to the same epitope as an anti-IL-6 antibody having CDRs corresponding to the amino acid sequences of SEQ ID NO.s 10-15.
  • the anti-IL-6 antibody, or derivative thereof is selected from, or derived from, the group consisting of 13A8, 9H4, 9C8, 8C8, 18D4 and 28D2.
  • the anti-IL-23 antibody, or derivative thereof may comprise particular motifs from the CDR regions of the 31A12 antibody.
  • the present invention provides an anti-IL-23 antibody, or derivative thereof, which comprises:
  • a CDR2 region comprising the amino acid sequence YYAX 1 WAX 2 G, wherein X′ is selected from the group consisting of serine, proline and aspartate, and X 2 is selected from the group consisting of lysine and glutamine;
  • SEQ ID 337 and/or a CDR5 region comprising the amino acid sequence AX 1 TLX 2 S, wherein X 1 is selected from the group consisting of serine and alanine X 2 is selected from the group consisting of alanine and threonine.
  • CDR1 refers to VH CDR1
  • CDR2 refers to VH CDR2
  • CDR3 refers to VH CDR3
  • CDR4 refers to VL CDR1
  • CDR5 refers to VL CDR2
  • CDR6 refers to VL CDR3.
  • the anti-IL-23 antibody, or derivative thereof may comprise at least one, at least two, at least three, at least four, at least five or six CDR regions whose amino acid sequences are selected from the group consisting of SEQ ID NOs 90-95.
  • the CDR region may be selected from the CDRs of the heavy chain of the variable region (VH) (i.e. SEQ ID NOs. 90-92 and/or from the CDRs of the light chain of the variable region (VL) (i.e. SEQ ID NO. 93-95).
  • the anti-IL-23 antibody, or derivative thereof may comprise all of the amino acid sequences of SEQ ID NO.s 90-95.
  • the anti-IL-23 antibody, or derivative thereof may comprise the entire VH and/or VL of an anti-IL-23 antibody or derivative thereof.
  • the anti-IL-23 antibody, or derivative thereof may comprise the VH of an anti-IL-23 antibody, the VH having the sequence of SEQ ID NO. 267 and/or the VL of an anti-IL-23 antibody, the VL having the sequence of SEQ ID NO. 269.
  • the bivalent, bispecific construct comprises an anti-IL-23 antibody, or derivative thereof, which is a scFv comprising a heavy chain comprising
  • the scFv may comprise the amino acid sequence of SEQ ID NO. 267 and a light chain comprising the amino acid sequence of SEQ ID NO. 269.
  • the invention also provides an anti-IL-23 antibody, or derivative thereof, or a bivalent bispecific construct comprises an anti-IL-23 antibody, or derivative thereof according to the above clauses based on antibodies 49B7, 16C6, 34E11 and 35H4 in which reference to SEQ ID NOs 90-95 is replaced by reference, respectively, to SEQ ID Nos 100-105, 110-115, 120-25 and 130-135.
  • the present invention also encompasses bivalent, bispecific constructs having anti-IL-23 antibodies, or derivatives thereof, which comprise at least one CDR region whose amino acid sequence has at least 90%, at least 95%, at least 98%, or at least 99% identity to an amino acid sequence selected from the group consisting of SEQ ID NO.s 90-95.
  • the present invention also encompasses bivalent, bispecific constructs comprising an anti-IL-23 antibody, or derivative thereof, which comprise at least one CDR region whose amino acid sequence comprises one or more amino acid additions, deletions or substitutions to an amino acid sequence selected from the group consisting of SEQ ID NOs. 90-95.
  • the CDR region comprises at least one conservative amino acid substitution to an amino acid sequence selected from the group consisting of SEQ ID NO.s 90-95.
  • the present invention also provides bivalent, bispecific construct comprising an anti-IL-23 antibody, or derivative thereof, which comprises at least one CDR region that binds to the same epitope as an anti-IL-6 antibody having CDRs corresponding to the amino acid sequences of SEQ ID NO.s 90-95.
  • the anti-IL-23 antibody is selected from, or derived from, the group consisting of 31A12, 34E11, 35H4, 49B7 and 16C6. It is likely that such antibodies bind to the p19 subunit of IL-23.
  • the anti-IL-23 antibody may also bind IL-12.
  • IL-12 binds to the p40 subunit that is shared by both IL-23 and IL-12.
  • Such antibodies are referred to herein as anti-IL-23/IL-12 antibodies.
  • antibodies bind to p40 and inhibit IL-23 yet do inhibit IL-12—such antibodies are included within the scope of “anti-IL-23 antibodies”.
  • the present invention provides anti_IL-23/IL-12 antibodies, or derivatives thereof, which may comprise particular motifs from the CDR regions of the 45G5 or 22H8 antibodies.
  • the present invention provides an anti-IL-23/IL-12 antibody, or derivative thereof, which inhibits both IL-12 and IL-23 which comprises:
  • a CDR2 region comprising the amino acid sequence sequence WX 1 KG, wherein X1 is alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine or tryptophan, and preferably is alanine or valine (SEQ ID NO. 358); and/or a CDR3 region comprising the amino acid sequence YAYX 1 GDAFDP, wherein X 1 is alanine or isoleucine; (SEQ ID NO. 339) and/or a CDR3 region comprising the amino acid sequence SDYFNX 1 , wherein X 1 is isoleucine or valine; (SEQ ID NO.
  • a CDR4 region comprising the amino acid sequence QX 1 SQX 2 , wherein X 1 is alanine or serine, and X 2 is selected from the group consisting of glycine, asparagine, glutamine, cysteine, serine, threonine, and tyrosine; preferably X 2 is serine or threonine; and/or (SEQ ID NO. 359) a CDR5 region comprising the amino acid sequence ASX 1 LA, wherein X 1 is lysine or threonine. (SEQ ID NO. 341) and/or a CDR6 region comprising the amino acid sequence QSYYDX 1 NAGYG, wherein X 1 is alanine or valine. (SEQ ID NO. 342)
  • the anti-IL-23/IL-12 antibody or derivative thereof may comprise at least one, at least two, at least three, at least four, at least five or six CDR regions whose amino acid sequences are selected from the group consisting of SEQ ID NO.s 140-145.
  • the CDR region may be selected from the CDRs of the heavy chain of the variable region (VH) (i.e. SEQ ID NOs. 140-142 and/or from the CDRs of the light chain of the variable region (VL) (i.e. SEQ ID NOs. 143-145.
  • the anti-IL-23/IL-12 antibody, or derivative thereof may comprise all of the amino acid sequences of SEQ ID NO.s 140-145.
  • the anti-IL-23/IL-12 antibody, or derivative thereof may comprise the entire VH and/or VL of an anti-IL-23/IL-12 antibody or derivative thereof.
  • the anti-IL-23/IL-12 antibody, or derivative thereof may comprise the VH of an anti-IL-23/IL-12 antibody, the VH having SEQ ID NO. 271 and/or the VL of an anti-IL-23/IL-12 antibody, the VL having SEQ ID NO. 273.
  • the bivalent, bispecific construct comprises an anti-IL-23/IL-12 antibody, or derivative thereof, which is a scFv comprising at least one, at least two or three CDR regions having the amino acid sequence of SEQ ID NOs. 140-142 and a light chain comprising at least one, at least two or three CDR regions having the amino acid sequence of SEQ ID NOs. 143-145.
  • the scFv may comprise a heavy chain comprising the amino acid sequence of SEQ ID NO. 271 and a light chain comprising the amino acid sequence of SEQ ID NO. 273.
  • the invention also provides an anti-IL-23/IL-12 antibody, or derivative thereof, or a bivalent bispecific construct comprises an anti-IL-23/IL-12 antibody, or derivative thereof according to the above clauses based on antibodies 45G5, 1H1, 4F3, 5C5 and 14B5 in which reference to SEQ ID NOs 140-145 is replaced by reference, respectively, to SEQ ID NOs 150-155, 160-165, 170-175, 180-185 and 190-195.
  • reference to SEQ ID NOs. 271 and 273 may be replaced by references to SEQ ID NOs. 275 and 277.
  • the present invention also encompasses bivalent, bispecific constructs comprising anti-IL-23/IL-12 antibodies, or derivatives thereof, which comprise at least one CDR region whose amino acid sequence has at least 90%, at least 95%, at least 98%, or at least 99% identity to an amino acid sequence selected from the group consisting of SEQ ID NO.s 140-145.
  • the present invention also encompasses bivalent, bispecific constructs comprising anti-IL-23/IL-12 antibodies, or derivatives thereof, which comprise at least one CDR region whose amino acid sequence comprises one or more amino acid additions, deletions or substitutions to an amino acid sequence selected from the group consisting of SEQ ID NO.s 140-145.
  • the CDR region comprises at least one conservative amino acid substitution to an amino acid sequence selected from the group consisting of SEQ ID NO.s 140-145.
  • the present invention also provides bivalent, bispecific construct having an anti-IL-23/IL-12 antibody, or derivative thereof, which comprises at least one CDR region that binds to the same epitope as an anti-IL-23/IL-12 antibody having CDRs corresponding to the amino acid sequences of SEQ ID NOs. 140-145.
  • the epitope is present on the p40 subunit that is common to both IL-12 and IL-23.
  • the anti-IL-23/IL-12 antibody is selected from, or derived from, the group consisting of 22H8, 45G5, 14B5, 4F3, 5C5, and 1H1.
  • the anti-IL-6 antibodies and derivatives thereof, described above that may form part of a bivalent bispecific construct may be independently combined with the anti-IL-23 antibodies, or derivatives thereof, (including the anti-IL-23/IL-12 antibodies, and derivatives thereof) described above in a single bivalent bispecific construct.
  • both the anti-IL-6 antibody, or derivative thereof, and the anti-IL-23 antibody, or derivative thereof may incorporate non-natural amino acids, through which the anti-IL-6 antibody, or derivative thereof, is coupled to the anti-IL-23 antibody, or derivative thereof.
  • the bivalent bispecific constructs of the present invention may further comprise a linker between a non-natural amino acid in each antibody, or derivative thereof.
  • the bivalent bispecific constructs of the present invention may further comprise polyethylene glycol molecules (PEG).
  • PEG polyethylene glycol molecules
  • the PEG molecule may optionally serve as a linker between the anti-IL-6 antibody, or derivative thereof, and the anti-IL-23 antibody, or derivative thereof, (including anti-IL-23/IL-12 antibodies, or derivatives thereof).
  • PEG polyethylene glycol molecules
  • the PEG molecule may optionally serve as a linker between the anti-IL-6 antibody, or derivative thereof, and the anti-IL-23 antibody, or derivative thereof, (including anti-IL-23/IL-12 antibodies, or derivatives thereof).
  • other water soluble polymers such as polyvinylalcohol, polysaccharides, polyalkylene oxides, hydroxyethyl starch, and polyols, may also be used.
  • anti-IL-6 and anti-IL-23 antibodies or derivatives thereof, (including anti-IL-23/IL-12 antibodies, or derivatives thereof) described herein are useful per se.
  • the present invention provides anti-IL-6 and anti-IL-23 antibodies or derivatives thereof, (including anti-IL-23/IL-12 antibodies, or derivatives thereof), that have particular utility in the manufacture of bivalent bispecific constructs of the invention and/or in combination therapeutics.
  • Said anti-IL-6 and anti-IL-23 or derivatives thereof, (including anti-IL-23/IL-12 antibodies, or derivatives thereof) may optionally be modified to increase half life (for instance through PEGylation).
  • Anti-IL-6 antibodies, or derivatives thereof, and anti-IL-23 antibodies, or derivatives thereof may incorporate non-natural amino acids to facilitate linkage of PEG groups.
  • An aspect of the invention provides a combination (for separate, sequential or separate administration) comprising an anti-IL-6 antibody or derivative thereof and an anti-IL-23 antibody or derivative thereof (which may, for example, by an anti-IL-23/IL-12 antibody or derivative thereof).
  • the present invention further encompasses bivalent bispecific constructs comprising anti-IL-6 and anti-IL-23 antibody derivatives (including anti-IL-23/IL-12 antibody derivatives) wherein said antibody derivatives are selected from Fab, Fab′, F(ab)′, Single Chain Antibodies (scFv), kappabodies, Minibodies and Janusins.
  • An anti-IL-6 antibody or derivative thereof, which comprises a heavy chain comprising the amino sequence of SEQ ID NO. 259 and a light chain comprising the amino sequence of SEQ ID NO. 261.
  • An anti-IL-23 antibody or derivative thereof which comprises a heavy chain comprising the amino sequence of SEQ ID NO. 267 and a light chain comprising the amino sequence of SEQ ID NO. 269.
  • polynucleotide encoding a portion of a bivalent, bispecific construct of the present invention is provided.
  • Such polynucleotides may encode an antibody, or derivative thereof, as disclosed herein
  • the present invention also provides vectors comprising such polynucleotides, host cell comprising such vectors (optionally the host cells are auxotrophic), oligonucleotide primers for cloning and expressing antibodies, or derivative thereof, as disclosed herein.
  • oligonucleotide primer of the present invention include oligonucleotide primers comprising one of the nucleotide sequences set out in any one of 200-258.
  • the method may comprise:
  • the method may comprise coupling the modified anti-IL-6 antibody, or modified derivative thereof, and the modified anti-IL-23 antibody, or modified derivative thereof, through a linkage comprising a linker portion, wherein one end of the linker portion is coupled to a non-natural amino acid of the modified anti-IL-6 antibody, or modified derivative thereof, and the other end of the linker portion is coupled to a non-natural amino acid of modified anti-IL-23 antibody, or modified derivative thereof.
  • suitable linkers are known in the art and include short peptide sequences.
  • the present invention also provides for the use of PEG as a linker.
  • the linker portion may be a PEG molecule.
  • the method may comprise the use of non-natural amino acids that contain a group selected from:
  • the non-natural amino acid may be azidohomoalanine, homopropargylglycine, homoallylglycine, p-bromophenylalanine, p-iodophenylalanine, azidophenylalanine, acetylphenylalanine or ethynylephenylalanine, amino acids containing an internal alkene such as trans-crotylalkene, serine allyl ether, allyl glycine, propargyl glycine, vinyl glycine, pyrrolysine, N-sigma-o-azidobenzyloxycarbonyl-L-Lysine (AzZLys), N-sigma-propargyloxycarbonyl-L-Lysine, N-sigma-2-azidoethoxycarbonyl-L-Lysine, N-sigma-tert-butyloxycarbonyl-L-Lysine (Bo
  • non-natural amino acid may be azidohomoalanine, homopropargylglycine, homoallylglycine, p-bromophenylalanine, p-iodophenylalanine, azidophenylalanine, acetylphenylalanine or ethynylephenylalanine, amino acids containing an internal alkene such as trans-crotylalkene, serine allyl ether, allyl glycine, propargyl glycine, vinyl glycine.
  • the method may comprise coupling the modified anti-IL-6 antibody, or modified derivative thereof, and the modified anti-IL-23 antibody, or modified derivative thereof using a [3+2] cycloaddition/[3+2] dipolar cycloaddition or azide-alkyne cycloaddition reaction commonly referred to as Click reaction (which may be catalyzed by copper(I), ruthenium, other metals, or promoted by strain and/or electron withdrawing groups), a Heck reaction, a Sonogashira reaction, a Suzuki reaction, a Stille coupling, a Hiyama/Denmark reaction, olefin metathesis, a Diels-alder reaction, carbonyl condensation with hydrazine, hydrazide, alkoxy amine or hydroxyl amine.
  • Click reaction which may be catalyzed by copper(I), ruthenium, other metals, or promoted by strain and/or electron withdrawing groups
  • a Heck reaction a Sonogashira reaction,
  • the method may also comprise:
  • the method may also comprise incorporating a non-natural amino acid (e.g. Aha) by incorporating it at a specific selected amino acid encoded position (typically a methionine encoded position), and if necessary mutating the polynucleotide sequence of the target protein to eliminate methionine (or other specific selected amino acid) codons at positions in which it is not desired to incorporate a non-natural amino acid and/or if necessary mutating the polynucleotide sequence of the target protein to provide one or more (typically one) new methionine (or other specific selected amino acid) codons at positions in which it is desired to incorporate a non-natural amino acid.
  • a non-natural amino acid e.g. Aha
  • a method of selecting parent antibodies suitable for inclusion in a bivalent, bispecific construct of the present invention comprising the steps of:
  • the bivalent, bispecific construct as described above is provided for use in therapy.
  • the such diseases include multiple sclerosis, psoriasis, psoriatic arthritis, pemphigus vulgaris, organ transplant rejection, Crohn's disease, inflammatory bowel disease (IBD), irritable bowel syndrome (IBS), lupus erythematosis, and diabetes.
  • the present invention also provides a combination therapeutic comprising an anti-IL-6 antibody and an anti-IL-23 antibody, for use in treating a T H 17, T H 22 and/or T H 17 and T H 1 mediated disease.
  • a combination therapeutic comprising an anti-IL-6 antibody and an anti-IL-23 antibody, for use in treating a T H 17, T H 22 and/or T H 17 and T H 1 mediated disease.
  • combinations are provided for use the treatment of inflammatory and autoimmune disorders.
  • FIG. 1 shows a B cell selection. Each well from one 96 well plate of B cells was assayed for both IL-6 neutralization by the B9 cell line proliferation assay and IL-6 binding by ELISA. The results from each assay are aligned for comparison.
  • FIG. 2 shows an outline of the experimental process from V-region rescue to scFv generation
  • FIGS. 3A and B show the human and primate IL-6 neutralization activity of selected anti-IL6 rabbit/human chimeric antibodies: Rabbit/human chimeric mAbs were expressed in mammalian cells. The mAbs were quantitated in the supernatants by ELISA. They were tested for neutralization of 50 pg/ml of human IL-6 or 100 pg/ml of primate IL-6, as indicated, using the B9 cell proliferation assay.
  • FIGS. 3C and D show the human and primate IL-6 neutralization activity of selected anti-IL6 rabbit/human chimeric antibodies: Rabbit/human chimeric mAbs were expressed in mammalian cells. The mAbs were quantitated in the supernatants by ELISA. They were tested for neutralization of 50 pg/ml of human IL-6 or 100 pg/ml of primate IL-6, as indicated, using the B9 cell proliferation assay.
  • FIG. 3E shows the human IL-6 neutralization activity of selected anti-IL6 rabbit/human chimeric antibodies: Rabbit/human chimeric mAbs were expressed in mammalian cells. The mAbs were quantitated in the supernatants by ELISA. They were tested for neutralization of 50 pg/ml of human IL-6 or 100 pg/ml of primate IL-6, as indicated, using the B9 cell proliferation assay.
  • FIG. 4 shows B cell selection Each well from one B cells 96-well plate was assayed for IL-23 neutralization and IL-23 binding. The results of the two assays for each well are aligned for comparison.
  • FIGS. 5A-5I show human IL-23 neutralization activity of selected anti-IL-23 rabbit/human chimeric antibodies.
  • Candidate mAbs were derived and tested for neutralization of heterodimeric recombinant IL-23 (eBio IL23)
  • FIGS. 6A and 6B show the neutralization activity of primate and human IL-23 Transfection supernatants of several mAbs were compared for neutralization of either heterodimeric human (panel B) or primate IL-23 (panel A) using the mouse splenocyte assay. Ig levels were measured in the transfection supernatants to allow comparison of specific activities.
  • FIG. 7A shows the structure of IL-12 and IL-23
  • FIG. 7B shows the IL-12 and IL-23 Receptors and their associated mechanisms.
  • FIG. 8A shows transfection supernatants of several mAbs were compared for neutralization of human IL-12 using the NK92 cell line assay.
  • FIG. 8B shows neutralization of Human IL-23 by selected mAbs mAbs that were positive in the primary rescue transfections from B cell clones, were expressed in HEK293 transient transfections, in which IgG concentration was quantitated for calculating EC50 values.
  • the mAbs were tested for neutralization of 1200 pg/ml of IL-23 (eBiosciences).
  • FIG. 78C shows neutralization of Human IL-23 by selected mAbs mAbs that were positive in the primary rescue transfections from B cell clones, were expressed in HEK293 transient transfections, in which IgG concentration was quantitated for calculating EC50 values.
  • the mAbs were tested for neutralization of 1200 pg/ml of IL-23 (eBiosciences).
  • FIG. 8D shows neutralization of Human IL-23 by selected mAbs mAbs that were positive in the primary rescue transfections from B cell clones, were expressed in HEK293 transient transfections, in which IgG concentration was quantitated for calculating EC50 values.
  • the mAbs were tested for neutralization of 1200 pg/ml of IL-23 (eBiosciences).
  • FIG. 8E shows neutralization of Human IL-23 by selected mAbs mAbs that were positive in the primary rescue transfections from B cell clones, were expressed in HEK293 transient transfections, in which IgG concentration was quantitated for calculating EC50 values.
  • the mAbs were tested for neutralization of 1200 pg/ml of IL-23 (eBiosciences).
  • FIGS. 8F-8G show neutralization of Human IL-12 by selected mAbs mAbs that were positive in the primary rescue transfections from B cell clones, were expressed in HEK293 transient transfections, in which IgG concentration was quantitated for calculating EC50 values.
  • the mAbs were tested for neutralization of 1000 pg/ml of human IL-12.
  • FIGS. 8H and 8I show neutralization of Primate IL-23 by selected mAbs mAbs that were positive in the primary rescue transfections from B cell clones, were expressed in HEK293 transient transfections, in which IgG concentration was quantitated for calculating EC50 values. The mAbs were tested for neutralization of 1000 pg/ml of primate IL-23.
  • FIG. 9A shows Rabbit scFvs were expressed in mammalian cells.
  • the scFvs were quantitated in the supernatants by SDS PAGE. They were tested for neutralization of 200 pg/ml of human IL-6 as indicated, using the B9 cell proliferation assay.
  • FIG. 9B shows 9C8 humanization.
  • 9C8 a high affinity and high potency chimeric mAb, was humanized by changing the framework regions of the VH and VL to human framework sequences, with limited back mutation to rabbit framework sequences. Different human framework sequences were compared for humanization of 9C8.
  • the humanized mAbs were expressed by transient transfection of HEK293 cells. Transfection supernatants were tested for the ability to neutralize 50 pg/ml of human IL-6 using the B9 cell proliferation assay.
  • FIG. 9C shows Humanized Anti-IL-6 mAbs.
  • 9C8 and 18D4 high affinity and high potency chimeric mAbs, were humanized by changing the framework regions of the VH and VL to human framework sequences, with limited back mutation to rabbit framework sequences.
  • the humanized mAbs were expressed by transient transfection of HEK293 cells. IgG in the transfection supernatants were quantitated and they were tested for the ability to neutralize 50 pg/ml of human IL-6 using the B9 cell proliferation assay.
  • FIG. 9D shows Humanized Anti-IL-6 mAbs and scFvs. 9C8, a high affinity and high potency chimeric mAb, was humanized by changing the framework regions of the VH and VL to human framework sequences, with limited back mutation to rabbit framework sequences. 2 humanized 9C8 scFv were generated from the rabbit scFv comparing 2 different VHCDR1 sequences. The rabbit scFv was directly expressed without humanization and the humanized mAb, and scFvs were expressed by transient transfection of HEK293 cells. Transfection supernatants were tested for the ability to neutralize 50 pg/ml of human IL-6 using the B9 cell proliferation assay.
  • FIG. 10 shows a PCR strategy for CDR grafting onto human V-region frameworks in scFv format.
  • VL light chain V-region
  • L leader (signal peptide)
  • FR framework region
  • CDR complementarity determining region
  • arrows indicate individual primers and their directionality in a PCR amplification
  • CDR-specific primers designated by chain and CDR number, H is VH, L is VL;
  • curved line between VL-FR4 and VH-FR1 represents the 20 aa (G4S) 4 linker that is added for scFv construction.
  • FIGS. 11A & 11B show IL-6 neutralization by humanized scFv 13A8.
  • Humanized 13A8 anti IL-6 scFv was expressed in mammalian cells. The scFv was purified from the supernatants by Ni affinity. Testing for neutralization of human IL-6 (B) or primate IL-6 (A) was carried out using the B9 cell proliferation assay.
  • FIG. 11C shows IL-6 neutralization by anti IL-6 Humanized scFv: Humanized 9C8 scFv v3-1 (from the multistep method) and 28D2 scFv were expressed in mammalian cells, purified by Ni chromatography, and compared for inhibition of IL-6 induced B9 cell proliferation.
  • FIG. 12 shows that Anti IL-23 31A12 scFv Neutralizes IL-23 31A12 mAb was converted into a humanized scFv and expressed in mammalian transfection along with the parental mAb. Both were tested for neutralization of 600 pg/ml of eBiosciences human II-23 using the mouse splenocyte assay for induction of IL-17.
  • FIG. 13 shows that Humanized Anti IL-23 45G5 scFv Neutralizes Human IL-23: Humanized 45G5 scFv was compared to the chimeric mAb 31A12 and 22H8. All mAbs were expressed in mammalian cells, purified and tested for the inhibition of 1.2 ng/ml of eBiosciences human IL-23 using the mouse splenocyte assay of IL-17 induction.
  • FIGS. 14A-14D show testing of Humanized 13A8 scFv mammalian expression constructs which were engineered to remove the 2 Met residues.
  • the various double mutant constructs, and the parental scFv (MM), were expressed in HEK cells and tested for inhibition of 50 pg/ml of human IL-6 using the in vitro B9 bioassay.
  • FIG. 14E shows testing of Humanized 31A12 scFv, with both Mets replaced (only H34L versions shown), and the parental Met containing scFv, which were expressed transiently in HEK293 cells. Supernatants were tested for inhibition of biological activity of 600 pg/ml of eBiosciences IL-23 in the mouse splenocyte assay.
  • FIGS. 14F and 14G show testing of Humanized 45G5 scFv with the H82 Met replaced with either L or V, both in combination with H34L, which were compared to the parental 45G5 chimeric mAb and Met free 31A12 scFv (31A12-LL). All were expressed transiently in HEK293 cells. scFv and mAbs were purified and tested for inhibition of biological activity of 1200 pg/ml of eBiosciences IL-23 in the mouse splenocyte assay.
  • FIG. 14H shows that Humanized Anti IL-23 scFvs neutralizes Human IL-23. Wild Type Anti IL-23 scFv 22H8 and 45G5, were compared to the 22H8 scFv with the Met at H34 replaced with either V or L, as indicated.
  • FIG. 15 shows neutralization of IL-6 by E. coli 28D2 scFv with Aha at the N or C term or in the Gly/Ser Linker:
  • FIG. 16A shows PEGylation of 13A8cAha with 20K linear PEG bis alkyne SDS PAGE (reducing, 4-20% Tris-Glycine) of the 13A8cAha PEGylation reaction with 20K PEG bis alkyne.
  • Lane 1 13A8cAha alone; Lane 2: ( ⁇ ) control—no Copper; Lane 3; 200 mL small scale reaction mixture; Lane 4: 600 mL reaction—centrifuged—sample supernatant.
  • FIG. 16B shows PEGylation of 31A12cAha with 20K linear PEG bis alkyne.
  • SDS PAGE reducing, 4-20% Tris-Glycine
  • Lane 1 Molecular weight markers
  • Lane 3 ( ⁇ ) control—no Copper
  • Lane 4 small scale reaction
  • Lane 5 400 mL reaction—centrifuged—sample supernatant.
  • Scanning Laser Densitometry indicated a 59% yield (lane 5) of the PEGylated 31A12cAha.
  • FIG. 16C shows PEGylation of 13A8cAha with 40K linear PEG bis alkyne.
  • SDS-PAGE reducing, 4-20% Tris-Glycine
  • Scanning Laser Densitometry indicated a 51% yield
  • FIG. 16D shows PEGylation of 13A8L Aha with 20K linear PEG Bis-Alkyne.
  • SDS-PAGE reducing, 4-20% Tris-Glycine.
  • Scanning Laser Densitometry indicated a 60% yield
  • FIG. 16E shows PEGylation of 45G5cAha with 20K linear PEG bis alkyne.
  • SDS PAGE reducing, 4-20% Tris-Glycine
  • Lane 1 ( ⁇ ) control—no Copper
  • Lane 2 small scale reaction
  • Lane 3 small scale reaction, no triazole ligand
  • Lane 4 160 mL reaction—centrifuged—sample supernatant
  • Lane 6 Molecular weight markers. Scanning Laser Densitometry indicated a 59% yield of the PEGylated 45G5c.-.
  • FIG. 17A shows IL-6 Neutralization with 28D2c-PEG. 2 samples of 28D2c-30 KPEG refolded under different conditions were assayed for IL-6 neutralization.
  • FIGS. 17B and C shows PEG-scFv Stability: 31A12-PEG has a Tm of 69° C. 13A8-PEG has a Tm of 66° C. This is reflected in the stability of these molecules in solution as shown. Each scFv-PEG was incubated in PBS (or Tween as indicated) and is assayed for potency relative to the parental scFv (from mammalian expression). Storage temperature, for 13 or 20 days, is indicated; 3 ⁇ FT indicates three cycles of freezing and thawing.
  • FIG. 18A shows the preparation of 13A8c-PEG-31A12c Bispecific SDS-PAGE (reducing, 4-20% Tris-Glycine).
  • Lane 1 Molecular weight markers
  • Lane 2 13A8-PEG alone
  • Lane 3 ( ⁇ ) control—no copper
  • Lane4 1000 mL reaction
  • FIG. 18B shows the preparation of 13A8n-PEG-45G5c Bispecific: SDS-PAGE (reducing, 4-20% Tris-Glycine). Lane 1: Large Scale Reaction 1, Lane 3: Large Scale Reaction 2
  • FIG. 18C shows the preparation of 13A8c-PEG-22H8c Bispecific SDS-PAGE reducing, 4-20% Tris-Glycine).
  • Lane 1 Molecular weight markers
  • Lane 2 13A8c-PEG alone
  • Lane 3 ( ⁇ ) control—no copper
  • Lane4 1150 mL reaction
  • FIG. 18E shows the preparation of 13A8L-PEG-31A12c Bispecific SDS-PAGE (reducing, 4-20% Tris-Glycine). Lane 1 reaction of 31A12-20 KPEG+13A8LAha to form bispecific. Reaction yield was found to be 37%.
  • FIG. 19A shows the functional Activity of 31A12c-PEG-13A8c for Neutralization of IL-6 and IL-23
  • FIG. 20A shows Rat PK of 28D2c scFv administered SC. Rats were treated SC with 1 mg/kg of anti IL-6 scFv 28D2c. Blood was collected at the indicated times, the presence of 28D2 in the plasma of the rats was measured using an anti IL-6 neutralization assay.
  • FIG. 20B shows Rat PK of 31A12c-PEG-13A8c bispecific administered subcutaneously: Rats were treated SC with 1 mg/kg of 31A12c-PEG-13A8c bispecific. Blood was collected at the indicated times, the presence of bispecific in the plasma of the rats was measured using an anti IL-6 neutralization assay. The PK data for the 28D2 scFv from FIG. 20B is included here for comparison.
  • FIG. 21 shows the bioactivity of the 13A8n-PEG-31A12c bispecific.
  • the neutralization of 50 pg/ml of IL-6 by the bispecific was measured in the B9 bioassay.
  • the mammalian 13A8 scFv protein is included for comparison.
  • FIG. 22 shows the functional activity of 13A8n-PEG-45G5 for IL-6 and IL-23.
  • the bioactivity of bispecific vs IL 6 and IL23 is shown.
  • the neutralization of 50 pg/ml of human IL-6 (22A) and 1200 pg/ml (22B) of human eBiosciences IL-23, by the 13A8n-PEG-45G5 bispecific was measured using the B9 cell line bioassay for IL-6 and the mouse splenocyte assay for IL-23. EC50 values were calculated from the curves.
  • FIG. 23 shows the activity of 13A8c-PEG-22H8c for IL-6 and IL-23. Bioactivity of 13A8c-PEG-22H8c for IL-6 and IL-23 is shown. The neutralization of 50 pg/ml of human IL-6 (A) and 1200 pg/ml (B) of human eBiosciences IL-23, by the 13A8c-PEG-22H8 bispecific was measured using the B9 cell line bioassay for IL-6 and the mouse splenocyte assay for IL-23. EC50 values were calculated from the curves.
  • FIG. 24 shows the activity of 13A8c-40 KPEG-31A12c for IL-6 and IL-23. Bioactivity of 13A8c-40kPEG31A12c for IL-6 and IL-23 is shown. The neutralization of 50 pg/ml of human IL-6 (A) and 1200 pg/ml (B) of human eBiosciences IL-23, by the bispecific was measured using the B9 cell line bioassay for IL-6 and the mouse splenocyte assay for IL-23. EC50 values were calculated from the curves.
  • FIG. 25 A Shows serum levels (as measured in the B9 assay) of the 13A8c-40 KPEG-31A12c bispecific, 13A8c-20 KPEG-31A12c bispecific, 13A8c-PEG and a naked scFv (28D2) after subcutaneous administration in rats.
  • FIG. 25 B Shows the results of pharmacokinetic analysis of serum levels of 13A8c-40 KPEG-31A12c bispecific, 13A8c-20 KPEG-31A12c bispecific, 13A8c-PEG and naked scFv (28D2) after subcutaneous administration in rats.
