WO2007019621A1 - Chimeric antibodies with new world primate regions - Google Patents

Chimeric antibodies with new world primate regions Download PDF

Info

Publication number
WO2007019621A1
WO2007019621A1 PCT/AU2006/001166 AU2006001166W WO2007019621A1 WO 2007019621 A1 WO2007019621 A1 WO 2007019621A1 AU 2006001166 W AU2006001166 W AU 2006001166W WO 2007019621 A1 WO2007019621 A1 WO 2007019621A1
Authority
WO
WIPO (PCT)
Prior art keywords
antigen
binding portion
antibody
chimeric antibody
cdr
Prior art date
Application number
PCT/AU2006/001166
Other languages
English (en)
French (fr)
Inventor
Philip Anthony Jennings
Anthony Gerard Doyle
Adam William Clarke
Original Assignee
Arana Therapeutics Limited
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from AU2005904406A external-priority patent/AU2005904406A0/en
Application filed by Arana Therapeutics Limited filed Critical Arana Therapeutics Limited
Priority to NZ565904A priority Critical patent/NZ565904A/en
Priority to BRPI0614867-0A priority patent/BRPI0614867A2/pt
Priority to AU2006281981A priority patent/AU2006281981A1/en
Priority to EP06774813A priority patent/EP1945669A4/en
Priority to JP2008526323A priority patent/JP2009504686A/ja
Priority to CA002619245A priority patent/CA2619245A1/en
Priority to MX2008002161A priority patent/MX2008002161A/es
Publication of WO2007019621A1 publication Critical patent/WO2007019621A1/en
Priority to NO20080800A priority patent/NO20080800L/no

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • 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
    • C07K2317/50Immunoglobulins 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]

