WO2008012543A1 - Single chain fc polypeptides - Google Patents

Single chain fc polypeptides Download PDF

Info

Publication number
WO2008012543A1
WO2008012543A1 PCT/GB2007/002842 GB2007002842W WO2008012543A1 WO 2008012543 A1 WO2008012543 A1 WO 2008012543A1 GB 2007002842 W GB2007002842 W GB 2007002842W WO 2008012543 A1 WO2008012543 A1 WO 2008012543A1
Authority
WO
WIPO (PCT)
Prior art keywords
domain
seq
terminus
linker
polypeptide according
Prior art date
Application number
PCT/GB2007/002842
Other languages
French (fr)
Inventor
Alastair David Griffiths Lawson
Paul Edward Stephens
Original Assignee
Ucb Pharma S.A.
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 Ucb Pharma S.A. filed Critical Ucb Pharma S.A.
Priority to JP2009521344A priority Critical patent/JP5384338B2/en
Priority to EP07789062.2A priority patent/EP2046832B1/en
Priority to ES07789062.2T priority patent/ES2564389T3/en
Priority to CA2658542A priority patent/CA2658542C/en
Priority to CN200780027807.2A priority patent/CN101495510B/en
Priority to US12/374,213 priority patent/US10479824B2/en
Publication of WO2008012543A1 publication Critical patent/WO2008012543A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • 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
    • A61P1/02Stomatological preparations, e.g. drugs for caries, aphtae, periodontitis
    • 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
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • 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
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • 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
    • A61P1/18Drugs for disorders of the alimentary tract or the digestive system for pancreatic disorders, e.g. pancreatic enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • 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
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • A61P19/10Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
    • 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
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • 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
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/10Antimycotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • 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
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/02Antithrombotic agents; Anticoagulants; Platelet aggregation inhibitors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/14Vasoprotectives; Antihaemorrhoidals; Drugs for varicose therapy; Capillary stabilisers
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/52Constant or Fc region; Isotype
    • 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
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • C07K2317/734Complement-dependent cytotoxicity [CDC]

Definitions

  • the present invention relates to single chain polypeptides comprising one or more immunoglobulin Fc domains.
  • the present invention relates to single- chain polypeptides in which at least one functional Fc domain is formed within the chain.
  • Immunoglobulins are bivalent Y-shaped molecules comprising two identical heavy chains and two identical light chains. Disulfide bonds link together the heavy and light chain pairs as well as the two heavy chains.
  • Each chain consists of one variable domain that varies in sequence and is responsible for antigen binding, these are known as the V H and V L domains for the heavy and light chains respectively.
  • Each chain also consists of at least one constant domain. In the light chain there is a single constant domain (C L ) and in the heavy chain there are at least three (C H I, C H 2 and C H 3), sometimes four (CH4) depending on the isotype.
  • IgA which includes IgAl and IgA2
  • IgD which includes IgAl and IgA2
  • IgE which includes subclasses IgGl, IgG2, IgG3 and IgG4
  • IgM immunoglobulins
  • the Fc domain of an antibody typically comprises at least the last two heavy chain constant region domains of each chain which dimerise to form the Fc domain.
  • the Fc domain is responsible for providing antibody effector functions, including determining antibody half-life and distribution throughout the body, ability to fix complement and binding to cell surface Fc receptors.
  • the properties of Fc domains have made them useful therapeutic agents and Fc domains have been fused to other non-antibody proteins, such as receptor proteins e.g etanercept.
  • Fc domain fusions have also been used as research reagents, 'Fc tags', which facilitate fusion protein detection and purification.
  • each chain comprises two Fc domains i.e. each antibody chain comprises in linear sequence CH2 CH3 CH2 CH3 and these domains dimerise to form two functional Fc domains to provide enhanced effector functions.
  • WO2005017148 and Hayden et al. supra describe single chain polypeptides comprising a single-chain Fv fused to half of an Fc domain i.e.
  • These polypeptides can exist as both monomers or dimers.
  • the binding specificity of antibodies has made these useful therapeutic agents, however, bivalent molecules such as antibodies are often inappropriate targeting agents for certain cell surface antigens. Bivalent binding can cause the target cell to undergo co-stimulation, activation and/or antigenic modulation, thereby offering the cell a means of evading complement and the various effector cells recruited by the Fc domain of the antibody.
  • antibodies have typically been conjugated to drugs or toxins that kill the cells upon internalization.
  • univalent antibody fragments do not cause antigenic modulation as no redistribution of surface antigen occurs and hence no co-stimulation and no internalization. It would be desirable therefore to retain the natural effector functions of an antibody in such fragments and thus avoid the need for costly and time consuming attachment of drugs or toxins.
  • An example of one such antibody fragment was produced by proteolytic cleavage of a rabbit IgG by Glennie and Stevenson, 1982, Nature, 295, 712-713. The fragment comprised only a single Fab binding site but retained the entire Fc domain.
  • the fragment was produced by papain digestion of a rabbit IgG antibody Al 2 allotypic variant which is glycosylated on one chain, making that chain resistant to papain digestion, thus allowing one Fab arm to be retained.
  • the fragment produced was demonstrated to be more effective in invoking complement mediated lysis of cells than the whole IgG.
  • Similar fragments have been produced from human IgG by proteolytic digestion (Michaelsen and Natvig, Scand. J. Immunology, 1973, 2, 299-312; Michaelsen and Natvig, Scand. J. Immunology, 1972, 1, 255-268) and by chemical cleavage (Wines and Easterbrook- Smith, Molecular Immunology, 1991, 28, 8, 855-863). These fragments are not practical to produce on a commercial scale as the use of proteolysis requires long preparation times and results in low yields and mixed products.
  • WO20050010125 describes hybrid proteins comprising two polypeptide chains, the first polypeptide chain comprising an Fc region and a biologically active molecule and the second polypeptide chain comprising an Fc region without the biologically active molecule of the first chain.
  • the two chains are produced separately and are either allowed to dimerise or are chemically conjugated together. Although this achieves the desired functional molecule, the preparation is complex and involves low yielding chromatographic procedures.
  • Surprisingly we have now found that it possible to produce a functional Fc domain as a single-chain polypeptide.
  • the polypeptides of the present invention therefore have the advantage that they can be produced recombinantly in large amounts and can be linked by any suitable means to any other molecule, such as a binding domain. Further, as the antibody constant domains form an Fc domain within the chain, the polypeptides of the present invention are not prone to dimerisation thus, where desired, bi-valent binding domains can be avoided.