  • FIG. 26A shows in vitro polarization of T H 17/22 cells
  • Different human T cell subsets including T H 17 and T H 22 cells, can be generated in both in vivo and in vitro systems.
  • FIG. 26B shows in vitro Human Th17 Development: Human PBMC were stimulated with anti-CD3/28 for 7 days in vitro either alone or in the presence of LPS and TGFb, as indicated. They were then restimulated with PMA+ ionomycin as indicated and stained for IL-17 and RORC
  • FIG. 26C shows T H 17 and T H 22Cells can be generated from cultured PBMC T H 17 are seen in the mixed lymphocyte reaction, while T H 17 and T H 22 are seen with anti CD3 stimulated PBMC.
  • PBMC peripheral blood mononuclear cells
  • FIG. 27 shows inhibition of Th17 and Th22 development in vitro with selected scFvs.
  • Human PBMC were cultured for 5 days in anti CD3+ anti CD28 and LPS+IL-1+TGFb in the presence of the indicated scFv. After 5 days, the cells were restimulated with PMA+ ionomycin and the % of CD4, IL-17 and IL-22 producing cells was determined by flow cytometry.
  • FIG. 28 shows a mixed lymphocyte reaction. Inhibitory effect of anti IL-6 and IL-23 scFv used alone or used in combination, on T H 17 differentiation is shown. The indicated anti IL-6 scFv and anti IL-23 scFv were added to PBMC cultures during stimulation with allogeneic PBMC. After 5 days, the cells were washed and restimulated with PMA+ lonomycin and stained for IL-17.
  • FIG. 29 shows the beneficial inhibitory effect of bispecific anti IL-6/IL-23 antibodies on T H 17 differentiation.
  • Anti IL-6 and anti IL-23 mAbs 13A8 and 31A12 were tested alone or in combination, as well as 31A12c-20 KPEG-13A8c bispecific.
  • the mAbs or the bispecific were added to PBMC cultures during stimulation with allogeneic PBMC. Molar concentration of binding domains added is indicated. After 5 days, the cells were washed and restimulated with PMA+ lonomycin and stained for IL-17.
  • FIG. 30 shows in-vivo polarization of T H 17/22 cells
  • Different human T cell subsets including T H 17 and T H 22 cells, can be generated in both in vivo and in vitro systems.
  • FIG. 31A shows treatment of humanized scid/hu mice with a combination of antagonists against IL-6 and IL-23.
  • NSG mice that were successfully engrafted with human immune cells were transplanted with human allogeneic skin and received 100 mg of 13A8c-PEG anti IL-6 and 31A12c-PEG anti IL-23 (scFv-PEGs) every 2 days.
  • scFv-PEGs 13A8c-PEG anti IL-6 and 31A12c-PEG anti IL-23
  • FIG. 31B shows intracellular cytokine expression in CD3+/CD4+ cells from spleens of humanized scid/hu mice treated with a combination of antagonists against IL-6 and IL-23.
  • the splenocytes form treated and untreated NSG mice with skin allografts
  • CD3+/CD4+ cells were analyzed for intracellular IL-17 and IL-22 by flow cytometry.
  • Data shows marked reductions in all populations of IL-17 and IL-22 positive CD4+ T cells in animals treated with anti IL-6 and anti IL-23. The data are plotted as the mean and SEM of the treated or untreated mice according to the indicated subset of TH17 or TH22 cells.
  • FIG. 32 shows the effect of the 13A8cPEG-31A12c bispecific on inhibition of Th17 and Th22 differentiation in Sci/hu allograft model:
  • mice with established human immune systems were transplanted with allogeneic human skin. After 4 weeks of EOD treatment, as indicated, the splenocytes were activated in vitro and the cytokines measured in each human CD4 T cell by multi-parameter flow cytometry. Each point indicates an individual treated or control mouse, and each mouse is represented 3 times (for each cytokine shown). Monospecific scFv anti-IL23 is 31A12cPEG; bispecific scFv anti IL6 IL23 is 13A8c-20 KPEG-31A12c. Untreated mice received placebo. 13A8c-20 KPEG-31A12c significantly reduced the differentiation of Th17 cells as measured by the inhibition of IL-17 ( FIG.
  • FIG. 33 shows histological analysis of a section of epidermis of placebo treated mice (A) compared to mice treated with 13A8c-20kPEG-31A12c anti IL-6/anti IL-23 bispecific (B), in which the 13A8c-20kPEG-31A12c anti IL-6/anti IL-23 bispecific significantly reduces the histological features of psoriasis, epidermal thickness in particular.
  • FIG. 34 shows Six experiments utilizing the scid/hu allograft model were completed and the clinical scores judged by a pathologist blinded during the treatment period and are summarized in FIG. 34A (clinical scores).
  • the analysis of histological sections enables a highly quantitative measurement of the most meaningful metrics of psoriasis, in particular, epidermal thickness which is an unbiased measure is shown if FIG. 34B (quantitative epidermal thickness).
  • the bispecific scFv has a highly significant and potent effect on the reduction of psoriasis clinical scores and epidermal thickness.
  • FIG. 35 A shows the readout of the ear hyperplasia mouse model: Mice received intra-dermal injections of rhIL-23 in the right ear (1 ⁇ g) in a volume of 20 ⁇ L on days 0, 1, 2 and 3. PBS was injected into contra-lateral ear as control. Ear thickness was measured on day 4. The first panel shows ear thickness of the IL-23 injected ear compared to the PBS injected ear. The second panel shows the increase in thickness of the IL-23 injected ear compared to the PBS injected ear for each animal.
  • FIG. 35 B shows the results of the ear hyperplasia model when mice were treated with vehicle or 13A8c-20 KPEG-31A12c (100 ug i.p.) on days ⁇ 1 and 2.
  • the first panel shows ear thickness of IL-23 injected ears compared to PBS injected ears in both the vehicle or 13A8c-20 KPEG-31A12c treated animals.
  • the second panel shows the increase in ear thickness when comparing the IL-23 injected ear to the PBS injected ear for each animal.
  • FIG. 35 C shows the results of the ear hyperplasia model when mice were treated with vehicle or 13A8c-20 KPEG-31A12c or 13A8c-40 KPEG-31A12c (100 ug i.p.) on day ⁇ 1 only.
  • the first panel shows ear thickness of IL-23 injected ears compared to PBS injected ears in both the vehicle, 13A8c-20 KPEG-31A12c or 13A8c-40 KPEG-31A12c treated animals.
  • the second panel shows the increase in ear thickness when comparing the IL-23 injected ear to the PBS injected ear for each animal.
  • FIG. 35 D shows the results of the ear hyperplasia model when mice were treated with vehicle or 13A8c-40 KPEG-31A12c (100 ug i.p.) or Ustekinumab (288 ug i.p.) on days ⁇ 1 and 2.
  • the first panel shows ear thickness of IL-23 injected ears compared to PBS injected ears in both the vehicle, 13A8c-40 KPEG-31A12c or Ustekinumab treated animals.
  • the second panel shows the increase in ear thickness when comparing the IL-23 injected ear to the PBS injected ear for each animal.
  • FIG. 36 A shows binding of anti IL-23 chimeric antibodies to IL-12 coated on ELISA plates.
  • An anti IL-6 antibody 13A8 is included as a negative control.
  • human IL-12 is coated on the plate. 22H8 shows strong binding while 31A12 and 49B7 show weaker, but still positive binding.
  • monkey IL-12 monkey IL-12 (macaque) is coated on the plates. Here 49B7, 31A12 and 22H8 all show strong binding to macaque IL-12.
  • the plates are coated with human IL-12 p40 subunit. 22H8 shows strong binding, and 49B7 and 31A12 weaker binding to the p40 subunit.
  • FIG. 36 B shows neutralization of macaque IL-12 induced Interferon ⁇ secretion in the NK92 cell bioassay. Both 31A12 and 22H8 show strong inhibition of the macaque IL-12.
  • FIG. 36 C shows the neutralization of human IL-12 induced Interferon ⁇ secretion in the NK92 cell bioassay.
  • human IL-12 is not neutralized by 31A12 or 49B7. 22H8 neutralizes both macaque and human IL-12.
  • the specification describes, inter alia, bivalent, bispecific constructs that bind to IL-6 and IL-23 and modulate their activity.
  • IL-6 and IL-23 are both known to play a role in the differentiation and activation of T H 17 cells.
  • the activated T H 17 cells are in turn involved in mediating immune responses through a variety of downstream pathways.
  • These two cytokines function at different stages of T H 17 differentiation with IL-6 acting very early in T cell commitment of the T H 17 pathway and IL-23 acting on committed T H 17 cells.
  • the present invention provides novel bivalent bispecific constructs that inhibit two distinct points in the T H 17 activation pathway and have additional inhibitory effects on some of the downstream inflammatory responses mediated by T H 17 products (e.g.
  • the bispecific molecules are able to inhibit T H 17 mediated responses at multiple points in the T H 17 pathway and potentially act with greater potency than the corresponding monospecific antibodies alone.
  • the person skilled in the art will appreciate that the successful production of stable bivalent, bispecific construct that retains the functional characteristics of its constituent antibodies, or has improved functional characteristics, represents a surprising and unexpected result given the uncertainties involved in generating bivalent, bispecific antibodies.
  • the bivalent bispecific constructs of the present invention can modulate (e.g. inhibit), T H 22 cell activation.
  • T H 22 represent a recently identified (Eyerich et al, 2009), distinct subset of T helper cells that are involved in inflammatory and wound healing processes and are particularly implicated in skin inflammation (Nograles et al, 2009). The mechanism of their activation and subsequent action remains the subject of investigation, but the cells themselves are characterized by the secretion of IL-22 and TNF- ⁇ but not IL-17 or Interferon ⁇ .
  • Th22 cells have not been fully characterized, but can be isolated from patients with psoriasis, and express a distinctive gene expression profile from that seen with other T cell subsets. IL-22 expression has been reported to be IL-23 dependent (Kreymborg et al, 2007). The studies conducted here further suggest that Th22 cells are IL-2 dependent in contrast to Th17 cells which rely on IL-21 for growth stimulation.
  • the antibodies and bivalent bispecific constructs of the present invention may be specific for either IL-23 or for both IL-23 and IL-12.
  • the present invention provides a subset of antibodies and bivalent bispecific constructs that bind IL-23, which also target IL-12 molecules.
  • this subset of anti-IL-23 antibodies referred to herein as anti-IL-23/IL-12 antibodies
  • Those that target the p40 subunit of IL-23 are likely to inhibit IL-12 in addition to IL-23.
  • antibodies against p40 may bind an epitope which impairs IL-23 activity without inhibiting IL-12 activity.
  • those antibodies that target the p19 subunit of IL-23 would not be expected to bind IL-12.
  • IL-12 is involved in T H 1 mediated immune responses and as such these particular bivalent bispecific constructs may be useful in modulating not only T H 17 cell mediated immune responses but also T H 1 cell mediated responses. This may be particularly advantageous in treating conditions that have both a T H 1 mediated and T H 17 mediated aspect to their aetiology.
  • the bivalent, bispecific constructs of the present invention comprise an anti-IL-6 antibody, or derivative thereof, and an anti-IL-23 antibody, or derivative thereof.
  • the bivalent, bispecific constructs of the present invention comprise an anti-IL-6 antibody, or derivative thereof, and an anti-IL-23/IL-12 antibody, or derivative thereof.
  • bivalent, bispecific constructs of the present invention can be assayed for their utility in modulating both IL-23 and IL-6 activity using both in vitro and in vivo methods.
  • the assays detailed below may be used.
  • the initial antibodies on which the antibodies, and derivatives thereof, in the bivalent bispecific constructs of the present invention are based can be identified by standard experimental techniques. These antibodies are referred to herein as parent antibodies.
  • the parent antibodies are selected on the basis of their ability to bind IL-6, IL-23 or IL-12.
  • the binding of the parent antibodies can be measured by determining their Kd values.
  • the parent antibodies are selected on the basis of their ability to modulate the activity of IL-6, IL-23 or IL-12.
  • the parent antibodies are selected on the basis of their ability to bind IL-6, IL-23 or IL-12, and on their ability to modulate the activity of IL-6, IL-23 or IL-12.
  • the parent antibodies may be selected for their ability to inhibit the biological activity of IL-6, IL-23 or II-12, or they may be selected for their ability to promote the biological activity of IL-6, IL-23 or IL-12.
  • the parent antibodies are selected for their ability to inhibit IL-6, IL-23 and IL-12.
  • parent antibodies, or derivatives thereof can be obtained from identical or separate animal species.
  • the parent antibodies may, for example, be obtained from an antibody produced in primate, rodent, lagomorph, tylopoda or cartilaginous fish.
  • the parent antibodies may be obtained from transgenic animals. For instance, they may be obtained from a transgenic mouse that has been genetically altered to possess a human immune system, e.g. a Xenomouse®. Antibodies produced in such transgenic animals may have the characteristics of antibodies produced by the exogenous immune system, e.g. antibodies from a Xenomouse may be regarded as human antibodies.
  • the rodent is advantageously a, mouse or a rat.
  • the lagomorph is advantageously a rabbit.
  • one or more of the parent antibodies are obtained from a tylopoda they be obtained from a camel, a llama or a dromedary.
  • This use of such “camelid” antibodies may be advantageous as these species are known to produce high affinity antibodies of only a single variable domain.
  • a tylopoda antibody it is advantageous to use the VHH domain or a modified variant thereof.
  • the cartilaginous fish is advantageously a shark.
  • the primate is advantageously a monkey or ape.
  • the parent antibodies may be immortalized by standard experimental techniques.
  • the present invention provides monoclonal antibodies generated from the parent antibodies that are suitable for incorporation into a bivalent bispecific construct according to the present invention.
  • the present invention also provides compositions comprising a combination of the antibodies and/or derivatives thereof.
  • the combinations comprise an IL-6 antibody, or derivative thereof, and an IL-23 antibody or derivative thereof.
  • the IL-23 antibody, or derivative thereof may also bind IL-12.
  • Preferred combinations of antibodies, and derivatives thereof comprise any one of the IL-6 antibodies defined below, combined with any one of the anti-IL-23 or anti-IL-23/IL-12antibodies defined below.
  • compositions comprising such combinations are expected to have greater activity than the individual antibodies when administered alone.
  • a particularly preferred combination of antibodies, or derivatives thereof is the anti-IL-6 antibody, 13A8, or a derivative based on 13A8 and the anti-IL-23 antibody, 31A12, or a derivative based on 31A12.
  • This combination of antibodies provides greater inhibition of T H 17 cell activity compared to either antigen alone.
  • the combination has greater T H 17 cell inhibitory activity than antibodies known in the art. Furthermore, it exhibits this inhibitory activity at advantageously low dosages.
  • a particularly preferred combination of antibodies or derivatives thereof comprises a PEGylated IL-6 antibody or derivative thereof combined with a PEGylated IL-23 antibody or derivative thereof.
  • the IL-23 antibody, or derivative thereof may also bind IL-12 (i.e. be an IL-23/IL-12 antibody).
  • the antibodies of the bivalent bispecific construct may be subjected to alteration to render them less immunogenic when administered to a human.
  • alteration may comprise one or more of the techniques commonly known as chimerization, humanization, CDR-grafting, deimmunization and/or mutation of framework region amino acids to correspond to the closest human germline sequence (germlining).
  • Subjecting antibodies to such alteration has the advantage that an antibody which would otherwise elicit a host immune response is rendered more, or completely “invisible” to the host immune system, so that such an immune response does not occur or is reduced.
  • Antibodies which have been altered as described according to this embodiment will therefore remain administrable for a longer period of time with reduced or no immune response-related side effects than corresponding antibodies which have not undergone any such alteration(s).
  • One of ordinary skill in the art will understand how to determine whether, and to what degree an antibody must be altered in order to prevent it from eliciting an unwanted host immune response.
  • the present invention provides humanized, or chimeric antibodies that have been altered such that they include amino acid sequences from one or more organisms, or contain synthetic amino acid sequences (e.g. a humanized or chimeric antibody according to the present invention may comprise human framework regions joined to CDR regions obtained from a rodent).
  • humanized anti-IL-6, anti-IL-23 and anti-IL-23/IL-12 antibodies are provided. These antibodies are based on parent antibodies that demonstrated the ability to both bind IL-6, IL-23 or p40 and to modulate (e.g. inhibit) their biological activity. Furthermore, the particular antibodies provided by the present invention retain, or substantially retain, these abilities following immortalization and humanization.
  • Particular humanized antibodies of interest include the following:
  • 31A12 (comprising the VH of SEQ ID NO. 267 and the VL of SEQ ID NO. 269); 34E11 (comprising the VH of SEQ ID NO. 116 and the VL of SEQ ID NO. 118); 35H4 (comprising the VH of SEQ ID NO. 126 and the VL of SEQ ID NO. 128); 49B7 (comprising the VH of SEQ ID NO. 96 and the VL of SEQ ID NO. 98); and 16C6 (comprising the VH of SEQ ID NO. 106 and the VL of SEQ ID NO. 108).
  • 45G5 (comprising the VH of SEQ ID NO. 275 and the VL of SEQ ID NO. 277); 14B5 (comprising the VH of SEQ ID NO. 186 and the VL of SEQ ID NO. 188) 4F3 (comprising the VH of SEQ ID NO. 166 and the VL of SEQ ID NO. 168) 5C5 (comprising the VH of SEQ ID NO. 176 and the VL of SEQ ID NO. 178) 22H8 (comprising the VH of SEQ ID NO. 271 and the VL of SEQ ID NO. 273); and 1H1 (comprising the VH of SEQ ID NO. 156 and the VL of SEQ ID NO. 158)
  • Particularly preferred humanized antibodies are humanized forms of 13A8, 31A12 and 22H8.
  • the present invention also provides antibody variants, for example, as components of the bivalent, bispecific construct.
  • the antibodies retain, or substantially retain, the binding affinity and ability to modulate the biological activity of IL-6, IL-23 or IL-12 (e.g. the Kd value of a variant antibody is at least 80% compared to its parent antibody, and its ability to modulate biological activity is at least 80% of that of its parent antibody as determined by the assays disclosed herein).
  • Variant antibodies or derivatives thereof may be obtained by mutating the variable domains of the heavy and/or light chains to alter a binding property of the antibody.
  • a mutation may be made in the nucleic acid molecule encoding one or more of the CDR regions to increase or decrease the Kd of the antibody for IL-6 or IL-23, to increase or decrease the ability of the antibody to modulate the biological activity of IL-6, IL-23 or IL-12, or to alter the binding specificity of the antibody.
  • Techniques for introducing such mutations using site-directed mutagenesis are well-known in the art.
  • variable domains of the heavy and/or light chains may be obtained by mutating the variable domains of the heavy and/or light chains to alter the isoelectric point (p1) to enhance protein stability at the pH 3-7.5 range of the final formulation to avoid disulphide bond shuffling.
  • p1 isoelectric point
  • SEQ ID NO 332 31A12 pl optimization where the following aminoacids were modified: Q26R, L56R, K109-G110insR, and Q142K
  • SEQ ID 334, 13A18 pl optimization where the following aminoacids were modified: Q26R, L56R, K112-G113insR, and Q145K.
  • stability may be enhanced by mutating the variable domains of the heavy and/or light chains to reduce aggregation of the product in solution, see for example SEQ ID NO 331, 31A12 F125 mutation predicted to enhance solubility and reduce aggregation of the product, and SEQ ID NO. 333, 31A12 combined pl optimization and F125 mutation
  • the nucleic acid molecules may be mutated in one or more of the framework regions.
  • a mutation may be made in a framework region or constant domain to increase the half-life of the anti-IL-6 or anti-IL-23 antibody.
  • a mutation in a framework region or constant domain may also be made to alter the immunogenicity of the antibody, to provide a site for covalent or non-covalent binding to another molecule, or to alter such properties as complement fixation.
  • mutations may be made in each of the framework regions, the constant domain and the variable regions in a single mutated antibody.
  • mutations may be made in only one of the framework regions, the variable regions or the constant domain in a single mutated antibody.
  • the present invention provides variant anti-IL-6 antibodies that have at least 90% sequence identity to the anti-IL-6 antibody prior to mutation.
  • the variant anti-IL-6 antibody has at least 95%, 96%, 97%, 98% or 99% sequence identity to the anti-IL-6 antibody prior to mutation.
  • the present invention provides variant anti-IL23 antibodies that have at least 90% sequence identity to the anti-IL-23 antibody prior to mutation.
  • the variant anti-IL-23 antibody has at least 95%, 96%, 97%, 98% or 99% sequence identity to the anti-IL23 antibody prior to mutation.
  • the present invention provides variant anti-IL-23/IL-12 antibodies that have at least 90% sequence identity to the anti-IL-23/IL-12 antibody prior to mutation.
  • the variant anti-IL-23/IL-12 antibody has at least 95%, 96%, 97%, 98% or 99% sequence identity to the anti-IL-23/IL-12 antibody prior to mutation.
  • Antibody derivatives may be generated using techniques and methods known to one of ordinary skill in the art. Antibody derivatives according to the present invention retain, or substantially retain, the binding affinity and ability to modulate the biological activity of IL-6, IL-23 or p40 of the antibodies from which they are derived. Examples of antibody derivatives include, Fab, Fab′, F(ab)′ and scFv constructs, Kappabodies, Minibodies, and Janusins derived from the anti-IL-6, anti-IL-23, and anti-IL-23/IL-12 antibodies disclosed herein.
  • Fab, Fab′, F(ab)′ fragments of the anti-IL-6 antibodies or variant anti-IL-6 antibodies are provided.
  • Fab, Fab′, F(ab)′ fragments of the anti-IL-23 antibodies or variant anti-IL-23 antibodies are provided.
  • Fab, Fab′, F(ab)′ fragments of the anti-IL-23/IL-12-23/IL-126 antibodies or variant anti-IL-23/IL-12 antibodies are provided.
  • scFv derivatives of the anti-IL-6 antibodies or variant anti-IL-6 antibodies are provided.
  • scFv derivatives of the anti-IL-23 antibodies or variant anti-IL-23 antibodies are provided.
  • scFv derivatives of the anti-IL-23/IL-12 antibodies or variant anti-IL-23/IL-12 antibodies are provided.
  • the VH- and VL-encoding DNA fragments are operatively linked to another fragment encoding a flexible linker such that the VH and VL sequences can be expressed as a contiguous single-chain protein, with the VL and VH regions joined by the flexible linker (see e.g., Bird et al. (1988) Science 242: 423-426; Huston et al. (1988) Proc. Natl. Acad. Sci. USA 85: 5879-5883; McCafferty et al., Nature (1990) 348: 552-554).
  • the single chain antibody may be monovalent, if only a single VH and VL are used, bivalent, if two VH and VL are used, or polyvalent, if more than two VH and VL are used.
  • the single chain antibody is prepared using one or more of the variable regions from an anti-IL-6 antibody. In another embodiment, the single chain antibody is prepared using one or more CDR regions from the anti-IL-6 antibody.
  • the single chain antibody is prepared using one or more of the variable regions from an anti-IL-23 antibody. In another embodiment, the single chain antibody is prepared using one or more CDR regions from the anti-IL-23 antibody.
  • the single chain antibody is prepared using one or more of the variable regions from an anti-IL-23/IL-12 antibody. In another embodiment, the single chain antibody is prepared using one or more CDR regions from the anti-IL-23/IL-12 antibody.
  • anti-IL-6 single chain antibodies are derived from the humanized anti-IL-6 antibodies described above
  • anti-IL-23 single chain antibodies are derived from the humanized anti-IL-23 antibodies described above
  • anti-IL-23/IL-12 single chain antibodies are derived from the humanized anti-IL-23/IL-12 antibodies described above.
  • a linker portion of the present invention may be the sequence GGGGS repeated 3 to 5 times, or a non integer repeat of the GGGGS sequence, see for instance SEQ ID NO. 330.
  • single chain antibodies of the invention are covalently linked to PEG.
  • modified antibodies may be prepared using anti-IL-6 antibody, anti-IL-23-antibody or anti-IL-23/IL-12 antibody encoding nucleic acid molecules.
  • “Kappa bodies” (ILI et al., Protein Eng 10: 949-57 (1997)), “Minibodies” (Martin et al., EMBO J 13: 5303-9 (1994)), or “Janusins” (Traunecker et al., EMBO J 10: 3655-3659 (1991) and Traunecker et al. “Janusin: new molecular design for bispecific reagents” Int J Cancer Suppl 7: 51-52 (1992)) may be prepared using standard molecular biological techniques.
  • CDRs Complementarity Determining Regions
  • Complementarity determining regions are relatively short amino acid sequence in the shape of a flexible loop, found in the variable (V) domains of antigen receptors (e.g. immunoglobulin and T cell receptor).
  • V variable domains of antigen receptors
  • the CDRs of both immunoglobulin and the T cell receptor are the parts of these molecules that determine their specificity and make contact with a specific ligand.
  • the CDRs are the most variable part of the molecule, and contribute to the diversity of these molecules, allowing the immunoglobulin and the T cell receptor to recognize a vast repertoire of antigens.
  • anti-IL-6, anti-IL-23 and anti-IL-23/IL-12 antibodies that make up the bivalent, bispecific constructs of the invention play a key role in determining the specificity of the antibodies, and antibodies that have particular CDRs regions in common would be expected to have the same or similar antigen specificity.
  • anti-IL-6, anti-IL-23 and anti-IL-23/IL-12 antibodies, or derivatives thereof comprise the CDR regions of the antibodies on which they are based.
  • the present invention provides for the antibodies and antibody derivatives that make up the bivalent, bispecific constructs of the invention to comprise at least one of CDR1, CDR2, CDR3, CDR4, CDR5 and CDR6 of a parent antibody.
  • the antibodies and antibody derivatives that that make up the bivalent, bispecific constructs comprise at least CDR3.
  • the mutated anti-IL-6 antibody has at least one complementarity determining region (CDR) that remains unchanged compared to the anti-IL-6 antibody prior to mutation.
  • the unchanged CDR may be CDR1, CDR2, CDR3, CDR4, CDR5 or CDR6.
  • the mutated anti-IL-23 antibody has at least one complementarity determining region (CDR) that remains unchanged compared to the anti-IL-23 antibody prior to mutation.
  • the unchanged CDR may be CDR1, CDR2, CDR3, CDR4, CDR5 or CDR6.
  • the mutated anti-IL-23/IL-12 antibody has at least one complementarity determining region (CDR) that remains unchanged compared to the anti-IL-6 antibody prior to mutation.
  • the unchanged CDR may be CDR1, CDR2, CDR3, CDR4, CDR5 or CDR6.
  • motifs regions that are particularly important in determining the specificity of a particular antibody or derivative thereof.
  • the specificity of these regions may be determined by a number of factors, such as their conformation and the location of charged amino acid residues within the region.
  • the person skilled in the art may identify these motifs through techniques known in the art including, epitope mapping and comparing the sequence of antibodies known to bind the same target.
  • the present invention provides antibodies or derivatives thereof that comprise CDRs having particular motifs.
  • the CDRs comprise at least 3, at least 4, at least 5 or at least 6 consecutive amino acids taken from the CDR regions of the following antibodies:
  • the CDRs comprise substituted consecutive amino acid sequences taken from the above mentioned CDRs.
  • the motifs may comprise at least 3, at least 4, at least 5, or at least 6 residues wherein the identity and position of the amino acid is fixed relative to the other amino acids in the sequence, and one or two amino acids may be substituted, compared to the corresponding amino acid of the CDR prior to substitution.
  • the substitutions are conservative substitutions.
  • An example of such a motif can be found within the CDR2 region of the 22H8 anti-IL-23/IL-12 antibody. The motif may be described by the following formula:
  • X1 is alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine or tryptophan, and preferably is alanine or valine;
  • a motif in the CDR3 region comprising the amino acid sequence YAYX 1 GDAFDP, wherein X 1 is alanine or isoleucine; (SEQ ID NO. 339) and/or a motif in the CDR3 region comprising the amino acid sequence SDYFNX 1 , wherein X 1 is isoleucine or valine; (SEQ ID NO.
  • X 1 is alanine or serine
  • X 2 is selected from the group consisting of glycine, asparagine, glutamine, cysteine, serine, threonine, and tyrosine; preferably X 2 is serine or threonine; and/or a motif in the CDR5 region comprising the amino acid sequence ASX 1 LA, wherein X 1 is lysine or threonine; (SEQ ID NO. 341) and/or a motif in the CDR6 region comprising the amino acid sequence QSYYDX 1 NAGYG, wherein X 1 is alanine or valine. (SEQ ID NO. 342)
  • Examples of common motifs found with the CDRs of the IL-23 antibodies of the present invention include:
  • a motif in the CDR2 region comprising the amino acid sequence YYAX 1 WAX 2 G, wherein X 1 is selected from the group consisting of serine, proline and aspartate, and X 2 is selected from the group consisting of lysine and glutamine;
  • SEQ ID NO. 337 and/or a motif in the CDR5 region comprising the amino acid sequence AX 1 TLX 2 S, wherein X 1 is selected from the group consisting of serine and alanine X 2 is selected from the group consisting of alanine and threonine. SEQ ID NO. 338)
  • Examples of common motifs found with the CDRs of the IL-6 antibodies of the present invention include:
  • a motif in the CDR2 region comprising the amino acid sequence YIYTDX 1 STX 2 YANWAKG, wherein X 1 is selected from the group consisting of glycine, asparagine, glutamine, cysteine, serine, threonine, tyrosine; and X 2 is selected from the group consisting of phenylalanine, tryptophan, and tyrosine; and preferably X 1 is serine or threonine and X2 is tryptophan or tyrosine; (SEQ ID NO. 335) and/or a motif in the CDR5 region comprising the amino acid sequence RX 1 STLX 2 S, wherein X 1 and X 2 are independently alanine or threonine. (SEQ ID NO. 336)
  • the present invention provides for the incorporation of non-natural amino acid residues into the anti-IL-6 anti-IL-23 and anti-IL-23/IL-12 antibodies, or derivatives thereof, to provide a point of the attachment for the anti-IL-6 antibodies, or derivatives thereof, to anti-IL-23 or anti-IL-23/IL-12 antibodies, or derivatives thereof.
  • non-natural amino acids including, for instance azidohomoalanine (Aha).
  • Additional non natural amino acids include azidonorleucine, 3-(1-naphthyl)alanine, 3-(2-naphthyl)alanine, p-ethynyl-phenylalanine, p-propargly-oxy-phenylalanine, m-ethynyl-phenylalanine, 6-ethynyl-tryptophan, 5-ethynyl-tryptophan, (R)-2-amino-3-(4-ethynyl-1H-pyrol-3-yl)propanic acid, p-bromophenylalanine, p-idiophenylalanine, p-azidophenylalanine, 3-(6-chloroindolyl)alanine, 3-(6-bromoindoyl)alanine, 3-(5-bromoindolyl)alanine, homoallylglycine, homopropargylglycine, and p-