Definitions

  • the present, invention relates to a chimeric antibody or antigen-binding portion thereof, wherein the antigen binding portion comprises at least two complementarity determining region (CDR) sequences and at least three framework regions, wherein at least one CDR is a New World primate CDR, and to the use of the antibody or antigen -binding portion thereof in treating diseases or disorders.
  • CDR complementarity determining region
  • Antibodies play an important role in the immune system of a mammal. They are produced by plasma cells which have developed from precursor B cells. Antibodies consist ⁇ f two identical light polypeptide chains and two identical heavy polypeptide chains which are joined by disulfide bridges. The light chains are referred to as either kappa or lambda light chains and the heavy chains as gamma, mu, delta, alpha or cpsilon. Each chain consists of a constant and variable region. The variable region gives the antibody its specificity. Within each variable region are regions of hypervari ability or complementarity determining regions (CDRs) which are flanked by more conserved regions referred to as framework regions. Within each variable region arc three CDRs and four framework regions.
  • CDRs complementarity determining regions
  • Antibodies are bifunctional molecules, the N-terminai variable segments from the heavy and light chains associate together in a specific manner to generate a three-dimensional structure with affinity for a particular epitope on the surface of an antigen.
  • the constant region segments are responsible for prolonged serum half-life and the effector functions of the antibody and relate to complement binding, stimulation of phagocytosis, antibody- dependent cellular cytotoxicity and triggering of granulocyte granule release.
  • hybridoma technology has facilitated the production of monoclonal antibodies of a particular specificity.
  • hybi ⁇ domas are murine hybridor ⁇ as.
  • Human/mouse chimeric antibodies have been created in which antibody variable region sequences from the mouse genome are combined with antibody constant region sequences from the human genome.
  • the chimeric antibodies exhibit the binding characteristics of the parental mouse antibody, and the effector functions associated with the human constant region.
  • the antibodies are produced by expression in a host cell, including for example Chinese Hamster Ovary (CHO), NSO myeloma cells, COS cells and SP2 cells.
  • CHO Chinese Hamster Ovary
  • NSO myeloma cells CHO
  • COS cells COS cells
  • SP2 cells a host cell
  • Such chimeric antibodies have been used in human therapy, however antibodies to these chimeric antibodies have been produced by the human recipient.
  • Such anti-chimcric antibodies are detrimental io continued therapy with chimeric antibodies.
  • human monoclonal antibodies are expected to be aii improvement over mouse monoclonal antibodies for in vivo human therapy. From work done with antibodies from Old World primates (rhesus monkeys and chimpanzees) it has been postulated that these non-human primate antibodies will be tolerated in humans because they arc structurally similar to human antibodies (Ehrlich PH el al., Human and primate monoclonal antibodies for in vivo therapy. Clin Chem. 34:9 pg 168J- 1688 (1988)). Furthermore, because human antibodies are non-immunogenic in Rhesus monkeys (Ehrich PH et al.. Rhesus monkey responses to multiple injections of human monoclonal antibodies.
  • Hybridoma 1987; 6:15.1-60 it is likely that the converse is also applicable and primate antibodies will be non-immunogenic in humans.
  • These monoclonal antibodies are secreted by hybr ⁇ domas constructed by fusing lymphocytes to a human x. mouse heteromyeloma.
  • EP 0 605442 discloses chimeric antibodies which bind human antigens. These antibodies comprise the whole variable region from an Old World monkey and the constant region of " a human or chimpanzee antibody.
  • One of the advantages suggested in this reference for these constructs is the ability to raise antibodies in Old World monkeys to human antigens which arc less immunogenic in humans compared with antibodies raised in a mouse host.
  • New World primates comprise at least 53 species commonly divided into two families, the CulUthricidae and Cebidae.
  • the Callilhricidae eonsist of marmosets and tainarins.
  • the Cebidae includes the squirrel monkey, titi monkey, spider monkey, woolly monkey, capuchin, uakaris, sakis, night or owl monkey and the howler monkey.
  • Evolulionarily distant primates such as New World primates, are not only sufficiently different from humans to allow antibodies against human antigens to be generated, but are sufficiently similar to humans to have antibodies similar to human antibodies so that the host does not generate an anti-antibody immune response when such primatc-derived antibodies arc introduced into a human.
  • Domain antibodies are functional binding units which can be created using antibody frameworks and correspond to the variable regions of either the heavy (VH) or light (V L ) chains of antibodies. Domain antibodies have a molecular weight of approximately 13 kDa, or less than one tenth the size of a full antibody.
  • Immunoglobulin light chains are referred to as either kappa or lambda tight chains and the heavy chains as gamma, mu, delta, alpha or epsilon.
  • the variable region gives the antibody its specificity, Within each variable region are regions of hypervari ability, otherwise known as complementarity determining regions (CDRs) which are flanked by more conserved regions referred to as framework regions. Within each light and heavy chain variable region are three CDRs and four framework regions.
  • domain antibodies are well expressed in bacterial, yeast and mammalian systems. Their small size allows for higher molar quantities per gram of product, thus providing a significant increase in potency.
  • domain antibodies can be used as a building block to create therapeutic products such as multiple targeting domain antibodies in which a construct containing two or more variable domains bind to two or more therapeutic targets, or domain antibodies targeted for pulmonary or oral administration.
  • the present invention provides a chimeric antibody or an antigen-binding portion thereof, wherein the antigen-binding portion comprises at least two complementarity determining regions (C-DR) and at least three framework regions, wherein at least one CDR is a New World p ⁇ imate CDR.
  • C-DR complementarity determining regions
  • the present invention provides a method of producing a chimeric antibody ⁇ r an antigen-binding portion thereof, the method comprising deleting a CDR from a human antibody variable region comprising id least two CDRs and at least three framework regions and replacing it with a New World primate CDR predicted to he of low immunogenic ity to produce a chimeric variable region,
  • the method further comprises the step of recovering the chimeric variable region.
  • the present invention provides a chimeric antibody or an antigen- binding portion thereof produced according to the method of the present invention.
  • the invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising an effective amount of tine antibody or antigen-binding portion thereof according to the present invention, together with a pharmaceutically acceptable excipitsnt or diluent.
  • the invention provides for die use of an antibody or antigen-binding portion thereof of the present invention in a diagnostic application for detecting an antigen associated with a particular disease or disorder.
  • the present invention provides a method for treating a disease or disorder characterised by human TNF- ⁇ activity in a human subject, comprising administering to the subject in need thereof an effective amount of a chimeric antibody as described herein, or a pharmaceutical composition thereof in which the antibody or antigen-binding portion thereof binds TNF- ⁇ .
  • FIGURES figure 1 demonstrates the binding of AB 138 to rat MOG present in rat spinal cord lysatc (lane 2) and not to CHOKISV lysate (lane 3).
  • J.ane 1 contains molecular weight markers.
  • Figure 2 demonstrates the lack of non-specific binding of an OMl-TTJFa monoclonal antibody to the same sample of rat MOG present in rat spinal cord lysate ⁇ lunts 2) and CHC)KlSV lysalc (lane 3).
  • Lane I contains molecular weight markers.
  • Figure 3 shows the acceptor domain antibody amino acid and nucleotide sequence (both strands).
  • CDR2 residues are indicated in underline.
  • Figure 4 is a sequence alignment of the domain antibody acceptor sequence with a panel of New World primate derived immunoglobulin sequences performed using AlignX (Vector NTJ, Invitr ⁇ gen, Australia). The CDR2 is highlighted in bold text. DETAILED DESCRIPTION OF THE INVENTION
  • the present invention provides a chimeric antibody or an antigen-binding portion thereof, wherein the antigen-binding portion comprises at least two complementarity determining regions (CDR) and at least three framework regions, wherein at least one CDR is a New World primate CDR. It is preferred that the antigen binding portion comprises three CDRs and four framework regions. It is also preferred that the antigen-binding portion comprises at least one, and preferably two human CDRs.
  • CDR complementarity determining regions
  • the chimeric antibody or antigen-bmdmg portion thereof comprises one New World primate CDR.
  • the chimeric antibody or antigen-binding portion thereof comprises two New World primate CDRs.
  • CDR2 of the antibody or antigen-binding portion is a New World primate CDR.
  • the at least one New World primate CDR is not from a sequence that binds a target antigen.
  • the framework regions are human sequences. Framework regions that arc human sequences include sequences derived from human framework regions, or synthetic sequences based on human framework regions.
  • sequence of the antigen hinding portion may be further subject to affinity maturation in order to improve its antigen binding characteristics such as antigen binding or potency.
  • An increase in binding is demonstrated by a decrease in K D (K aff /k nn ) for the antibody or antigen binding portion thereof.
  • ⁇ n increase in potency is demonstrated in biological assays.
  • assays thaL can be used to measure the potency of the antibody or antigen-binding portion thereof include the TNF ⁇ -induced L929 cytotoxicity neutralisation assay, IL-12-induced human PH ⁇ -activaled peripheral blood mononuclear cell (PBMC) proliferation assay, and RANKL mediated osteoclast differentiation of mouse splcnocyles (Stem, Proc. Natl. Acad. Sci. USA 87:6808 - 6812 (1990); Kong, Y-Y. et a Nature 397:315 - 323 (1990); Matthews, N. and ML. Neale in Lymphokines and Interferons, a Practical Approach, 1987, MJ. Clemens, A.G. Morris and AJ.H. Gearing, cds., JRL Press, p. 22J)
  • At least one framework region is modified to increase binding and/or to reduce predicted immunogcnicity in humans
  • At least one CDR sequence is modified to increase binding or potency and or to reduce predicted immunogenicity in humans. It is preferred that where at least one CDR sequence which is modified it is not the New World primate CDR.
  • At least one framework region is modi Tied, in addition to at least one CDR sequence, to increase binding and or to reduce predicted immunogenicity in humans. It is preferred that the at the least one CDR sequence which is modified it is not a New World primate CDR sequence.
  • the antigen-binding portion is a domain antibody.