  • the present invention provides a single chain polypeptide comprising two CH2 domains and two CH3 domains characterized in that said CH2 and CH3 domains form a functional Fc domain within the chain.
  • the functional Fc domain in the single-chain polypeptides of the present invention is not formed by dimerisation of two chains i.e. the two CH2 domains and two CH3 domains are present in a single chain and form a functional Fc domain within the single chain.
  • the present invention provides a single chain polypeptide comprising two CH2 domains and two CH3 domains characterized in that said CH2 and CH3 domains form a functional Fc domain within the chain and not by dimerisation with another polypeptide chain. Accordingly, in the single chain polypeptide of the present invention a first CH2 domain is dimerised with a second CH2 domain and a first CH3 domain is dimerised with a second CH3 domain within the polypeptide chain.
  • the term 'functional' as used herein refers to the ability of the Fc domain formed within the single chain polypeptide to provide one or more effector functions usually associated with Fc domains although it will be appreciated that other functions maybe engineered into such domains.
  • effector functions include determining the half-life and/or distribution of the Fc polypeptide throughout the body, the ability of the Fc polypeptide to fix complement and the ability of the Fc polypeptide to bind to cell surface Fc receptors.
  • effector functions include but are not limited to, antibody-dependent cellular cytotoxicity (ADCC), antibody-dependent cellular phagocytosis (ADCP) and complement-dependent cytotoxicity (CDC).
  • the Fc domain of the present invention comprises four or more constant domains which may be derived from any suitable species and/or class of antibody.
  • the constant domains are human.
  • IgA which includes IgAl and IgA2
  • IgD which includes IgAl
  • IgE IgG
  • IgM IgM
  • Any suitable Fc domain may be used depending on the effector functions required.
  • Fc domain refers to the last two constant region immunoglobulin domains of IgA, IgD and IgG (CH2 and CH3) and the last three constant region domains of IgE and IgM (CH2, CH3 and CH4), although it will be appreciated that in certain circumstances not all the domains may be required, for example in the case of IgE or IgM only CH2 and CH3 domains may be sufficient. It will also be appreciated that more than one Fc domain may be formed within the single chain Fc polypeptide and that these Fc domains may be derived from the same or different isotypes.
  • the Fc domain is derived from IgA and the single-chain Fc polypeptide comprises two C ⁇ 2 domains and two C ⁇ 3 domains.
  • the Fc domain is derived from IgM and the single-chain Fc polypeptide comprises two C ⁇ 2 domains, two C ⁇ 3 domains and two C ⁇ 4 domains.
  • the Fc domain is derived from IgD and the single-chain Fc polypeptide comprises two C ⁇ 2 domains and two C ⁇ 3 domains.
  • the Fc domain is derived from IgE and the single-chain Fc polypeptide comprises two C ⁇ 2 domains, two C ⁇ 3 domains and two C ⁇ 4 domains.
  • the Fc domain of the present invention is derived from an IgG and the single-chain Fc polypeptide comprises two C ⁇ 2 and two C ⁇ 3 domains.
  • the preferred sequences for the C ⁇ 2 domain of IgGl, IgG2, IgG3 and IgG4 for use in the present invention are provided in SEQ ID NOS: 2, 15, 28 and 41 respectively and the preferred sequences for the C ⁇ 3 domain of IgGl, IgG2, IgG3 and IgG4 for use in the invention are provided in SEQ ID NOS: 3, 16, 29 and 42 respectively.
  • the present invention provides a single chain polypeptide comprising two C ⁇ 2 domains and two C ⁇ 3 domains characterized in that said C ⁇ 2 and C ⁇ 3 domains form a functional Fc domain within the chain i.e. a first C ⁇ 2 domain dimerises with a second C ⁇ 2 domain and a first C ⁇ 3 domain dimerises with a second C ⁇ 3 domain within the polypeptide chain
  • the constant region domains for use in producing the Fc domain of the present invention may include variants of the naturally occurring constant domains described herein above. Such variants may comprise one or more amino acid variations compared to wild type constant domains.
  • the Fc domain of the present invention comprises at least one constant domain which varies in sequence from the wild type constant domain. It will be appreciated that the variant constant domains may be longer or shorter than the wild type constant domain. Preferably the variant constant domains are at least 50% identical or similar to a wild type constant domain.
  • Identity indicates that at any particular position in the aligned sequences, the amino acid residue is identical between the sequences.
  • Similarity indicates that, at any particular position in the aligned sequences, the amino acid residue is of a similar type between the sequences.
  • leucine may be substituted for isoleucine or valine.
  • Other amino acids which can often be substituted for one another include but are not limited to:
  • variant constant domains are at least 60% identical or similar to a wild type constant domain. In another example the variant constant domains are at least 70% identical or similar. In another example the variant constant domains are at least 80% identical or similar. In another example the variant constant domains are at least 90% identical or similar. In another example the variant constant domains are at least 95% identical or similar.
  • the variant constant domains provide equivalent Fc effector functions compared to the wild type Fc domain. In one embodiment the variant constant domains provide improved effector functions. In one embodiment the variant constant domains provide altered effector functions. In one example the Fc domain provides no effector functions other than extended half-life. In one example the Fc domain provides FcR binding but not CIq binding. In one example the Fc domain provides CIq binding but not FcR binding.
  • Fc variant polypeptides are known in the art, see for example Idusogie et ah, Journal of Immunology, 2000, 164, 4178-4184 and Shields et ah, Journal of Biological Chemistry, 2001, 276, 9, 6591-6604.
  • a comprehensive list of Fc variants is provided by WO2005077981 (see in particular, paragraph number 80) and these are incorporated herein by reference.
  • Fc variants include in IgGl N314Q (or N297Q), T318A (T299A), A349S (A330S) with P350A (P331A), L247A (L234A) with L248A (L235A) or P348A (P329A) (number in brackets is EU numbering).
  • IgG4 Fc domain the S241P (S228P) mutation may be used (Angal et al., Molecular Immunology, 1993, 30 (1), 105-108). It will be appreciated that any suitable variant may be produced and tested using routine methods known in the art.
  • the CH2, CH3 and where present CH4 domains of the single-chain Fc polypeptide of the present invention are linked in the single polypeptide chain such that they can still form a functional Fc domain within the chain.
  • any suitable amino acid linkers may be used to link these constant domains provided they allow a functional Fc domain to form within the single-chain polypeptide.