  • a non-natural amino acid may be located at the N-terminus of an antibody, or derivative thereof, as disclosed herein.
  • a non-natural amino acid may be located at the C-terminus of an antibody, or derivative thereof, as disclosed herein.
  • the non-natural amino acid may be located in the linker region between the VH and VL portions of an scFv as disclosed herein (e.g. within SEQ ID NO. 327).
  • each antibody to be incorporated into the bivalent, bispecific construct.
  • examples of antibodies, or derivatives thereof, scFvs, and/or portions of the bivalent bispecific constructs of the present invention include SEQ ID No. 287 to 312.
  • non-natural amino acids is achieved by expressing the antibodies in auxotrophic host cells that incorporate a non-natural amino acid (such as Aha) in place of methionine (Met).
  • a non-natural amino acid such as Aha
  • the antibody nucleotide sequences must be engineered to remove any naturally occurring codons for methionine not located at the desired site of attachment. This may be achieved by substituting them with codons for other amino acids (typically natural amino acids). Since 1-2 methionine residues are frequently found within framework regions and CDRs of immunoglobulin VH-regions, and infrequently in VL regions, it is necessary to find suitable replacements for these residues where they occur without impacting the expression, stability or function (e.g.
  • methionine-free scFv can then be optimized for expression in a methionine auxotrophic bacterial strain, purified, refolded and tested for biologic activity.
  • more than one methionine codon can be left in the sequence to allow for incorporation of more than one non-natural amino acid (such as Aha).
  • a methionine codon can be introduced that serves as an insertion site for a non-natural amino acid with a chemically reactive site for attachment.
  • the antibodies modified to include non-natural amino acids may be attached to one or more separate entities. These entities include linker groups and/or other similarly modified antibodies. Examples of suitable linkers are known in the art and include short peptide sequences.
  • the present invention also provides for the use of PEG as a linker.
  • an anti-IL-6 antibody, or derivative thereof, incorporating a non-natural amino acid may be covalently linked to a PEG linker group, which PEG linker group is in turn attached to an anti-IL-23 or anti-IL-23/IL-12 antibody, or derivative thereof, incorporating a non-natural amino acid.
  • Such bi-specific, PEGylated constructs can then be purified and refolded to yield a stable, biologically active therapeutic protein.
  • the antibodies or derivatives thereof in the present invention modified to include non-natural aminoacids may directly (e.g. without the use of linker groups) be linked to other similarly modified molecules, including but not limited to, other antibodies or derivatives thereof, dyes, drugs or toxins.
  • a bivalent bispecific construct or antibody of the invention can be derivatized or linked to another molecule.
  • the bivalent bispecific construct is derivatized such that binding and biological activity of the constituent antibodies or derivatives thereof is not affected adversely by the derivatization or labelling.
  • a bivalent bispecific construct of the invention can be functionally linked (by chemical coupling, genetic fusion, noncovalent association or otherwise) to one or more other molecular entities, such as a detection agent, a cytotoxic agent, a pharmaceutical agent, and/or a protein or peptide that can mediate association of the antibody or antibody portion with another molecule (such as a streptavidin core region or a polyhistidine tag).
  • a detection agent such as a cytotoxic agent, a pharmaceutical agent, and/or a protein or peptide that can mediate association of the antibody or antibody portion with another molecule (such as a streptavidin core region or a polyhistidine tag).
  • a type of derivatized bivalent bispecific construct is a labelled bivalent bispecific construct.
  • Useful detection agents with which bivalent bispecific construct of the invention may be derivatized include fluorescent compounds, including fluorescein, fluorescein isothiocyanate, rhodamine, 5-dimethylamine-1-napthalenesulfonyl chloride, phycoerythrin, lanthanide phosphors and the like.
  • An bivalent bispecific construct y may also be labelled with enzymes that are useful for detection, such as horseradish peroxidase, -galactosidase, luciferase, alkaline phosphatase, glucose oxidase and the like.
  • enzymes that are useful for detection, such as horseradish peroxidase, -galactosidase, luciferase, alkaline phosphatase, glucose oxidase and the like.
  • a bivalent bispecific construct may also be labelled with biotin, and detected through indirect measurement of avidin or streptavidin binding.
  • a bivalent bispecific construct may also be labelled with a predetermined polypeptide epitopes recognized by a secondary reporter (e.g. leucine zipper pair sequences, binding sites for secondary antibodies, metal binding domains, epitope tags).
  • labels are attached by spacer arms of various lengths to reduce potential steric hindrance.
  • the bivalent bispecific construct may also be labelled with a radiolabelled amino acid.
  • the radiolabel may be used for both diagnostic and therapeutic purposes.
  • the radio-labelled bivalent bispecific construct may be used diagnostically, for example, for determining IL-6 and/or IL-23 levels in a subject. Further, the radio-labelled bivalent bispecific construct may be used therapeutically for treating diseases mediated by the T H 17 pathway.
  • Examples of radiolabels include, but are not limited to, the following radioisotopes or radionuclides-3H, 14C, 15N, 35S, 90Y, 99Tc, 111In, 125I, 131I.
  • Radioisotopes may also be bound to the antibody or bispecific by derivitization with a chelation moiety such as DOTA. Several of the useful imaging and therapeutic radioisotopes bind tightly to these chelators.
  • a bivalent bispecific construct may also be derivatized with a chemical group such as polyethylene glycol (PEG), a methyl or ethyl group, or a carbohydrate group. These groups may be useful to improve the biological characteristics of the antibody, e.g. to increase serum half-life or to increase tissue binding.
  • PEG polyethylene glycol
  • methyl or ethyl group a methyl or ethyl group
  • carbohydrate group may be useful to improve the biological characteristics of the antibody, e.g. to increase serum half-life or to increase tissue binding.
  • bivalent, bispecific constructs of the present invention, and the antibodies, and derivatives thereof, that make up the bivalent, bispecific constructs can be expressed using conventional recombinant technology.
  • constructs and/or antibodies, and derivatives thereof comprise non-natural amino acids
  • recombinant methods as described in WO 2007/130453 may be used.
  • the nucleotide sequences, vectors, host cells etc. used to express the bivalent bispecific constructs and the antibodies, and derivatives thereof are objects of the present invention
  • the present invention provides nucleotide sequences encoding the bivalent, bispecific constructs and the antibodies, and derivatives thereof, that make up the bivalent, bispecific constructs as defined above.
  • the present invention encompasses nucleotide sequences encoding (1) monoclonal antibodies according to the invention (2) humanized antibodies according to the invention (3) variant antibodies based on (1) & (2) according to the invention (4) derivatives of the antibodies of (1) to (3) according to the invention, and (5) bivalent, bispecific constructs according to the invention.
  • nucleotide sequences encode portions of anti-IL-6 antibodies. Examples of such sequences are given in SEQ ID NOs. 7 and 9, which are the nucleotide sequences of the VH and VL regions of the IL-6 antibody designated 13A8.
  • nucleotide sequences encode portions of anti-IL-23 antibodies. Examples of such sequences are given in SEQ ID NOs. 87 and 89, which are the nucleotide sequences of the VH and VL regions of the anti-IL-23 antibody designated 31A12.
  • nucleotide sequences encode portions of anti-IL-23/IL-12 antibodies. Examples of such sequences are given in SEQ ID NOs. 137 and 139, which are the nucleotide sequences of the VH and VL regions of the anti-IL-23/IL-12 antibody designated 22H8.
  • bivalent, bispecific construct may be expressed as a single product.
  • nucleotide sequences of the present invention are operably linked to a promoter sequence.
  • suitable promoters include, but are not limited to, T5/Lac promoter: T7/Lac or modified T7/lac promoters, Trc or tac promoters, phage pL or pR temperature inducible promoters, tetA promoter/operator, araBAD (pBAD) promoter, rhaPBAD promoter and lac UV5 promoter.
  • suitable promoters may be identified from Terpe, K. (2006) (Appl Microbiol Biotechnol 72:211-222).
  • the promoter is a T5/Lac promoter.
  • the present invention provides a vector comprising a nucleotide e sequence of the present invention optionally, operably linked to a promoter sequence.
  • the present invention provides a host cell transfected with a vector of the present invention and capable of expressing the nucleotide sequences contained within the vectors.
  • the host cell is an auxotrophic cell, capable of incorporating a non-natural amino acid in place of a particular natural amino acid (e.g. AHA in place of Met).
  • the host cell may be a prokaryotic cell or an eukaryotic cell. Suitable eukaryotic cells include yeast cells, mammalian cells and insect cells.
  • the host cells are prokaryotic, in particular, E. coli B384 which are methionine auxotrophic cells.
  • the cells are mammalian cells, more preferably they are human cells, yet more preferably they are human embryonic kidney cells (e.g. HEK293 or HEK 293c18 cells) or CHO cells.
  • primers for the cloning and expression of the anti-IL-6, anti-IL-23 antibodies and anti-IL-23/IL-12 antibodies, and derivatives thereof are provided. These primers vary in length between 10 and 40 nucleotides, preferably they are between 15 and 30 nucleotides in length. The person skilled in the art will be able to determine suitable primer sequences given the disclosure of the nucleic acid sequences of the antibodies, and derivatives thereof, disclosed herein. Particular primer sequences of interest are given in SEQ ID NOs 0.200-258, which are useful for the cloning and expression of the antibodies and scFvs disclosed herein.
  • the first step in the protein engineering process is usually to select a set of non-natural amino acids that have the desired chemical properties.
  • the selection of non-natural amino acids depends on pre-determined chemical properties and the modifications one would like to make in the target molecule or target protein.
  • Non-natural amino acids once selected, can either be purchased from vendors, or chemically synthesized. Any number of non-natural amino acids may be incorporated into the target molecule and may vary according to the number of desired chemical moieties that are to be attached. The chemical moieties may be attached to all or only some of the non-natural amino acids. Further, the same or different non-natural amino acids may be incorporated into the molecule, depending on the desired outcome.
  • At least two different non-natural amino acids are incorporated into the molecule and one chemical moiety, such as PEG, is attached to one of the non-natural amino acid residues, while another chemical moiety, such as a cytotoxic agent, is attached to the other non-natural amino acid.
  • one chemical moiety such as PEG
  • another chemical moiety such as a cytotoxic agent
  • non-natural amino acids can be used in the methods of the invention.
  • the non-natural amino acids of use in the invention are selected or designed to provide additional characteristics unavailable in the twenty natural amino acids.
  • non-natural amino acids are optionally designed or selected to modify the biological properties of a molecule, including a protein, e.g., into which they are incorporated.
  • the following properties are optionally modified by inclusion of an non-natural amino acid into a molecule, such as a protein: toxicity, biodistribution, solubility, stability, e.g., thermal, hydrolytic, oxidative, resistance to enzymatic degradation, and the like, facility of purification and processing, structural properties, spectroscopic properties, chemical and/or photochemical properties, catalytic activity, ability to function as a vaccine, redox potential, half-life, ability to react with other molecules, e.g., covalently or noncovalently, and the like.
  • toxicity e.g., biodistribution, solubility, stability, e.g., thermal, hydrolytic, oxidative, resistance to enzymatic degradation, and the like, facility of purification and processing, structural properties, spectroscopic properties, chemical and/or photochemical properties, catalytic activity, ability to function as a vaccine, redox potential, half-life, ability to react with other molecules, e.g., covalently or
  • an “non-natural amino acid” refers to any amino acid, modified amino acid, or amino acid analogue other than selenocysteine and the following twenty genetically encoded alpha-amino acids: alanine, arginine, asparagine, aspartic acid, cysteine, glutamine, glutamic acid, glycine, histidine, isoleucine, leucine, lysine, methionine, phenylalanine, proline, serine, threonine, tryptophan, tyrosine, valine.
  • the generic structure of an alpha-amino acid is illustrated by Formula I:
  • a non-natural amino acid is typically any structure having Formula I wherein the R group is any substituent other than one used in the twenty natural amino acids. See, e.g., any biochemistry text such as Biochemistry by L. Stryer, 3rd ed. 1988, Freeman and Company, New York, for structures of the twenty natural amino acids. Note that the non-natural amino acids disclosed herein may be naturally occurring compounds other than the twenty alpha-amino acids above. Because the non-natural amino acids disclosed herein typically differ from the natural amino acids in side chain only, the non-natural amino acids form amide bonds with other amino acids, e.g., natural or non-natural, in the same manner in which they are formed in naturally occurring proteins.
  • R in Formula I optionally comprises an alkyl-, aryl-, aryl halide, vinyl halide, beta-silyl alkenyl halide, beta-silyl alkenyl sulfonates, alkyl halide, acetyl, ketone, aziridine, nitrile, nitro, nitrile oxide, halide, acyl-, keto-, azido-, ketal, acetal, hydroxyl-, hydrazine, cyano-, halo-, hydrazide, alkenyl, alkynyl, ether, thioether, epoxide, sulfone, boronic acid, boronate ester, borane, phenylboronic acid, thiol, seleno-, sulfonyl-, borate, boronate, phosphonate, phosphoric acid, boronate ester, boronate, phosphoric
  • unnatural amino acids include, but are not limited to, p-acetyl-L-phenylalanine, O-methyl-L-tyrosine, an L-3-(2-naphthyl)alanine, a 3-methyl-phenylalanine, an O-4-allyl-L-tyrosine, a 4-propyl-L-tyrosine, a tri-O-acetyl-GlcNAc.beta.-serine, .beta.-O-GlcNAc-L-serine, a tri-O-acetyl-GalNAc-.alpha.-threonine, an .alpha.-GalNAc-L-threonine, an L-Dopa, a fluorinated phenylalanine, an isopropyl-L-phenylalanine, a p-azido-L-phenylalanine, a p-acyl-L-phenyl
  • p-acetyl-L-phenylalanine O-methyl-L-tyrosine, an L-3-(2-naphthyl)alanine, a 3-methyl-phenylalanine, an O-4-allyl-L-tyrosine, a 4-propyl-L-tyrosine, a tri-O-acetyl-GlcNAc.beta.-serine, .beta.-O-GlcNAc-L-serine, a tri-O-acetyl-GalNAc-.alpha.-threonine, an .alpha.-GalNAc-L-threonine, an L-Dopa, a fluorinated phenylalanine, an isopropyl-L-phenylalanine, a p-azido-L-phenylalanine, a p-acyl
  • Aryl substitutions may occur at various positions, e.g. ortho, meta, para, and with one or more functional groups placed on the aryl ring.
  • Other non-natural amino acids of interest include, but are not limited to, amino acids comprising a photoactivatable cross-linker, spin-labeled amino acids, dye-labeled amino acids, fluorescent amino acids, metal binding amino acids, metal-containing amino acids, radioactive amino acids, amino acids with novel functional groups, amino acids with altered hydrophilicity, hydrophobocity, polarity, or ability to hydrogen bond, amino acids that covalently or noncovalently interact with other molecules, photocaged and/or photoisomerizable amino acids, amino acids comprising biotin or a biotin analogue, glycosylated amino acids such as a sugar substituted serine, other carbohydrate modified amino acids, keto containing amino acids, amino acids comprising polyethylene glycol or a polyether, a polyalcohol, or a polysaccharide, amino acids that can undergo metathesis, amino
  • non-natural amino acids also optionally comprise modified backbone structures, e.g., as illustrated by the structures of Formula II and III:
  • Z typically comprises OH, NH.sub.2, SH, NH.sub.20-, NH—R′, R′NH—, R′S—, or S—R′—;
  • X and Y which may be the same or different, typically comprise S, N, or O, and R and R′, which are optionally the same or different, are typically selected from the same list of constituents for the R group described above for the non-natural amino acids having Formula I as well as hydrogen or (CH.sub.2).sub.x or the natural amino acid side chains.
  • non-natural amino acids disclosed herein optionally comprise substitutions in the amino or carboxyl group as illustrated by Formulas II and III.
  • Non-natural amino acids of this type include, but are not limited to, .alpha.-hydroxy acids, .alpha.-thioacids .alpha.-aminothiocarboxylates, or .alpha.-.alpha.-disubstituted amino acids, with side chains corresponding e.g. to the twenty natural amino acids or to non-natural side chains. They also include but are not limited to .beta.-amino acids or .gamma.-amino acids, such as substituted .beta.-alanine and .gamma.-amino butyric acid.
  • substitutions or modifications at the .alpha.-carbon optionally include L or D isomers, such as D-glutamate, D-alanine, D-methyl-O-tyrosine, aminobutyric acid, and the like.
  • L or D isomers such as D-glutamate, D-alanine, D-methyl-O-tyrosine, aminobutyric acid, and the like.
  • Other structural alternatives include cyclic amino acids, such as proline analogs as well as 3-, 4-, 6-, 7-, 8-, and 9-membered ring proline analogs.
  • Some non-natural amino acids such as aryl halides (p-bromo-phenylalanine, p-iodophenylalanine, provide versatile palladium catalyzed cross-coupling reactions with ethyne or acetylene reactions that allow for formation of carbon-carbon, carbon-nitrogen and carbon-oxygen bonds between aryl halides and a wide variety of coupling partners.
  • aryl halides p-bromo-phenylalanine, p-iodophenylalanine
  • non-natural amino acids are based on natural amino acids, such as tyrosine, glutamine, phenylalanine, and the like.
  • Tyrosine analogs include para-substituted tyrosines, ortho-substituted tyrosines, and meta substituted tyrosines, wherein the substituted tyrosine comprises an acetyl group, a benzoyl group, an amino group, a hydrazine, an hydroxyamine, a thiol group, a carboxy group, an isopropyl group, a methyl group, a C6-C20 straight chain or branched hydrocarbon, a saturated or unsaturated hydrocarbon, an O-methyl group, a polyether group, a nitro group, or the like.
  • Glutamine analogs include, but are not limited to, .alpha.-hydroxy derivatives, .beta.-substituted derivatives, cyclic derivatives, and amide substituted glutamine derivatives.
  • exemplary phenylalanine analogs include, but are not limited to, meta-substituted phenylalanines, wherein the substituent comprises a hydroxy group, a methoxy group, a methyl group, an allyl group, an acetyl group, or the like.
  • non-natural amino acids include, but are not limited to, o, m and/or p forms of amino acids or amino acid analogs (non-natural amino acids), including homoallylglycine, cis- or trans-crotylglycine, 6,6,6-trifluoro-2-aminohexanoic acid, 2-aminopheptanoic acid, norvaline, norleucine, O-methyl-L-tyrosine, o-, m-, or p-methyl-phenylalanine, O-4-allyl-L-tyrosine, a 4-propyl-L-tyrosine, a tri-O-acetyl-GlcNAc.beta.-serine, an L-Dopa, a fluorinated phenylalanine, an isopropyl-L-phenylalanine, a p-azidophenylalanine, a p-acyl-L-phenylalanine,
  • Tyrosine analogs include para-substituted tyrosines, ortho-substituted tyrosines, and meta substituted tyrosines, wherein the substituted tyrosine comprises an acetyl group, a benzoyl group, an amino group, a hydrazine, an hydroxyamine, a thiol group, a carboxy group, an isopropyl group, a methyl group, a C6-C20 straight chain or branched hydrocarbon, a saturated or unsaturated hydrocarbon, an O-methyl group, a polyether group, a nitro group, or the like.
  • multiply substituted aryl rings are also contemplated.
  • Glutamine analogs of the invention include, but are not limited to, .alpha.-hydroxy derivatives, .beta.-substituted derivatives, cyclic derivatives, and amide substituted glutamine derivatives.
  • Example phenylalanine analogs include, but are not limited to, meta-substituted phenylalanines, wherein the substituent comprises a hydroxy group, a methoxy group, a methyl group, an allyl group, an acetyl group, or the like.
  • Lysine analogs include N-sigma substituted such as pyrrolysine, N-sigma-o-azidobenzyloxycarbonyl-L-Lysine (AzZLys), N-sigma-propargyloxycarbonyl-L-Lysine, N-sigma-2-azidoethoxycarbonyl-L-Lysine, N-sigma-tert-butyloxycarbonyl-L-Lysine (BocLys), N-sigma-allyloxycarbonyl-L-Lysine (AlocLys), N-sigma-acetyl-L-Lysine (AcLys), N-sigma-benzyloxycarbonyl-L-Lysine (ZLys), N-sigma-cyclopentyloxycarbonyl-L-Lysine (CycLys), N-sigma-D-prolyl-L-Lysine, N-sigma-
  • other examples optionally include (but are not limited to) an non-natural analog of a tyrosine amino acid; an non-natural analog of a glutamine amino acid; an non-natural analog of a phenylalanine amino acid; an non-natural analog of a serine amino acid; an non-natural analog of a threonine amino acid; an alkyl, aryl, acyl, azido, cyano, halo, hydrazine, hydrazide, hydroxyl, alkenyl, alkynl, ether, thiol, sulfonyl, seleno, ester, thioacid, borate, boronate, phospho, phosphono, phosphine, heterocyclic, enone, imine, aldehyde, hydroxylamine, keto, ketal, acetal, strained cyclooctyne, strained cycloalkene, cyclopropene, norbornenes
  • non-natural amino acids utilized herein for certain embodiments may be selected or designed to provide additional characteristics unavailable in the twenty natural amino acids.
  • non-natural amino acid are optionally designed or selected to modify the biological properties of a protein, e.g., into which they are incorporated.
  • the following properties are optionally modified by inclusion of an non-natural amino acid into a protein: toxicity, biodistribution, solubility, stability, e.g., thermal, hydrolytic, oxidative, resistance to enzymatic degradation, and the like, facility of purification and processing, structural properties, spectroscopic properties, chemical and/or photochemical properties, catalytic activity, redox potential, half-life, ability to react with other molecules, e.g., covalently or noncovalently, and the like.
  • amino acid analogs optionally include (but are not limited to) an non-natural analog of a tyrosine amino acid; an non-natural analog of a glutamine amino acid; an non-natural analog of a phenylalanine amino acid; an non-natural analog of a serine amino acid; an non-natural analog of a threonine amino acid; an alkyl, aryl, acyl, azido, cyano, halo, hydrazine, hydrazide, hydroxyl, alkenyl, alkynl, ether, thiol, sulfonyl, seleno, ester, thioacid, borate, boronate, phospho, phosphono, phosphine, heterocyclic, enone, imine, aldehyde, hydroxylamine, keto, ketal, acetal, strained cyclooctyne, strained cycloalkene, cyclopropene,
  • Non-natural amino acids suitable for use in the methods of the invention also include those that have a saccharide moiety attached to the amino acid side chain.
  • an non-natural amino acid with a saccharide moiety includes a serine or threonine amino acid with a Man, GalNAc, Glc, Fuc, or Gal moiety.
  • the invention includes unprotected and acetylated forms of the above.
  • meta-substituted phenylalanines are synthesized in a procedure as outlined in WO 02/085923 (see, e.g., FIG. 14 of the publication).
  • NBS N-bromosuccinimide
  • Typical substituents used for the meta position include, but are not limited to, ketones, methoxy groups, alkyls, acetyls, and the like.
  • 3-acetyl-phenylalanine is made by reacting NBS with a solution of 3-methylacetophenone.
  • a similar synthesis is used to produce a 3-methoxy phenylalanine.
  • the R group on the meta position of the benzyl bromide in that case is—OCH.sub.3. (See, e.g., Matsoukas et al., J. Med. Chem., 1995, 38, 4660-4669, incorporated by reference in its entirety).
  • the design of non-natural amino acids is biased by known information about the active sites of synthetases, e.g., external mutant tRNA synthetases used to aminoacylate an external mutant tRNA.
  • synthetases e.g., external mutant tRNA synthetases used to aminoacylate an external mutant tRNA.
  • three classes of glutamine analogs are provided, including derivatives substituted at the nitrogen of amide (1), a methyl group at the .gamma.-position (2), and a N-Cy-cyclic derivative (3).
  • the analogs were designed to complement an array of side chain mutations of residues within a 10.ANG. shell of the side chain of glutamine, e.g., a mutation of the active site Phe233 to a small hydrophobic amino acid might be complemented by increased steric bulk at the Cy position of Gln.
  • N-phthaloyl-L-glutamic 1,5-anhydride (compound number 4 in FIG. 23 of WO 02/085923) is optionally used to synthesize glutamine analogs with substituents at the nitrogen of the amide.
  • N-phthaloyl-L-glutamic 1,5-anhydride (compound number 4 in FIG. 23 of WO 02/085923) is optionally used to synthesize glutamine analogs with substituents at the nitrogen of the amide.
  • the anhydride is typically prepared from glutamic acid by first protection of the amine as the phthalimide followed by refluxing in acetic acid. The anhydride is then opened with a number of amines, resulting in a range of substituents at the amide. Deprotection of the phthaloyl group with hydrazine affords a free amino acid as shown in FIG. 23 of WO 2002/085923.
  • Substitution at the .gamma.-position is typically accomplished via alkylation of glutamic acid.
  • a protected amino acid e.g., as illustrated by compound number 5 in FIG. 24 of WO 02/085923, is optionally prepared by first alkylation of the amino moiety with 9-bromo-9-phenylfluorene (PhflBr) (see, e.g., Christie & Rapoport, J. Org. Chem.
  • N-Cy cyclic analog as illustrated by Compound number 3 in FIG. 25 of WO 02/085923, is optionally prepared in 4 steps from Boc-Asp-Ot-Bu as previously described.
  • Boc-Asp-Ot-Bu See, e.g., Barton et al., Tetrahedron Lett. 43, 4297-4308, 1987, and Subasinghe et al., J. Med. Chem. 35 4602-7, 1992, each is hereby incorporated by reference).
  • Generation of the anion of the N-t-Boc-pyrrolidinone, pyrrolidinone, or oxazolidone followed by the addition of the compound 7, as shown in FIG. 25 results in a Michael addition product.
  • Deprotection with TFA then results in the free amino acids.
  • Trifluoroleucine (Tfl) and hexafluoroleucine (Hfl), may be synthesized by various methods known in the art.
  • 5′,5′,5′-trifluoro-DL-leucine may be synthesized in step-wise fashion by first diluting commercial trifluoromethyl crotonic acid with ethanol and hydrogenating it in the presence of a catalyst. Next, the mixture may be refluxed, and the ester distilled. Next, .alpha.-oximino-5′,5′,5′-trifluoroisocaproic acid may be derived by reflux and distillation, followed by recrystallization of 5′,5′,5′-trifluoro-DL-leucine.
  • (S)-5,5,5,5′,5′,5′-Hexafluoroleucine may be prepared from hexafluoroacetone and ethyl bromopyruvate in multiple steps, including a highly enantioselective reduction of the carbonyl group in an .alpha.-keto ester by bakers' yeast or by catecholborane utilizing an oxazaborolidine catalyst.
  • homoproparglyglycine HPG
  • AHA azidohomoalanine
  • Bispecific constructs of the present invention may also be made by methods known in the art. These include somatic hybridization, chemical coupling and recombinant techniques
  • Somatic hybridization involves the fusion of two hybridomas and purification of the bispecific secreted by the resulting quadromas. Two different methods have been described: (1) fusion of two established hybridomas generates a quadroma (Milstein and Cuello, 1983; Suresh et al., 1986), and (2) fusion of one established hybridoma with lymphocytes derived from a mouse immunized with a second antigen generates trioma (Nolan and Kennedy, 1990). Somatic hybridization for development of bsMAb involves methods similar to those for preparing hybridomas. However, the production and random association of two different heavy chains and two different light chains within one cell leads to the assembly of a substantial proportion of non-functional molecules. Elaborate purification techniques need to be developed to purify the bispecific with the required specificity, and this mostly precludes large scale manufacture for clinical use. Nonetheless, the present invention provides a bivalent bispecific construct as disclosed above manufactured using somatic hybridization.
  • DTNB 5,5-Dithiobis(2-nitrobenzoic acid)
  • O-PDM o-phenylenedimaleimide
  • DTNB acts to regenerate disulfide bonds between the two Fabs
  • O-PDM acts to form a thioether bond between the two Fab′.
  • the thioether bond of O-PDM could be more stable than the disulfide bond regenerated by DTNB.
  • Heterobifunctional reagents can introduce a reactive group onto a protein that will enable it to react with a second protein.
  • SPDP N-Succinimidyl-3-(2-pyridyldithio) propionate
  • bispecific antibodies can also be used to make bispecific antibodies.
  • Such bispecific antibodies derived by genetic engineering offer several advantages over conventional bispecific antibodies made by chemical cross-linking or fusion of two hybridoma clones, including greater control over the size, and affinity of the bispecific.
  • By using only the variable domains as building blocks, recombinant antibodies lack the Fc-region of an antibody, and thus do not induce Fc-mediated immune effector function.
  • a wide variety of different recombinant bispecific antibody formats have been developed over the past years. Amongst them tandem single-chain Fv molecules and diabodies and various derivatives thereof are the most widely used formats for the construction of recombinant bispecific antibodies.
  • tandem scFv molecules represent a straightforward format simply connecting the two scFv molecules with an additional peptide linker.
  • the two scFv fragments present in these tandem scFv molecules form separate folding entities.
  • various linkers can be used to connect the two scFv fragments and linkers with a length of up to 63 residues have been reported.
  • present invention provides a bivalent bispecific construct as disclosed above manufactured using recombinant techniques as detailed above.
  • the non-natural amino acid contains an azide, cyano, nitrile oxides, alkyne, alkene, strained cyclooctyne, strained cycloalkene, cyclopropene, norbornenes or aryl, alkyl or vinyl halide, ketone, aldehyde, ketal, acetal, hydrazine, hydrazide, alkoxy amine, boronic acid, organotin, organosilicon, beta-silyl alkenyl halide, beta-silyl alkenyl sulfonates, pyrones, tetrazine, pyridazine, aryl sulfonates, thiosemicarbazide, semicarbazide, tetrazole, alpha-ketoacid group prior to linkage.
  • the non-natural amino acid may be azidohomoalanine, homopropargylglycine, homoallylglycine, p-bromophenylalanine, p-iodophenylalanine, azidophenylalanine, acetylphenylalanine or ethynylephenylalanine, amino acids containing an internal alkene such as trans-crotylalkene, serine allyl ether, allyl glycine, propargyl glycine, or vinyl glycine, pyrrolysine, N-sigma-o-azidobenzyloxycarbonyl-L-Lysine (AzZLys), N-sigma-propargyloxycarbonyl-L-Lysine, N-sigma-2-azidoethoxycarbonyl-L-Lysine, N-sigma-tert-butyloxycarbonyl-L-Lysine (Bo