  • the domain antibody may be multirncrised, as for example, hctero- or.homodimers (e.g., V H / V H , V L / V L or V H /VL), netero- or homolrimcrs (e.g., V II / V H / V H , V L / V L / V L , V H / V II / V L or V H / V L / V L ), hetcro- or homotetramers (e.g., V II /V H / V H / V II , V L / V L / V L / V L V H /V II /V H ,V L , V II / V H / V L / V L or V H /V L /V L /V L ), or higher order hetero- or homomultimers.
  • hctero- or.homodimers e.g., V H / V H , V
  • the invention provides a domain antibody wherein the domain antibody is linked, to at least one further domain antibody.
  • Each domain antibody may bind to the same or different antigens.
  • the domain antibody multimers may further comprise o ⁇ c or more domain antibodies which are linked and wherein each domain antibody binds to a different antigen, multi- specific Iigands including so-called "dual-specific ljgands".
  • the dual specific Kenya may comprise a pair of V 11 domains or a pair of V L domains.
  • dual-speci fie Iigands are described in WO 2004/003019 (PCT/GB2003/002804) in the name of Domanlis Ltd incorporated by reference herein in its entirety.
  • the antibody or antigen-binding portion further comprises a human or non- human primate constant region sequence.
  • non-human primates include, but are not limited to, chimpanzees, ⁇ rang ⁇ atangs and baboons.
  • the present invention also provides a method of producing a chimeric antibody or an antigen -binding portion thereof, the method comprising deleting a CDR from a human antibody variable region comprising at least two CDRs and at least three framework regions and replacing it with a New World primate CDR predicted to be of low immunogcnicity to produce a chimeric variable region. Jn a related aspect the method further comprises the step of recovering the chimeric variable region.
  • the selected New World prunate CDR is CDR2. It is preferred that the CDR2 sequence is selected from KVSNR ⁇ S, RVSNRAS, KVSTRGP, ⁇ ASKR ⁇ S, TSSNLQA, DASSLQP and YASFLQG. Particularly preferred sequences are KVSNRAS, A ⁇ SNRAS, TSSNLQ ⁇ and KVSTRGP due to their predicted lower immunogemcity.
  • the method further comprises modifying the sequence of the chimeric variable region to increase binding and/or to decrease immuraogenicity in humans. It is preferred that the New World primate CDR sequence is not modified. Where two or more New World primate CDR sequences arc present then it is preferred that at least one New World primate CDR. is not modified.
  • At least one framework region is modified in addition to at least one CDR sequence, to increase binding and or to reduce predicted immunogemcity in humans. It is preferred that the at the least one CDR sequence which is modified it is not a New World primate CDR. scquence.Thc present invention also provides a chimeric antibody or an antigen-binding portion thereof produced by the method of the present invention.
  • the term "antibody” as used herein, is intended to refer to immunoglobulin molecules comprised ot four polypeptide chains, two heavy (H) chains and two light (Iy) chains interconnected by disulfide bonds. Each heavy chain is comprised of a heavy chain variable region (HCVR or Vn) and a heavy chain constant region.
  • the heavy chain constant region comprises three domains, C H I, C H 2 and Cn3>
  • Each light chain is comprised of a light chain variable region (LCVR or V L ) and a light chain constant region.
  • the light chain constant region is comprised of one domain, C L .
  • the V H and V L regions can be further subdivided into regions of hypcrvariabilily, termed complementarity determining regions (CDR), interspersed with regions that arc more conserved, termed framework regions (FR).
  • Each VH and V L i.s composed of three CDRs and four FRs, arranged from ammo-terminus to carboxy-tcrminutJ in the following order: FR 1 , CDRl, FR2, CDR2, FR3, CDR3, FR4,
  • antigen-binding portion of an antibody refers to one or more components or derivatives of an immunoglobulin thai exhibit the ability to bind to an antigen. It has been shown that the antigen-binding function of an antibody can be performed by fragments of a full length antibody.
  • E ⁇ ampl ⁇ s of binding fragments encompassed within the term "antigen-binding portion" of an antibody include (i) a Fab fragment, a monovalent fragment consisting of the V L , V H , C L and C H 1 domains; (ii) a F(ab')2 fragment, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) a Fd fragment consisting of the V H and C H 1 domains; (iv) a Fv fragment consisting of the V t , and VH domains of a single arm of an antibody; (v) a dAb fragment (Ward et al, 1989, Nature 341 :544-54G) which consists of a single V H domain, or a V L domain (van don Beuken T et al, 2(X)I, J.
  • a Fab fragment a monovalent fragment consisting of the V L , V H , C L and C H
  • single chain Fvs are also intended to be encompassed within the term "antigen-binding portion" of an anti body.
  • Other forms of single chain Fvs and related molecules such as diabodies or triabodies are also encompassed, Diabodies arc bivalent antibodies in which V H and V L domains arc expressed on a single polypeptide chain, buL using a linker that is too short to allow for pairing between the two domains on the same chain, thereby forcing the domains to pair with complementary domains of another chain and creating two antigen binding sites (see e.g., JJolliger, P., et al. r 1993, Proc. NaU. ⁇ cad. Sci.
  • the domain antibody comprises a human framework regions and at letust one New World primate CJDRs, more preferably marmoset CDRs.
  • the New World primate is selected from the group consisting of marmosets, lamatins, squirrel monkey, titi monkey, spider monkey, woolly monkey, capuchin, uakaris, saTds. night or owl monkey and the howler monkey. More preferably, the New Wortd primate is a marmoset.
  • the antibody or antigen-binding portion thereof according to the present invention has predicted low immunogenic] ty in a human host.
  • low immunogenioily il is meant that the antibody docs not raise an antibody response in at. least the majority of individuals receiving the antibody of sufficient magnitude to reduce the effectiveness of continued administration of the antibody for a sufficient time to achieve therapeutic efficacy.
  • the level of immunogenicity in humans may predicted using the MHC class II binding 0 prediction program Propred (http://www.imtech.res.in/raghava/propred) using a 1% threshold value analysis of all alleles.
  • Other programs which may bo used include:
  • Low immunogenicity molecules will contain no or low numbers of peptides predicted to 5 bind to MlJC class Il alleles that are highly expressed in the target population (Flower DR, Doytchinova IA. (2004) Immunoinformatics and the prediction of immunogenicity, Drug Discov Today, 9(2): 82-90).
  • Reduced immunogenicity molecules will contain no or a reduced numbers of peptides predicted to bind to MHC class Il alleles that are highly expressed in the target population, 0 relative to the starting donor molecule.
  • Functional analysis of MHC class ⁇ I binding can be performed by generating overlapping peptides corresponding to lhc protein of interest and testing these for their ability to evoke T cell activation (T ce)! proliferation assay) or displace a reporter peptide, a known Ml IC class l!-binding peptide (Hammer J el at., 1994, J. Kxp. Med., 180:2353).
  • the present invention is further based on a method for amplification of New World primate immunoglobulin genes, for example by polymerase chain reaction (PCR) from nucleic acid extracted from New World primate lymphocytes using primers specific for heavy and light chain variable region gene families.
  • PCR polymerase chain reaction
  • the amplified variable region is then cloned into an expression vector containing a human or primate constant region gene for the production of New World primate chimeric recombinant antibody.
  • Standard recombinant DNA methodologies are used to obtain antibody heavy and light chain genes, incorporate these genes into recombinant expression vectors and introduce the vectors into host cells, such as those described in Sambrook, Krilsch and Maniotis (eds), Molecular Cloning: a laboratory manual, second edition, Cold Spring Harbor, N.Y (1989). Suitable expression vectors will be familiar to those skilled in the art.
  • the New World pi imalc lymphocytes producing the immunoglobulins are typically immortalised by fusion with a myeloma cell line to generate a hybridoma.
  • Preferred mammalian host cells for expressing the recombinant antibodies of the invention include Chinese I lamster Ovary (CHO), NSO myeloma cells, COS cells and SP2 cells.
  • CHO Chinese I lamster Ovary
  • NSO myeloma cells include Chinese I lamster Ovary (CHO), NSO myeloma cells, COS cells and SP2 cells.
  • the present invention also contemplates the use of non-mammalian expression systems such as those which arc plant or prokaryotic (bacterial) derived. Such expression systems would be familiar to persons skilled in the art.
  • the repertoire of Vn, V L and constant region domains can be a naturally occurring repertoire of immunoglobulin sequences or a synthetic repertoire.
  • a nanirally occurring repertoire is one prepared, for example, from immunoglobulin expressing cells harvested from one or more primates. Such repertoires can be naive ie. prepared from newborn immunoglobulin expressing cells, or rearranged ie. prepared from, for example, adult primate B cells. If desired, clones identified from a natural repertoire, or any repertoire that bind the target antigen are then subject to mutagenesis and further screening in order to produce and select variants with improved binding characteristics.
  • Synthetic repertoires of immunoglobulin variable domains are prepared by artificially introducing diversity into a cloned variable domain.
  • affinity maturation techniques will be familiar to persons skilled in lhe art (Irving R. A. et ai (2001) Ribosorne display and affinity maturation: from antibodies Io single V-domains and steps towards cancer therapeutics, Journal of Immunological Methods, 248: 31-45).
  • variable region, or a CDR thereof, of a New World primate antibody gene may be cloned by providing nucleic acid eg. cDN ⁇ , providing a primer complementary to the cDN A sequence encoding a 5' leader sequence of an antibody gene, contacting that cDNA and the primer to form a hybrid complex and amplifying the cDNA to produce nucleic acid encoding the variable region (or CDR region) of the New World primate antibody gene.
  • non-New World primate variable region sequence may be used as an acceptor for grafting New World primate sequences, in particular, CDR sequences using standard recombinant techniques.
  • CDR sequences using standard recombinant techniques.
  • US Patent No. 5,585,089 describes methods for creating low immuiiogenicity chimeric antibodies that retain the high affinity of the non-human parent antibody and contain one or more CDRs from a donor immunoglobulin and a framework region from a human immunoglobulin.
  • 20030039649 describes a humanisation method for creating low immiinogenicity chimeric antibodies containing CDR sequences from a non-human antibody and framework sequences of human antibodies based on using canonical CDR structure types of the non-human antibody in comparison to germline canonical CDR structure types of human antibodies as the basis for selecting the appropriate human framework sequences for a humanised antibody. Accordingly, these principles can be applied to the grafting of one Or more New World primate CDRs into a -non-New World primate acceptor variable region.