  • Suitable amino acids for use in linkers of the present invention include, but are not limited to, small flexible amino acids such as GIy, Ser, Ala and Thr.
  • the linker comprises or consists of glycine residues.
  • the linker comprises or consists of serine residues.
  • the linker comprises or consists of alanine residues.
  • the linker comprises or consists of threonine residues. In one embodiment the linker comprises or consists of glycine and serine residues. In one embodiment the linker comprises or consists of glycine, serine and alanine residues. In one embodiment the linker comprises or consists of glycine, serine, alanine and threonine residues. For the avoidance of doubt, it is understood that all permutations of glyine and/or serine and/or alanine and/or threonine residues are included. In one example the linker comprises or consists of 30-80% glycine residues and 20-70% serine residues.
  • the linker comprises or consists of 35-50% glycine residues; 30-40% serine residues; 5-15% threonine residues and 10-20% alanine residues.
  • the amino acid residues are randomly distributed within the linker.
  • suitable linkers include glycine-serine polymers comprising for example repeats of sequences such as GS, GSGGS, GGGGS and GGGS.
  • the Fc domain of the single chain polypeptide of the present invention comprises two CH2 domains and two CH3 domains.
  • the present invention provides a single chain Fc polypeptide comprising two CH2 domains and two CH3 domains wherein in N- to C-terminal sequence, a first CH2 domain is linked at its C-terminus to the N- terminus of a first CH3 domain and said first CH3 domain is linked at its C-terminus via a linker to the N-terminus of a second CH2 domain which is linked at its C- terminus to the N-terminus a second CH3 domain (as shown in Figure 1).
  • the first CH2 domain is directly linked i.e. genetically fused at its C-terminus to the N-terminus of the first CH3 domain.
  • the second CH2 domain is directly linked i.e. genetically fused at its C-terminus to the N-terminus of the second CH3 domain.
  • CH2 domains genetically fused to CH3 domain(s) are given in SEQ ID NOS: 5, 18, 31 and 44.
  • a linker is employed to link the C-terminus of the first CH2 domain to the N-terminus of the first CH3 domain.
  • a linker is employed to link the C-terminus of the second CH2 domain to the N-terminus of the second CH3 domain.
  • linker is used to link (i) the C-terminus of the first CH2 domain to the N-terminus of the first CH3 domain and/or (ii) the C-terminus of the second CH2 domain to the N-terminus of the second CH3 domain
  • the linker will be of sufficient length to allow a functional Fc domain to form within the chain.
  • the linker will only be a few amino acids in length, preferably less than 10 amino acids in length.
  • a linker is used in both (i) and (ii) above the two linkers may be the same or different.
  • the linkers will be approximately the same length.
  • the linker used to link the C-terminus of the first CH3 domain to the N- terminus of the second CH2 domain will be sufficiently long to allow a functional Fc domain to form within the chain i.e. it will be sufficiently long to allow a first CH2 domain to dimerise with a second CH2 domain and a first CH3 domain to dimerise with a second CH3 domain within the polypeptide chain.
  • the linker is around 30-100 amino acids in length, in another embodiment the linker is around 40 to 100 amino acids in length. In one embodiment the linker is around 40 to 90 amino acids in length. In one embodiment the linker is around 40 to 80 amino acids in length, preferably 40 to 70 amino acids in length. Suitable amino acids for use in these linkers are described herein above.
  • the linker comprises or consists of 35-50% glycine residues; 30-40% serine residues; 5-15% threonine residues and 10-20% alanine residues.
  • the amino acid residues are randomly distributed within the linker.
  • An example of a suitable linker is provided in SEQ ID NO:62.
  • the linker may comprise, preferably towards its C-terminus one or more cysteine residues.
  • the linker may comprise at its C-terminus the sequence of all or part of the hinge region of an antibody or variant thereof comprising one or more cysteine residues.
  • linker of SEQ ID NO:62 further comprises at its C-terminus any one of the sequences provided in SEQ ID NOs: 53-57.
  • the linker is around 30 to 130 amino acids in length. In one embodiment the linker is around 50 to 100 amino acids in length. In one embodiment the linker is around 50 to 80 amino acids in length.
  • the present invention provides a single chain Fc polypeptide comprising two CH2 domains and two CH3 domains wherein in N- to C-terminal sequence a first CH2 domain is linked at its C-terminus via a linker to the N-terminus of a second CH2 domain and said second CH2 domain is linked at its C- terminus to the N-terminus of a first CH3 domain and said first CH3 domain is linked at its C-terminus to the N-terminus of a second CH3 domain via a linker (as shown in Figure 2).
  • the second CH2 domain is genetically fused at its C- terminus to the N-terminus of the first CH3 domain.
  • the C-terminus of the second CH2 domain is connected by a linker to the N-terminus of the first CH3 domain.
  • linker When a linker is used to link the C-terminus of the second CH2 domain to the N-terminus of the first CH3 domain the linker will be of sufficient length to allow a functional Fc domain to form within the chain. Typically the linker will only be a few amino acids in length, preferably less than 10 amino acids in length.
  • linker used to link the C-terminus of the first CH2 domain to the N- terminus of the second CH2 domain and the linker used to link the C-terminus of the first CH3 domain to the N-terminus of the second CH3 domain will be sufficiently long to allow a functional Fc domain to form within the chain. It will be appreciated that these two linkers may be the same or different in both composition and/or length. In one embodiment one or both of the linkers is between 15 and 50 amino acids in length. In one embodiment one or both of the linkers is between 15 and 40 amino acids in length. In one embodiment one or both of the linkers is between 20 and 40 amino acids in length. In another embodiment one or both of the linkers is between 20 and 35 amino acids in length.
  • linker comprises or consists of 50-80% glycine residues and 10-30% serine residues. In one example the amino acid residues are randomly distributed within the linker.
  • the linker between the first CH2 domain and the second CH2 domain comprises, preferably towards its C-terminus, one or more cysteine residues.
  • the linker comprises all or part of an antibody hinge sequence or variant thereof as described herein above. Accordingly, in one embodiment the linker between the C-terminus of the first CH2 domain and the N- terminus of the second CH2 domain having the sequence given in SEQ ID NO: 63 further comprises at its C-terminus any one of the hinge sequences provided in SEQ ID NOs 53-57.