  • the non-natural amino acid contains an azide, alkyne, alkene, or aryl, alkyl or vinyl halide, ketone, aldehyde, hydrazine, hydrazide, alkoxy amine, boronic acid, organotin, organosilicon group prior to linkage.
  • the non-natural amino acid may be azidohomoalanine, homopropargylglycine, homoallylglycine, p-bromophenylalanine, p-iodophenylalanine, azidophenylalanine, acetylphenylalanine or ethynylephenylalanine, amino acids containing an internal alkene such as trans-crotylalkene, serine allyl ether, allyl glycine, propargyl glycine, or vinyl glycine.
  • the reaction for coupling the first portion to the second portion is a [3+2] dipolar cycloaddition or Click reaction, a Heck reaction, a Sonogashira reaction, a Suzuki reaction, a Stille coupling, a Hiyama/Denmark reaction, olefin metathesis, a Diels-alder reaction, or a carbonyl condensation with hydrazine, hydrazide, alkoxy amine or hydroxylamine.
  • Diabodies, single-chain diabodies and tandem-scFv molecules have a molecular weight of 50-60 kDa., which can cause rapid clearance of these entities from the circulation by extravasation, proteolysis and renal elimination. Exemplary initial half-lives of these entities (t1/2 ⁇ ) are below 30 min.
  • Several approaches have been undertaken to improve the pharmacokinetics of recombinant antibodies. One approach is to increase the size of these molecules. Dimeric single-chain diabody molecules with a molecular weight of 100-115 kDa can also be generated by varying the length of the linkers connecting the variable domain.
  • bispecific antibodies to human serum albumin (HSA), HSA binding peptides, or to peptides derived from hormones that have naturally long half-lives.
  • HSA human serum albumin
  • PEG polyethylene-glycol polymers
  • PEG has several chemical properties which are desirable in a final bispecific product and solve problems endemic with scFvs. PEGylation should improve protein solubility and increase scFv stability, thereby reducing scFv aggregation and precipitation.
  • a long and flexible linker such as PEG increases the physical separation of the two antibody fragments, allowing them to refold independently from each other. This solves one of the problems that often occurs in the refolding of bispecific antigen binding domains linked by genetic fusion, (i.e. uncontrolled and undesirable cross linking between the two constituent antibodies).
  • PEG polymers are traditionally covalently linked to biomolecules through reactive sites such as lysine, cysteine and histidine residues.
  • the amount of polymer attached to the target molecule needs to be tightly controlled. Conjugation of PEG polymers to reactive sites in the protein often results in a heterogeneous mixture of PEG-modified proteins, which may result in sub-optimal stabilization and half life extension, as well as potential loss of bioactivity of the polymer-modified protein when the PEG reactive sites are important for the protein activity (e.g. they are located in or near a receptor binding site).
  • the present invention provides a solution to this problem, by engineering the constituent antibodies of the bivalent bispecific construct to include non-natural amino acids at specific locations and reacting PEG with these non-natural amino acids.
  • PEG linker provides yet more advantages to those detailed above due to the versatility of the chemical syntheses that may be used.
  • PEG can be easily functionalized to be a complementary reaction partner with any non-natural amino acid that is incorporated into the scFv proteins.
  • PEG can also be functionalized with multiple sites of conjugation which enables construction of multivalent protein hybrids.
  • the PEG functionalization can be made with homo-bifunctional or hetero-bifunctional PEG's depending on the desired conjugation chemistry.
  • the structure of PEG can be tailored for linear or branched variations, which can impact pharmacokinetics and bioactivity.
  • Bispecific scFvs may be constructed by the conjugation of two different scFv antigen binding domains to each other by way of a linker. This strategy may be realized in a two-step process in which each scFv is conjugated to the bifunctional linker.
  • the two scFvs, comprising the bispecific conjugate each contain at least one non-natural amino acid (e.g. Aha) at a position which serves as a specific site of conjugation.
  • the linker can be homo-bifunctional or hetero-bifunctional and contain a complimentary functional group (e.g. alkyne) that is reactive with the non-natural amino acid contained in the scFv (Aha).
  • the following reaction scheme can then be applied by to generate bispecific scFv (Scheme 1 below).
  • scFv-PEG conjugates The chemistry used to conjugate scFvs to the linker is orthogonal to the 20 natural amino acids.
  • Azide-alkyne copper mediated cycloaddition is used here, in the preparation of scFv-PEG conjugates and bispecifics.
  • an scFv containing azidohomoalanine (Aha) is reacted with an excess amount of a homo-bifunctional PEG linker functionalized with alkynes.
  • the predominant product at limiting excess of PEG is a monovalent PEGylated scFv, which is then purified.
  • the free pendant alkyne of the PEG linker undergoes a second copper mediated azide-alkyne cycloaddition with a second scFv containing Aha to afford the bispecific.
  • Azide-alkyne copper mediated cycloadditions (Meldal and Torn ⁇ e, 2008, Kolb et al 2001), as well as alkene-aryl halide palladium mediated Heck reactions, have been extensively applied to the site specific conjugation of polymers, toxins or peptides to target proteins.
  • the copper mediated cycloaddition reaction is completely orthogonal with all natural amino acids, such that this chemistry cannot be used to modify biological molecules, unless a non natural azide or alkyne containing moiety can be introduced. When this is done, the chemistry occurs only at the position of that azide or alkyne.
  • Azides and alkynes can be introduced into proteins as analogs of natural amino acids, providing a specific position for bioconjugation.
  • anti-IL-6 and anti-IL-23 or derivatives thereof may optionally be modified through PEGylation to increase half life.
  • PEGylation of anti-IL-6 and anti-IL-23 or derivatives thereof may be achieved through similar methods.
  • PEG groups and PEG linkers of use in bispecific constructs and antibodies of the invention have weight 2-100 kDa for example 5-60 kDa e.g. 10-40 kDa such as around 20 or around 40 kDa.
  • PEG groups and linkers may be straight chain or branched.
  • the invention provides pharmaceutical compositions and kits comprising the bivalent, bispecific constructs of the invention and a pharmaceutically acceptable carrier.
  • the pharmaceutical composition or kit further comprises another component, such as an imaging reagent or therapeutic agent.
  • the pharmaceutical composition or kit is used in diagnostic or therapeutic methods.
  • compositions may for example be aqueous formulations e.g. aqueous solutions comprising conventional excipients such as sodium chloride, sugars, amino acids, surfactants and the like.
  • compositions may also be lyophilized products suitable for reconstitution by addition of water or saline.
  • the invention provides for the use of the bivalent, bispecific constructs of the invention in therapy.
  • the present invention provides for the treatment of T H 17, T H 22, and T H 1 mediated diseases, as well as diseases mediated by combinations of these T H cells.
  • inflammatory and autoimmune disorders such as multiple sclerosis, psoriasis, psoriatic arthritis, pemphigus vulgaris, organ transplant rejection, Crohn's disease, inflammatory bowel disease (IBD), irritable bowel syndrome (IBS), lupus erythematosis, and diabetes.
  • T H 17 mediated diseases include Amyotrophic lateral sclerosis or ALS (Lou Gehrig's disease), Ankylosing spondylitis, Asperger's, Back pain, Barrett's esophagus, Bipolar disorder, Cardiac arrhythmia, Celiac disease, Chronic fatigue syndrome (CFS/CFIDS/, E), Chronic Lyme disease (borreliosis), Crohn's disease, Diabetes insipidus, Diabetes type I, Diabetes type II, Dementia, Depression, Epilepsy, Fibromyalgia (FM), Gastroesophageal reflux disease (GERD), Hashimoto's thyroiditis, Irritable Bowel Syndrome (IBS), Interstitial cystitis (IC), Inflammatory bowel disease, Irritable bowel syndrome, Kidney stones, Löfgren's syndrome, Lupus erythematosis, Mania, Multiple Chemical Sensitivity (MCS), Migraine headache, Morgellon's, Multiple s, Multiple
  • Further diseases include cytokine storm from sepsis or hemorrhagic fever, biliary cirrhosis, Still's Disease, COPD, Grave's Opthalmopathy, perionditis, Behcet Disease, asthma, atopic dermatitis, Hidradenitis suppurativa, Giant cell arteritis and cardiac fibrosis.
  • T H 22 mediated diseases include chronic inflammatory diseases such as eczema, scleroderma, asthma and COPD.
  • the present invention provides a method of treatment of T H 17 mediated diseases comprising administering a therapeutically effective amount of the bivalent bispecific construct of the present invention to a patient.
  • the present invention provides a bivalent, bispecific construct of the present invention for the treatment of diseases mediated by T H 17.
  • the present invention provides for the use of a bivalent, bispecific construct of the present invention for the manufacture of a medicament for the treatment of diseases mediated by T H 17.
  • the present invention provides a method of treatment of diseases mediated by T H 22 cells comprising administering a therapeutically effective amount of the bivalent, bispecific construct of the present invention to a patient.
  • the present invention provides a bivalent, bispecific construct of the present invention for the treatment of diseases mediated by T H 22 cells.
  • the present invention provides for the use of a bivalent, bispecific construct of the present invention for the manufacture of a medicament for the treatment of diseases mediated by both T H 22 cells.
  • the present invention provides a method of treatment of diseases mediated by both T H 17 and T H 1 cells comprising administering a therapeutically effective amount of the bivalent, bispecific construct of the present invention to a patient
  • the present invention provides a bivalent, bispecific construct of the present invention for the treatment of diseases mediated by both T H 17 and T H 1.
  • the present invention provides for the use of a bivalent, bispecific construct of the present invention for the manufacture of a medicament for the treatment of diseases mediated by both T H 17 and T H 1.
  • the present invention provides a method of treatment of T H 17 mediated diseases comprising administering a therapeutically effective amount of a combination of anti-IL-6 and anti-IL-23 antibodies or derivatives thereof according to the present invention to a patient.
  • the present invention provides a combination of anti-IL-6 and anti-IL-23 antibodies or derivatives thereof according to the present invention for the treatment of T H 17 mediated diseases.
  • the present invention provides for the use of a combination of anti-IL-6 and anti-IL-23 antibodies or derivatives thereof according to the present invention for the manufacture of a medicament for the treatment of T H 17 mediated diseases.
  • the present invention provides a method of treatment of diseases mediated by T H 22 cells comprising administering a therapeutically effective amount of a combination of anti-IL-6 and anti IL-23 antibodies or derivatives thereof according to the present invention to a patient.
  • the present invention provides a combination of anti-IL-6 and anti-IL-23 antibodies or derivatives thereof according to the present invention for the treatment of diseases mediated by T H 22 cells.
  • the present invention provides for the use of a combination of anti-IL-6 and anti-IL-23 antibodies or derivatives thereof according to the present invention for the manufacture of a medicament for the treatment of diseases mediated by T H 22 cells.
  • the present invention provides a method of treatment of diseases mediated by both T H 17 and T H 1 comprising administering a therapeutically effective amount of a combination of anti-IL-6 and anti-IL-23 antibodies or derivatives thereof according to the present invention to a patient.
  • the present invention provides a combination of anti-IL-6 and anti-IL-23 antibodies or derivatives thereof according to the present invention for the treatment of diseases mediated by both T H 17 and T H 1.
  • the present invention provides for the use of a combination of anti-IL-6 and anti-IL-23 antibodies or derivatives thereof according to the present invention for the manufacture of a medicament for the treatment of diseases mediated by both T H 17 and T H 1.
  • the invention also provides methods of treatment of diseases that have both a T H 17 and T H 22 component to their aetiology. Additionally, the aetiology of the diseases to be treated according to the present invention may involve all three of T H 17, T H 22 and T H 1 cells.
  • the anti-IL-23 antibody, or derivative thereof may be an anti-II-23/IL-12 antibody
  • the bivalent bispecific constructs, antibodies, and antibody combinations for use in therapy according to the present invention may be administered to patients at advantageously low doses whilst still achieving the same therapeutic effect , as compared to therapies currently available in the art.
  • the lower doses are facilitated by higher activity of the antibodies disclosed herein, and potentially reduce the incidence of side effects.
  • bivalent bispecific constructs, antibodies, and antibody combinations for use in therapy according to the present invention may be administered to patients at equivalent or higher doses as compared to therapies currently available in the art. Such higher dosages may facilitate reduced frequency of administering bivalent bispecific constructs, antibodies, and antibody combinations to a patient
  • bivalent bispecific constructs, antibodies, and antibody combinations of the present invention may be administered monthly, bi-monthly, weekly, bi-weekly, daily, bi-daily.
  • Antibody affinities may be determined using methods well known to the person skilled in the art.
  • Equilibrium dissociation constants may be determined by surface Plasmon resonance using a SensiQ Pioneer (ICx Nomadics, Stillwater, Okla.) and a carboxylated COOH1 sensor (Ibid) amenable for amine coupling.
  • Protein G (6510-10, Biovision, Mountain View, Calif.) is coupled to the COOH1 sensor using amine coupling reagents (Sigma Aldrich (N-Hydroxysuccinimide (NHS, 56480), N-(3-Dimethylaminopropyl)-B′-ethylcarbodiimide hydrochloride (EDC, E7750), Ethanolamine (398136), St. Louis, Mo.) or with the Biacore Amine Coupling Kit (BR-1000-50, GE Healthcare, Waukesha, Wis.).
  • amine coupling reagents Sigma Aldrich (N-Hydroxysuccinimide (NHS, 56480), N-(3-Dimethylaminopropyl)-B′-ethylcarbodiimide hydrochloride (EDC, E7750), Ethanolamine (398136), St. Louis, Mo.) or with the Biacore Amine Coupling Kit (BR-1000-50,
  • the carboxylated surface is activated with 2 mM EDC and 0.5 mM NHS for a contact time ranging between 2-10 minutes.
  • Protein G in variable concentrations ranging between 20-400 ug/mL, is diluted into 10 mM acetate buffer, pH 4.3 (sodium acetate, BP334-1; glacial acetic acid, A490-212; Thermo Fisher Scientific, Waltham, Mass.), and injected over the activated sensor for variable contact times ranging between 5 and 10 minutes at a rate ranging from 5-10 ⁇ L/min.
  • Quantities of Protein G immobilized to the COOH1 sensor chip range from 400-2000 response units (RU). Remaining activated sites should be capped with 100 ⁇ L of ethanolamine at a flow rate of 25 ⁇ L/min.
  • Equilibrium constants for rabbit human chimeric mAbs may be determined by binding the mAb to the protein G coated chip followed by binding of each analyte (IL-6 or IL-23) to its respective mAb.
  • each analyte IL-6 or IL-273
  • the surface densities of the mAbs for each analyte should be adjusted so that as analyte binding approaches saturation its corresponding RU falls between 200 and 300.
  • the on/off rates (ka/kd) and dissociation constants (KD) may be determined using the format described above along with pseudo-first-order 1:1 interaction model software (Qdat, ICx Nomadics, Stillwater, Okla.).
  • Equilibrium constants for scFvs may be determined as previously described; with the proviso that the protocol should be modified so that an epitope tagged IL-6 is captured on the chip surface and the dissociation of anti-IL-6 scFvs from IL-6 is monitored. Briefly, anti-FLAG® M2 antibody (200472, Agilent Technologies, Santa Clara, Calif.) is bound to Protein G, and then 3 ⁇ FLAG-IL-6 is captured by the anti-FLAG antibody. The anti-IL-6 scFvs should be assayed over a range of concentrations between 1 and 100 nM.
  • IL-23 binding by the bispecific was performed by first immobilizing the bispecific with IL-6 as described, at a constant density ( ⁇ 240 RU). Binding and dissociation of a recombinant human dimeric IL-23 (34-8239, eBiosciences, San Diego, Calif.) may be assayed in concentrations ranging from 3 to 25 nM using the same parameters detailed above
  • the ability of the antibodies and derivatives thereof to modulate IL-6 activity may be assayed using methods well known to the person skilled in the art. For the purposes of determining whether antibodies have the desired ability to modulate IL-6 activity that would render them potentially suitable for inclusion in the bivalent bispecific antibodies of the present invention, the following detailed assay procedure is provided, but it will be appreciated that minor variations to the methodology (e.g. in the use of different, but similar, pieces of apparatus, or different brands of common reagents) will allow for the same determination to be made.
  • An ELISA may be used to evaluate IL-6 binding.
  • Recombinant IL-6 (See Example 1) is added to an ELISA plate in 100 ⁇ l PBS at 0.25 ⁇ g/ml. Plates should be incubated 1 hour at 37C, or overnight at 4C.
  • 100 ⁇ l/well PBS containing 10% goat serum (Cat #16210-072, Invitrogen, USA) should be added to each well. Plates should then be incubated 1 hour at room temperature. Plates should then be rinsed 5 times with de-ionized water.
  • To each well is added 50 ⁇ l PBS/10% goat serum. Test samples are then added at 50 ⁇ l/well. Plates should then be incubated 1 hour at room temperature.
  • Plates should then be rinsed 5 times with de-ionized water. To each well is added 100 ⁇ l peroxidase-conjugated goat anti-rabbit IgG (Cat. #111-035-008, Jackson Immuno Research) diluted 1:5000 in PBS/10% goat serum. Plates should then be incubated 1 hour at room temperature, then washed 5 times with de-ionized water. TMB substrate (Thermo Scientific, Rockford, Ill., USA) is added at 100 ⁇ l/well. The reaction should then be stopped with 100 ⁇ l 1N H2SO4 (JT Baker, Phillipsburg, N.J., USA). Absorbance can then be measured at 450 nm using a Molecular Devices M2 plate reader.
  • a bioassay using an IL-6 dependant murine B-cell hybridoma cell line may be used to evaluate IL-6 inhibition ( FIG. 3 ).
  • Samples to be tested for neutralizing activity should be diluted in 100 ⁇ l assay medium (RPMI 1640 w/L-glutamine, 10% FBS, Non-Essential Amino Acids, Sodium Pyruvate, 50 ⁇ M 2-mercaptoethanol) in a 96-well tissue culture plate. This is followed by the addition of 50 ⁇ l of IL-6 (Cat. # CYT-274 Prospec-Tany Technogene) containing assay medium, and 30 minutes of incubation at room temperature.
  • assay medium RPMI 1640 w/L-glutamine, 10% FBS, Non-Essential Amino Acids, Sodium Pyruvate, 50 ⁇ M 2-mercaptoethanol
  • B9 cells are then recovered from flasks and centrifuged for 7 min at 180 ⁇ g, and the pellet resuspended in IL-6-free culture medium (RPMI 1640 w/L-glutamine, 10% FBS, Non-Essential Amino Acids, Sodium Pyruvate, 50 ⁇ M 2-mercaptoethanol). Cells should be centrifuged and resuspended three times to remove IL-6. Following viability determination by trypan blue exclusion, cells should be adjusted to 1 ⁇ 105 cells/ml. A volume of 50 ⁇ l of B9 cells, corresponding to 5 ⁇ 10 3 cells, should be added to each well along with appropriate control wells containing IL-6-free medium.
  • IL-6-free culture medium RPMI 1640 w/L-glutamine, 10% FBS, Non-Essential Amino Acids, Sodium Pyruvate, 50 ⁇ M 2-mercaptoethanol. Cells should be centrifuged and resuspended three times to remove
  • the plates should then be incubated for 48 h at 37° C. 5% CO2. Subsequently, 20 ⁇ l of Alamar Blue (Cat # DAL1100, Invitrogen, USA) should be added to each well, and the plates incubated for an additional 18 h. The plates can then be read on a Molecular Devices (Sunnyvale, Calif., USA) M2 plate reader at 570 and 600 nm.
  • Alamar Blue Cat # DAL1100, Invitrogen, USA
  • An ELISA assay may be used to evaluate IL-23 binding (Aggarwal et al., 2003). ELISA plates are coated using either a direct or indirect method of binding IL-23.
  • anti-His antibody (Cat # A00613, GenScript Corp., New Jersey, USA) should be added to the plates in 100 ml/well of PBS at 0.01-0.02 ug/ml. Plates should then be incubated for 1 hour at 37C, or overnight at 4C.
  • 100 ml/well PBS containing 10% goat serum (Cat #16210-072, Invitrogen, USA) should be added to each well, after which plates should be rinsed 5 times with de-ionized water.
  • IL-23 p40-p19-His (SEQ ID 4) in 100 ml/well PBS/10% goat serum at 0.5 mg/ml should be added and the plates incubated for 1 hour at room temperature.
  • IL-23 p40-p19-His (SEQ ID NO. 4) should be added to an ELISA plate in 100 ml PBS at 0.5 mg/ml. Plates should then be incubated for 1 hour at 37C, or overnight at 4C. To block non-specific binding 100 ml/well PBS containing 10% goat serum (Cat #16210-072, Invitrogen, USA) should be added to each well. Plates should then be incubated for 1 hour at room temperature.
  • plates should be rinsed 5 times with de-ionized water.
  • To each well should be added 50 ml PBS/10% goat serum. Test samples should then be added at 50 ml/well. Plates should then be incubated for 1 hour at room temperature and rinsed 5 times with de-ionized water.
  • To each well should then be added 100 ml peroxidase-conjugated goat anti-rabbit IgG (Cat. #111-035-008, Jackson Immuno Research) diluted 1:5000 in PBS/10% goat serum. Plates should then be incubated 1 hour at room temperature, then washed 5 times with de-ionized water.
  • TMB substrate (Thermo Scientific, Rockford, Ill., USA) should be added at 100 ml /well. The reaction should then be stopped with 100 ml 1N H2SO4 (JT Baker, Phillipsburg, N.J., USA). Absorbance can then be measured at 450 nm using a Molecular Devices M2 plate reader.
  • a bioassay based on the detection of IL-23-induced IL-17 expression by mouse spleen cells, may be used to detect antibody mediated inhibition of IL-23 binding to the IL-23 receptor and resulting bioactivity.
  • 5 ⁇ 10 5 C57Bl/6 spleen cells should be cultured in the wells of a 96-well plate in 200 ml containing a dilution of the heterodimeric IL-23 (eBioscience cat. #14-8239 or Humanzyme, Chicago, USA cat. #HZ-1049) and the plates incubated for 2-3 days at 37° C.
  • the culture medium used should be RPMI 1640, 10% FBS, 50 uM 2-mercaptoethanol, non-Essential Amino Acids, pyruvate, gentamicin and 10 ng/ml human IL-2 (Cat # CYT-209, Prospec-Tany Technogene). After 3 days, the culture supernatants should be assayed by ELISA for IL-17A, as described below.
  • An ELISA assay may be used used to detect mouse IL-17. Plates are coated with anti-mlL-17A (eBioscience #14-7178) 1 mg/ml in 100 ml PBS, and incubated overnight at 4° C. or 1 hr at 37° C. Plates should be washed in deionized water and blocked for 1 h with 100 ml of PBS, 10% goat serum. After washing the plates, 50 ml of PBS/10% goat serum and 50 ml of culture supernatant should be added to the plates, and incubated for 1 hr.
  • anti-mlL-17A eBioscience #14-7178
  • the plates should then be washed, 100 ml/well of anti-mlL-17A-Biotin (eBioscience #13-7179) at 0.5 mg/ml in PBS/10% goat serum added and the plates incubated for 1 h at room temperature.
  • the plates should then be washed, and reacted with 100 ml/well Streptavidin-HRP (Jackson Labs) at 1:1000 in PBS/10% goat serum. Plates should be washed again, and the signal detected by adding 100 ml/well TMB substrate (Thermo Scientific, IL, USA). After stopping the reaction with 100 ml/well 1N H2SO4, the optical density can be read at 450 nM.
  • an assay to measure their neutralizing capacity against IL-12 utilizes their ability to modulate the level of IFN- ⁇ (a product of T H 1 cell activity) are disclosed herein.
  • the person skilled in the art will be aware of suitable assay methods to determine the neutralizing effect of the antibodies and their effect on IFN- ⁇ production, however, the following assay is provided as an example of a suitable assay.
  • Antibodies may be assayed for p40 neutralizing capacity using the IL-12 responsive cell line NK-92 (CRL-2407, ATCC, Manassas, Va., USA). 50 ml of culture supernatant from the B cell cloning plates or 50 ml of supernatant from antibody transfection should be transferred to a 96 well tissue culture plate. 50 ml of human IL-12 (Cat. # Cyt-362, Prospec-Tany Technogene, Rehovot, Israel) should be added to each well at 4 ng/ml. Plates should then be incubated for 30-60 minutes at room temperature, after which 5 ⁇ 10 4 NK-92 cells should be added to each well in 100 ml.
  • the assay medium should be RPMI 1640, 10% FBS, NEAA, pyruvate, 50 mM 2-mercaptoethanol, gentamicin and 10 ng/ml human IL-2 (Cat # Z00368, GeneScript Corporation, Piscataway, N.J., USA).
  • An ELISA assay may be used to detect human Interferon- ⁇ . Plates are coated with anti-human Interferon-g (Cat. # Mab 1-D1K, Mabtech, Cincinnati, Ohio, USA) 1 mg/ml in 100 ml PBS, overnight @4° C. or 1 hr @ 37° C. Plates should then be washed in de-ionized water and blocked for 1 h with 100 ⁇ l of PBS, 10% goat serum. After washing the plates, 50 ml of PBS/10% goat serum and 50 ml of culture supernatant were added to the plates, and incubated for 1 hr.
  • anti-human Interferon-g Cat. # Mab 1-D1K, Mabtech, Cincinnati, Ohio, USA
  • the plates should then be washed, 100 ml/well of anti-human Interferon- ⁇ -Biotin (Cat # Mab 7b6-1-biotin, Mabtech) at 0.5 mg/ml in PBS/10% goat serum added and the plates then incubated for 1 h at room temperature.
  • the plates should then be washed and reacted with 100 ml/well Streptavidin-HRP (Jackson Labs) at 1:1000 in PBS/10% goat serum. Plates should then be washed again, and the signal detected by adding 100 ml/well TMB substrate (Thermo Scientific, IL, USA). After stopping the reaction with 100 ml/well 1N H2SO4, the optical density can be read at 450 nM.
  • the assays against primate interleukins are identical to those for the measurement of activity against the human assays, save for the use of the primate version of the cytokine being assayed.
  • cytokine antagonists for human therapy will require initial toxicology testing. Toxicology is most efficiently demonstrated in non human species. In order to facilitate initial toxicology studies the antibodies of the present invention may be screened for their ability to neutralize IL-6 from the species being considered for the studies.
  • immunoglobulin is a tetrameric molecule.
  • each tetramer is composed of two identical pairs of polypeptide chains, each pair having one“light” (about 25 kDa) and one “heavy” chain (about 50-70 kDa).
  • the amino-terminal portion of each chain includes a variable region of about 100 to 110 or more amino acids primarily responsible for antigen recognition.
  • the carboxy-terminal portion of each chain defines a constant region primarily responsible for effector function.
  • Human light chains are classified as K and X light chains.
  • Heavy chains are classified as, a, or E, and define the antibody's isotype as IgM, IgD, IgG, IgA, and IgE, respectively.
  • the variable and constant regions are joined by a “J” region of about 12 or more amino acids, with the heavy chain also including a “D” region of about 10 more amino acids. See generally, Fundamental Immunology Ch. 7 (Paul, W., ed., 2nd ed. Raven Press, N.Y. (1989)) (incorporated by reference in its entirety for all purposes).
  • the variable regions of each light/heavy chain pair form the antibody binding site such that an intact immunoglobulin has two binding sites.
  • Immunoglobulin chains exhibit the same general structure of relatively conserved framework regions (FR) joined by three hypervariable regions, also called complementarity determining regions or CDRs.
  • the CDRs from the two chains of each pair are aligned by the framework regions, enabling binding to a specific epitope.
  • FR1, CDR1, FR2, CDR2, FR3, CDR3 and FR4 From N-terminus to C-terminus, both light and heavy chains comprise the domains FR1, CDR1, FR2, CDR2, FR3, CDR3 and FR4.
  • the assignment of amino acids to each domain is in accordance with the definitions of Kabat Sequences of Proteins of Immunological Interest (National Institutes of Health, Bethesda, Md. (1987 and 1991)), or Chothia & Lesk J. Mol. Biol. 196: 901-917 (1987); Chothia et al. Nature 342: 878-883 (1989).
  • an “antibody” refers to an intact immunoglobulin, or to an antigen-binding portion thereof that competes with the intact antibody for specific binding.
  • Antigen-binding portions may be produced by recombinant DNA techniques or by enzymatic or chemical cleavage of intact antibodies.
  • Antigen-binding portions include, inter alia, Fab, Fab′, F (ab′)2, Fv, dAb, and complementarity determining region (CDR) fragments, single-chain antibodies (scFv), chimeric antibodies, diabodies and polypeptides that contain at least a portion of an immunoglobulin that is sufficient to confer specific antigen binding to the polypeptide.
  • An Fab fragment is a monovalent fragment consisting of the VL, VH, CL and CH I domains;
  • a F (ab′) 2 fragment is a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region;
  • a Fd fragment consists of the VH and CH 1 domains;
  • an Fv fragment consists of the VL and VH domains of a single arm of an antibody;
  • a dAb fragment (Ward et al., Nature 341: 544-546, 1989) consists of a VH domain.
  • a single-chain antibody is an antibody in which a VL and VH regions are paired to form a monovalent molecules via a synthetic linker that enables them to be made as a single protein chain (Bird et al., Science 242: 423-426, 1988 and Huston et al., Proc. Natl. Acad. Sci. USA 85: 5879-5883, 1988).
  • An antibody may have one or more binding sites. If there is more than one binding site, the binding sites may be identical to one another or may be different. For instance, a naturally-occurring immunoglobulin has two identical binding sites, a single-chain antibody or Fab fragment has one binding site, while a “bispecific” antibody has two different binding sites. Bispecific antibodies can be produced by a variety of methods including fusion of hybridomas or linking of Fab′ fragments. See, e.g., Songsivilai & Lachmann Clin. Exp. Immunol. 79: 315-321 (1990), Kostelny et al. J. Immunol. 148: 1547-1553 (1992).
  • isolated antibody is an antibody that (1) is not associated with naturally-associated components, including other naturally-associated antibodies, that accompany it in its native state, (2) is free of other proteins from the same species, (3) is expressed by a cell from a different species, or (4) does not occur in nature.
  • isolated antibodies include an anti-IL-6 antibody that has been affinity purified using IL-6, an anti-IL-6 antibody that has been synthesized by a hybridoma or other cell line in vitro, and a human anti-IL-6 antibody derived from a transgenic mouse.
  • human antibody includes all antibodies that have one or more variable and/or constant regions derived from human immunoglobulin sequences. These antibodies may be prepared in a variety of ways, by way of example two are described below.
  • a humanized antibody is an antibody that is derived from a non-human species, in which certain amino acids in the framework and constant domains of the heavy and light chains have been mutated so as to avoid or abrogate an immune response in humans.