  • the CDR sequences may be obtained from the genomic UNA isolated from an antibody, or from sequences present in a database e.g. The National Centre for Biotechnology Information protein and nucleotide databases, The Kabat Database of Sequences of Proteins of Immunological Interest
  • the CDR sequence may be a genomic DNA or a cDNA.
  • Methods for grafting a replacement CDR(s) into an acceptor variable sequence will be familiar to persons skilled in the an of the present invention.
  • the CDRs will be grafted into acceptor variable region sequences for each of a variable light chain and a variable heavy chain or a sitiglc chain in the case of a domain antibody.
  • the preferred method of the present invention involves replacement of either CDRl or, more preferably, CDR2 in a variable region sequence via primer directed mutagenesis.
  • the method consists of annealing a synthetic oligonucleotide encoding a desired mutation to a target region where it servea as a primer for initiation of DNA synthesis in vitr ⁇ , extending the oligonucleotide by a DN ⁇ polymerase io generate a double-stranded I ) NA that carries the desired mutation, and ligating and cloning the sequence inlo an appropriate expression vector (Sambroolc, Joseph; and David W. Russell (2001). Molecular Cloning: A Laboratory Manual, 3rd cd., Cold Spring Harbor, N. Y.: Cold Spring Harbor Laboratory Press)..
  • an antibody or antigen-binding portion thereof may be part of a larger immunoadhesio ⁇ molecule, formed by covalent or noncovalcnt association of the antibody ⁇ or antibody portion with one or more other proteins or peptides.
  • immunoadhesio ⁇ molecule formed by covalent or noncovalcnt association of the antibody ⁇ or antibody portion with one or more other proteins or peptides.
  • irnmunoadhesion molecules include use of the streplavidin core region to make a tctrameric seFv molecule (Kipriyanov, S. M., et al.
  • the constant region sequence (Fe portion) is preferably obtained from a human or non- human primate immunoglobulin sequence.
  • the primate sequence may be a New World primate or an Old World primate sequence. Suitable Old World primates include chimpanzee, or other hominid ape eg. gorilla or orang utan, which because of their close phylogenetic proximity to humans, share a high degree of homology with the human constant region sequence. Sequences which encode for human or primate constant regions are available from databases including e.g. 'Che National Centre for Biotechnology Information protein and nucleotide databases, The Kabat Database of " Sequences of Proteins of Immunological Interest.
  • the antibody or antigen-binding portion according to the invention is capable of binding to a human or non-human antigen.
  • the antigen to which the chimeric antibody or antigen-binding portion thereof binds is peptide, protein, carbohydrate, glycoprotein, lipid or glycolipid in nature, selected from a tumour-associated antigen including carcinoembryonic antigen, EpCAM, Lewis-Y, I-ewis-Y/b, PMSA, CD20, CD30, CD33, CD38, CD52, CD154, EGF-R, Her-2, TRAIL and VEGF receptors, an antigen involved in an immune or inflammatory disease or disorder including CD3, CD4, CD25, CD40, CD49d, MHC class I 7 MHC.: class II, GM- CSF, intcrfcron- ⁇ .
  • IL-I IL-12, IL-13, IL-23, TNF- ⁇ , and IgE
  • an antigen expressed on a host cell including glycoprotein Ub/AIIa, P-glycoprotcin, purinergjc receptors and adhesion receptors including CDlIa, CDl Ib, CDl Ic, CD18, CD56, CD58, CD62 or CD144, an antigen comprising a cytokine, chemokinc, gi'owth factor or other soluble physiological modulator or a receptor thereof including eotaxi ⁇ , 11 ⁇ 6, IL-8, TGF-p 1 , C3ii, C5n, VEGF, NGF and their receptors, an antigen involved in central nervous system diseases or disorders including ⁇ -amyloid and prions, an antigen of non-human origin such as microbial, nanobial or viral antigens or toxins including respiratory sync y tial vims protein F, anthrax toxin, rattle snake venom and
  • the antigen is TNF ⁇ , preferably human TNFo.
  • the chimeric antibody or antigen-binding portion thereof may bind a non-human antigen
  • the non-human antigen is selected from the group consisting of respiratory syncytial virus F protein, cytomegalovirus, snake venoms and digoxin.
  • the term "binds to” as used herein, is intended to refer to the binding of an antigen by an immunoglobulin variable region of an antibody with a dissociation constant (K ⁇ ) of l ⁇ lv ⁇ or lower as measured by surface plasmon resonance analysis using, for example a EI ⁇ eorcTM surface plasmon resonance system and BI ⁇ corcTM kinetic evaluation software (eg. version 2.1).
  • the affinity or dissociation constant (Ku) for a specific binding interaction is preferably about 500 nM to about 50 pM, more preferably about 500 nM or lower, more preferably about 300 nM or lower and preferably at least about 300 nM to about 50 pM, about 200 nM to about 50 pM, and more preferably at least about 100 nM to about 50 pM, about 75 nM lo about 50 pM, about 10 nM to about 50 pM.
  • the antibodies of the present invention are advantageous in human therapy because the likelihood of induction of a human anti-antibody response will be reduced.
  • Recombinant antibodies produced according to the invention that hind a target antigen can be identified and isolated by screening a combinatorial immunoglobulin library (eg a phage display library) to isolate library members that exhibit the desired binding specificity and l ⁇ mctional behaviour.
  • a combinatorial immunoglobulin library eg a phage display library
  • the phage display technique has bcen described extensively in lhe art and examples of methods and compounds for generating and screening such libraries and affinity maturing the products of them can be found in, for example, Barbus et al. (1991) PNAS 88:7978-7982: Clarkson et al. (1991) Nature 352:624:628; Dower ct al. PCT. 91/17271 , U.S. Patent No. 5,427,908, U.S. Patent N ⁇ . 5,580,717 and EP 527,839; Fuchs et a!. (1991) Bio/Technology 9:1370-1372; Garrad et al.
  • Recombinant libraries expressing the antibodies of the invention can be expressed on the surface of microorganisms eg. yeast or bacteria (sec PCT publications WO99/36569 and 98/49286),
  • the Selected Lymphocyte Antibody Method or SLAM as it is referred to in the state of the art, is another means of generating high affinity antibodies rapidly. Unlike phage display approaches all antibodies are fully divalent.
  • New World primates arc immunised with a human antigen eg. a TNFo: polypeptide. Following immunisation cells are removed and selectively proliferated in individual micro wells. Supematants arc removed from wells and tested for both binding and function.
  • Gene sequences can be recovered for subsequent manipulations eg, humanisaliofl, Fab fragment, scFv or domain antibody generation.
  • SLAM derivation of the ligand of the invention by SLAM and its derivatives (Babcook, J.S. et al 1996, Proc. Natl. Acad. Sci, US ⁇ 93; 7843-7848, US Patent 5,627,052 and PCT publication WO92/0255t).
  • Adaptations of SLAM such as the use of alternatives to testing supernatants s ⁇ cli as panning, also lie within the scope of this invention.
  • JLti one expression system the recombinant pepfide/protein library is displayed on ribosomes (for examples see Roberts, RW and Szostak, J.W.I 997. Proc.NatJ.Acad.Sci.USA. 94:12297 - 123202 and PCT Publication No. WO98/31700).
  • a DNA library eg of antibodies and derivatives
  • translation of the library such that the protein and "immunised" mRN ⁇ s stay on the ribosome
  • affinity selection eg by binding to RSP
  • mRN ⁇ isolation e.g by reverse translation and subsequent amplification
  • reverse translation and subsequent amplification eg by polymerase chain reaction or related technology. Additional rounds of selection and amplification can be coupled as necessary ⁇ to affinity maturation through introduction of somatic mutation in this system or by other methods of affinity maturation as known in the state of the art.
  • the antibody or antigen-binding portion thereof according to the invention can be derivatised or linked to another functional molecule.
  • the antibody or antigen- binding portion can be functionally linked by chemical coupling, genetic fusion, noncovalenl association or otherwise, to one or more other molecular entities, such as another antibody, a detectable agent, a cytotoxic agent, a pharmaceutical agent, and/or a protein or peptide that can mediate association of the antibody or anli gen-binding portion thereof with another molecule (such as a streptavidin core region or a polyhistidine tag).
  • Useful delectable agents with which an antibody or antigen-binding portion thereof may be derivatised include fluorescent compounds.
  • Exemplary fluorescent delectable agents include fluorescein, fluorescein isothiocyanate, rhodamine, 5-dimethylamine- ⁇ - napthalenesulfonyl chloride, phycoeiythrin and the like.
  • An antibody may also be derivatised wilh delectable enzymes such as alkaline phosphatase, horseradish peroxidase, glucose oxidase and the like. When an antibody is derivatized with a detectable enzyme, it is detected by adding additional reagents that the enzyme uses to produce a detectable reaction product.
  • An antibody may also be dcrivatised with biolin, and detected through indirect measurement of avidin or streptavidin binding.
  • the present invention also extends to PEGylatcd antibodies or antibody-binding portion which provide increased half-life and resistance to degradation without a loss in activity (eg. binding affinity) relative to non-PEGylatcd antibody polypeptides.
  • the antibody or antibody-binding portion as described herein can be coupled, using methods known in the art, to polymer molecules (preferably PEG) useful for achieving the increased half-life and degradation resistance properties.
  • polymer molecules preferably PEG
  • Polymer moieties which can be utilised in the invention can be synthetic or naturally occurring and include, but are not limited to, straight or branched chai n polyaikylcnc, polyalkeny lene or polyoxyalkylcne polymeis, or a branched or unbranched polysaccharide such as a horno-or heterapolysaccharidc.
  • Preferred examples of synthetic polymers which can be used in the invention include straight or branched chain polyethylene glycol) (PEG), polypropylene glycol), or poly(v ⁇ nyl alcohol) and derivatives or substituted forms thereof.
  • Particularly preferred substituted polymers for linkage to antibodies as described herein include substituted PEG, including methoxy(polyethylene glycol).
  • Naturally occurring polymer moieties which can be used in addition to or in place ⁇ f PEG include lactose, amylose, dextran, or glycogen, as well as derivatives thereof which would be recognised by persons skilled in the art.
  • Dcrivatised forms of polymer molecules include, for example, derivatives which have additional moieties or reactive groups present therein to permit interaction with amino acid residues of the antibody polypeptides described herein.
  • Such derivatives include N- hydraxylsuccinimidc (NIIS) active esters, succinimidyl propionate polymers, and sulfhydryl-sclcctivc reactive agents such as maleimide, vinyl sulfonc, and thiol.
  • NIIS N- hydraxylsuccinimidc
  • Particularly preferred derivatized polymers include, but are not limited to PEG polymeis having the formulae: PEG-O-CH 2 CH 2 CH 2 -CO 2 -N HS; PEG-O-CH 2 -NHS; PEO-O- CH 2 CH Z -CO 2 -NHS; PKG-S-CH 2 CH 2 -CO-NI IS; PKG-O 2 CNH-CH(R)-CO 2 -NHS; PKG- NHCO-CH 2 CH 2 -CO-NHS; and PEG-O-CH 2 -CO 2 -NHS; where R is (CH 2 ) 4 )NHC0 2 (mPEG), PBG polymers can be linear molecules, or can be branched wherein multiple PEG moieties are present in a single polymer.