  • the linker between the C-terminus of the first CH2 domain and the N-terminus of the second CH2 domain is between 25 and 90 amino acids in length. In one embodiment this linker is between 25 and 80 amino acids in length.
  • this linker is between 25 and 70 amino acids in length. In one embodiment this linker is between 25 and 60 amino acids in length. In one embodiment this linker is between 25 and 50 amino acids in length. In one embodiment this linker is between 25 and 40 amino acids in length.
  • the Fc domain of the single chain Fc polypeptide of the present invention comprises two CH2 domains, two CH3 domains and one or two CH4 domains, preferably two.
  • the present invention provides a single chain Fc polypeptide comprising two CH2 domains, two CH3 domains and two CH4 domains wherein in N- to C-terminal sequence, a first CH2 domain is linked at its C-terminus to the N-terminus of a first CH3 domain, said first CH3 domain is linked at its C- terminus to the N-terminus of a first CH4 domain and said first CH4 domain is linked at its C-terminus via a linker to the N-terminus of a second CH2 domain which is linked at its C-terminus to the N-terminus of a second CH3 domain which is linked at its C-terminus to the N-terminus of a second CH4 domain (see for example Figure 3 a).
  • one or more of the following domains are directly linked i.e. genetically fused (i) the C-terminus of the first CH2 domain to the N-terminus of the first CH3 domain (ii) the C-terminus of the second CH2 domain to the N- terminus of the second CH3 domain (iii) the C-terminus of the first CH3 domain to the N-terminus of the first CH4 domain (iv) the C-terminus of the second CH3 domain to the N-terminus of the second CH4 domain.
  • one or more of the following domains are connected by a linker (v) the C-terminus of the first CH2 domain to the N-terminus of the first CH3 domain (vi) the C-terminus of the second CH2 domain to the N-terminus of the second CH3 domain (vii) the C-terminus of the first CH3 domain to the N-terminus of the first CH4 domain (viii) the C-terminus of the second CH3 domain to the N- terminus of the second CH4 domain.
  • a linker is present between any one of (v, vi, vii or viii) this linker will be of sufficient length to allow a functional Fc domain to form within the chain.
  • the linker will only be a few amino acids in length. Where there is more than one linker it will be appreciated that these may be the same or different.
  • the linkers will be approximately the same length.
  • the linker used to link the C-terminus of the first CH4 domain to the N- terminus of the second CH2 domain will be sufficiently long to allow a functional Fc domain to form within the chain.
  • the linker is around 50-100 amino acids in length, in another embodiment the linker is around 60 to 100 amino acids in length. In one embodiment the linker is around 70 to 100 amino acids in length, preferably 80 to 100 amino acids in length. Suitable amino acids for use in these linkers have been described herein above.
  • the linker may comprise, preferably towards its C-terminus, one or more cysteine residues.
  • the linker comprises all or part of an antibody hinge sequence or variant thereof as described herein above.
  • the present invention provides a single chain Fc polypeptide comprising two CH2 domains, two CH3 domains and two CH4 domains wherein in N- to C-terminal sequence a first CH2 domain is linked at its C-terminus via a linker to the N-terminus of a second CH2 domain and said second CH2 domain is linked at its C-terminus to the N-terminus of a first CH3 domain and said first CH3 domain is linked at its C-terminus to the N-terminus of a first CH4 domain which is linked at its C-terminus, via a linker, to the N-terminus of a second CH3 domain which is linked at its C-terminus to the N-terminus of a second CH4 domain (see for example Figure 3b).
  • one or more of the following domains are directly linked i.e. genetically fused (i) the C-terminus of the second CH2 domain to the N-terminus of the first CH3 domain (ii) the C-terminus of the first CH3 domain to the N- terminus of the first CH4 domain (iii) the C-terminus of the second CH3 domain to the N-terminus of the second CH4 domain.
  • one or more of the following domains are connected by a linker (i) the C-terminus of the second CH2 domain to the N-terminus of the first CH3 domain (ii) the C-terminus of the first CH3 domain to the N-terminus of the first CH4 domain (iii) the C-terminus of the second CH3 domain to the N-terminus of the second CH4 domain.
  • linker is present between one or more of (i) the C-terminus of the second CH2 domain and the N-terminus of the first CH3 domain (ii) the C-terminus of the first CH3 domain and the N-terminus of the first CH4 domain (iii) the C- terminus of the second CH3 domain and the N-terminus of the second CH4 domain (iii) the C- terminus of the second CH3 domain and the N-terminus of the second CH4 domain the linker will be of sufficient length to allow a functional Fc domain to form within the chain.
  • the linker will only be a few amino acids in length. Where there is more than one linker it will be appreciated that these may be the same or different.
  • the linkers will be approximately the same length.
  • linker between the C-terminus of the first CH2 domain and the N- terminus of the second CH2 domain and the linker between the C-terminus of the first CH4 domain and the N-terminus of the second CH3 domain will be sufficiently long to allow a functional Fc domain to form within the chain. Suitable amino acids for these linkers have been described herein above.
  • the linker between the C-terminus of the first CH2 domain and the N- terminus of the second CH2 domain is typically between 15 and 40 amino acids in length. In another embodiment the linker is between 20 and 35 amino acids in length. In one embodiment the linker between the C-terminus of the first CH2 domain and the N-terminus of the second CH2 domain comprises the sequence (GGGGS)n where n-5 (SEQ ID NO:63).
  • the linker between the first CH2 domain and the second CH2 domain comprises, preferably towards its C-terminus, one or more cysteine residues.
  • the linker comprises all or part of an antibody hinge sequence or variant thereof as described herein above which may comprise one or more cysteine residues. Suitable hinge sequences include those provided in SEQ ID NOs 53-57.
  • the linker between the C-terminus of the first CH4 domain and the N- terrninus of the second CH3 domain is typically around 30-100 amino acids in length, in another embodiment the linker is around 40 to 100 amino acids in length. In one embodiment the linker is around 40 to 80 amino acids in length, preferably 40 to 70 amino acids in length.
  • An example of a suitable linker is provided in SEQ ID NO:62.
  • the present invention provides a single chain Fc polypeptide comprising two CH2 domains, two CH3 domains and two CH4 domains wherein in N- to C-terminal sequence a first CH2 domain is linked at its C-terminus to the N-terminus of a first CH3 domain and said first CH3 domain is linked at its C- terminus via a linker to the N-terminus of a second CH2 domain and said second CH2 domain is linked at its C-terminus to the N-terminus of a second CH3 domain which is linked at its C-terminus to the N-terminus of a first CH4 domain which is linked at its C-terminus, via a linker, to the N-terminus of a second CH4 domain (see for example Figure Ic).