  • a humanized antibody may be produced by fusing the constant domains from a human antibody to the variable domains of a non-human species. Examples of how to make humanized antibodies may be found in U.S. Pat. Nos. 6,054,297, 5,886,152 and 5,877,293.
  • chimeric antibody refers to an antibody that contains one or more regions from one antibody and one or more regions from one or more other antibodies.
  • Koff refers to the off rate constant for dissociation of an antibody from the antibody/antigen complex.
  • Kd refers to the dissociation constant of a particular antibody-antigen interaction.
  • epitopic determinants includes any protein determinant capable of specific binding to an immunoglobulin or T-cell receptor.
  • Epitopic determinants usually consist of chemically active surface groupings of molecules such as amino acids or sugar side chains and usually have specific three dimensional structural characteristics, as well as specific charge characteristics.
  • An antibody is said to specifically bind an antigen when the dissociation constant is, preferably less than 10 nM and most preferably 0 nM.
  • Preferred amino- and carboxy-termini of fragments or analogs occur near boundaries of functional domains.
  • Structural and functional domains can be identified by comparison of the nucleotide and/or amino acid sequence data to public or proprietary sequence databases.
  • IL-12 is a heterodimer consisting of two subunits, p35 and p40, linked by a disulfide bond.
  • Antigen presenting cells primarily of the myeloid lineage, express IL-12, which participates in cell-mediated immunity by binding to a receptor complex expressed on the surface of T cells or natural killer cells. It is believed that the p40 subunit of IL-12 binds the IL-12 receptor beta 1 (IL-12R ⁇ 1) receptor and the p35 subunit binds to the second receptor chain (IL-12R ⁇ 2), resulting in intracellular signaling.
  • IL-23 is a heterodimer, consisting of the same p40 protein subunit of IL-12, covalently linked to a p19 protein. IL-23 binds to a receptor related to the IL-12R, that shares the IL-12R ⁇ 1 chain and also has a unique IL-23R chain.
  • IL-6 is a pleiotropic cytokine with various biological activities in immune regulation including hematopoiesis, inflammation, and oncogenesis. IL-6 activates a receptor complex consisting of the IL-6 receptor (IL-6R) and the signal-transducing receptor subunit gp130. IL-6R exists in both a transmembrane form and a soluble form. IL-6 binds to both of these forms, which can then interact with gp130 to trigger downstream signal transduction and gene expression.
  • IL-6R IL-6 receptor
  • gp130 the signal-transducing receptor subunit
  • T H 1 cells are T regulatory cells (also known as T helper cells) involved in mammalian immune responses. They are characterized by the production of IFN- ⁇ .
  • T H 17 cells are T regulatory cells (also known as T helper cells) involved in mammalian immune responses. They are characterized by the production of IL-17.
  • T H 17 mediated diseases are diseases in which T H 17 cells play a role in the aetiology of the disease.
  • T H 22 cells are T regulatory cells (also known as T helper cells) involved in mammalian immune responses. They are characterized by the production of IL-22.
  • T H 22 mediated diseases are diseases in which T H 22 cells play a role in the aetiology of the disease.
  • computerized comparison methods are used to identify sequence motifs or predicted protein conformation domains that occur in other proteins of known structure and/or function. Methods to identify protein sequences that fold into a known three-dimensional structure are known. Bowie et al. Science 253: 164 (1991).
  • Preferred amino acid substitutions are those which: (1) reduce susceptibility to proteolysis, (2) reduce susceptibility to oxidation, (3) alter binding affinity for forming protein complexes, (4) alter binding affinities, and (4) confer or modify other physicochemical or functional properties of such analogs.
  • Analogs can include various muteins of a sequence other than the naturally-occurring peptide sequence. For example, single or multiple amino acid substitutions (preferably conservative amino acid substitutions) may be made in the naturally-occurring sequence (preferably in the portion of the polypeptide outside the domain (s) forming intermolecular contacts.
  • a conservative amino acid substitution should not substantially change the structural characteristics of the parent sequence (e.g., a replacement amino acid should not tend to break a helix that occurs in the parent sequence, or disrupt other types of secondary structure that characterizes the parent sequence).
  • Examples of art-recognized polypeptide secondary and tertiary structures are described in Proteins, Structures and Molecular Principles (Creighton, Ed., W. H. Freeman and Company, New York (1984)); Introduction to Protein Structure (C. Branden and J. Tooze, eds., Garland Publishing, New York, N.Y. (1991)); and Thornton et al. Nature 354: 105 (1991), which are each incorporated herein by reference.
  • Examples of unconventional amino acids include: 4-hydroxyproline, -carboxyglutamate, s-N,N,N-trimethyllysine, s-N-acetyllysine, O-phosphoserine, N-acetylserine, N-formylmethionine, 3-methylhistidine, 5-hydroxylysine, s-N-methylarginine, and other similar amino acids and imino acids (e.g., 4-hydroxyproline).
  • the lefthand direction is the amino terminal direction and the righthand direction is the carboxy-terminal direction, in accordance with standard usage and convention.
  • polynucleotide as referred to herein means a polymeric form of nucleotides of at least 10 bases in length, either ribonucleotides or deoxynucleotides or a modified form of either type of nucleotide.
  • the term includes single and double stranded forms of DNA.
  • isolated polynucleotide shall mean a polynucleotide of genomic, cDNA, or synthetic origin or some combination thereof, which by virtue of its origin the“isolated polynucleotide” (1) is not associated with all or a portion of a polynucleotide in which the“isolated polynucleotide” is found in nature, (2) is operably linked to a polynucleotide which it is not linked to in nature, or (3) does not occur in nature as part of a larger sequence.
  • oligonucleotide includes naturally occurring, and modified nucleotides linked together by naturally occurring, and non-naturally occurring oligonucleotide linkages.
  • Oligonucleotides are a polynucleotide subset generally comprising a length of 200 bases or fewer. Preferably oligonucleotides are 10 to 60 bases in length and most preferably 12, 13, 14, 15, 16, 17, 18, 19, or 20 to 40 bases in length. Oligonucleotides are usually single stranded, e.g. for probes; although oligonucleotides may be double stranded, e.g. for use in the construction of a gene mutant. Oligonucleotides of the invention can be either sense or antisense oligonucleotides.
  • nucleotides includes deoxyribonucleotides and ribonucleotides.
  • modified nucleotides includes nucleotides with modified or substituted sugar groups and the like.
  • oligonucleotide linkages includes oligonucleotides linkages such as phosphorothioate, phosphorodithioate, phosphoroselenoate, phosphorodiselenoate, phosphoroanilothioate, phoshoraniladate, phosphoroamidate, and the like. See e.g., LaPlanche et al. Nucl. Acids Res.
  • oligonucleotide can include a label for detection, if desired.
  • the left hand end of single-stranded polynucleotide sequences is the 5′ end; the left hand direction of double-stranded polynucleotide sequences is referred to as the 5′ direction.
  • the direction of 5′ to 3′ addition of nascent RNA transcripts is referred to as the transcription direction; sequence regions on the DNA strand having the same sequence as the RNA and which are 5′ to the 5′ end of the RNA transcript are referred to as “upstream sequences”; sequence regions on the DNA strand having the same sequence as the RNA and which are 3′ to the 3′ end of the RNA transcript are referred to as“downstream sequences”.
  • “Operably linked” sequences include both expression control sequences that are contiguous with the gene of interest and expression control sequences that act in trans or at a distance to control the gene of interest.
  • expression control sequence refers to polynucleotide sequences which are necessary to effect the expression and processing of coding sequences to which they are ligated. Expression control sequences include appropriate transcription initiation, termination, promoter and enhancer sequences; efficient RNA processing signals such as splicing and polyadenylation signals; sequences that stabilize cytoplasmic mRNA ; sequences that enhance translation efficiency (i.e., Kozak consensus sequence); sequences that enhance protein stability; and when desired, sequences that enhance protein secretion.
  • control sequences differs depending upon the host organism; in prokaryotes, such control sequences generally include promoter, ribosomal binding site, and transcription termination sequence; in eukaryotes, generally, such control sequences include promoters and transcription termination sequence.
  • control sequences is intended to include, at a minimum, all components whose presence is essential for expression and processing, and can also include additional components whose presence is advantageous, for example, leader sequences and fusion partner sequences.
  • vector is intended to refer to a nucleic acid molecule capable of transporting another nucleic acid to which it has been linked.
  • plasmid refers to a circular double stranded DNA loop into which additional DNA segments may be ligated.
  • viral vector Another type of vector is a viral vector, wherein additional DNA segments may be ligated into the viral genome.
  • Certain vectors are capable of autonomous replication in a host cell into which they are introduced (e.g., bacterial vectors having a bacterial origin of replication and episomal mammalian vectors).
  • Other vectors e.g., non-episomal mammalian vectors
  • vectors are capable of directing the expression of genes to which they are operatively linked. Such vectors are referred to herein as “recombinant expression vectors” (or simply, “expression vectors”).
  • expression vectors of utility in recombinant DNA techniques are often in the form of plasmids.
  • plasmid and vector may be used interchangeably as the plasmid is the most commonly used form of vector.
  • the invention is intended to include such other forms of expression vectors, such as viral vectors (e.g., replication defective retroviruses, adenoviruses and adeno-associated viruses), which serve equivalent functions.
  • recombinant host cell (or simply “host cell”), as used herein, is intended to refer to a cell into which a recombinant expression vector has been introduced. It should be understood that such terms are intended to refer not only to the particular subject cell but to the progeny of such a cell. Because certain modifications may occur in succeeding generations due to either mutation or environmental influences, such progeny may not, in fact, be identical to the parent cell, but are still included within the scope of the term “host cell” as used herein.
  • selectively hybridize means to detectably and specifically bind.
  • Polynucleotides, oligonucleotides and fragments thereof in accordance with the invention selectively hybridize to nucleic acid strands under hybridization and wash conditions that minimize appreciable amounts of detectable binding to nonspecific nucleic acids.
  • “High stringency” or “highly stringent” conditions can be used to achieve selective hybridization conditions as known in the art and discussed herein.
  • high stringency or “highly stringent” conditions is a method of incubating a polynucleotide with another polynucleotide, wherein one polynucleotide may be affixed to a solid surface such as a membrane, in a hybridization buffer of 6 ⁇ SSPE or SSC, 50% formamide, 5 ⁇ Denhardt's reagent, 0.5% SDS, 100 p, g/ml denatured, fragmented salmon sperm DNA at a hybridization temperature of 42 C for 12-16 hours, followed by twice washing at 55 C using a wash buffer of 1 ⁇ SSC, 0.5% SDS. See also Sambrook et al., supra, pp. 9.50-9.55.
  • Two amino acid sequences are homologous if there is a partial or complete identity between their sequences. For example, 85% homology means that 85% of the amino acids are identical when the two sequences are aligned for maximum matching. Gaps (in either of the two sequences being matched) are allowed in maximizing matching; gap lengths of 5 or less are preferred with 2 or less being more preferred. Alternatively and preferably, two protein sequences (or polypeptide sequences derived from them of at least 30 amino acids in length) are homologous, as this term is used herein, if they have an alignment score of at more than 5 (in standard deviation units) using the program ALIGN with the mutation data matrix and a gap penalty of 6 or greater. See Dayhoff, M.
  • nucleotide sequence “TATAC” corresponds to a reference sequence “TATAC” and is complementary to a reference sequence“GTATA”.
  • reference sequence is a defined sequence used as a basis for a sequence comparison; a reference sequence may be a subset of a larger sequence, for example, as a segment of a full-length cDNA or gene sequence given in a sequence listing or may comprise a complete cDNA or gene sequence. Generally, a reference sequence is at least 18 nucleotides or 6 amino acids in length, frequently at least 24 nucleotides or 8 amino acids in length, and often at least 48 nucleotides or 16 amino acids in length.
  • two polynucleotides or amino acid sequences may each (1) comprise a sequence (i.e., a portion of the complete polynucleotide or amino acid sequence) that is similar between the two molecules, and (2) may further comprise a sequence that is divergent between the two polynucleotides or amino acid sequences
  • sequence comparisons between two (or more) molecules are typically performed by comparing sequences of the two molecules over a “comparison window” to identify and compare local regions of sequence similarity.
  • a “comparison window”, as used herein, refers to a conceptual segment of at least 18 contiguous nucleotide positions or 6 amino acids wherein a polynucleotide sequence or amino acid sequence may be compared to a reference sequence of at least 18 contiguous nucleotides or 6 amino acid sequences and wherein the portion of the polynucleotide sequence in the comparison window may comprise additions, deletions, substitutions, and the like (i.e., gaps) of 20 percent or less as compared to the reference sequence (which does not comprise additions or deletions) for optimal alignment of the two sequences.
  • Optimal alignment of sequences for aligning a comparison window may be conducted by the local homology algorithm of Smith and Waterman Adv. Appl. Math.
  • sequence identity means that two polynucleotide or amino acid sequences are identical (i.e., on a nucleotide-by-nucleotide or residue-by-residue basis) over the comparison window.
  • percentage of sequence identity is calculated by comparing two optimally aligned sequences over the window of comparison, determining the number of positions at which the identical nucleic acid base (e.g., A, T, C, G, U, or I) or residue occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the comparison window (i.e., the window size), and multiplying the result by 100 to yield the percentage of sequence identity.
  • substantially identical denotes a characteristic of a polynucleotide or amino acid sequence, wherein the polynucleotide or amino acid comprises a sequence that at least 90 to 95 percent sequence identity, more preferably at least 98 percent sequence identity, more usually at least 99 percent sequence identity as compared to a reference sequence over a comparison window of at least 18 nucleotide (6 amino acid) positions, frequently over a window of at least 24-48 nucleotide (8-16 amino acid) positions, wherein the percentage of sequence identity is calculated by comparing the reference sequence to the sequence which may include deletions or additions which total 20 percent or less of the reference sequence over the comparison window.
  • the reference sequence may be a subset of a larger sequence.
  • the term “substantial identity” means that two peptide sequences, when optimally aligned, such as by the programs GAP or BESTFIT using default gap weights, share at least 80 percent sequence identity, preferably at least 90 percent sequence identity, more preferably at least 95 percent sequence identity, even more preferably at least 98 percent sequence identity and most preferably at least 99 percent sequence identity.
  • residue positions which are not identical differ by conservative amino acid substitutions.
  • Conservative amino acid substitutions refer to the interchangeability of residues having similar side chains.
  • a group of amino acids having aliphatic side chains is glycine, alanine, valine, leucine, and isoleucine
  • a group of amino acids having aliphatic-hydroxyl side chains is serine and threonine
  • a group of amino acids having amide-containing side chains is asparagine and glutamine
  • a group of amino acids having aromatic side chains is phenylalanine, tyrosine, and tryptophan
  • a group of amino acids having basic side chains is lysine, arginine, and histidine
  • a group of amino acids having sulfur-containing side chains is cysteine and methionine.
  • Preferred conservative amino acids substitution groups are: valine-leucine-isoleucine, phenylalanine-tyrosine, lysine-arginine, alanine-valine, glutamate-aspartate, and asparagine-glutamine.
  • amino acid sequences of antibodies or immunoglobulin molecules are contemplated as being encompassed by the present invention, providing that the variations in the amino acid sequence maintain at least 90%, more preferably 95%, and most preferably 99% sequence identity.
  • conservative amino acid replacements are contemplated. Conservative replacements are those that take place within a family of amino acids that are related in their side chains.
  • More preferred families are: serine and threonine are aliphatic-hydroxy family; asparagine and glutamine are an amide-containing family; alanine, valine, leucine and isoleucine are an aliphatic family; and phenylalanine, tryptophan, and tyrosine are an aromatic family.
  • serine and threonine are aliphatic-hydroxy family
  • asparagine and glutamine are an amide-containing family
  • alanine, valine, leucine and isoleucine are an aliphatic family
  • phenylalanine, tryptophan, and tyrosine are an aromatic family.
  • an isolated replacement of a leucine with an isoleucine or valine, an aspartate with a glutamate, a threonine with a serine, or a similar replacement of an amino acid with a structurally related amino acid will not have a major effect on the binding or properties of the resulting molecule, especially
  • the terms “label” or “labelled” refers to incorporation of another molecule in the antibody.
  • the label is a detectable marker, e.g., incorporation of a radiolabelled amino acid or attachment to a polypeptide of biotinyl moieties that can be detected by marked avidin (e.g., streptavidin containing a fluorescent marker or enzymatic activity that can be detected by optical or colorimetric methods).
  • the label or marker can be therapeutic, e.g. a drug conjugate or toxin.
  • Various methods of labeling polypeptides and glycoproteins are known in the art and may be used.
  • labels include, but are not limited to, the following: radioisotopes or radionuclides (e.g., 3H, 14C, 15N, 35S, 90Y, 99Tc, 111In, 125I, 131I), fluorescent labels (e.g., FITC, rhodamine, lanthanide phosphors), enzymatic labels (e.g., horseradish peroxidase, -galactosidase, luciferase, alkaline phosphatase), chemiluminescent markers, biotinyl groups, predetermined polypeptide epitopes recognized by a secondary reporter (e.g., leucine zipper pair sequences, binding sites for secondary antibodies, metal binding domains, epitope tags), magnetic agents, such as gadolinium chelates, toxins such as pertussis toxin, taxol, cytochalasin B, gramicidin D, ethidium bromide, emetine, mito
  • patient includes human and veterinary subjects.
  • Antigen binding domains are generated from hyperimmunized rabbits from cloned antigen specific B cells.
  • Antigen specific B cells are selected from rabbit blood by antigen panning and cloned under culture conditions that favor the expansion of activated B cells.
  • Human IL-6 (DQ891463; ABM82389.1, SEQ ID NOs. 343 and 344) was expressed as a 3 ⁇ FLAG-IL6-Avi fusion.
  • the expression construct was generated by a two-step polymerase chain reaction (PCR) amplification.
  • the product was inserted into the plasmid p3xFLAG-CMV-23 (Sigma), downstream of the CMV promoter and in frame with the pre-pro-trypsin leader sequence and triple FLAG tag.
  • the expressed IL-6 (SEQ ID NO. 1) contained an amino terminal triple FLAG tag (amino acid sequence dhdgdykdhdidykdddd; SEQ ID NO.
  • the IL6 coding region was amplified by PCR and inserted into p3xFLAG-CMV-23 to express 3 ⁇ FLAG-IL6 and 3 ⁇ FLAG-IL6-myc fusions. (SEQ ID NO. 2)
  • the coding region of the macaca mulatta IL-6 (mmIL-6; NM — 001042733.1; NP — 001036198; SEQ ID NO. 346 and SEQ ID NO. 347) was generated by overlapping oligomers and PCR (DNA2.0).
  • the synthesized sequence encodes mmIL-6 containing an amino terminal signal sequence (mnsfstsafgpvafslglllylpaafpap; SEQ IS NO. 349) and a carboxyl terminal FLAG tag.
  • This construct was inserted into the mammalian expression vector pCEP4 (Invitrogen) downstream of the CMV promoter (ALLO66-MM4-143). (SEQ ID NO. 3)
  • the human IL-23 cytokine is a functional heterodimer of the p19 (IL23A; accession number NT — 029419) and p40 (IL12B; NT 023133) proteins.
  • the IL-23 dimer was expressed as a p40:p19 fusion with the individual proteins separated by the elastin linker (accession number NM — 001081755; SEQ ID NO. 351) gttcctggagtaggggtacctggggtgggc encoding the amino acid sequence VPGVGVPGVG SEQ ID NO. 352).
  • a two step PCR amplification strategy was used to generate a p40:p19 genetic fusion and also introduce a 6 ⁇ HIS tag on each of the domains.
  • the resulting constructs encoding p40:p19-6 ⁇ HIS (SEQ ID NO. 4) and p40-6 ⁇ HIS: p19 were introduced into the mammalian expression plasmid pCEP4 (Invitrogen)
  • IL23 heterodimer was expressed as a p40 (NP — 001038190; NM — 001044725):p19 (BV209310) fusion with the elastin linker between the two proteins and containing a 3′ 6 ⁇ HIS tag.
  • the construct was synthesized by overlapping oligomers and PCR and cloned into the p3xFLAG-CMV-13 plasmid, downstream of the CMV promoter and in frame with the Pre-pro-trypsin signal sequence (ALLO87-MM-5-74) (SEQ ID NO. 5).
  • cytokines The expression of mammalian cytokines was performed in HEK293 or HEK293c18 cells. Transfections were performed using 3 ⁇ l/mg DNA of lipofectamine2000, or 293fectin (Invitrogen) on cells plated at a density of approx. 200,000 cells/cm 2 , and using 2-2.5 ⁇ g DNA per million cells. Cells were incubated at 37° C. for 3-4 days in DMEM containing 10% fetal calf serum and the growth medium collected for purification of the target protein.
  • Proteins bound to the Ni-NTA beads were eluted by elution buffer (50 mM Phosphate buffer pH7.5, 300 mM NaCl, 500 mM imidazole). Fractions containing the target protein were identified by SDS-PAGE and Coomassie staining and quantified by densitometry. Peak fractions were combined and dialysed to PBS and used directly in in vitro assays or SPR analyses.
  • elution buffer 50 mM Phosphate buffer pH7.5, 300 mM NaCl, 500 mM imidazole.
  • FLAG tagged proteins were purified from the expression medium using M2-conjugated beads (Invitrogen). In short, M2 beads were added directly to the expression medium and incubated at 4° C. for 4-16 hours. The beads were washed in FLAG wash buffer (20 mM Tris, pH7.4, 150 mM NaCl, 0.1% Tween-20, 1 mM ethylenediaminetetraacetic acid) and bound protein collected by washes with 0.1M glycine (pH2.5) or using 3 ⁇ FLAG peptide. Acid elutions were immediately neutralized using 1/20 th volume of 1 M Tris base. Peak fractions were identified by SDS-PAGE and Coomassie staining and pooled. These were dialysed to PBS and used directly for in vitro assays or SPR analyses.
  • FLAG wash buffer 20 mM Tris, pH7.4, 150 mM NaCl, 0.1% Tween-20, 1 mM ethylenediaminetetraacetic acid
  • Equilibrium dissociation constants were determined by surface Plasmon resonance using a SensiQ Pioneer (ICx Nomadics, Stillwater, Okla.) and a carboxylated COOH1 sensor (Ibid) amenable for amine coupling.
  • Protein G (6510-10, Biovision, Mountain View, Calif.) was coupled to the COOH1 sensor using amine coupling reagents (Sigma Aldrich (N-Hydroxysuccinimide (NHS, 56480), N-(3-Dimethylaminopropyl)-B′-ethylcarbodiimide hydrochloride (EDC, E7750), Ethanolamine (398136), St. Louis, Mo.) or with the Biacore Amine Coupling Kit (BR-1000-50, GE Healthcare, Waukesha, Wis.).
  • the carboxylated surface is activated with 2 mM EDC and 0.5 mM NHS for a contact time ranging between 2-10 minutes.
  • Protein G in variable concentrations ranging between 20-400 ug/mL, was diluted into 10 mM acetate buffer, pH 4.3 (sodium acetate, BP334-1; glacial acetic acid, A490-212; Thermo Fisher Scientific, Waltham, Mass.), and injected over the activated sensor for variable contact times ranging between 5 and 10 minutes at a rate ranging from 5-10 ⁇ L/min.
  • Quantities of Protein G immobilized to the COOH1 sensor chip range from 400-2000 response units (RU). Remaining activated sites were capped with 100 ⁇ L of ethanolamine at a flow rate of 25 ⁇ L/min.
  • Equilibrium constants for rabbit human chimeric mAbs were determined by binding the mAb to the protein G coated chip followed by binding of each analyte (IL-6 or IL-23) to its respective mAb.
  • each analyte IL-6 or IL-273
  • the surface densities of the mAbs for each analyte were adjusted so that as analyte binding approached saturation its corresponding RU fell between 200 and 300.
  • scFvs Equilibrium constants for scFvs were determined as previously described; however, the protocol was modified so that an epitope tagged IL-6 was captured on the chip surface and the dissociation of anti-IL-6 scFvs from IL-6 was monitored. Briefly, anti-FLAG® M2 antibody (200472, Agilent Technologies, Santa Clara, Calif.) was bound to Protein G, and then 3 ⁇ FLAG-IL-6 was captured by the anti-FLAG antibody. The anti-IL-6 scFvs were assayed over a range of concentrations between 1 and 100 nM.
  • IL-23 binding by the bispecific was performed by first immobilizing the bispecific with IL-6 as described, but at a constant density ( ⁇ 240 RU). Binding and dissociation of a recombinant human dimeric IL-23 (34-8239, eBiosciences, San Diego, Calif.) was assayed in concentrations ranging from 3 to 25 nM using the same parameters detailed above.
  • IL-6 protein Recombinant E. Coli -derived human IL-6, Ref. Seq. accession NP000591.1, obtained from ProSpec-Tany TechnoGene Ltd., Rehovot, Israel (Cat. # CYT-213i)
  • Sigma Adjuvant System Sigma S6322
  • the rabbit was boosted not less than 10 days prior to bleeding. Rabbits were maintained at R & R Research Laboratories (Stanwood, Wash., USA) in accordance with NIH, USDA and IACUC guidelines.
  • PBMC Peripheral Blood Mononuclear cells
  • the neutralizing activity of immune rabbit serum against human IL-6 was assayed after 3 immunizations.
  • Human IL-6 is titered from 1 ng/ml with and without a 1:3200 dilution of immune rabbit serum.
  • 6 cm tissue culture petri dishes were coated with IL-6 as follows: His-tagged human IL-6 (Cat. # CYT-484, Prospec-Tany Technogene, Rehovot, Israel) was captured on an anti-His antibody coated plate.
  • Anti-His antibody (Cat # A00613, GeneScript Corp., New Jersey, USA) at 2 ⁇ g/ml in PBS, was incubated overnight at 4 C or 1 hour at 37 C in a 6 cm plastic petri dish. The antibody solution was then removed and 4 ml of PBS+5% BSA was added for 1 hour.
  • IL-6 was captured by incubating 3 ml of His-IL-6 at 2 ⁇ g/ml in PBS for 1 hour, followed by 4 washes with PBS.
  • PBMC were suspended in 2 ml of PBS containing 5% BSA, and plated on the antigen-coated dishes for 40 minutes at 4 C. The plates were subsequently washed 4 to 8 times with PBS, and the adherent cells were removed by gentle scraping. These cells were resuspended in complete medium (RPMI 1640, 10% FBS, Non-Essential Amino Acids, Pyruvate, 50 uM beta-mercaptoethanol) at 100 to 500 ⁇ 10 3 cells/ml.
  • complete medium RPMI 1640, 10% FBS, Non-Essential Amino Acids, Pyruvate, 50 uM beta-mercaptoethanol
  • 100 ⁇ l of cell suspension was added to each well of a 96-well plate, in addition to 100 ⁇ l complete medium containing Mitomycin-c treated EL4-B5 cells (Zubler et al 1985) at 5 ⁇ 10 5 cells/well, recombinant human IL-2 (GenScript Corp, Piscataway, N.J., USA) at 20 ng/ml, and 5% conditioned media from rabbit spleen cells.
  • EL4-B5 cells were suspended at a density of 1 ⁇ 107 cells/ml in RPMI containing 50 ⁇ g/ml mitomycin-c (Cat # M0503, Sigma-Aldrich) for 40 minutes and washed 6 times in complete medium.
  • the rabbit conditioned media was prepared as follows: Rabbit spleen cells were mechanically dissociated, filtered through a 70 ⁇ m mesh, resuspended at 1 ⁇ 10 6 cells/ml, in complete medium (RPMI 1640, 10% FBS, Non-Essential Amino Acids, Pyruvate, 50 uM beta-mercaptoethanol). The cells were stimulated with 500 ng/ml of lonomycin and either Concanavalin A (Cat # C 5275, Sigma-Aldrich) at 5 ⁇ g/ml or PMA at 40 ng/ml for 48 h in a CO2 (5%) incubator at 37° C. The conditioned medium was sterile filtered and stored at ⁇ 20 C for subsequent experiments. For some experiments mitomycin-c treated (as per EL4-B5) normal rabbit spleen cells were added to the cloning plates at 1 ⁇ 2 ⁇ 10 5 cells/well.
  • complete medium RPMI 1640, 10% FBS, Non-Ess
  • the plates containing antigen-selected cells were incubated for 7 to 10 days at 37° C., 5% CO2.
  • the culture supernatants were then harvested to be assayed for IL-6 binding (ELISA) as well as inhibition of IL-6 activity.
  • IL-6 binding was evaluated using Recombinant IL-6 (See Example 1) was added to an ELISA plate in 100 ⁇ l PBS at 0.25 ⁇ g/ml. Plates were incubated 1 hour at 37 C, or overnight at 4 C. To block 100 ⁇ l/well PBS containing 10% goat serum (Cat #16210-072, Invitrogen, USA) was added to each well. Plates were incubated 1 hour at room temperature. Plates were rinsed 5 times with de-ionized water. To each well was added 50 ⁇ l PBS/10% goat serum. Test samples were then added at 50 ⁇ l/well. Plates were incubated 1 hour at room temperature. Plates were rinsed 5 times with de-ionized water.
  • a bioassay using an IL-6 dependant murine B-cell hybridoma cell line (B9cell line; Aarden et al., 1987) was used to evaluate IL-6 inhibition, by means of measuring B9 cell proliferation in response to E. Coli or CHO cell derived human IL-6 (Prospec-Tany Technogene, Rehovot, Israel)
  • Samples to be tested for neutralizing activity were diluted in 100 ⁇ l assay medium (RPMI 1640 w/L-glutamine, 10% FBS, Non-Essential Amino Acids, Sodium Pyruvate, 50 ⁇ M 2-mercaptoethanol) in a 96-well tissue culture plate. This was followed by the addition of 50 ⁇ l of IL-6 (Cat.
  • B9 cells were recovered from flasks and centrifuged for 7 min at 180 ⁇ g, and the pellet was resuspended in IL-6-free culture medium (RPMI 1640 w/L-glutamine, 10% FBS, Non-Essential Amino Acids, Sodium Pyruvate, 50 ⁇ M 2-mercaptoethanol). Cells were centrifuged and resuspended three times to remove IL-6. Following viability determination by trypan blue exclusion, cells were adjusted to 1 ⁇ 10 5 cells/ml. A volume of 50 ⁇ l of B9 cells, corresponding to 5 ⁇ 10 3 cells, was added to each well along with appropriate control wells containing IL-6-free medium.
  • IL-6-free culture medium RPMI 1640 w/L-glutamine, 10% FBS, Non-Essential Amino Acids, Sodium Pyruvate, 50 ⁇ M 2-mercaptoethanol. Cells were centrifuged and resuspended three times to remove IL-6
  • FIG. 1 illustrates an exemplary experiment as carried out to select cells producing antibodies suitable for further characterization: each supernatant was tested for both IL-6 binding (lower panel) and IL-6 neutralization (upper panel). Supernatants suitable for further characterization were positive in both assays (arrow and star).