  • the reactive group (e.g., MAL, NHS, SP ⁇ , VS, or Thiol) may be attached directly to the PRG polymer or may be attached to PEG via a linker molecule.
  • the size of polymers useful in the invention can be in the range of between 500 Da to 60 kDa, for example, between 1000 Da and 60 kDa, 10 kDa and 60 kDa, 20 kDa and 60 kDa, 30 kDa and 60 kDa, 40 kDa and 60 kDa, and up to between 50 kDa and 60 kDa.
  • the polymers used in the invention, particularly PEG can be straight chain polymers or may possess a branched conformation.
  • the polymer (PEG) molecules useful in the invention can be attached to an antibody or antigen-binding portion thereof using methods which are well known in the art.
  • the first step in the attachment of PEG or other polymer moieties to an antibody polypeptide monomer or multimer of the invention is the substitution of the hydroxyl end-groups of the PEG polymer by clectrophiie-containing functional groups.
  • PEG polymers are attached to cither cysteine or lysine residues present in the antibody polypeptide monomers or multimers.
  • the cysteine and lysine residues can be naturally occurring, or can be engineered into the antibody polypeptide molecule.
  • cysteine residues can be recombinantly engineered at the C-terminus of an antibody polypeptide, or residues at specific solvent accessible locations in an antibody polypeptide can be substituted with cysteine or lysine.
  • the antibody may be linked to one or more molecules which can increase its half -life in vivo. These molecules arc linked to the antibody at a site on the antibody other than the antigen binding site, so that they do not interfcrc/sterically hinder the antigen-binding site. Typically, such molecules are polypeptides which occur naturally in viv ⁇ and which resist degradation or removal by endogenous mechanisms. It will be obvious to one skilled in the art that fragments or derivatives of such naturally occurring molecules may be used, and that some may not be polypeptides.
  • Molecules which increase half life may be selected from the following: (a) proteins from the extracellular matrix, eg. collagen, Jaminin, integrin and fibronectin;
  • proteins found in blood eg, fibrin ⁇ -2 macroglobulin, serum albumin, fibrinogen A, fibrinogen B, scrum amyloid protein A, hcptaglobin, protein, ubiquitin, utcroglobulin, ⁇ -2 microglobulin, plasminogen, lysozymc, cystatin C, alpha-1-antitrypsin and pancreatic kypsin inhibitor;
  • immune serum proteins eg. IgE, IgG, IgM;
  • transport proteins eg. rctinol binding protein, ⁇ -1 microglobulin;
  • defensins eg. beta-dcfcnsin 1, Neutrophil defcnsins 1, 2 and 3;
  • proteins found at the blood brain barrier or in neural tissues eg. melanocortin receptor, myelin, ascorbatc transporter;
  • transferrin receptor specific ligand-neuropharmaccutJcai agent fusion proteins see US5977307; brain capillary endothelial cell receptor, transferrin, transferrin receptor, insulin, insulin- like growth factor 1 (IGF 1) receptor, i ⁇ sulin- ⁇ ke growth factor 2 (IGF 2) receptor, insulin receptor;
  • proteins localised to the kidney eg. polyeyslin, type IV collagen, organic anion transporter Kl, Heymann's antigen;
  • proteins localised to the lung eg. secretory component (binds IgA);
  • proteins localised to the skin eg, keratin
  • bone specific proteins such as bone morphogeny proteins (BM Ps) eg. BMP-2, -4, -5, -6, -7 (also referred to as osteogenic protein (OP-I) and -8 (OP-2);
  • BM Ps bone morphogeny proteins
  • BMP-2, -4, -5, -6, -7 also referred to as osteogenic protein (OP-I) and -8 (OP-2);
  • tumour specific proteins eg. human trophoblast antigen, herceptin receptor, oestrogen receptor, cathepsi ⁇ s eg cathepsin B (found in liver and spleen);
  • disease-specific proteins eg. antigens expressed only on activated T- cells: including LAG-3 (lymphocyte activation gene), ostcoprotegerin ligand (OPGL) sce Natuie 402, 304-309, 199S); OX40 (a member of the TNF ⁇ receptor family, expressed on activated T cells and the only costimulatory T cell molecule known to be specifically up-regulated in human '1' cell leukaemia virus type ⁇ l (HTLV-I)-producing cells - see J. Immunol.
  • LAG-3 lymphocyte activation gene
  • OPGL ostcoprotegerin ligand
  • OX40 a member of the TNF ⁇ receptor family, expressed on activated T cells and the only costimulatory T cell molecule known to be specifically up-regulated in human '1' cell leukaemia virus type ⁇ l (HTLV-I)-producing cells - see J. Immunol.
  • angiogenic giowth factors including acidic fibroblast growth factor (FGF-I), basic fibroblast growth factor (FGF-2), Vascular endothelial growth factor/vascular permeability factor (VEGF/VPK), transforming growth factor- ⁇ (TGF- ⁇ ), tumor necrosis factor-alpha (TNF ⁇ ), angiogenin, inierlcukin-3 (IL-3), intcrleukin-8 (IL-8), platelet derived endothelial growth factor (PD- BCGF), placental growth factor (PlGF), midkine platelet-derived growth factor-BJ ⁇ (FDGF), fractalkine;
  • FGF-I acidic fibroblast growth factor
  • FGF-2 basic fibroblast growth factor
  • VEGF/VPK Vascular endothelial growth factor/vascular permeability factor
  • TGF- ⁇ tumor necrosis factor-alpha
  • TNF ⁇ tumor necrosis factor-alpha
  • IL-8 inierlcukin-3
  • IL-8 platelet derived endot
  • stress proteins heat shock proteins
  • the invention provides a pharmaceutical composition comprising an effective amount of the chimeric antibody or antigen-binding portion thereof according to the present invention, together with a pharmaceutically acceptable excipient or diluent.
  • pharmaceutically acceptable excipient or diluent includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and Lhe like that are physiologically compatible.
  • pharmaceutically acceptable carriers include one or more of water, Baline, phosphate buffered saline, dextrose, glycerol, ethan ⁇ l, and the like as well as combinations thereof.
  • isotonic agents for example, sugars, polyalcohols such as r ⁇ an ⁇ itol, sorbitol, or sodium chloride in the composition.
  • Pharmaceutically acceptable substances such as wetting or minor amounts of auxiliary substances such as wetting or emulsifying agents, preservatives! or buffers.
  • effective amount refers to an amount of an antibody or antigen binding portion thereof (including pharmaceutical compositions comprising the antibody or antigen binding portion thereof) sufficient to treat a specified disease of disorder or one or more of its symptoms and/or to prevent the occurrence of the disease or disorder.
  • diagnosis refers to an amount of a antibody or antigen binding portion thereof (including pharmaceutical compositions comprising the antibody or antigen binding portion thereof) sufficient to diagnose a specified disease or disorder and/or one or more of its rnamfestutiona, where diagnosis includes identification of the existence of the disease or disorder and/or detection of the extent or severity of the disease or disorder. Often, diagnosis will be carried out with reference to a baseline or background detection level observed for individuals without the disease or disorder. Levels of detection above background or baseline levels (elevated levels of detection) are indicative of the presence and, in some cases, The severity of the condition.
  • an individual “in need thereof” may be an individual who has been diagnosed with or previously treated for the disease or disorder to be treated.
  • an individual "in need thereof may be an individual who is suspected to have a disease or disorder, is at risk for a disease or disorder, or has previously been diagnosed with the disease or disorder (e.g., diagnosis can include monitoring of the severity (e.g., progression/regression) of the disease or disorder over time and/or in conjunction with therapy).
  • the chimeric antibody or antigen-binding portion thereof blocks or stimulates receptors functions or neutralizes active soluble products, such as one or more of the inlerleukins, TN Fa or C5a. More preferably, the active soluble product is human TN Fa.
  • the composition may be in a variety of forms, including liquid, semi-solid or solid dosage forms, such as liquid solutions (eg injectable and infusible solutions), dispersions or suspensions, tablets, pills, powders, liposomes or suppositories.
  • lhe composition is in the form of an injectable solution for immunization.
  • the administration may be intravenous, subcutaneous, intraperitoneal, intramuscular, transdermal, intrathecal, and intraarterial.
  • the dosage form is in the range of from about 0.001 nig to about 10 mg/lfg body weight administered daily, weekly, bi- or tri-wcekly or monthly, mote preferably about 0.05 to about 5 mg/kg body weight weekly.
  • composition may also be formulated as a sterile powderlbr the preparation of sterile injectable solutions.
  • the active compound may be prepared with a carder that will protect the compound against rapid release, such as a controlled release formulation, including implants, transdermal patches, and microencapsulated delivery systems.
  • a carder that will protect the compound against rapid release, such as a controlled release formulation, including implants, transdermal patches, and microencapsulated delivery systems.
  • Compatible polymers may be used such as ethylene vinyl acetate, polyanbydrides, polyglycolic acid, collagen, potyorthocsters or polylactic acid.
  • composition may also be formulated for oral administration.
  • the antibody may be enclosed in a hard or soft shell gelatin capsule, compressed into tablets, or incorporated directly into the subject's diet.
  • composition may also be formulated for rectal administration.
  • the antibody r ⁇ ay be administered in order to bind to and identify selected cells in vitro and in vivo, to bind to and destroy selected cells i? ⁇ vlv ⁇ , or in order to penetrate into and destroy selected cells in vivo.
  • the antibody may be used as an immunotoxin to deliver a cytotoxic agent eg. a toxin or cliemolherapcutie agent IO a particular cell type such as a tumour cell. Production of irnrnunotoxins would be familiar to persons sltilled in the art.
  • Cytotoxic agents commonly used to generate immunotoxins include radioactive isotopes such as 111 In or Y, selenium, libonucleascs, binding domain - deleted truncated microbial toxins such as Pseudomonas exotoxin or Diphtheria toxin, tubulin inhibitors such as calioheamicin (ozagamicin), maytansinoids (including DM-I), auristatins, and taxoids, ribosome inactivating proteins such as ricin, ebulin 1, saporin and gclonin, and prodrugs such as melphalan,
  • radioactive isotopes such as 111 In or Y, selenium, libonucleascs, binding domain - deleted truncated microbial toxins such as Pseudomonas exotoxin or Diphtheria toxin
  • tubulin inhibitors such as calioheamicin (ozagamicin
  • the composition is administered- to a human.
  • the present invention also provides Tor Lhe use of the chimeric antibody or antigen-binding portion thereof in a diagnostic application for detecting an antigen associated with a particular disease or disorder,
  • the invention provides for the use of the chimeric antibody or antigen-binding portion thereof in a method for diagnosing a subject having an antigen associated with a particular disease or disorder, comprising administering to said subject a diagnostically effective amount of a pharmaceutical composition according to the third aspect.
  • a subject is a human.