  • one or more of the following domains are connected by a linker (i) the C-terminus of the first CH2 domain and the N-terminus of the first CH3 domain (ii) the C-terminus of the second CH2 domain and the N-terminus of the second CH3 domain (iii) the C-terminus of the second CH3 domain and the N- terminus of the first CH4 domain.
  • linker will be of sufficient length to allow a functional Fc domain to form within the chain.
  • linker will only be a few amino acids in length. Where there is more than one linker it will be appreciated that these may be the same or different. Preferably the linkers will be approximately the same length.
  • the linker between the C-terminus of the first CH3 domain and the N- terminus of the second CH2 domain and the linker between the C-terminus of the first CH4 domain and the N-terminus of the second CH4 domain will be of sufficient length to allow a functional Fc domain to form within the chain.
  • the linker between the C-terminus of the first CH3 domain and the N- terminus of the second CH2 domain is typically around 30-100 amino acids in length, in another embodiment the linker is around 40 to 100 amino acids in length. In one embodiment the linker is around 40 to 80 amino acids in length, preferably 40 to 70 amino acids in length.
  • An example of a suitable linker is provided in SEQ ID NO:62.
  • the linker between the first CH3 domain and the second CH2 domain comprises, preferably towards its C-terminus, one or more cysteine residues.
  • this linker comprises all or part of an antibody hinge sequence or variant thereof as described herein above which may comprise one or more cysteine residues.
  • Suitable hinge sequences include SEQ ID NOs 53-57.
  • the single chain Fc polypeptide of the present invention further comprises an amino acid linker genetically fused to the N-terminus of the first CH2 domain.
  • the linker may comprise any suitable amino acids and be of any suitable length.
  • the linker comprises one or more cysteine residues.
  • the linker genetically fused to the N-terminus of the first CH2 domain comprises all or part of an antibody hinge sequence or variant thereof as described herein above.
  • the linker comprises the sequence given in any one of SEQ ID NOs:53-57.
  • one or more of the cysteine residues present in the linker are disulphide linked to one or more cysteine residues present in any one of the following linkers where present (i) the linker which connects the C-terminus of the first CH3 domain and the N-terminus of the second CH2 domain (see for example Figure 1 a) (ii) the linker which connects the C-terminus of the first CH2 domain to the N-terminus of the second CH2 domain (see for example Figure 2a) or (iii) the linker which connects the C-terminus of the first CH4 domain to the N-terminus of the second CH2 domain.
  • linker fused to the N-terminus may comprise all or part of the hinge region of an antibody or variant thereof in which one or more cysteines have been substituted for another amino acid, preferably serine.
  • suitable linkers of this type are provided in SEQ ID NOs: 58-61.
  • single chain Fc polypeptides are provided for IgGl, 2, 3 and 4 in SEQ ID NOS: 8-13, 21-26, 34-39 and 47-52 respectively. See also Figure 6 for examples of IgGl sequences. The invention also extends to variants of these sequences as set out herein above.
  • the present invention provides a single chain Fc polypeptide comprising a sequence having at least 70% identity or similarity to any one of the sequences given in SEQ ID NOS: 8-13, 21-26, 34-39 and 47-52.
  • a single chain Fc polypeptide of the present invention comprises a sequence having at least 80% identity or similarity to any one of the sequences given in SEQ ID NOS: 8-13, 21-26, 34-39 and 47-52. In another example a single chain Fc polypeptide of the present invention comprises a sequence having at least 90% identity or similarity to any one of the sequences given in SEQ ID NOS: 8-13, 21-26, 34-39 and 47-52. In another example a single chain Fc polypeptide of the present invention comprises a sequence having at least 95% or 98% identity or similarity to any one of the sequences given in SEQ ID NOS: 8-13, 21-26, 34-39 and 47-52.
  • the single chain Fc polypeptide of the present invention further comprises a CHl domain fused optionally via a hinge to the N-terminus of the first CH2 domain.
  • CHl domains are provided in SEQ ID NOs I, 14, 27 and 40.
  • the single chain Fc polypeptides of the present invention can be used in a number of applications, including therapeutic, diagnostic and research applications.
  • the single chain Fc polypeptides of the present invention further comprise one or more other molecules which may be fused or otherwise linked at the N and/or C-terminus and/or elsewhere on the polypeptide.
  • Such molecules include, but are not limited to, nucleic acids, small molecules, carbohydrates, proteins and peptides, including for example receptor proteins, antibodies and antibody fragments.
  • the single-chain Fc polypeptide may be linked to another molecule, optionally via a linker (amino acid or chemical), by any suitable means known in the art, including for example, chemical conjugation, chemical cross-linking or genetic fusion.
  • the single chain Fc polypeptide comprises a cysteine containing linker, such as an antibody hinge, at its N-terminus and one of these free cysteines is used as a site of attachment for another molecule, preferably a biologically active molecule as described below.
  • a cysteine containing linker such as an antibody hinge
  • the single-chain Fc polypeptides of the present invention are used as an Fc tag, for example to aid protein purification and/or protein detection. Accordingly in one embodiment the single-chain Fc polypeptide further comprises at its N-terminus all or part of another protein.
  • Such Fc fusions advantageously do not dimerise unlike currently available Fc fusions thus ensuring that the fusion protein remains monomelic.
  • the single-chain Fc polypeptide may be linked to another protein via a cleavable linker.
  • the single-chain Fc polypeptide of the present invention is linked at its N and/or C-terminus to a biologically active molecule.
  • the biologically active molecule may be any protein or other suitable molecule, including nucleic acids, small molecules, carbohydrates, receptor proteins or immunoglobulins.
  • biologically active molecules include enzymes, antibody fragments, domain antibodies, single chain antibodies, aptamers, MicrobodiesTM, binding agents based on protein scaffolds (see for example Nygren and Uhlen, 1997, Current Opinion in Structural Biology, 7, 463-469) versabodies, avimers, adnectins, anticalins, phylomers, aptamers, cyclic peptides, peptides, antiviral agents, hemostatic agents and cytokines and growth factors such as EPO, RANTES, interleukins such as IL-I, IL-2, IL-3, IL-4, IL-5, IL-6, IL-8, IL-12, IL-13, IL-16 or IL-17, interferons such as interferon ⁇ , interferon ⁇ or interferon ⁇ , tumour necrosis factor- ⁇ , tumor necrosis factor- ⁇ , colony stimulating factors such as G-CSF or GM- CSF.