  • the V-region rescue process is summarized in FIG. 2 .
  • IgG variable heavy and light chains from the supernatants positive for both IL-6 neutralization and IL-6 binding tests were captured by amplification using reverse transcriptase coupled polymerase chain reaction (RT-PCR).
  • RT-PCR reverse transcriptase coupled polymerase chain reaction
  • the VH and VL cDNAs thus obtained, were cloned and ligated onto human constant region constructs, such that the final cDNA construct encoded a chimeric rabbit human IgG as shown in FIG. 3 .
  • Primers for rescuing immunoglobulin V-regions from activated rabbit B-cells were designed for both cDNA synthesis from mRNA captured after cell lysis as well as the subsequent PCR amplification steps in which the final PCR adds restriction enzyme sites for cloning into an expression vector. Since one rabbit would have a b9 allotype background (Rader et al, 2000), it was necessary to design various cDNA primers and nested J-region primers as J-kappa and Ckappa region usage differ between rabbit allotypes (Sehgal et al., 1990). A list of selected DNA oligonucleotide primers used in both the RT and PCR steps is shown in Table 1, with their heavy/light chain specificity designated in the right hand column.
  • Selected positive B-cells were lysed and mRNA prepared using the mRNA DIRECT Micro Kit, from Dynabeads (Cat. #610.21) according to the manufacturer's instructions.
  • mRNA generated from a single antigen positive well is used in a one-step RT/PCR (Qiagen One Step RT-PCR Kit, cat. N. 210212) reaction for both the heavy and light chains.
  • RT/PCR Qiagen One Step RT-PCR Kit, cat. N. 210212
  • gene specific primers located in the constant regions of the heavy and light chains of the rabbit IgG molecule are used to generate a single strand cDNA, followed by nested J-region primers together with Leader peptide-specific primers for first round PCR generation
  • Primer sets for V-region rescue of the rabbit variable-regions are.
  • Primer Specificity SEQ Primer (Heavy Chain/Light ID name Sequence 5′-3′ Chain) RT/1st Rnd 66 rCH1R1 GCCAGTGGGAAGACTGACGGAG H 67 rVHL-F ATGGAGACTGGGCTGCGCTGG H 68 rJH-R1 GGAGACGGTGACCAGGGTGCCTGGG H 69 rJH6R TGAAGAGACGGTGACGAGGGTC H 70 rCk1R1 GCAGCTGGTGGGAAGATGAGGAC L 71 rVK5UTR GCCAGGCAGGACCCAGCATGGAC L 72 rJK2-R ACCACCACCTYGGTCCCTCCGCC L 73 rJK1-R GATTTCYACCTTGGTGCCAGCTCC L 74 rJK24R GTTTGATCTCCACCTTGGTCCCCGCACCG L 75 rJK2b9R ACT
  • a second round of PCR is performed to add restriction sites to the rescued V-regions for subcloning into vectors containing the constant regions of either the heavy or light chain of human IgG1. Separate PCRs are performed for heavy and light chains. Restrictions sites added to the V-regions are HindIII/XhoI and NcoI/BsiW1 for heavy and light chains respectively. Vectors containing constant regions were obtained from InvivoGen (pFUSE-CHIg-hG1 #08E07-SV and pFUSE2-CLIg-hk #08F19-SV). Both vectors were modified in-house for the sub cloning strategy. After addition of the restriction sites, the PCR products were subjected to the relevant Restriction enzymes digestion, gel purified and ligated into the appropriate vector.
  • the ligated DNA was transformed into DH5 ⁇ E. coli . (Invitrogen). The entire transformation mixture was cultured over night in medium containing the appropriate antibiotic resistance. The cultured bacteria were harvested and plasmid DNA was isolated and purified (Qiagen kit) for use in transient HEK293 expression of chimeric antibodies. At this time the isolated DNA may or may not be homogenous for one specific V-region, as selected wells may contain one or more different B-cell clones. To generate the chimeric antibodies, HEK293 cells were co-transfected with the DNA of both heavy and light chain from a selected well.
  • Supernatant was harvested after three to five days of cell culture and assayed for IgG and antigen binding by ELISA., as well as IL-6 neutralization (see above for methods).
  • an ELISA immunoassay is done which utilizes an anti-human IgG Fc capture antibody coated to an ELISA plate, followed by the supernatants and human IgG standard. Detection of Fc-captured antibody is obtained using an anti-human IgG (H&L)-HRP reagent and TMB substrate.
  • DNA sequencing was used to screen each ELISA-positive well to determine how many unique heavy and light chain combinations were rescued under the assumption that there would likely be more than one unique clone present per well.
  • DNA sequencing the DNA isolated previously for transfection is retransformed into DH5 ⁇ E. coli and plated on agar plates containing the appropriate antibiotic. Multiple colonies from each transformation are picked and processed for DNA production using a rolling circle DNA amplification kit (Templiphy, GE Healthcare) following manufacturer's instructions. The DNA generated from the Templiphy reactions is sequenced and subsequently analyzed to determine the complexity of V-regions for each well. In addition to making DNA, bacteria used for the Templiphy reactions are saved for future DNA isolation since each DNA now represents a unique clone.
  • Vh Variable region Heavy Chain
  • VI Variable region Light chain
  • the 13A8 clone demonstrated high potency, with an EC50 (calculated as concentration necessary to inhibit bioactivity of 50 pg/ml of IL-6) of 34 pg/ml ( FIGS. 3A and B), and high affinity antigen binding properties determined by SPR analysis: K d 1.38 ⁇ 10 ⁇ 4 (s ⁇ 1 ); K a 6.33 ⁇ 10 5 (M ⁇ 1 s ⁇ 1 ), and K D 218 pM
  • Vh Variable region light chain identified as SEQ ID 16, aminoacid sequence and SEQ ID 17, nucleotide sequence.
  • V Variable region Light chain identified as SEQ ID 18, aminoacid sequence and SEQ ID 19, nucleotide sequence.
  • the 28D2 clone demonstrated high potency, with an EC50 (calculated as concentration necessary to inhibit bioactivity of 50 pg/ml of IL-6) of 65 pg/ml ( FIGS. 3A and B).
  • Vh Variable region light chain identified as SEQ ID 26, aminoacid sequence and SEQ ID 27, nucleotide sequence.
  • V Variable region Light chain identified as SEQ ID 28, aminoacid sequence and SEQ ID 29, nucleotide sequence
  • the 18D4 clone demonstrated high potency, with an EC50 (calculated as concentration necessary to inhibit bioactivity of 50 pg/ml of IL-6) of 54 pg/ml, and high affinity antigen binding properties determined by SPR analysis: K d 8.49 ⁇ 10 ⁇ 5 (s ⁇ 1 ); K a 6.66 ⁇ 10 5 (M ⁇ 1 s ⁇ 1 ), and K D 128 pM
  • Vh Variable region light chain identified as SEQ ID 36, aminoacid sequence and SEQ ID 37, nucleotide sequence.
  • V Variable region Light chain identified as SEQ ID 38, aminoacid sequence and SEQ ID 39, nucleotide sequence
  • the initial transfection supernatants of the 8C8 clone demonstrated high potency for inhibition of bioactivity.
  • scFv were derived directly from the rabbit IgG and these demonstrated high potency, with an EC50 (calculated as concentration necessary to inhibit bioactivity of 200 pg/ml of IL-6) of 510 pg/ml ( FIG. 12A ).
  • Vh Variable region light chain identified as SEQ ID 46, aminoacid sequence and SEQ ID 47, nucleotide sequence.
  • V Variable region Light chain identified as SEQ ID 48, aminoacid sequence and SEQ ID 49, nucleotide sequence
  • the 9H4 clone demonstrated high potency, with an EC50 (calculated as concentration necessary to inhibit bioactivity of 50 pg/ml of IL-6) of 109 pg/ml ( FIGS. 3C , D, E), and high affinity antigen binding properties determined by SPR analysis: K d 4.75 ⁇ 10 ⁇ 5 (s ⁇ 1 ); K a 8.16 ⁇ 10 5 (M ⁇ 1 s ⁇ 1 ), and K D 58 pM
  • Vh Variable region light chain identified as SEQ ID 56, aminoacid sequence and SEQ ID 57, nucleotide sequence.
  • V Variable region Light chain
  • the 9C8 clone demonstrated high activity and antigen binding properties, with an EC50 (calculated as concentration necessary to inhibit bioactivity of 50 pg/ml of IL-6) of 400 pg/ml ( FIG. 3E ), and K d 3.17 ⁇ 10 ⁇ 5 (s ⁇ 1 ); K a 7.65 ⁇ 10 5 (M ⁇ 1 s ⁇ 1 ), and K D 42 pM
  • cytokine antagonists for human therapy will require initial toxicology testing. Toxicology is most efficiently demonstrated in non human species. In order to conduct toxicology studies it is first necessary to demonstrate the capacity of the antibodies to neutralize IL-6 from the species being considered for the studies.
  • FIG. 3 Several of the anti-IL6 chimeric antibodies that have been tested for neutralization of non-human primate IL-6 activity are shown in FIG. 3 .
  • IL-23 protein Baculovirus-derived recombinant human IL-23 composed of the p40 chain, accession NM — 002187 and the p19 chain, accession NM — 016584, from eBiosciences, San Diego Calif., USA (Cat. #34-8239)
  • Sigma Adjuvant System Sigma S6322
  • the animals were bled at least 10 days after immunization. Rabbits were maintained at R & R Research Laboratories (Stanwood, Wash., USA) and Spring Valley Laboratories (Woodbine, Md., USA) in accordance with NIH, USDA and IACUC guidelines.
  • the NZW and B9 rabbits express different immunoglobulin gene allotypes which correspond to differences in the framework and CDR regions and corresponding differences in the structures of the mAbs isolated.
  • the b9 allotype rabbits are reported to be better for use in phage display cloning of mAbs, due to the absence of Cys residues in the V region. All the anti IL-6 mAbs were cloned from NZW rabbits, and none presented Cys residues in the V region.
  • the IL-23 neutralization activity of high titer sera from B9 and NZW rabbits immunized with human IL-23 was measured. Serum from immune rabbits is able to fully neutralize the IL-17A secretion induced by 600 pg/ml of human IL-23 from mouse splenocytes at dilutions approaching 1:10,000.
  • B-Cells specific for IL-23 were selected as in Example 3.
  • PBMC Peripheral Blood Mononuclear cells
  • IL-23 coated plates were produced by incubating IL23 (eBioscience) at 2 ⁇ g/ml in PBS overnight at 4 C, or 1 hour at 37 C, washed 4 times with PBS and used for capturing B-cells.
  • PBMC were suspended in 2 ml of PBS containing 5% BSA, and plated on the antigen-coated dishes for 40 minutes at 4 C. The plates were subsequently washed 4 to 8 times with PBS, and the adherent cells were removed by gentle scrapping.
  • ELISA assay was used to evaluate IL-23 binding (Aggarwal et al., 2003). ELISA plates were coated using either a direct or indirect method of binding IL-23.
  • anti-His antibody (Cat # A00613, GenScript Corp., New Jersey, USA) was added to the plates in 100 ⁇ l/well of PBS at 0.01-0.02 ug/ml. Plates were incubated 1 hour at 37 C, or overnight at 4 C.
  • 100 ⁇ l/well PBS containing 10% goat serum (Cat #16210-072, Invitrogen, USA) was added to each well, after which plates were rinsed 5 times with de-ionized water.
  • IL-23 p40-p19-His (SEQ ID 4) in 100 ⁇ l/well PBS/10% goat serum at 0.5 ⁇ g/ml was added and incubated for 1 hour at room temperature.
  • IL-23 p40-p19-His (SEQ ID 4) was added to an ELISA plate in 100 ⁇ l PBS at 0.5 ⁇ g/ml. Plates were incubated 1 hour at 37 C, or overnight at 4 C. To block non-specific binding 100 ⁇ l/well PBS containing 10% goat serum (Cat #16210-072, Invitrogen, USA) was added to each well. Plates were incubated 1 hour at room temperature.
  • a bioassay based on the detection of IL-23-induced IL-17 expression by mouse spleen cells, was used to detect antibody mediated inhibition of IL-23 binding to the IL-23 receptor and resulting bioactivity.
  • 5 ⁇ 10 5 C57Bl/6 spleen cells were cultured in the wells of a 96-well plate in 200 ⁇ l containing a dilution of the heterodimeric IL-23 (eBioscience cat. #14-8239 or Humanzyme, Chicago, USA cat. #HZ-1049) and the plates incubated for 2-3 days at 37 C.
  • Culture medium is RPMI 1640, 10% FBS, 50 uM 2-mercaptoethanol, non-Essential Amino Acids, pyruvate, gentamicin and 10 ng/ml human IL-2 (Cat # CYT-209, Prospec-Tany Technogene). After 3 days, the culture supernatants were assayed by ELISA for IL-17A, as described below.
  • test mAb samples were added at various dilutions to the cultures of mouse spleen cells containing 150-1200 pg/ml IL-23 and the secretion of IL-17A was compared to cultures not treated with mAb.
  • ELISA assay was used to detect mouse IL-17. Plates were coated with anti-mlL-17A (eBioscience #14-7178) 1 ⁇ g/ml in 100 ⁇ l PBS, overnight at 4° C. or 1 hr at 37° C. Plates were washed in deionized water and blocked for 1 h with 100 ⁇ l of PBS, 10% goat serum. After washing the plates, 50 ⁇ l of PBS/10% goat serum and 50 ⁇ l of culture supernatant were added to the plates, and incubated for 1 hr.
  • anti-mlL-17A eBioscience #14-7178
  • the plates were washed and 100 ⁇ l/well of anti-mIL-17A-Biotin (eBioscience #13-7179) at 0.5 ⁇ g/ml in PBS/10% goat serum was added and the plates were incubated for 1 h at RT, washed, and reacted with 100 ⁇ l/well Streptavidin-HRP (Jackson Labs) at 1:1000 in PBS/10% goat serum. Plates were washed again, and the signal was detected by adding Add 100 ⁇ l/well TMB substrate (Thermo Scientific, IL, USA). After stopping the reaction with 100 ⁇ l/well 1N H 2 SO 4 , the optical density was read at 450 nM. Data were plotted and analyzed with Graphpad (Prism, Mountainview, Calif.) software.
  • B cells were cloned from the IL-23 immunized rabbits and the B cell clone supernatants were tested for IL-23 neutralization and IL-23 binding.
  • FIG. 7 illustrates an example 96 well plate from an experiment where each supernatant was tested for both IL-23 binding (lower panel) and IL-23 neutralization (upper panel). Supernatants suitable for further characterization were positive in both tests.
  • the IgG variable heavy and light chains from the B cells positive for both IL-23 neutralization and IL-23 binding assays were captured by RT-PCR essentially as in Example 3.
  • FIGS. 5A-I show examples of human IL-23 neutralization activity by several anti IL-23 neutralizing mAbs obtained.
  • IL-23 Several antibodies have been further characterized in their binding to primate IL-23, as depicted in FIG. 6 . Monoclonal antibodies neutralizing IL-23 were further tested for neutralization of human IL-12, as shown in FIG. 8A . mAb 31A12 neutralizes specifically IL-23 while 45G5 and 22H8 neutralize both IL-23 and IL-12.
  • Mapping of epitopes recognized by antibodies of the present invention may be achieved through several experimental methods, such as cross competition binding assays, or binding to linear peptides.
  • LCMS Liquid Chromatography Mass Spectroscopy
  • Vh Variable region Heavy Chain identified as SEQ ID 86, aminoacid sequence ; SEQ ID 87, nucleotide sequence;
  • V Variable region Light chain
  • the 31A12 mAb demonstrated high potency and antigen binding properties, with an EC50 (calculated as concentration necessary to inhibit bioactivity of 600 pg/ml of IL-23) of 3286 pg/ml, and high affinity antigen binding properties determined by SPR analysis: K d 2.02 ⁇ 10 ⁇ 4 (s ⁇ 1 ); K a 4.79 ⁇ 10 5 (M ⁇ 1 s ⁇ 1 ), and K D 422 pM.
  • Vh Variable region light chain identified as SEQ ID 96, aminoacid sequence and SEQ ID 97, nucleotide sequence.
  • V Variable region Light chain
  • the 49B7 mAb demonstrated high potency, with an EC50 (calculated as concentration necessary to inhibit bioactivity of 600 pg/ml of IL-23) of 988 pg/ml.
  • Vh Variable region light chain identified as SEQ ID 106, aminoacid sequence and SEQ ID 107 nucleotide sequence.
  • V Variable region Light chain
  • the 16C6 mAb demonstrated high potency, with an EC50 (calculated as concentration necessary to inhibit bioactivity of 150 pg/ml of IL-23) of 219 pg/ml.
  • Vh Variable region light chain identified as SEQ ID 116, aminoacid sequence and SEQ ID 117, nucleotide sequence.
  • V Variable region Light chain
  • the 34E11 clone demonstrated high potency, with an EC50 (calculated as concentration necessary to inhibit bioactivity of 150 pg/ml of IL-23) of 50 pg/ml.
  • Vh Variable region light chain identified as SEQ ID 126, aminoacid sequence and SEQ ID 127, nucleotide sequence.
  • V Variable region Light chain
  • the 35H4 clone demonstrated high potency in the transfection supernatants of the cloned mAb relative to other mAbs isolated in the same B cell cloning experiment ( FIG. 5I ).
  • IL-12 and IL-23 share a common p40 polypeptide and differ in the second chain, covalently linked to p40 ( FIG. 7A ).
  • the p19 chain of IL-23 and the p35 chain of IL-12 are both four helix bundle, cytokine like polypeptides.
  • the p19 and p40 subunits are linked to the common p40 subunit via a disulfide bond.
  • Antibodies neutralizing both IL-12 and IL-23 occur due to the sharing of the p40 chain between the two molecules.
  • IL-12 and IL-23 receptors share a common chain (IL-12R ⁇ 1) and in addition, each have a unique receptor component (IL-23R and IL-12R ⁇ 2) ( FIG. 7B ). These differences result in significant differences in the target cell and signaling pathways used by IL-12 and IL-23. These receptors have transmembrane signaling domains that pair with JAK2 or TYK2 tyrosine kinases for STAT activation.
  • Antibodies binding both IL-23 and IL-12 were isolated from rabbits immunized with recombinant IL-23 (see Example 4). B cell clones exhibiting binding and functional activity towards both IL-23 and IL-12 were selected for further characterization.
  • Vh Variable region Heavy Chain identified as SEQ ID 136, aminoacid sequence; SEQ ID 137, nucleotide sequence;
  • V Variable region Light chain
  • mAb 22H8 demonstrated high potency and antigen binding properties, with an EC50 (calculated as concentration necessary to inhibit bioactivity of 50 pg/ml of IL-6) of 603 pg/ml and high affinity antigen binding properties determined by SPR analysis: K d 8.94 ⁇ 10 ⁇ 5 (s ⁇ 1 ); K a 4.03 ⁇ 10 5 (M ⁇ 1 s ⁇ 1 ), and K D 221 pM.
  • Vh Variable region Heavy Chain identified as SEQ ID 146, aminoacid sequence ; SEQ ID 147, nucleotide sequence;
  • V Variable region Light chain
  • mAb 45G5 demonstrated high potency and antigen binding properties, with an EC50 (calculated as concentration necessary to inhibit bioactivity of 50 pg/ml of IL-6) of 385 pg/ml.
  • Vh Variable region Heavy Chain identified as SEQ ID 156, aminoacid sequence ; SEQ ID 157, nucleotide sequence;
  • V Variable region Light chain identified as SEQ ID 158, aminoacid sequence; SEQ ID 159, nucleotide sequence.
  • mAb 1H1 demonstrated high potency and antigen binding properties, with an EC50 (calculated as concentration necessary to inhibit bioactivity of 50 pg/ml of IL-6) of 603 pg/ml.
  • Vh Variable region Heavy Chain identified as SEQ ID 166 aminoacid sequence ; SEQ ID 167, nucleotide sequence;
  • V Variable region Light chain
  • mAb 4F3 demonstrated high potency and antigen binding properties, with an EC50 (calculated as concentration necessary to inhibit bioactivity of 50 pg/ml of IL-6) of 2339 pg/ml.
  • Vh Variable region Heavy Chain identified as SEQ ID 176, aminoacid sequence ; SEQ ID 177, nucleotide sequence;
  • V Variable region Light chain
  • mAb 5C5 demonstrated high potency and antigen binding properties, with an EC50 (calculated as concentration necessary to inhibit bioactivity of 50 pg/ml of IL-6) of 1907 pg/ml.
  • VH Variable region Heavy Chain identified as SEQ ID 186, amino acid sequence; SEQ ID 187, nucleotide sequence;
  • VL Variable region Light chain identified as SEQ ID 188, aminoacid sequence; SEQ ID 189, nucleotide sequence.
  • mAb 14B5 demonstrated high potency and antigen binding properties, with an EC50 (calculated as concentration necessary to inhibit bioactivity of 50 pg/ml of IL-6) of 767 pg/ml.
  • Antibodies were assayed for IL-12 neutralizing capacity using the IL-12 responsive cell line NK-92 (CRL-2407, ATCC, Manassas, Va., USA). 50 ⁇ l of culture supernatant from the B cell cloning plates or 50 ⁇ l of supernatant from antibody transfection was transferred to a 96 well tissue culture plate. 50 ⁇ l of human IL-12 (Cat. # Cyt-362, Prospec-Tany Technogene, Rehovot, Israel) was added to each well at 4 ng/ml. Plates were incubated for 30-60 minutes at room temperature, after which 5 ⁇ 10 4 NK-92 cells were added to each well in 100 ⁇ l.
  • human IL-12 Cat. # Cyt-362, Prospec-Tany Technogene, Rehovot, Israel
  • Assay medium is RPMI 1640, 10% FBS, NEAA, pyruvate, 50 ⁇ M 2-mercaptoethanol, gentamicin and 10 ng/ml human IL-2 (Cat # Z00368, GeneScript Corporation, Piscataway, N.J., USA).
  • the plates were washed and 100 ⁇ l/well of anti-human Interferon- ⁇ -Biotin (Cat # Mab 7b6-1-biotin, Mabtech) at 0.5 ⁇ g/ml in PBS/10% goat serum was added and the plates were incubated for 1 h at RT, washed, and reacted with 100 ⁇ l/well Streptavidin-HRP (Jackson Labs) at 1:1000 in PBS/10% goat serum. Plates were washed again, and the signal was detected by adding 00 ⁇ l/well TMB substrate (Thermo Scientific, IL, USA). After stopping the reaction with 100 ⁇ l/well 1N H 2 SO 4 , the optical density was read at 450 nM.
  • V-regions Rabbit immunoglobulin variable regions (V-regions) are captured from mRNA isolated from peripheral blood B-cells from immunized rabbits. These rabbit B-cells were plated at low density in 96-well plates and activated as previously described. V-region cDNAs are amplified from the mRNA of each well using reverse-transcriptase-PCR (RT-PCR) with a gene-specific primer from the constant region for first strand synthesis and a nested J-region-specific primer at the 3′ end with a 5′ leader primer for the PCR step.
  • RT-PCR reverse-transcriptase-PCR
  • V-regions are then cloned into either a human IgG heavy chain, kappa or lambda light chain vector cassette , transiently expressed in HEK 293 cells and 72-hour post-transfection supernatants tested for both total IgG expression and neutralization of respective targets. Potent neutralizers were then sequenced to determine the level of complexity present in the well from which they were subcloned. Once the number of unique light and heavy chains was determined, all possible combinations present were again transiently expressed into HEK 293, assayed for neutralization and IgG content. Potent neutralizers were then further assayed for other desirable activities.
  • Anti-human IL-6 antibodies were tested for neutralization of IL-6 from non-human primate.
  • Anti-human IL-23 antibodies are assayed not only for neutralization of non-human primate IL-23, but also neutralization of human IL-12 since both IL-12 and IL-23 dimers share the same p19 chain.
  • the inventors followed two strategies: they constructed rabbit scFvs directly from selected heavy chain (VH) and light chain (VK or VL) by PCR genetically fusing the heavy and light chain V-regions in either the VLVH or VHVL orientation by introducing a 20 amino acid linker composed of four tandem repeats of the sequence gly-gly-gly-gly-ser (G4S) between the two domains.
  • Rabbit scFvs are helpful in assessing whether or not the conversion from a chimeric antibody to an scFv format has had an adverse effect on the functional or biophysical properties of the V-region pair.
  • ScFvs were then transiently expressed in HEK 293 and assayed for function as illustrated in FIG. 12A . Potent neutralizers were selected for humanization (see section 6.1)
  • Immunoglobulin V-regions can be humanized in many different formats including both a full length antibody and a single-chain Fv (scFv). Since the described invention relies on prokaryotic recombinant protein expression, a full length antibody structure is not desirable. However, the invention does describe successful humanization of rabbit V-regions in an antibody format. Regardless of the format, the current invention involves removal of any naturally occurring methionine residues, substituting them with other amino acids. Since methionine residues are frequently found within framework regions and CDRs of immunoglobulin V-regions, it is necessary to find suitable replacements for these residues where they occur without impacting the expression, stability or function of the desired protein. This methionine-free scFv can then be optimized for expression in a methionine auxotrophic bacterial strain, purified, refolded and tested for biologic activity.
  • scFv single-chain Fv
  • Successful humanization and subsequent methionine substitution provides part of a therapeutic vehicle that can be chemically modified by insertion of a single methionine codon that serves as an insertion site for a non-natural amino acid with a chemically reactive site for covalently linking other complementary molecules such as an activated PEG moiety.
  • This PEGylated scFv can then be further modified by covalent linkage to another such scFv through a similarly reactive group at the remaining terminus of the PEG polymer.
  • This bi-specific, PEGylated product can then be purified and refolded to yield a stable, biologically active therapeutic protein.
  • Rabbit-human chimeric monoclonal antibodies can be humanized as full length antibodies. This entails the exchange of human VH and VL framework regions for the rabbit frameworks with the retention of the rabbit CDRs and often includes retaining particular rabbit framework residues. Just as there are multiple strategies for humanizing rodent V-regions there are other possible methods by which a rabbit-human chimeric antibody might be partially or fully humanized.
  • 9C8 a high affinity and high potency chimeric mAb, was humanized by changing the framework regions of the VH and VL to human framework sequences, with limited back mutation to rabbit framework sequences.
  • Humanizing NZW rabbit V-regions was accomplished by first comparing their primary amino acid sequence to those found in human V-regions (Altschul et al., 1990). Selection of potentially compatible human V-region frameworks were made based on sequence similarity within framework regions (FR1, FR2, FR3 and FR4), sequence length and content within the complementarity determining regions (CDR1, CDR2 and CDR3), as well as key FR residues that are known to be critical for supporting IgV canonical loop structures. Using these data human frameworks were chosen for both light and heavy chain V-regions and the rabbit CDRs were grafted onto these frameworks as illustrated in FIG. 15 by PCR using overlapping oligonucleotide primers (Table 19).
  • FIG. 9B illustrates a side by side comparison of the parental chimeric 9C8 mAb with the humanized 9C8 mAbs (9C8 mAbv1 and 9C8 mAbv2) containing the 2 different rabbit back mutations proximal to CDR1 of the VH region (at positions H23-H30).
  • the humanized monoclonal antibodies 9C8 mAbv1 (containing TV at VH residues 23-24) and 9C8 mAbv2 (AA at 23-24) were expressed by transient co-transfection of both heavy and light chain DNAs into HEK293 cells as described previously.
  • the humanized monoclonal antibodies were tested for neutralization of 50 pg/ml IL-6, as indicated These changes, TVSGIDLS (mAbv2) or AASGIDLS (mAbv1) do not affect functional activity ( FIG. 9B ). 9C8 mAbv1 was further compared to humanized mAb 18D4 in FIG. 9C .
  • Oligonucleotide Primers used to graft rabbit CDRs on selected human V-region frameworks AZ_ID SEQ_ID primer sequence scFv-NotF 200 GCGATAGCGGCCGCACCACCATGGAGGCTCCC JHXhoR 201 GCTATACTCGAGACGGTGACCAGGGTGCCCTGGCCCC DPK8F1 202 GACATCCAGTTGACCCAGTCTCCATCCTTTCTGTCTGCATCTGTAGGAGACAG DPK8-AgeF 203 GACACAACCGGTGACATCCAGTTGACCCAGTC 9C8-H1F 204 GAATCGACCTCAGTAGCTACGACATGAGCTGGGTCCGTCAGGCACCTG 9C8-H1R1 205 GTAGCTACTGAGGTCGATTCCAGAAGCTGCACAGGAGAGGCGCAGGG 9C8-H1R2 206 GTAGCTACTGAGGTCGATTCCAGAGAGACAGTACAGGAGAGGCGCAGGG 9C8-H2F 207 ACT
  • the primers used for amplifying the humanized V-regions code for restriction enzyme sites identical to those used to capture rabbit V-regions in previous Examples and illustrated in FIG. 10 .
  • the humanized light chain was ligated to the Ckappa containing expression vector and the humanized heavy chain V-region was ligated to the Cgamma1 containing expression vector and transformed into E. coli as described previously. Isolated colonies were then screened and sequenced.
  • Humanized 9C8 scFvs were derived from the humanized mAbv1 (TV) and mAbv2 (AA). The resulting scFvs, retained the rabbit framework 1 residues (23-30) proximal to VH1CDR1 These scFvs also retained the endogenous methionine residues at H34 and H82. Final versions of the humanized scFv for 9C8 were then made from these scFvs, but substituting new frameworks, DPK5,6/DP47. These new humanized scFvs, 9C8 Hum scFv v3-1 and 9C8 Hum scFv v3-2 showed potent anti IL-6 neutralizing activity ( FIG. 9D ).
  • Rabbit V-regions can also be humanized directly in an scFv format. Although the humanization methods used may be generally the same as those used for humanizing monoclonal antibodies, not all humanized antibodies are easily converted to an scFv. Moreover, humanization of an antibody carries the requirement to account for Constant region interactions with the grafted CDR.
  • the sequences of both heavy and light chain V-regions were compared to human germline and expressed sequences using both V-base (http://vbase.mrc-cpe.cam.ac.uk/) as well as IgBLAST (http://www.ncbi.nlm.nih.gov/) as described in Example 6.1.
  • Humanized scFvs were designed to encode a 5′ Not I restriction enzyme site, followed by a Kozak box (Kozak, 1987), an IgVK3 leader (L2), a human VK1-JK4 framework, a 20 amino acid flexible (gly4ser)4 linker, human VH3-JH4 framework, and a 3′ Xho I restriction site nested within the last two serine residues at the C-terminus of the VH3-FR4 ( FIG.
  • the pcDNA3.1( ⁇ ) vector cassette had been modified to encode a short proline-rich linker followed by a 6 ⁇ His tag (gly-pro-pro-pro-pro-his-his-his-his-his-his) in frame with the C-terminus of the scFv.
  • Ligated pcDNA3.1-6 — 13A8 was transformed into competent E. coli TOP 10 (Invitrogen, Carlsbad, Calif.) and selected on LB agar +100 ⁇ g/ml carbenicillin plates (Teknova, Hollister, Calif.) at 37° C. overnight.
  • each scFv was transfected into HEK293 cells using Lipofectamine 2000 (Invitrogen) using the manufacturer's protocol. Briefly, the day before transfection, log phase HEK293 cells were plated into 12 well culture plates (Corning, Lowell, Mass.) at a density of 500,000 cells per well in complete media (DMEM+Glutamax+Non-Essential Amino Acids+Pen-Strep+10% FBS-Life Sciences) and incubated overnight in a 37° C. CO 2 incubator.
  • complete media DMEM+Glutamax+Non-Essential Amino Acids+Pen-Strep+10% FBS-Life Sciences
  • Anti IL-6 scFvs humanized using the one step method, retained IL-6 neutralization activity when expressed in mammalian cells, as shown for 13A8 ( FIGS. 11A-B ), 28D2 and 9C8 v3-1 ( FIG. 11C ).
  • Measurement of binding affinities by Surface Plasmon resonance (SPR) also demonstrated successful humanization of the 13A8 and 9C8 anti IL-6 scFvs (Table 21).
  • the humanized 13A8 and 9C8 scFvs used for affinity testing contained a 6 ⁇ -Histidine tag and purified from transfected HEK supernatants (using methods described in previous examples) and were tested by SPR carried on essentially as described in Example 2.
  • Human immunoglobulin (Ig) V regions often contain methionine residues in CDR1 of both the light and heavy chain, at relatively conserved residues in VH-FR3 (human VH3 family, amino acid H82) as well as at position L4 of the kappa light chain. Since these humanized scFvs will ultimately be linked covalently using a methionine analog, all methionine residues within the mature scFvs must be replaced by another naturally occurring amino acid. This amino acid substitution must have minimal or no impact on either function or stability of the resulting scFv.
  • CDRs were grafted into the human germline framework DPK8 (GenBank X93626), which has a leucine residue at that position.
  • degenerate oligonucleotide primers were designed such that methionine would be changed to either isoleucine (ile), leucine (leu), valine (val) or phenylalanine (phe). These four amino acids can be found in IgVH regions from other species at position H82 as well as in some expressed human antibodies.
  • These new methionine-free scFvs were transiently expressed in HEK 293 cells, then neutralization activity compared to those of their parental scFv.
  • methionine-free DNA sequences were optimized for expression in E. coli by altering codon usage and potential secondary structure that could interfere with translation efficiency