  • the chimeric antibody ⁇ r antigen-binding fragment thereof can be used to detect the presence ⁇ f an antigen, or elevated levels of an antigen (e.g. TNFu) in a biological sample, such as serum or plasma using a convention immunoassay, such as an enzyme linked immunosorbent assay (ELISA), a radioimmunoassay (RIA) or tissue immunohistoehemistry.
  • an antigen e.g. TNFu
  • a biological sample such as serum or plasma
  • a convention immunoassay such as an enzyme linked immunosorbent assay (ELISA), a radioimmunoassay (RIA) or tissue immunohistoehemistry.
  • the antigen to which the chimeric antibody or antigen-binding portion thereof binds is peptide, protein, carbohydrate, glycoprotein, lipid or glycolipid in nature, sclccTcd from a tumour-associated antigen including carcinoembryonic antigen, EpC ⁇ M, L ⁇ wis-Y, Lewis- Y/b, PMS ⁇ , CD20, CD30, CD33, CD38, CD52, CD154, EGF-R, Her-2, TRAIL and VEGF receptors, an antigen involved in an immune or tnflanimatory disease or disorder including CD3, CD4, CD25, CD40, CD49d, MHC class I, MHC class II, GM- CSF, inlerferon- ⁇ , IL-I, IL- 12, IL-13, IL-23, TNh'- ⁇ , and IgR, an antigen expressed on a host cell including glycoprotein llb/llla, P-glycoprotcin, purinergic receptors and adh.
  • anthrax toxin rattle snake venom and digoxin
  • the chimeric antibody acts as an agonist or antagonist or is active Io either deplete (kill or eliminate) undesired cells (eg. a ⁇ ti-CD4) by acting with complement, or killer cells (eg. NK cells) oris active as a cytotoxic agent or to cause Fe-receptor binding by a phagocyte or neutralizes biological activity of its target.
  • the anti-human TNF ⁇ chimeric antibody or antigen binding portion thereof according to the invention may also be used in cell culture applications where it is desired to inhibit TNFn activity.
  • the present invention also provides a method for treating a disease or disorder characterised by human TNF ⁇ activity in a human subject, comprising administering to the subject in need thereof a pharmaceutical composition according to the present invention in which the chimeric antibody or antigen-binding portion thereof binds TNFre.
  • disease or disorder characterised by human TNF ⁇ activity is intended to include diseases or disorders in which the presence of TNF ⁇ in a subject suffering from the disease or disorder has been shown to be or is suspected of being either responsible for the pathophysiology of the disease or disorder or a factor that contributes to the worsening of the disease or disorder. Accordingly, a disease or disorder in which TNF ⁇ activity is detrimental is a disease or disorder in which inhibition of TNFre activity is expected to al leviate symptoms and/or progression of the disease or disorder.
  • Such diseases or disorders may be evidenced, for example, by an increase in the concentration of TNF ⁇ in a biological fluid of a subject suffering from the disease or disorder (c.g., an increase in the concentration of TNF ⁇ in serum, plasma, synoviid fluid etc of the subject), which can be detected, for example, using a chimeric antibody of the invention specific for TNF ⁇
  • a disease or disorder characterised by human TNF ⁇ activity is intended to include diseases or disorders and other disease or disorder in which the presence of TNF ⁇ in a subject suffering from the disease or disorder has been shown to be, or is suspected of being, cither responsible for the pathophysiology of the disease or disorder or a factor which contributes to a worsening of the disease or disorder.
  • the disease or disorder characterised by human TNF ⁇ activity is selected from the group consisting of sepsis, including septic shock, endotoxic shock, gram negative sepsis and toxic shock syndrome; autoimmune disease, including rheumatoid arthritis, rheumatoid spondylitis, osteoarthritis, psoriasis and gouty arthritis, allergy, multiple sclerosis, autoimmune diabetes, autoimmune uveitis and nephrotic syndrome; infectious disease, including fever and myalgias due to infection and cachexia secondary to infection: graft versus host disease; tumour growth or metastasis; pulmonary disease including adult respiratory distress syndrome, shock lung, chronic pulmonary inflammatory disease, pulmonary sarcoidosis, pulmonary fibrosis and silicosis; inflammatory bowel disease including Crohn's disease and ulcerative colitis; cardiac disease; inflammatory bone disease, hepatitis, coagulation disturbances, burns, reperfusion injury, keloid formation and
  • Supplementary active compounds can also be incorporated into lhc composition.
  • the ' antibody or antibody-binding fragment may be co-formulalcd with and/or administered simultaneously, separately or sequentially with one or more additional therapeutic agents eg. antibodies that bind to other targets s ⁇ ch as cytokines or cell surface molecules or alternatively one or more chemical agents that inhibit human TNfFa production or activity.
  • lhe invention provides a kit comprising a therapeutically effective amount of a chimeric antibody or antigen-binding portion of lhc invention, or a pharmaceutical composition comprising a therapeutically effective amount of a chimeric antibody or antigen-binding portion thereof, together with packaging and instructions for use.
  • the instructions for use include instructions for how to effectively administer a therapeutic amount of the chimeric antibody or antigen-binding portion of flic invention,
  • V K chain (Accession Number: AAM54057.
  • SEQ LD NO: 1 ) of lhe MOG specific marmoset derived antibody was expressed with a human constant region (human IgGI heavy chain C H 1., hinge, Cn2 & C H 3 domains (such as NCBI accession number POl 857) (SEQ ID NO; 2».
  • Vr. chain (Accession Number: A AM54058, SEQ ID NO: 3) of the MOG specific marmoset derived antibody was expressed with a human kappa light chain constant region (such as NCBI accession number AAA58989) (SEQ ID NO: 4).
  • DNA encoding the light chain (V L - Kappa) amino acid sequence was prepared as described above for tlic heavy chain.
  • the supernatant was passed over a HiTrap Protein A column (Amersham Bioseienecs, Cat No; 17-0402-01) three times at a flow rate of 1 mL/min. The column was then washed with 20 mlVt sodium phosphate for 40 niins at I mL/min. The antibody was eluted with 0.1 M citric acid pH 3.5 with fractions collected and immediately neutralised with IM Tris- I ICl ptf 9.0. Antibody samples were then desalted on a PD-10 column ( ⁇ mcrsham Biosciences, Cat No: 17-0851-01). Analysis of the antibody by SDS-P ⁇ GE and size- exclusion HPLC confirmed the correct molecular weight, presence of assembled antibody and the concentration of antibody.
  • the pellet and supernatant was diluted into SDS-P ⁇ GE sample buffer (125 ⁇ uM Tris-HCl pH 6.8, 5% SDS, 0.25% bromophenol blue, 25% glycerol).
  • SDS-P ⁇ GE sample buffer 125 ⁇ uM Tris-HCl pH 6.8, 5% SDS, 0.25% bromophenol blue, 25% glycerol.
  • SIGMA CeILytic M Cell J.ysis Reagent
  • rat MOG could be produced using recombinant DNA technology and the ability of AR 138 to bind rat MOG determined in binding assays such as JELIS ⁇ or Biacorc analysis.
  • Standard recombinant DNA technology can be used to produce a locally engineered domain antibody by substitution of the CDR2 of an acceptor anti-TNF ⁇ domain antibody (Basran el al. WO 2004/081026; SRQ ID NO: 7; Figure 3) with a CDR2 from a donor New World primate immunoglobulin.
  • the CDR2 is identified on the acceptor anti-TNT- ⁇ domain antibody (SASELQS).
  • SASELQS acceptor anti-TNT- ⁇ domain antibody
  • the domain antibody acceptor sequence is then aligned against a panel of New World primate immunoglobulin sequences. These sequences arc derived from the Ma's night monkey ( ⁇ otus nancymaac) (SEQ !D NOs:.8 - I 8) and from the common marmoset (Callithrix jacchus) (SEQ IT) NOs: 19 - 24 ) ( Figure 4).
  • the CDR2 sequences of the New World primate immunoglobulins that differ from that of Lhe acceptor CDR2 sequence can be identified as SASTLQT, DASSLQP, GASTRAT, KVSNRAS, RVSNRAS, KVSTRGF, A ⁇ SNRAS, TSSNLQA, KASTLQS, AASTLQS, YASSLQS, YASFLQG) (Table I).
  • BLAST analysis http://www.ncbi.nlm.nih.gov/BLAST/ oil each of these donor New World primate CDR2 sequences is performed to remove sequences that are exact matches for human immunoglobulin sequences. Sequences unique to New World primates were KVSNRAS, RVSNRAS, KVSTRCGP, A ⁇ SNRAS, TSSNLQA, DASSLQP, YASFLQG (Table 1 ).
  • Table 1 New World primate CDR2 sequences and their suitability as donor sequences.
  • the acceptor CDR2 and the potential donor CDR2s are examined for their predicted immunogenicity in humans by the MHC class U binding prediction program Proprcd (http://www.imleeh.res.in/raghava/prot>rcd) using a 1% threshold value analysis of all alleles. From this analysis the acceptor CDR2, SASELQS, forms part of the peptide, LIYSASF ⁇ LQ, which is predicted to bind MiIC class II encoded by 1 1 alleles (DRBl .0306, DRBl_0307.
  • DRB1_O3O8 DRB1_O311, DRBl-0401 , DRB1_O426, DRB1_0806, DRBl_0813, DKBlJ 50 L, DRB I .1502, DRB1_1506).
  • the donor CDR2 sequence, KVSNRAS forms part ⁇ f a sequence, LIYK VSNRAS, which is predicted to bind MHC class !l encoded by 9 alleles (DRB 1 _0309, DRB1_O4O2, DRB I _0802, DRB 1_0804, DRB 1. 0806, DRB l_0813, DRB 1 _1301 , DRB IJ 327, DRB I J328).
  • the donor CDR2 sequence, ⁇ ASNRAS forms part of a sequence, LlY AASNRA, which is predicted to bind MHC class Il encoded by 6 alleles (DRBl_0402, DRBl_0404, DRBl_040S, DRB1_O423, DIIB1_O813, DRB IJ 506).
  • the donor CDR2 sequence, TSSNLQA forms part of a sequence, Ll YTSSNLQA, which is predicted to bind MHC class l[ encoded by 10 alleles (DRBlJMO 1 , DRB l_0402, DRB1_0404, DRB I JWlO 7 DRBl JH23, DRBl J)42(>, DRBl_0813, DRB1J50I , DRBl J502, DRBl J 506).
  • the donor CDR2 sequence, KVSTRGP forms part of a sequence LUYKVSTR, which is predicted to bind MUC class II encoded by 8 alleles (DRB1_0309, DRBI_0802, DR.B1J0804, DRB1JJ806, DRB1JJ813, DRB 1 J301, DRB1_1327, DRB1_1328).
  • the acceptor CDR2 can be replaced with a donor CDR2 of lower predicted immunogenicity, including KVSNRAS, A ASNRAS, TSSNLQ ⁇ and KVSTRGP.
  • the acceptor CDR2 is replaced with the donor CDR2 sequences, generating the locally engineered domain antibodies (SEQ ID No: 25 - 31),
  • Examples of recombinant DN ⁇ technology include those described by Winter et al. (US 5,225,539), and include, but is not limited to, techniques such as sile-dirceled mutagenesis and oligo annealing.
  • Protein expression of the domain antibodies is then performed in E .colt BL21 (DE3) pLys (Novagcn, Germany) using a suitable vector for expression such as pBT21d(+) (Novagen, Germany), or by other such methods known in the art such as those describe by Basran et al.
  • domain antibodies are purified using Protein L (Pierce, USA) chromatography. Following purification the engineered domain antibodies are analysed for retention of TNF ⁇ binding ability by methods known in the art, such as the L929 neutralisation assay or the TNF ⁇ receptor I binding assay.
  • affinity maturation could be performed by amino acid substitution of the framework residues surrounding and stabilising CDR2 or by other methods known in the art.(Winter et al. (US 5,225,539); Griffiths et al (US 5,885,793); Rajpal, A. et at (2005) A general method for greatly improving the affinity ot antibodies by using combinatorial libraries, PrM Natl Acad Sci U S A , 102(24) 8466-71 ; Irving R.A. el al. (2001) Rtbosome display and affinity maturation: from antibodies to single V-d ⁇ mains and steps towards cancer therapeutics, Journal of Immunological Methods, 248: 31 -45).
PCT/AU2006/001166 2005-08-15 2006-08-15 Chimeric antibodies with new world primate regions WO2007019621A1 (en)