  • interferons such as interferon
  • the biologically active molecule brings the single-chain Fc polypeptide of the present invention into contact with a desired target, for example a target protein.
  • a desired target protein for example a target protein.
  • the biologically active molecule binds to a desired target protein.
  • the target protein is a cell-associated protein, for example a cell surface protein on cells such as bacterial cells, yeast cells, T-cells, endothelial cells or tumour cells, or it may be a soluble protein.
  • Target proteins may also be any medically relevant protein such as those proteins upregulated during disease or infection, for example receptors and/or their corresponding ligands.
  • cell surface proteins include adhesion molecules, for example integrins such as ⁇ l integrins e.g.
  • the target protein is a soluble protein.
  • Soluble proteins include interleukins such as IL-I, IL-2, IL-3, IL-4, IL-5, IL-6, IL-8, IL-12, IL-13, IL- 16 or IL- 17, viral proteins for example respiratory syncytial virus or cytomegalovirus proteins, immunoglobulins, such as IgE, interferons such as interferon ⁇ , interferon ⁇ or interferon ⁇ , tumour necrosis factor- ⁇ , tumor necrosis factor- ⁇ , colony stimulating factors such as G-CSF or GM-CSF, and platelet derived growth factors such as PDGF- ⁇ , and PDGF- ⁇ and where appropriate receptors thereof.
  • the single-chain Fc polypeptides of the present invention may be used to functionally alter the activity of a particular protein to which the biologically active molecule binds.
  • the single-chain Fc polypeptide may neutralize, antagonize or agonise the activity of a protein.
  • the binding of the single-chain Fc polypeptide to a cell via the biologically active molecule results in cell killing e.g via complement mediated cytotoxicity.
  • the biologically active molecule is a monovalent binding domain, in particular, a monovalent protein such as a receptor or fragment thereof or an immunoglobulin or fragment thereof.
  • the biologically active molecule is a receptor that may be naturally expressed on a cell surface or inside the cell.
  • suitable receptors include, but are not limited to, viral receptors, cytokine receptors, growth factor receptors, hormone receptors and bacterial receptors.
  • the term 'receptor' as used herein also includes suitable fragments of such receptors, an example of which includes the extracellular domain of a receptor.
  • the receptor is the human gpl30 receptor or a cytokine binding fragment thereof, such as domain 1, 2 and/or 3.
  • the biologically active molecule comprises domain 1 of the gpl30 receptor or a fragment thereof.
  • the biologically active molecule comprises amino acids 1 to 125 of SEQ ID NO:91.
  • the biologically active molecule comprises domain 2 and domain 3 of the gpl30 receptor. In one example the biologically active molecule comprises domain 1, domain 2 and domain 3 of the gp 130 receptor. It will also be appreciated that the term 'receptor' as used herein includes modified forms of naturally occurring receptors, including for example amino acid substitutions, additions or deletions.
  • a receptor comprising two chains may be produced as a single chain and linked to a single chain Fc polypeptide of the present invention.
  • the receptor may comprise all or part of the extracellular domains of the alpha and beta chains of the T cell receptor (TCR). Preferably these alpha and beta extracellular domains are linked in a single chain by a suitable linker which is in turn linked to a single chain Fc polypeptide of the present invention.
  • the monovalent binding protein is an antibody fragment.
  • suitable antibody fragments include but are not limited to, scFv, Fab, Fab', V HH , FV, VK, VH, V ⁇ , epitope-binding fragments of any of the above.
  • suitable antibody fragments include those described in Adair and Lawson, 2005. Therapeutic antibodies. Drug Design Reviews - Online 2(3):209-217, WO2005003169, WO2005003170 and WO2005003171.
  • An antibody fragment for use in the present invention can be derived from any class (e.g. IgG, IgE, IgM, IgD or IgA) or subclass of immunoglobulin molecule and may be obtained from any species including for example mouse, rat, shark, rabbit, pig, hamster, camel, llama, goat or human.
  • class e.g. IgG, IgE, IgM, IgD or IgA
  • subclass of immunoglobulin molecule e.g. IgG, IgE, IgM, IgD or IgA
  • subclass of immunoglobulin molecule e.g. IgG, IgE, IgM, IgD or IgA
  • subclass of immunoglobulin molecule e.g. IgG, IgE, IgM, IgD or IgA
  • subclass of immunoglobulin molecule e.g. IgG, IgE, IgM, IgD or I
  • the antibody fragment is a monoclonal, humanized and/or chimeric antibody fragment.
  • Humanized antibodies are antibody molecules having one or more complementarity determining regions (CDRs) from a non-human species and a framework region from a human immunoglobulin molecule which optionally comprises one or more donor residues from the non-human species (see, for example, US 5,585,089).
  • CDRs complementarity determining regions
  • Chimeric antibodies have been genetically engineered so that the light and heavy chain genes are composed of immunoglobulin gene segments belonging to different species.
  • the heavy and light chain constant regions are human and the variable regions are derived from another species.
  • Monoclonal antibodies may be prepared by any method known in the art such as the hybridoma technique (Kohler & Milstein, Nature, 1975, 256, 495-497), the trioma technique, the human B-cell hybridoma technique (Kozbor et al, Immunology Today, 1983, 4, 72) and the EBV-hybridoma technique (Cole et al, "Monoclonal Antibodies and Cancer Therapy", pp. 77-96, Alan R. Liss, Inc., 1985). Antibodies may also be obtained by any other suitable method such as those described in Babcook, J. et al, Proc. Natl. Acad. Sd. USA, 1996, 93 (15), 7843- 7848, WO 92/02551, WO2004/051268 and WO2004/106377.
  • An antibody fragment for use in the present invention may be obtained from any whole antibody, especially a whole monoclonal antibody, using any suitable enzymatic cleavage and/or digestion techniques, for example by treatment with pepsin.
  • antibody fragments may be prepared by the use of recombinant DNA techniques involving the manipulation and re-expression of DNA encoding antibody variable and/or constant regions. Standard molecular biology techniques may be used to modify, add or delete amino acids or domains as desired. Any alterations to the variable or constant regions are still encompassed by the terms 'variable' and 'constant' regions as used herein.
  • Antibody fragments for use in the present invention may possess a native or a modified hinge region comprising one or more cysteines.