  • overlapping degenerate primers (primers 54, 55 above) were designed to introduce leucine, valine, isoleucine or phenylalanine at position H82 by PCR along with flanking primers (primers 1, 2 above seq ID's 200 and 201 respectively). PCR products were cloned as described above and the DNA sequenced to determine which amino acid was encoded by each selected clone.
  • the humanized anti IL-6 scFv 13A8 (as several other scFvs) has 2 methionine residues at VH positions H34 and H82. These residues were replaced by PCR using degenerate oligonucleotide primers resulting in the substitution of methionine with either leucine, isoleucine, valine or phenylalanine as described above.
  • H34L was generally a well tolerated replacement. Replacements of H82 with different amino acids, in combination with H34L, in the anti IL-23 humanized scFv, 31A12, resulted in full retention of activity, compared to the parental Met containing scFv ( FIG. 14E ). In a similar fashion, replacement of 45G5 H82 with L or V, in combination with H34L resulted in full retention of activity compared to the original chimeric mAb ( FIG. 14F ). Replacement Mets in the 9C8 humanized scFv with H82L and H34L also retained potent activity ( FIG. 14G ). 22H8 scFv naturally does not have a Met residue at position H82.
  • FIG. 14H Changing the Met at H34 with either L or V resulted in full retention of potent IL-23 neutralizing activity ( FIG. 14H ).
  • the specific VH mutations for each lead candidate VH region are shown in Table 25 ⁇ , showing the rabbit back mutations at H23-30, and H49, as well as the H34 and H82 methionine replacements.
  • Leucine was chosen as the amino acid that was substituted at both positions H82 and H34.
  • Lead candidate scFv DNA sequences were optimized for expression in E. coli by modifying the codon usage according to those preferred by E. coli .
  • placement of the single Met residue for substitution in vivo by a methionine analog, such as Aha was investigated.
  • DNA was synthesized by PCR (as described above) was cloned into appropriate expression vector such as pQE vectors.
  • the synthetic DNA gene sequence in the expression vectors were confirmed by DNA sequence.
  • the synthetic gene in the expression vector was transformed into Methionione auxotrophic E. coli host such such B834 for recombinant protein production.
  • the scFv must have a single Met residue introduced at the position where the non natural amion acid (NNAA) may be incorporated during protein production in E. coli .
  • This NNAA will become the specific site of bioconjugation.
  • the NNAA of choice for these products is azidohomoalanine (Aha) which allows the use of copper catalyzed cycloaddition bioconjugation.
  • Aha azidohomoalanine
  • scFv DNA containing a single methionine codon was codon optimized for E. coli expression and synthesized and cloned into a methionine auxotrophic E. coli strain such as B384.
  • IB Inclusion bodies
  • Bispecific scFvs are constructed by the conjugation of two different scFv antigen binding domains to each other by way of a linker. This strategy is realized in a two-step process in which each scFv is conjugated to the bifunctional linker.
  • the two scFvs, comprising the bispecific conjugate contain each a single non natural amino acid (Aha or other) at a position which serves as a specific site of conjugation.
  • the linker can be homo-bifunctional or hetero-bifunctional and contain a complementary functional group (Alkyne) that is reactive with the unnatural amino acid contained in the scFv (Aha).
  • Alkyne complementary functional group
  • the linker employed in these examples is PEG (polyethylene glycol).
  • PEGs have several chemical properties which are desirable in a final bispecific product and solve problems endemic with scFvs. PEGylation improves protein solubility and increase scFv stability, reducing scFv aggregation and precipitation. In addition, PEGylation has been shown to increase serum half life of scFv bispecific product.
  • a long and flexible linker such as PEG increases the physical separation of the two antibody fragments, allowing them to refold independently from each other. This solves one of the critical problems that occurs in the refolding of bispecific antigen binding domains linked by genetic fusion, for which there often tends to be uncontrolled and undesirable cross linking of the two domains.
  • PEG linker has additional advantages due to the flexibility of chemical synthesis.
  • PEG can be easily functionalized to be a complementary reaction partner with any unnatural amino acid that is incorporated into the scFv proteins.
  • PEG can also be functionalized with multiple sites of conjugation which enables construction of multivalent protein hybrids.
  • the PEG functionalization can be made with homo-bifunctional or hetero-bifunctional PEG's depending on the desired conjugation chemistry.
  • the structure of PEG can be tailored for linear or branched variations, which can impact pharmacokinetics and bioactivity.
  • scFv-PEG conjugates The chemistry used to conjugate scFvs to the linker is orthogonal to the 20 natural amino acids.
  • Azide-alkyne copper mediated cycloadditions is used here, in the preparation of scFv-PEG conjugates and bispecifics.
  • an scFv containing azidohomoalanine (Aha) is reacted with an excess amount of a homo-bifunctional PEG linker functionalized with alkynes.
  • the monovalent PEGylated material is purified and then the free pendant alkyne of the PEG linker undergoes a second copper mediated azide-alkyne cycloaddition with a second scFv containing Aha to afford the bispecific.
  • the first step in the preparation of bispecifics is the site specific PEGylation of an scFv containing a non-natural amino acid, such as azidohomoalanine (Aha), with PEG that is either homo-bifunctional (eg. Bis-alkyne) or hetero-bifunctional (eg at least mono-alkyne).
  • PEG that is either homo-bifunctional (eg. Bis-alkyne) or hetero-bifunctional (eg at least mono-alkyne).
  • Monovalent PEGylated scFvs are purified by a series of CHT and SEC chromatography prior to the second step of the process. The monovalent materials are also assessed for their ability to be refolded. Finally, the refolded materials are evaluated by bioassay for activity.
  • the PEGylation of scFvs containing the non-natural amino acid azidohomoalanine (Aha) proceeds with an excess of PEG bis-alkyne (2-100 equivalents).
  • PEG molecular weights have been used.
  • the azide-alkyne cycloaddition used for conjugation is mediated by Copper (I), originating from a copper (I) source such as CuI or derived by reducing a copper (II) source (CuSO4) with a reducing agent such as DTT, cysteine, beta-mercaptoethanol, glutathione, cystamine, tris-carboxyethylphosphine.
  • a ligand such as Tris[(1-benzyl-1H-1,2,3-triazol-4-yl)methyl]amine, TBTA is also included in the reaction mixture.
  • Ligands such as TBTA have been shown to stabilize the reactive copper species and improve reaction yields.
  • the reaction pH is held between 3-10 or optionally between 6-9 by the addition of buffering reagents, such as sodium phosphate buffer, Tris or HEPES. Additional excipients such as SDS may be used to enhance reaction conditions and protein dynamics.
  • buffering reagents such as sodium phosphate buffer, Tris or HEPES.
  • Additional excipients such as SDS may be used to enhance reaction conditions and protein dynamics.
  • the monovalent PEGylated scFv can be separated from the unreacted scFv and PEG. This prevents the formation of side products in the subsequent bispecific preparation step.
  • the mixture is typically centrifuged or filtered to remove solid particulates and the solution treated with excess reducing agent such as DTT. The solution then undergoes a series of chromatography steps.
  • the first step in the purification of the monovalent scFv-PEG is loading the reduced reaction mixture onto CHT column which captures the reacting scFv and the scFv-PEG product, but does not bind all unreacted PEG.
  • the reacting scFv and product scFv-PEG can be partially or fully resolved by phosphate elution from the CHT column.
  • the desired fractions are pooled and subsequently loaded onto a size exclusion column (SEC), which can separate the residual unreacted scFv .
  • SEC size exclusion column
  • reaction mixture was poured into 50 mL centrifuge tubes and centrifuged (12,000 g, 10 min). The supernatant was poured onto DTT (1.5 g) and stirred under nitrogen for 1 h. Purification was accomplished by a combination of CHT and SEC chromatography.
  • reaction mixture was poured into a 50 mL centrifuge tube and centrifuged (12,000 g, 10 min). The supernatant was added to DTT (462 mg) and stirred under nitrogen for 1 h before storage at ⁇ 20° C.
  • reaction mixture was poured into a centrifuge bottle, centrifuged (10000 g, 15 min). Poured off supernatant into 250 mL bottle added DTT (3.4 g) and stirred under nitrogen for 1 h. Further purification was accomplished by ceramic hydroxyapatite (CHT-I, Bio-Rad) chromatography followed by size exclusion chromatography (SEC) (Superdex 200).
  • CHT-I ceramic hydroxyapatite
  • SEC size exclusion chromatography
  • reaction mixture was poured into a pair of centrifuge bottles and centrifuged (10000 g, 15 min). The supernatant was poured off into a 2 L glass bottle with screw cap. DTT was added (9 g), the vessel was blanketed with nitrogen and stirred for 1 h. Purification of the reaction mixture was accomplished by a combination of CHT and SEC chromatography as in Example 7.1.3.
  • the stir bar was removed from the reaction vessel and the mixture was centrifuged at high speed (10000 g, 15 min). The supernatant was poured off into a 500 mL polycarbonate bottle. DTT was added (3 g), the vessel was blanketed with nitrogen and stirred for 1 h. Purification of the reaction mixture was accomplished by a combination of CHT and SEC chromatography as in previous examples.
  • reaction mixture was transferred to a centrifuge bottle (250 mL) and centrifuged (12000 g, 15 min). The supernatant was poured off into a 250 mL bottle and DTT was added (1.5 g). The vessel was blanketed with nitrogen and stirred until the solids dissolved. Purification of the reaction mixture was accomplished by a combination of CHT and SEC chromatography as in previous preparations.
  • the material was allowed to settle ( ⁇ 5 min) and a solution of copper sulfate (80 mM, 12 mL) was added and stirring resumed.
  • the bottle was capped and the mixture stirred for 16 h at room temperature.
  • the reaction mixture was analyzed by SDS-PAGE (reducing). and the resulting gel was analyzed by densitometry which indicated a 59% conversion of starting material to the desired PEGylated product ( FIG. 16B ).
  • reaction mixture was transferred to a centrifuge bottle (500 mL) and centrifuged (10,000 g, 15 min). The resulting supernatant was transferred to a sterile polycarbonate bottle, dithiothreitol was added (6.3 g) and the solution stirred for 1 h under nitrogen. Additional purification was accomplished by CHT and SEC chromatography as done in example 7.1.3.
  • reaction mixture was transferred to a centrifuge bottle, centrifuged on tilt rotor (10,000 g, 15 min). The supernatant was poured into a new polycarbonate bottle, a stir bar and DTT (2.4 g) were added and stirred under nitrogen for 1 h. Purification was achieved by a CHT chromatography followed size exclusion chromatography as done in example 7.1.3.
  • Folding can occur by taking denatured scFv-PEG (e.g., in 8M urea) and exchanging it (e.g., by dialysis or tangential flow filtration) into a partially denaturing buffer (e.g., 3M urea) that contains a redox system (e.g., cysteine/cystine), followed by exchanging it into non-denaturing buffer (e.g., phosphate buffered saline).
  • a partially denaturing buffer e.g., 3M urea
  • a redox system e.g., cysteine/cystine
  • the scFv 28D2 with 30 kDa linear PEG bis alkyne conjugated to the C terminus was folded.
  • 28D2c-PEG was first purified and buffer exchanged into a buffer containing 9M urea and dithiothreitol (DTT), pH 7.2.
  • 28D2c-PEG was then diluted to starting concentrations of 0.05-1 mg/mL protein.
  • the starting material was then dialyzed overnight at room temperature into a first folding buffer consisting of 3M urea, 30 mM Tris pH 8.5, cysteine 2-6 mM, and cystine 1-3 mM.
  • the material was then dialyzed overnight at room temperature into the final buffer consisting of 20 mM sodium phosphate and 150 mM NaCl, pH 7.4.
  • Refolded material is seen as a monomer both by nonreducing SDS-PAGE and by SEC.
  • the recovery of monomeric 28D2c-PEG was highest at a protein folding concentration of 0.05-0.25 mg/mL protein. Similar results were achieved with cysteine:cystine concentrations ranging from 6:1 to 2:3 mM.
  • cysteine:cystine concentrations ranging from 6:1 to 2:3 mM.
  • the EC50 of the product was 116 pg/mL FIG. 17A
  • the binding affinity measured by SPR (carried on essentially as in example 1.4) is given in Table 27.
  • Folding can also occur by taking denatured scFv-PEG and rapidly diluting it into the partially denaturing buffer and then exchanging it into the non-denaturing buffer.
  • the starting material for this method can comprise scFV-PEG denatured in urea or guanidine, or denatured in SDS.
  • 28D2c-PEG in a buffer containing 9M urea and DTT, pH 7.2, 1 mg/mL was rapidly diluted to 0.05-0.1 mg/mL into a first folding buffer consisting of 3M urea, 30 mM Tris pH 8.5, cysteine 2-6 mM, and cystine 1-3 mM, and then dialyzed overnight at room temperature in the same buffer.
  • the material was then dialyzed overnight at room temperature into the final buffer consisting of 20 mM sodium phosphate and 150 mM NaCl, pH 7.4.
  • the final buffer consisting of 20 mM sodium phosphate and 150 mM NaCl, pH 7.4.
  • the EC50 of the product was 138 pg/mL.
  • 28D2c-PEG at 0.52 mg/mL protein in buffer containing 0.1% SDS and DTT, pH 7.25 was rapidly diluted into a first folding buffer consisting of 3M urea, 30 mM Tris pH 8.5, cysteine 2-6 mM, and cystine 1-3 mM, and then dialyzed overnight in the same buffer. A 200 ⁇ dilution was used, reducing the final SDS concentration to 0.0005%.
  • the folding buffer also contained 400 mM arginine and/or 150 mM NaCl.
  • the folding buffer contained 2 mM glutathinone and 2 mM oxidized glutathione in lieu of cysteine/cystine.
  • the material was then dialyzed for 3 days at 5 C into the final buffer consisting of 20 mM sodium phosphate and 150 mM NaCl, pH 7.4 The material was then concentrated 20 fold with a Millipore Centriprep concentrator (10,000 MWCO).
  • Folding can also occur by exchange and/or dilution from a starting material denatured in guanidine.
  • 28D2c-PEG was prepared in a buffer containing 6M guanidine hydrochloride and DTT, pH 8.0. The material was then dialyzed into, or rapidly diluted and then dialyzed into, folding buffers and then PBS.
  • the protein concentration in the fold was 0.05-0.25 mg/mL, and the fold buffer consisted of 3M urea, 30 mM Tris pH 8.5, cysteine 2 mM, and cystine 2 mM.
  • the fold buffer also contained 400 mM arginine, and optionally also contained 150 mM NaCl.
  • the protein concentration was 0.25 mg/mL, there was 22% monomer recovery by SEC and the EC50 of the product was 150 pg/mL
  • the scFv-PEGs were prepared in buffer containing 8M urea and DTT and diluted to 0.05-0.5 mg/mL total protein. They were then dialyzed at room temperature into fold buffer containing 3M urea, 30 mM Tris pH 8.5, 2-6 mM cysteine, and 1-3 mM cystine. Alternately, pH 8 or 9, 4M or 2M urea, 1% polysorabate 80, and/or dialysis at 4 C were also used.
  • the proteins could also be folded by dialysis into fold buffer containing no urea, with or without the addition of polysorbate 80.
  • the folding was completed by dialysis into PBS, or PBS with the redox system (2-6 mM cysteine and 1-3 mM cystine). Specific examples are given in Table 29.
  • Tm measurements of the PEGylated scFv's further confirmed the stability of these molecules. 31A12-PEG was found to have a Tm of 69.9° C. 13A8-PEG was found to have a Tm of 66.1° C.
  • Similar folding methods could be used for folding unPEGylated scFvs [e.g. 13A8c and 22H8c]. These methods could be useful for folding the proteins prior to conjugation, if desired.
  • 3 batches of 13A8c were folded in a buffer containing 3M urea, 4 mM cysteine, 2 mM cystine, 30 mM Tris, pH 8.5, with 0.1 mg/mL total protein, followed by dialysis into PBS.
  • the refolding protocol is reproducible, and the monomeric 13A8c recovery yields from 3 batches by SDS-PAGE were 37%, 35%, and 44%, respectively.
  • Refolded unPEGylated 22H8c scFv retained high potency compared to the parental mAb.
  • the next step in the generation of Anti IL-6/Anti IL-23 scFv PEG conjugates is the conjugation of an scFv containing an unnatural amino acid such as Aha to the scFv-PEG alkyne conjugate prepared in example 7.
  • the mixture is purified by a combination of chromatographies prior to undergoing a refolding process to afford the desired scFv-PEG-scFv bispecific.
  • the final materials are assessed for bioactivity and pharmacokinetic properties as well as efficacy in disease models.
  • the second chemical step in the bispecific preparation is the conjugation of the purified monovalent (scFv-PEG) to the second scFv.
  • the coupling is achieved by the reaction of the free pendant alkyne of the monovalent scFv-PEG to Aha of the second scFv via a copper mediated Huisgen cycloaddition.
  • Several monovalent scFv-PEG conjugates have been made successfully and either anti-IL-6-scFv-PEG or anti-IL-23 scFv-PEG can be used.
  • the Aha containing protein can either be an anti-IL-6 scFv or an anti-IL-23 scFv.
  • the reaction conditions differ from the copper mediated cycloaddition in the first step.
  • the reaction conditions employed an excess of PEG-bis alkyne and additives such as SDS to assist the reaction.
  • PEG-bis alkyne and additives such as SDS
  • the second step uses a much tighter ratio of alkyne to azide (1:1 to 1:3 alkyne:azide) reaction components.
  • the second step conjugation works best at higher dilution.
  • the TBTA triazole ligand utilized in the first step of the process was eventually dropped.
  • reaction mixture proceeds via a mixture of chromatography, similar to that used in example 7.
  • the Reaction mixture is first loaded onto a CHT column and eluted with a phosphate gradient.
  • the desired fractions are pooled and then loaded onto a SEC column. This material can then be further processed for refolding conditions.
  • the conjugation precedes the folding.
  • the presence of the PEG linker facilitates the subsequent refolding step and the scFvs refold independently with minimal interchain crosslinking.
  • interchain crosslinking is a serious impediment often occurring with bispecific constructs linked by genetic fusion and have no PEG linker to prevent the interaction of the antigen binding domains.
  • the reaction mixture was split into two centrifuge bottles (500 mL) and centrifuged (10000 g, 30 min) and the supernatant was disposed.
  • the bottle was nutated and swirled till materials were dissolved or thoroughly suspended. Contents were transferred to the second centrifuge bottle/pellet and rinsed out the first centrifuge bottle with 2 portions of sodium phosphate buffer (10 mM, 12.5 mL).
  • the second centrifuge bottle was swirled until the pellet was dissolved.
  • the material was centrifuged (10,000 g, 5 min). The supernatant was retained for further purification.
  • the pooled 6500 mL reaction volume was processed as follows. Into two centrifuge bottles (500 mL) placed approximately 450 mL of reaction mixture into each bottle. Centrifuged in swinging bucket centrifuge (5000 g, 15 min). Disposed of supernatant, added additional reaction mixture to each collection centrifuge bottle and centrifuge material again. Repeated sequence until all pooled reaction volume had been centrifuged and the pellet ( ⁇ 2) retained. To each bottle added a stir bar and the following buffer (700 ml): 250 mM DTT, 2% SDS. 10 mM sodium phosphate buffer. Stirred at room temperature for 30 min. Placed in water bath (40° C.) and stirred for 10 min. Stirred an additional 30 min at which point no solids remained. The two solutions were pooled before being loaded onto a CHT column. Elution with a phosphate gradient. The desired fractions are pooled with additional purification by size exclusion column.
  • the three reaction volumes were combined and processed as follows. Into two centrifuge bottles (250 mL) placed approximately 200 mL of reaction mixture into each bottle. Centrifuged in swing bucket centrifuge (5000 g, 15 min). Disposed of supernatant, added additional reaction mixture to each collection centrifuge bottle and centrifuge material again. Repeated sequence until all reaction mixture from the three reactions has been centrifuged and the pellet retained. To each bottle added a stir bar and the following buffer (220 ml): 250 mM DTT, 2% SDS. 10 mM sodium phosphate buffer. Stirred at room temperature for 30 min. Placed in water bath (40° C.) and stirred for 10 min. Solids remained.
  • the combined 6900 mL reaction volume was processed analogously to that previously described.
  • Into two 500 mL centrifuge bottles (500 mL) was placed approximately 450 mL ( ⁇ 2) of reaction volume. The mixture was centrifuged in a swinging bucket centrifuge (5000 g, 15 min). The supernatant was disposed, and additional reaction mixture was added to each collection centrifuge bottle and the centrifuged again. The was repeated the entire 6900 mL was processed.
  • To each pellet was added the following buffer (700 mL): 250 mM DTT, 2% SDS. 10 mM sodium phosphate buffer. Stirred at room temperature for 30 min. The solids were broken up with a spatula and stirring was resumed for an additional 1 h. The two solutions were combined and loaded onto a CHT column with elution by a phosphate gradient. Additional purification of the semi-pure bispecific was accomplished by SEC chromatography.
  • a sodium phosphate buffer (5 mM stock solution, 2100 mL).
  • MES buffer 80 mM stock, 141 mL
  • a solution of dithiothreitol 250 mM stock, 12 mL
  • the pooled 8500 mL reaction volume was centrifuged to collect all solids.
  • the solids were dissolved in the following workup buffer (1700 mL): 250 mM DTT, 2% SDS. 10 mM sodium phosphate buffer.
  • the final solution was purified by a combination of CHT and SEC chromatography.
  • 31A12 conjugated to 13A8 via a linear 20 kDa PEG linker (both conjugated at the C termini) was folded by methods similar to those given above.
  • the bispecific molecule was prepared in a buffer containing 8M urea and DTT, pH 7.3, at 0.05-0.1 mg/mL total protein.
  • the material, at this stage, might contain some amount of residual unreacted 31A12-PEG scFv in addition to the bispecific molecule.
  • the material was then folded by dialyzing overnight at room temperature into 3M urea, 30 mM Tris pH 8.5, 4 mM cysteine, 2 mM cystine.
  • 1% polysorbate 80, 500 mM Tris, or 500 mM Arginine were also added to the fold buffer, or the folding could be run at 4 C.
  • the folding reaction was further dialyzed into 20 mM sodium phosphate, 150 mM NaCl, pH 7.4 (PBS).
  • PBS sodium phosphate
  • 4 batches of 0.1 mg/mL of 31A12c-PEG-13A8c were refolded in 3M urea, 30 mM Tris pH 8.5, 4 mM cysteine, 2 mM cystine at room temperature, followed by dialysis into PBS.
  • the refolding protocol is reproducible, resulting in similar monomeric bispecific scFv recovery yields and EC50s (Table 30, FIGS.
  • the in vivo pharmacokinetics of the bispecific, made with a 20 kDa linear PEG linker was compared to the PK of a naked scFv.
  • the scFv alone was excreted very rapidly, with a terminal t 1/2 of about 2 h, Cmaxof 500 pg/ml and tmax of 1-2 h, being nearly completely cleared by 8 h ( FIG. 20A ),
  • the bispecific shows a much longer half life in vivo with a terminal t 1/2 of about 24 h, Cmaxof 1500 pg/ml, tmax of 24 h, and detectable levels in the serum at 100 h ( FIG. 20B ).
  • This improvement in the pharmacokinetic behavior of the bispecific scFv will make it a much more potent and effective therapeutic than a simple scFv.
  • 13A8n-PEG-45G5c was folded by similar methods to those for the 31A12 based bispecific. 0.1 mg/mL of protein was folded in 3M urea, 30 mM Tris pH 8.5, 4 mM cysteine, 2 mM cystine, followed by dialysis into PBS. This resulted in 29% monomer recovery by SEC, and EC50s of 933 pg/mL against IL-6 and 5,662 pg/mL against IL-23 ( FIGS. 22 A and B).
  • 13A8c-PEG-22H8c was folded by similar methods as above.
  • the 13A8c-PEG-22H8c was prepared in buffer containing 8M urea and DTT and diluted to 0.05-0.1 mg/mL total protein. They were then dialyzed at room temperature into fold buffer containing 3M urea, 30 mM Tris pH 8.5, 2-6 mM cysteine, and 1-3 mM cystine. Alternately, pH 8 or 9, 0.01-1% polysorabte 80, and/or dialysis at 4 C were also used. The folding was completed by dialysis into PBS.
  • 0.1 mg/mL of 13A8c-PEG-22H8c was folded in 3M urea, 30 mM Tris pH 8.5, 4 mM cysteine, 2 mM cystine, 0.05% polysorabte 80, and at room temperature, followed by dialysis into PBS, containing 0.05% polysorbate 80. This resulted in 35% monomer recovery by SEC, and EC50 of 246 pg/mL for neutralization of IL-6 and 234 pg/mL for neutralization of IL-23 ( FIGS. 23 A and B).
  • 13A8c-40 KPEG-31A12c was folded by similar methods to those for the 20K bispecifics. 0.1 mg/mL of protein was folded in 3M urea, 30 mM Tris pH 8.5, 4 mM cysteine, 2 mM cystine at 4° C., followed by dialysis into PBS at 4° C. This resulted in 56.3% recovery by SEC, and EC50s of 137.5 pg/mL against IL-6 and 2699 pg/mL against IL-23 ( FIGS. 24 A and B).
  • the 13A8c-40 KPEG-31A12c could also be refolded at higher concentration (0.5 mg/mL) by dialysis into refolding buffer containing 0.5M guanidine hydrochloride at both room temperature and 4° C.
  • the in vivo pharmacokinetics of the 13A8c-40 KPEG-31A12c bispecific was compared to the PK of the 13A8c-20 KPEG-31A12c, 13A8c-PEG and 28D2 naked ( FIGS. 25A and B).
  • the test articles were dosed subcutaneously in rats at 1 mg/kg (bispecifics and naked scFv) or 0.5 mg/kg (13A8c-PEG). Blood was collected at time intervals after dosing, and serum was assayed for the presence of test article using the B9 IL-6 neutralization assay. The results show that naked scFv is rapidly cleared, while the bispecifics and 13A8c-PEG have a significantly greater half-life and AUC. 13A8c-40 KPEG-31A12c also shows a significantly enhanced AUC relative to 13A8c-20 KPEG-31A12c ( FIG. 25B ).
  • Th17 and Th22 T cell subsets can be differentiated in vitro either by stimulation of whole PBMC or purified T cells with anti-CD3 plus anti CD28, or by allogeneic cells in mixed lymphocyte cultures ( FIG. 26A ).
  • the differentiation of such T cells requires further addition of a number of key regulatory cytokines that are especially well characterized for Th17 cells. These regulatory cytokines are primarily derived from myeloid cells and their addition can be replaced with the addition of myeloid cells along with a compound that stimulates those cells to release their regulatory cytokines. LPS was used with anti CD3 to stimulate whole PBMC to differentiate into Th17 cells.
  • Th17 and Th22 T cells can also be differentiated in allogeneic mixed lymphocyte cultures (MLC) with the addition of a stimulant to induce the myeloid cells to release their regulatory cytokines.
  • MLC allogeneic mixed lymphocyte cultures
  • Peptidoglycan was added to the MLC as that stimulant, as it is known to induce the secretion of IL-1, IL-6, TNF and other regulatory and proinflammatory cytokines. Addition of IL-2 was also required when the goal was to study the induction of Th22 cells.
  • PBMC peripheral blood mononuclear cells
  • PBMC peripheral blood mononuclear cells
  • PMA+ ionomycin to induce cytokine secretion and analyzed by flow cytometry for the expression of IL-17.
  • Th22 cells are also seen in the anti CD3/28 stimulation ( FIG. 26C ).
  • the in vitro stimulation of human T cells in vitro using allogeneic leukocytes also induced high levels of IL-17 producing T cells ( FIG. 26D ).
  • a xenograft model was employed in which human hematopoietic stem cells are transplanted into immunodeficient mice which in turn acquire a human immune system.
  • These humanized NOD-scid IL2 null (NSG) mice are transplanted with human skin allogeneic with the human immune cells populating the mice ( FIG. 30 ). They are then treated with a mixture of PEGylated scFv antagonists for IL-6 and IL-23 (13A8c-PEG and 31A12c-PEG). The human immune system will then reject this allogeneic human skin via the differentiation of human T cells into effector cells.
  • the IL-6 and IL-23 antagonists inhibited the differentiation of Th17 cells which is one consequence of allogeneic skin transplantation, but these antagonists did not inhibit the rejection of the skin allograft, reflecting their targeted immunosuppressive effects.
  • newborn NSG mice were irradiated and injected with human hematopoietic stem cells derived from umbilical cord blood and then screened for engraftment levels in the peripheral blood at 12 weeks (Brehm et al, 2010). Mice that were successfully engrafted were transplanted with human allogeneic skin and received 100 ⁇ g of anti IL-6 and anti IL-23 (13A8c-PEG and 31A12c-PEG) every 2 days.
  • CD3+/CD4+ cells were analyzed for IL-17 and IL-22 production by flow cytometry.
  • mice that were untreated with cytokine antagonists very significant levels of TH17 and TH22 cells developed as shown in the flow cytometry profiles ( FIG. 31A ) and in the compiled data representing the numbers of Th cells in each subset ( FIG. 31B ).
  • Mice were treated for 30 days, after skin transplantation, with a combination of anti IL-6 (13A8c scFv-PEG) and anti IL-23 (31A12c-PEG). The differentiation of TH17 and TH22 cells in treated mice was completely inhibited. These data clearly demonstrate, for the first time, that IL-6 and IL-23 are required for the in vivo differentiation of these TH17 and TH22 cells.
  • FIG. 32 A-C Similar results were obtained with the 13A8c-20kPEG-31A12c anti IL-6/anti IL-23 bispecific. As shown in FIG. 32 A-C, the bispecific molecule is more effective at inhibiting Th17 differentiation than the monovalent anti IL-23 reagent. However, FIG. 32D-L demonstrate that the bispecific is not generally immunosuppressive as leukocyte markers for cell types other than TH17/22 were not significantly reduced.
  • a scid/hu psoriasis model was used, in which human psoriatic skin was implanted onto immunodeficient scid mice. The skin engrafts and the psoriatic inflammation persists for up to 2 months. The mice are treated for two weeks with drugs and effects on the inflammation are measured by histological analysis, as shown in FIG. 33 , in which the effects of the 13A8c-20kPEG-31A12c anti IL-6/anti IL-23 bispecific can be clearly seen in the significant reduction in epidermal thickness. The effect can also be quantitated from the histological sections.
  • mice with 13A8c-PEG-31A12c made with either a 20 kDa PEG or a 40 kDa PEG were very effective inhibitors of ear swelling, even when only administered on the day before the IL-23 treatment began ( FIG. 35D ).
  • the 31A12 mAbs binds a unique epitope that has not been previously described. All of the mAbs used bind to human IL-12 ( FIG. 36B ) even though 31A12 and 49B7 are specific for IL-23 inhibition and do not inhibit human IL-12 ( FIG. 36B ). These data clearly indicate that these mAbs bind to the p40 chain. 31A12 and 49B7 bind relatively weakly to human IL-12 compared to 22H8, which also inhibits IL-12. However, all three mAbs bind strongly to monkey IL-12 ( FIG. 36B ) and also inhibit monkey IL-12 bioactivity ( FIG. 36C ). Thus 31A12 and 49B7 distinguish human and monkey IL-12 activity.
  • 31A12 and 49B7 see a p40 epitope that is partially masked in human IL-12, and exposed in monkey IL-12 as well as IL-23 from both species. Moreover, AZ17 does not inhibit the binding of Ustekinumab, a p40 specific mAb that inhibits both human IL-12 and IL-23.
US13/791,312 2010-09-10 2013-03-08 Antibody derivatives Abandoned US20140127209A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US13/791,312 US20140127209A1 (en) 2010-09-10 2013-03-08 Antibody derivatives
US14/886,978 US10202447B2 (en) 2010-09-10 2015-10-19 Antibody derivatives