Priority Applications (8)

Application Number Priority Date Filing Date Title
NZ565904A NZ565904A (en) 2005-08-15 2006-08-15 Chimeric antibodies with New World primate CDR regions
BRPI0614867-0A BRPI0614867A2 (pt) 2005-08-15 2006-08-15 anticorpo quimérico ou uma porção de ligação de antìgeno do mesmo, método para produzir um anticorpo quimérico ou uma porção de ligação de antìgeno do mesmo, e, kit
AU2006281981A AU2006281981A1 (en) 2005-08-15 2006-08-15 Chimeric antibodies with new world primate regions
EP06774813A EP1945669A4 (en) 2005-08-15 2006-08-15 CHIMERIC ANTIBODIES WITH NEUWELTPRIMATENREGIONEN
JP2008526323A JP2009504686A (ja) 2005-08-15 2006-08-15 新世界霊長類領域を用いるキメラ抗体
CA002619245A CA2619245A1 (en) 2005-08-15 2006-08-15 Chimeric antibodies with new world primate regions
MX2008002161A MX2008002161A (es) 2005-08-15 2006-08-15 Anticuerpos quimericos con regiones de primate del nuevo mundo.
NO20080800A NO20080800L (no) 2005-08-15 2008-02-14 Kimaere antistoffer med New World primat regioner