  • the native hinge region is the hinge region normally associated with the C H I domain of the antibody molecule.
  • a modified hinge region is any hinge that differs in length and/or composition from the native hinge region.
  • Such hinges can include hinge regions from other species, such as human, mouse, rat, rabbit, shark, pig, hamster, camel, llama or goat hinge regions.
  • Other modified hinge regions may comprise a complete hinge region derived from an antibody of a different class or subclass from that of the CHI domain.
  • a C H I domain of class ⁇ l may be attached to a hinge region of class ⁇ 4.
  • the modified hinge region may comprise part of a natural hinge or a repeating unit in which each unit in the repeat is derived from a natural hinge region.
  • the natural hinge region may be altered by converting one or more cysteine or other residues into neutral residues, such as serine or alanine, or by converting suitably placed residues into cysteine residues. By such means the number of cysteine residues in the hinge region may be increased or decreased.
  • Other modified hinge regions may be entirely synthetic and may be designed to possess desired properties such as length, cysteine composition and flexibility.
  • modified hinge regions have already been described for example, in US5,677,425, WO9915549, WO9825971 and WO2005003171 and these are incorporated herein by reference.
  • the protein for use in the present invention is a Fab' fragment with a native or a modified hinge region.
  • one or more cysteines may be engineered into the antibody fragments of the present invention, for example to create surface exposed cysteine(s) (US 5,219,996).
  • surface exposed cysteine(s) US 5,219,996
  • the number of cysteines in an antibody fragment may be modified in order to provide a specific number of sites for example for effector molecule attachment.
  • the single chain-Fc polypeptide of the present invention further comprises an antibody fragment.
  • the antibody fragment is a single chain-Fv polypeptide.
  • the single chain-Fc polypeptide of the present invention further comprises a single-chain Fv polypeptide.
  • the C-terminus of the VH domain of the sc-Fv is genetically fused to the N-terminus of the first CH2 domain, optionally via one of the linkers described herein above.
  • the C-terminus of the VL domain of the sc-Fv is genetically fused to the N-terminus of the first CH2 domain, optionally via one of the linkers described herein above.
  • the biologically active molecule is a Fab or Fab'(See for example Figure 1).
  • the single chain-Fc polypeptide of the present invention further comprises an antibody Fab or Fab' fragment.
  • the C-terminus of the VH-CHl chain of the Fab or Fab' is genetically fused to the N-terminus of a single-chain Fc polypeptide of the present invention.
  • the VL-CL chain of the Fab or Fab' is linked to the VH-CHl chain by a disulphide bond, preferably the native interchain disulphide bond.
  • the C-terminus of the VL-CL chain of the Fab or Fab' is genetically fused to the N-terminus of a single-chain Fc polypeptide of the present invention.
  • the VH-CHl chain of the Fab or Fab' is linked to the VL-CL by a disulphide bond, preferably the native interchain disulphide bond.
  • the single chain Fc polypeptide of the present invention may have one or more effector molecules attached. Effector molecules may be attached by any suitable method, for example by chemical conjugation or genetic fusion.
  • effector molecule' as used herein includes, for example, antineoplastic agents, drugs, toxins (such as enzymatically active toxins of bacterial or plant origin and fragments thereof e.g. ricin and fragments thereof) biologically active proteins, for example enzymes, other antibody or antibody fragments, synthetic or naturally occurring polymers, nucleic acids and fragments thereof e.g. DNA, RNA and fragments thereof, radionuclides, particularly radioiodide, radioisotopes, chelated metals, nanoparticles and reporter groups such as fluorescent compounds or compounds which may be detected by NMR or ESR spectroscopy.
  • an effector molecule may comprise a single effector molecule or two or more such molecules so linked as to form a single moiety that can be attached to a protein using the process of the present invention.
  • Particular antineoplastic agents include cytotoxic and cytostatic agents for example alkylating agents, such as nitrogen mustards (e.g. chlorambucil, melphalan, mechlorethamine, cyclosphophamide, or uracil mustard) and derivatives thereof, triethylenephosphoramide , triethylenethiophosphor-amide, busulphan, or cisplatin; antimetabolites, such as methotrexate, fluorouracil, floxuridine, cytarabine, mercaptopurine, thioguanine, fluoroacetic acid, or fluorocitric acid, antibiotics, such as bleomycins (e.g.
  • bleomycin sulphate doxorubicin, daunorubicin, mitomycins (e.g. mitomycin C), actionmycins (e.g. dactinomycin) plicamyin, calicheamicin and derivatives thereof, or esperamicin and derivatives thereof; mitotic inhibitors, such as etoposide, vincristine or vinblastine and derivatives thereof; alkaloids such as ellipticine; polyols such as taxicin-I or taxicin-II; hormones, such as androgens (e.g. dromostanolone or testolactone), progestins (e.g.
  • megestrol acetate or medroxyprogesterone acetate examples include estrogens (e.g. dimethylstilbestrol diphosphate, polyestradiol phosphate or estramustine phosphate) or antiestrogens (e.g. tamoxifen); anthraquinones, such as mitoxantrone, ureas, such as hydroxyurea; hydrazines, such as procarbazine; or imidazoles, such as dacarbazine.
  • estrogens e.g. dimethylstilbestrol diphosphate, polyestradiol phosphate or estramustine phosphate
  • antiestrogens e.g. tamoxifen
  • anthraquinones such as mitoxantrone, ureas, such as hydroxyurea
  • hydrazines such as procarbazine
  • imidazoles such as dacarbazine.
  • Chelated metals include chelates of di- or tripositive metals having a coordination number from 2 to 8 inclusive.
  • Particular examples of such metals include technetium (Tc), rhenium (Re), cobalt (Co), copper (Cu), gold (Au), silver (Ag), lead (Pb), bismuth (Bi), indium (In), gallium (Ga), yttrium (Y), terbium (Tb), gadolinium (Gd), and scandium (Sc).
  • the metal is preferably a radionuclide.
  • radionuclides include 99m Tc, 186 Re, 188 Re, 58 Co, 60 Co, 67 Cu, 195 Au, 199 Au, ⁇ o Ag, 203 Pb, 206 Bi, 207 Bi, 111 In, 67 Ga, 68 Ga, 88 Y, 90 Y, 160 Tb, 153 Gd and 47 Sc.