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US38178910P 2010-09-10 2010-09-10
PCT/EP2011/065697 WO2012032181A2 (fr) 2010-09-10 2011-09-09 Nouveaux dérivés d'anticorps
US13/791,312 US20140127209A1 (en) 2010-09-10 2013-03-08 Antibody derivatives

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2011/065697 Continuation WO2012032181A2 (fr) 2010-09-10 2011-09-09 Nouveaux dérivés d'anticorps

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US14/886,978 Continuation US10202447B2 (en) 2010-09-10 2015-10-19 Antibody derivatives

Publications (1)

Publication Number Publication Date
US20140127209A1 true US20140127209A1 (en) 2014-05-08

Family

ID=44583080

Family Applications (2)

Application Number Title Priority Date Filing Date
US13/791,312 Abandoned US20140127209A1 (en) 2010-09-10 2013-03-08 Antibody derivatives
US14/886,978 Expired - Fee Related US10202447B2 (en) 2010-09-10 2015-10-19 Antibody derivatives

Family Applications After (1)

Application Number Title Priority Date Filing Date
US14/886,978 Expired - Fee Related US10202447B2 (en) 2010-09-10 2015-10-19 Antibody derivatives

Country Status (5)

Country Link
US (2) US20140127209A1 (fr)
EP (1) EP2640745B1 (fr)
JP (1) JP6173911B2 (fr)
CN (1) CN103261222B (fr)
WO (1) WO2012032181A2 (fr)

Cited By (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20140371133A1 (en) * 2011-06-22 2014-12-18 Apellis Pharmaceuticals, Inc. Methods of treating chronic disorders with complement inhibitors
US20150017187A1 (en) * 2013-07-10 2015-01-15 Sutro Biopharma, Inc. Antibodies comprising multiple site-specific non-natural amino acid residues, methods of their preparation and methods of their use
US9682934B2 (en) 2012-08-31 2017-06-20 Sutro Biopharma, Inc. Modified amino acids
US9732161B2 (en) 2012-06-26 2017-08-15 Sutro Biopharma, Inc. Modified Fc proteins comprising site-specific non-natural amino acid residues, conjugates of the same, methods of their preparation and methods of their use
US9738724B2 (en) 2012-06-08 2017-08-22 Sutro Biopharma, Inc. Antibodies comprising site-specific non-natural amino acid residues, methods of their preparation and methods of their use
US9981908B2 (en) 2013-08-05 2018-05-29 Medimmune Limited Amino acid derivatives
US20180201926A1 (en) * 2016-12-23 2018-07-19 Visterra, Inc. Binding polypeptides and methods of making the same
WO2019075487A1 (fr) * 2017-10-13 2019-04-18 Ursure, Inc. Harvard Life Lab Produits et procédés pour surveiller l'adhésion à une thérapie par inhibiteurs nucléosidiques de la transcriptase inverse (inti)
US10435479B2 (en) 2014-05-10 2019-10-08 Sorrento Therapeutics, Inc. Chemically-locked bispecific antibodies
US10738111B2 (en) 2014-12-19 2020-08-11 Chugai Seiyaku Kabushiki Kaisha Anti-myostatin antibodies, polypeptides containing variant Fc regions, and methods of use
WO2020257586A2 (fr) 2019-06-20 2020-12-24 Baxalta Incorporated Méthode de traitement avec une thérapie génique à base de virus
US10906990B2 (en) 2015-08-19 2021-02-02 Riken Antibody with non-natural amino acid introduced therein
US10919953B2 (en) 2012-08-24 2021-02-16 Chugai Seiyaku Kabushiki Kaisha FcgammaRIIB-specific Fc region variant
US11046784B2 (en) 2006-03-31 2021-06-29 Chugai Seiyaku Kabushiki Kaisha Methods for controlling blood pharmacokinetics of antibodies
US11053308B2 (en) 2016-08-05 2021-07-06 Chugai Seiyaku Kabushiki Kaisha Method for treating IL-8-related diseases
US11180548B2 (en) 2015-02-05 2021-11-23 Chugai Seiyaku Kabushiki Kaisha Methods of neutralizing IL-8 biological activity
US11236168B2 (en) 2012-08-24 2022-02-01 Chugai Seiyaku Kabushiki Kaisha Mouse FcγammaRII-specific Fc antibody
US11248053B2 (en) 2007-09-26 2022-02-15 Chugai Seiyaku Kabushiki Kaisha Method of modifying isoelectric point of antibody via amino acid substitution in CDR
US11267868B2 (en) 2013-04-02 2022-03-08 Chugai Seiyaku Kabushiki Kaisha Fc region variant
US11359009B2 (en) 2015-12-25 2022-06-14 Chugai Seiyaku Kabushiki Kaisha Anti-myostatin antibodies and methods of use
US11359194B2 (en) 2008-04-11 2022-06-14 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule capable of binding two or more antigen molecules repeatedly
US11400213B2 (en) 2015-03-04 2022-08-02 Repro-Med Systems, Inc. Precision variable flow rate infusion system and method
CN115957319A (zh) * 2022-10-14 2023-04-14 北京东方百泰生物科技股份有限公司 一种抗nkg2a单克隆抗体的注射制剂
US11708413B2 (en) 2016-01-27 2023-07-25 Sutro Biopharma, Inc. Anti-CD74 antibody conjugates, compositions comprising anti-CD74 antibody conjugates and methods of using anti-CD74 antibody conjugates
US11891434B2 (en) 2010-11-30 2024-02-06 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule capable of binding to plurality of antigen molecules repeatedly

Families Citing this family (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2707840A1 (fr) 2007-08-20 2009-02-26 Allozyne, Inc. Molecules substituees par des acides amines
CA3017116A1 (fr) 2010-11-04 2012-05-10 Boehringer Ingelheim International Gmbh Anticorps anti-il-23
MX343324B (es) 2011-07-06 2016-11-01 Nestec Sa Ensayos para detectar auto anticuerpos neutralizadores de la terapia biologica con tnf alpha.
EP2583979B1 (fr) 2011-10-19 2015-12-16 Effimune Méthodes de preparation d'anticorps dirigés contre la sous-unité p19 de l'IL-23 humaine
ES2908474T3 (es) 2012-05-03 2022-04-29 Boehringer Ingelheim Int Anticuerpos anti-IL-23p19
US9670521B2 (en) * 2012-09-24 2017-06-06 Medimmune Limited Amino acid derivatives
EP2970949B1 (fr) 2013-03-15 2018-05-30 MedImmune Limited Nouvelles molécules d'acide nucléique
EP3708679A1 (fr) 2014-07-24 2020-09-16 Boehringer Ingelheim International GmbH Biomarqueurs utiles dans le traitement de maladies liées à l'il-23a
TWI711629B (zh) 2014-09-03 2020-12-01 德商包林格因蓋爾漢國際股份有限公司 靶向IL-23A與TNF-α之化合物及其用途
BR112017011545A2 (pt) 2014-12-05 2018-03-13 Nestec Sa ensaio de desvio de mobilidade homogênea indireto para a detecção de produtos biológicos nas amostras do paciente
CN106924754B (zh) * 2015-12-30 2021-11-09 北京大学 聚氨基酸偶联物
CA3060225A1 (fr) * 2017-05-09 2018-11-15 Cyano Biotech Gmbh Microcystines et nodularines modifiees
CN110283249A (zh) * 2018-03-19 2019-09-27 江苏莱博德医疗科技有限公司 一种抗白介素6的人ScFv单链抗体
US20190292310A1 (en) * 2018-03-20 2019-09-26 Northeastern University Synthesis of high density molecular dna brushes via organic-phase ring-opening metathesis (co)polymerization
SG11202010025YA (en) 2018-05-31 2020-11-27 Glyconex Inc Therapeutic antibodies binding to biantennary lewis b and lewis y antigens
US20210395298A1 (en) * 2018-10-11 2021-12-23 Amgen Inc. Downstream processing of bispecific antibody constructs
CN113203863B (zh) * 2021-04-28 2022-01-21 北京美联泰科生物技术有限公司 一种适用于白介素-6检测的缓冲液
CN113150122B (zh) * 2021-05-19 2023-03-17 上海儒克生物科技有限公司 高通量全兔源单克隆抗体的制备方法
WO2022251119A2 (fr) 2021-05-24 2022-12-01 Vir Biotechnology, Inc. Polypeptides modifiés

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007130453A2 (fr) * 2006-05-02 2007-11-15 Allozyne, Inc. Molécules substituées par des acides aminés

Family Cites Families (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1991016449A1 (fr) 1990-04-16 1991-10-31 The Trustees Of The University Of Pennsylvania Compositions saccharides et procedes et appareil servant a les synthetiser
US5151510A (en) 1990-04-20 1992-09-29 Applied Biosystems, Inc. Method of synethesizing sulfurized oligonucleotide analogs
GB9014932D0 (en) 1990-07-05 1990-08-22 Celltech Ltd Recombinant dna product and method
WO1994004679A1 (fr) 1991-06-14 1994-03-03 Genentech, Inc. Procede pour fabriquer des anticorps humanises
JPH05244982A (ja) 1991-12-06 1993-09-24 Sumitomo Chem Co Ltd 擬人化b−b10
GB9915247D0 (en) 1999-07-01 1999-09-01 Amat Limited Improvements relating to tyre degradation
EP1339427A4 (fr) 2000-11-01 2004-09-15 Elusys Therapeutics Inc Procede de production de molecules bispecifiques par transepissage de proteines
EP2796546B1 (fr) 2001-04-19 2017-08-09 The Scripps Research Institute Incorporation d'acides aminés artificiels
US20060073141A1 (en) 2001-06-28 2006-04-06 Domantis Limited Compositions and methods for treating inflammatory disorders
EP2292271A3 (fr) 2001-10-10 2011-09-14 BioGeneriX AG Remodelage et glycoconjugation des anticorps
WO2004035605A2 (fr) 2002-10-16 2004-04-29 The Scripps Research Institute Synthese de glycoproteines
JP2006523090A (ja) * 2002-12-27 2006-10-12 ドマンティス リミテッド リガンドに、そしてリガンド受容体に特異的な二重特異性単一ドメイン抗体
US20050287639A1 (en) 2004-05-17 2005-12-29 California Institute Of Technology Methods of incorporating amino acid analogs into proteins
WO2006009901A2 (fr) 2004-06-18 2006-01-26 Ambrx, Inc. Nouveaux polypeptides de liaison a l'antigene et leurs utilisations
JP2008504356A (ja) * 2004-06-30 2008-02-14 ドマンティス リミテッド 炎症性疾患を治療するための組成物及び方法
TW200708792A (en) 2005-08-24 2007-03-01 Asia Optical Co Inc Retractable lens system and turnover lens-receiving mechanism thereof
EP2064242A1 (fr) * 2007-02-23 2009-06-03 Schering Corporation Anticorps obtenus par génie génétiques dirigés contre l'il-23p19
NZ579297A (en) * 2007-02-28 2012-03-30 Schering Corp Combination therapy comprising an il-23 antagonist and a cytokine antagonist for treatment of immune disorders
US8188235B2 (en) * 2008-06-18 2012-05-29 Pfizer Inc. Antibodies to IL-6 and their uses
US20110158992A1 (en) * 2008-08-27 2011-06-30 Schering Corporation Engineered Anti-IL-23R Antibodies
TWI440469B (zh) * 2008-09-26 2014-06-11 Chugai Pharmaceutical Co Ltd Improved antibody molecules
CN102301235B (zh) * 2008-11-28 2014-11-19 Abbvie公司 稳定的抗体组合物和用于稳定其的方法
SG172354A1 (en) * 2009-01-29 2011-07-28 Medimmune Llc Human anti-il-6 antibodies with extended in vivo half-life and their use in treatment of oncology, autoimmune diseases and inflammatory diseases

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007130453A2 (fr) * 2006-05-02 2007-11-15 Allozyne, Inc. Molécules substituées par des acides aminés

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
Casset et al. BBRC(2003) 307, 198-205. *
Janeway et al., Immunology Third Edition, Garland Publishing Inc. 1997, Chapter 3, Structure of the Antibody Molecule and Immunoglobulin Genes, pages 3:1-3:11. *
Portolano et al., Journal of Immunology, 1993 150:880-887. *
Rudikoff et al. PNAS 1982 Vol 79 page 1979-1983. *
William E. Paul, M.D. FUNDAMENTAL IMMUNOLOGY, 3rd edition. 1993, page 242. *

Cited By (44)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11046784B2 (en) 2006-03-31 2021-06-29 Chugai Seiyaku Kabushiki Kaisha Methods for controlling blood pharmacokinetics of antibodies
US11248053B2 (en) 2007-09-26 2022-02-15 Chugai Seiyaku Kabushiki Kaisha Method of modifying isoelectric point of antibody via amino acid substitution in CDR
US11371039B2 (en) 2008-04-11 2022-06-28 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule capable of binding to two or more antigen molecules repeatedly
US11359194B2 (en) 2008-04-11 2022-06-14 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule capable of binding two or more antigen molecules repeatedly
US11891434B2 (en) 2010-11-30 2024-02-06 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule capable of binding to plurality of antigen molecules repeatedly
US10039802B2 (en) * 2011-06-22 2018-08-07 Apellis Pharmaceuticals, Inc. Methods of treating chronic disorders with complement inhibitors
US11712460B2 (en) 2011-06-22 2023-08-01 Apellis Pharmaceuticals, Inc. Methods of treating chronic disorders with complement inhibitors
US20140371133A1 (en) * 2011-06-22 2014-12-18 Apellis Pharmaceuticals, Inc. Methods of treating chronic disorders with complement inhibitors
US11013782B2 (en) 2011-06-22 2021-05-25 Apellis Pharmaceuticals, Inc. Methods of treating chronic disorders with complement inhibitors
US10669347B2 (en) 2012-06-08 2020-06-02 Sutro Biopharma, Inc. Antibodies comprising site-specific non-natural amino acid residues, methods of their preparation and methods of their use
US11958909B2 (en) 2012-06-08 2024-04-16 Sutro Biopharma, Inc. Antibodies comprising site-specific non-natural amino acid residues, methods of their preparation and methods of their use
US9738724B2 (en) 2012-06-08 2017-08-22 Sutro Biopharma, Inc. Antibodies comprising site-specific non-natural amino acid residues, methods of their preparation and methods of their use
US10501558B2 (en) 2012-06-26 2019-12-10 Sutro Biopharma, Inc. Modified Fc proteins comprising site-specific non-natural amino acid residues, conjugates of the same, methods of their preparation and methods of their use
US9732161B2 (en) 2012-06-26 2017-08-15 Sutro Biopharma, Inc. Modified Fc proteins comprising site-specific non-natural amino acid residues, conjugates of the same, methods of their preparation and methods of their use
US11236168B2 (en) 2012-08-24 2022-02-01 Chugai Seiyaku Kabushiki Kaisha Mouse FcγammaRII-specific Fc antibody
US10919953B2 (en) 2012-08-24 2021-02-16 Chugai Seiyaku Kabushiki Kaisha FcgammaRIIB-specific Fc region variant
US10112900B2 (en) 2012-08-31 2018-10-30 Sutro Biopharma, Inc. Modified amino acids
US9994527B2 (en) 2012-08-31 2018-06-12 Sutro Biopharma, Inc. Modified amino acids
US9682934B2 (en) 2012-08-31 2017-06-20 Sutro Biopharma, Inc. Modified amino acids
US10730837B2 (en) 2012-08-31 2020-08-04 Sutro Biopharma, Inc. Modified amino acids
US11548852B2 (en) 2012-08-31 2023-01-10 Sutro Biopharma, Inc. Modified amino acids
US11267868B2 (en) 2013-04-02 2022-03-08 Chugai Seiyaku Kabushiki Kaisha Fc region variant
US20150017187A1 (en) * 2013-07-10 2015-01-15 Sutro Biopharma, Inc. Antibodies comprising multiple site-specific non-natural amino acid residues, methods of their preparation and methods of their use
US11344626B2 (en) 2013-07-10 2022-05-31 Sutro Biopharma, Inc. Antibodies comprising multiple site-specific non-natural amino acid residues, methods of their preparation and methods of their use
US9764039B2 (en) * 2013-07-10 2017-09-19 Sutro Biopharma, Inc. Antibodies comprising multiple site-specific non-natural amino acid residues, methods of their preparation and methods of their use
US9981908B2 (en) 2013-08-05 2018-05-29 Medimmune Limited Amino acid derivatives
US10435479B2 (en) 2014-05-10 2019-10-08 Sorrento Therapeutics, Inc. Chemically-locked bispecific antibodies
US11454633B2 (en) 2014-12-19 2022-09-27 Chugai Seiyaku Kabushiki Kaisha Anti-myostatin antibodies, polypeptides containing variant Fc regions, and methods of use
US10738111B2 (en) 2014-12-19 2020-08-11 Chugai Seiyaku Kabushiki Kaisha Anti-myostatin antibodies, polypeptides containing variant Fc regions, and methods of use
US11180548B2 (en) 2015-02-05 2021-11-23 Chugai Seiyaku Kabushiki Kaisha Methods of neutralizing IL-8 biological activity
US11400213B2 (en) 2015-03-04 2022-08-02 Repro-Med Systems, Inc. Precision variable flow rate infusion system and method
US11787877B2 (en) 2015-08-19 2023-10-17 Riken Antibody with non-natural amino acid introduced therein
US10906990B2 (en) 2015-08-19 2021-02-02 Riken Antibody with non-natural amino acid introduced therein
US11359009B2 (en) 2015-12-25 2022-06-14 Chugai Seiyaku Kabushiki Kaisha Anti-myostatin antibodies and methods of use
US11708413B2 (en) 2016-01-27 2023-07-25 Sutro Biopharma, Inc. Anti-CD74 antibody conjugates, compositions comprising anti-CD74 antibody conjugates and methods of using anti-CD74 antibody conjugates
US11780912B2 (en) 2016-08-05 2023-10-10 Chugai Seiyaku Kabushiki Kaisha Composition for prophylaxis or treatment of IL-8 related diseases
US11053308B2 (en) 2016-08-05 2021-07-06 Chugai Seiyaku Kabushiki Kaisha Method for treating IL-8-related diseases
CN110366557A (zh) * 2016-12-23 2019-10-22 威特拉公司 结合多肽及其制备方法
US11820979B2 (en) * 2016-12-23 2023-11-21 Visterra, Inc. Binding polypeptides and methods of making the same
US20180201926A1 (en) * 2016-12-23 2018-07-19 Visterra, Inc. Binding polypeptides and methods of making the same
US11795239B2 (en) 2017-10-13 2023-10-24 Orasure Technologies, Inc. Products and methods for monitoring adherence to nucleoside reverse transcriptase inhibitor therapy
WO2019075487A1 (fr) * 2017-10-13 2019-04-18 Ursure, Inc. Harvard Life Lab Produits et procédés pour surveiller l'adhésion à une thérapie par inhibiteurs nucléosidiques de la transcriptase inverse (inti)
WO2020257586A2 (fr) 2019-06-20 2020-12-24 Baxalta Incorporated Méthode de traitement avec une thérapie génique à base de virus
CN115957319A (zh) * 2022-10-14 2023-04-14 北京东方百泰生物科技股份有限公司 一种抗nkg2a单克隆抗体的注射制剂

Also Published As

Publication number Publication date
EP2640745B1 (fr) 2018-11-07
WO2012032181A3 (fr) 2012-05-24
EP2640745A2 (fr) 2013-09-25
WO2012032181A9 (fr) 2013-06-13
WO2012032181A2 (fr) 2012-03-15
US10202447B2 (en) 2019-02-12
CN103261222B (zh) 2017-07-28
CN103261222A (zh) 2013-08-21
US20160194391A1 (en) 2016-07-07
JP6173911B2 (ja) 2017-08-09
JP2013545438A (ja) 2013-12-26

Similar Documents

Publication Publication Date Title
US10202447B2 (en) Antibody derivatives
CA2396212C (fr) Anticorps anti-interleukine-1 beta humain
TWI501976B (zh) 抗TNF-α之抗體及其用途
EP2326668B1 (fr) Anticorps anti-il-12/il-23
KR100603899B1 (ko) 인간 mcp-1에 대한 항체
KR20030034140A (ko) 인간 IL-1β에 대한 항체
TW201531484A (zh) 抗TNF-α之抗體及其用途
JP6865826B2 (ja) インターロイキン17aを標的とする抗体、その製造方法及び応用
JP6604204B2 (ja) 新規抗ヒトbdca−2抗体
KR20180089514A (ko) Tnf-알파, il-17a 및 il-17f에 대해 특이성을 갖는 다중특이적 항체 분자
JP5971238B2 (ja) 新規抗ヒトil−23受容体抗体
EP2796550B1 (fr) Nouvel anticorps anti ctgf humain
KR20210102946A (ko) 항il-17a 항체 및 이의 응용
CN116554323A (zh) 人源化抗il21抗体的开发和应用
CN115960240A (zh) 一种同时靶向人bcma和人cd3的双特异性抗体
WO2022247826A1 (fr) Protéine de liaison spécifique ciblant pd-l1 et cd73
WO2024018242A1 (fr) Molécules de liaison ciblant il-35r

Legal Events

Date Code Title Description
AS Assignment

Owner name: ALLOZYNE, INC., WASHINGTON

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:GRABSTEIN, KENNETH;BRADY, WILLIAM;KING, GORDON;AND OTHERS;SIGNING DATES FROM 20130509 TO 20130527;REEL/FRAME:030614/0720

STCB Information on status: application discontinuation

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

AS Assignment

Owner name: MEDIMMUNE LIMITED, UNITED KINGDOM

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:ALLOZYNE, INC.;REEL/FRAME:037333/0842

Effective date: 20140506