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
AU2005904406A AU2005904406A0 (en) 2005-08-15 New World Antibodies
AU2005904406 2005-08-15

Publications (1)

Publication Number Publication Date
WO2007019621A1 true WO2007019621A1 (en) 2007-02-22

Family

ID=37757238

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/AU2006/001166 WO2007019621A1 (en) 2005-08-15 2006-08-15 Chimeric antibodies with new world primate regions

Country Status (13)

Country Link
EP (1) EP1945669A4 (es)
JP (1) JP2009504686A (es)
KR (1) KR20080068005A (es)
CN (2) CN101287763A (es)
AU (1) AU2006281981A1 (es)
BR (2) BRPI0614867A2 (es)
CA (1) CA2619245A1 (es)
MX (1) MX2008002161A (es)
NO (1) NO20080800L (es)
NZ (1) NZ565904A (es)
RU (1) RU2008110058A (es)
WO (1) WO2007019621A1 (es)
ZA (2) ZA200802247B (es)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7846439B2 (en) 2006-02-01 2010-12-07 Cephalon Australia Pty Ltd Domain antibody construct
JP2011530297A (ja) * 2008-08-14 2011-12-22 セファロン・オーストラリア・ピーティーワイ・リミテッド バリアントドメイン抗体
CN103214577A (zh) * 2007-03-22 2013-07-24 拜奥根Idec马萨诸塞公司 特异性结合cd154 的包括抗体、抗体衍生物和抗体片段在内的结合蛋白及其用途
US8961976B2 (en) 2004-07-26 2015-02-24 Biogen Idec Ma Inc. Anti-CD154 antibodies

Families Citing this family (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
RU2516931C2 (ru) * 2010-08-06 2014-05-20 Олег Ильич Эпштейн Способ и композиция для профилактики инфицирования вич, профилактики и лечения заболеваний, вызываемых вич или ассоциированных с вич, в том числе спида
RU2577299C2 (ru) * 2011-07-04 2016-03-10 Олег Ильич Эпштейн Способ лечения инфекционных заболеваний и комплексное лекарственное средство для лечения инфекционных заболеваний
RU2535033C2 (ru) * 2010-08-06 2014-12-10 Олег Ильич Эпштейн Лекарственное средство и способ профилактики инфицирования вич, профилактики и лечения заболеваний, вызываемых вич или ассоциированных с вич, в том числе спида
RU2521392C2 (ru) * 2010-08-06 2014-06-27 Олег Ильич Эпштейн Комплексное лекарственное средство для лечения вирусных заболеваний и способ лечения вирусных заболеваний
RU2505312C2 (ru) * 2010-08-06 2014-01-27 Олег Ильич Эпштейн Комплексное лекарственное средство для лечения гриппа различных типов
RU2519862C2 (ru) * 2010-08-06 2014-06-20 Олег Ильич Эпштейн Комплексное лекарственное средство и способ профилактики и лечения инфекционных вирусных заболеваний
RU2535034C2 (ru) * 2010-08-06 2014-12-10 Олег Ильич Эпштейн Лекарственное средство и способ профилактики инфицирования вич, профилактики и лечения заболеваний, вызываемых вич или ассоциированных с вич, в том числе спида
RU2502521C2 (ru) * 2010-08-06 2013-12-27 Олег Ильич Эпштейн Комплексное лекарственное средство для лечения бактериальных инфекций и способ лечения бактериальных инфекций
RU2517085C2 (ru) * 2010-08-06 2014-05-27 Олег Ильич Эпштейн Комплексное лекарственное средство и способ профилактики инфицирования вич, профилактики и лечения заболеваний, вызываемых вич или ассоциированных с вич, в том числе спида
RU2579262C1 (ru) * 2014-10-02 2016-04-10 Мапикс Эс.Эй.Ар.Эл. Лекарственное средство для лечения инфекционных заболеваний
CL2015002152A1 (es) * 2015-07-31 2016-06-03 Pontificia Universidad Católica De Chile Anticuerpos monoclonales específicos para el antígeno p del virus respiratorio sincicial humano (vrsh), producidos y secretados por hibridomas celulares, útiles para la detección y el diagnostico de la infección causada por vrsh

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SK285046B6 (sk) * 1991-07-25 2006-05-04 Idec Pharmaceuticals Corporation Chimérna protilátka, ktorá sa špecificky viaže na ľudský antigén, farmaceutický prostriedok s jej obsahom, spôsob jej výroby a použitie
US6569431B2 (en) * 2000-10-17 2003-05-27 The Regents Of The University Of California Recombinant antibody fragments as autoantibody antagonists
US7235643B2 (en) * 2000-11-07 2007-06-26 Morphotek, Inc. Antibodies and methods for generating genetically altered antibodies with high affinity
DK1517921T3 (da) * 2002-06-28 2006-10-09 Domantis Ltd Immunglobulin-enkeltvariable antigen-bindende domæner og dobbeltspecifikke konstruktioner deraf
WO2005047325A2 (en) * 2003-11-07 2005-05-26 Amgen Inc. Monkey immunoglobulin sequences

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
LANGERMANS J. ET AL.: "Reactivity of human T-lymphocyte-specific antibodies with peripheral blood mononuclear cells and spleen of Aotus azarae ssp. boliviensis (owl monkey)", J. MED. PRIMATOL., vol. 29, 2000, pages 397 - 401, XP008077203 *
See also references of EP1945669A4 *

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8961976B2 (en) 2004-07-26 2015-02-24 Biogen Idec Ma Inc. Anti-CD154 antibodies
US7846439B2 (en) 2006-02-01 2010-12-07 Cephalon Australia Pty Ltd Domain antibody construct
CN103214577A (zh) * 2007-03-22 2013-07-24 拜奥根Idec马萨诸塞公司 特异性结合cd154 的包括抗体、抗体衍生物和抗体片段在内的结合蛋白及其用途
CN103214577B (zh) * 2007-03-22 2015-09-02 生物基因Ma公司 特异性结合cd154的包括抗体、抗体衍生物和抗体片段在内的结合蛋白及其用途
JP2011530297A (ja) * 2008-08-14 2011-12-22 セファロン・オーストラリア・ピーティーワイ・リミテッド バリアントドメイン抗体

Also Published As

Publication number Publication date
MX2008002161A (es) 2008-04-22
BRPI0614430A2 (pt) 2011-03-29
EP1945669A1 (en) 2008-07-23
NO20080800L (no) 2008-05-09
BRPI0614867A2 (pt) 2012-01-31
ZA200802247B (en) 2009-08-26
KR20080068005A (ko) 2008-07-22
EP1945669A4 (en) 2009-07-22
RU2008110058A (ru) 2009-09-27
CN101287763A (zh) 2008-10-15
AU2006281981A1 (en) 2007-02-22
ZA200802246B (en) 2009-09-30
CA2619245A1 (en) 2007-02-22
CN101287762A (zh) 2008-10-15
JP2009504686A (ja) 2009-02-05
NZ565904A (en) 2012-02-24

Similar Documents

Publication Publication Date Title
EP1945669A1 (en) Chimeric antibodies with new world primate regions
US20080095767A1 (en) Engineered antibodies with new world primate framework regions
US20080255343A1 (en) Chimeric antibodies
JP5030782B2 (ja) Tnfr1に対する単一ドメイン抗体およびその使用方法
AU2007211829B2 (en) Domain antibody construct
CN107849136B (zh) 抗TfR抗体及其在治疗增殖性和炎性疾病中的用途
AU2006326937B2 (en) Anti-inflammatory dAb
TW200848427A (en) Ligand
AU2008219666A1 (en) Antagonist OX40 antibodies and their use in the treatment of inflammatory and autoimmune diseases
JP2009525031A5 (es)
MX2008002162A (es) Anticuerpos diseñados con regiones de estructura de primate del nuevo mundo
NZ569405A (en) Anti-inflammatory domain antibody binding to TNF-alpha

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 200680038256.5

Country of ref document: CN

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

Ref document number: 12008500362

Country of ref document: PH

WWE Wipo information: entry into national phase

Ref document number: 2006281981

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2619245

Country of ref document: CA

Ref document number: 621/KOLNP/2008

Country of ref document: IN

Ref document number: 565904

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: MX/a/2008/002161

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: 2008526323

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2006281981

Country of ref document: AU

Date of ref document: 20060815

Kind code of ref document: A

WWP Wipo information: published in national office

Ref document number: 2006281981

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2006774813

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 1020087006270

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: 2008110058

Country of ref document: RU

WWP Wipo information: published in national office

Ref document number: 2006774813

Country of ref document: EP

ENP Entry into the national phase

Ref document number: PI0614867

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20080215