  • the chelated metal may be for example one of the above types of metal chelated with any suitable polyadentate chelating agent, for example acyclic or cyclic polyamines, polyethers, (e.g. crown ethers and derivatives thereof); polyamides; porphyrins; and carbocyclic derivatives.
  • any suitable polyadentate chelating agent for example acyclic or cyclic polyamines, polyethers, (e.g. crown ethers and derivatives thereof); polyamides; porphyrins; and carbocyclic derivatives.
  • chelating agent in conjugates according to the invention, is acyclic and cyclic polyamines, especially polyaminocarboxylic acids, for example diethylenetriaminepentaacetic acid and derivatives thereof, and macrocyclic amines, e.g. cyclic tri-aza and tetra-aza derivatives (for example as described in International Patent Specification No. WO 92/22583); and polyamides, especially desferriox-amine and derivatives thereof.
  • polyaminocarboxylic acids for example diethylenetriaminepentaacetic acid and derivatives thereof
  • macrocyclic amines e.g. cyclic tri-aza and tetra-aza derivatives (for example as described in International Patent Specification No. WO 92/22583)
  • polyamides especially desferriox-amine and derivatives thereof.
  • effector molecules include other proteins, peptides and enzymes.
  • Enzymes of interest include, but are not limited to, proteolytic enzymes, hydrolases, lyases, isomerases, transferases.
  • Proteins, polypeptides and peptides of interest include, but are not limited to, immunoglobulins, albumin, toxins such as abrin, ricin A, pseudomonas exotoxin, or diphtheria toxin, a protein such as insulin, tumour necrosis factor, ⁇ -interferon, ⁇ -interferon, nerve growth factor, platelet derived growth factor or tissue plasminogen activator, a thrombotic agent or an anti- angiogenic agent, e.g.
  • angiostatin or endostatin or, a biological response modifier such as a lymphokine, interleukin-1 (IL-I), interleukin-2 (IL-2), interleukin-6 (IL-6), granulocyte macrophage colony stimulating factor (GM-CSF), granulocyte colony stimulating factor (G-CSF), nerve growth factor (NGF) or other growth factors.
  • IL-I interleukin-1
  • IL-2 interleukin-2
  • IL-6 interleukin-6
  • GM-CSF granulocyte macrophage colony stimulating factor
  • G-CSF granulocyte colony stimulating factor
  • NGF nerve growth factor
  • effector molecules may include detectable substances useful for example in diagnosis.
  • detectable substances include various enzymes, prosthetic groups, fluorescent materials, luminescent materials, bioluminescent materials, radioactive nuclides, positron emitting metals (for use in positron emission tomography), and nonradioactive paramagnetic metal ions. See generally U.S. Patent No. 4,741,900 for metal ions which can be conjugated to antibodies for use as diagnostics.
  • Suitable enzymes include horseradish peroxidase, alkaline phosphatase, beta-galactosidase, or acetylcholinesterase; suitable prosthetic groups include streptavidin, avidin and biotin; suitable fluorescent materials include umbelliferone, fluorescein, rhodamine red, rhodamine green, B-phycoerythrin, R-phycoerythrin, allophycosyanin, Texas red, Pacific blue, Marina blue, Oregon green and the Alexa Fluor series 350, 405, 430, 488, 500, 514, 532, 546, 555, 568, 594, 610, 633, 647, 660, 680, 700 and 750; suitable luminescent materials include luminol; suitable bioluminescent materials include luciferase, luciferin, and aequorin; and suitable radioactive nuclides include 125 I, 131 1, 111 In and 99 Tc.
  • Synthetic or naturally occurring polymers for use as effector molecules include, for example optionally substituted straight or branched chain polyalkylene, polyalkenylene, or polyoxyalkylene polymers or branched or unbranched polysaccharides, e.g. a homo- or hetero- polysaccharide such as lactose, amylose, dextran, starch or glycogen.
  • Particular optional substituents which may be present on the above-mentioned synthetic polymers include one or more hydroxy, methyl or methoxy groups.
  • synthetic polymers include optionally substituted straight or branched chain poly(ethyleneglycol), poly(propyleneglycol), poly(vinylalcohol) or derivatives thereof, especially optionally substituted poly(ethyleneglycol) such as methoxypoly(ethyleneglycol) or derivatives thereof.
  • Derivatives as used herein is intended to include reactive derivatives, for example thiol-selective reactive groups such as an ⁇ -halocaraboxylic acid or ester, e.g. iodoacetamide, an imide, e.g. maleimide, a vinyl sulphone or disulphide malemides and the like.
  • the reactive group may be linked directly or through a linker segment to the polymer. It will be appreciated that the residue of such a group will in some instances form part of the product as the linking group between the protein and the polymer.
  • one or more other domains or biologically active molecules maybe genetically fused or otherwise conjugated to the C-terminus of the single-chain polypeptide.
  • the single-chain Fc polypeptide further comprises a transmembrane domain fused to the C-terminus of the single chain Fc polypeptide.
  • the transmembrane domain allows the single-chain Fc polypeptides to be expressed on the surface of a cell. Accordingly, appropriate transmembrane domains may be used depending on the cell type of interest. A number of different transmembrane domains have been described, see for example WO97/23613, WO99/00494, WO99/57268, WO00/63374 and WO00/63373.
  • transmembrane domains include the natural transmembrane domains with which immunoglobulins are expressed on the surface of B cells, see for example the sequences given in SEQ ID NOs: 65, 68, 71, 74, 77, 80, 83 and 86.
  • the transmembrane domains are connected to the C-terminus of the single chain Fc polypeptide via a linker.
  • this is the natural linker with which immunoglobulins are expressed on the surface of B cells, see for example the sequences given in SEQ ID NOs: 64, 67, 70, 73, 76, 79, 82 and 85.
  • the present invention provides a single-chain Fc polypeptide which further comprises a transmembrane domain and one or more signaling domains.
  • the present invention provides a single-chain Fc polypeptide further comprising a transmembrane domain fused to the C-terminus, optionally via a linker, which is in turn fused at its C-terminus to one or more signaling domains.
  • Suitable signaling domains are well known in the art and appropriate signaling and transmembrane domains may be chosen in order to obtain the desired expression and/or signaling in the cell in which the single-chain Fc is expressed.
  • the intracellular domains are the natural intracellular domains with which immunoglobulins are expressed on the surface of B cells, see for example the sequences given in SEQ ID NOs: 66, 69, 72, 75, 78, 81, 84 and 87.
  • the single chain Fc polypeptide also comprises a biological molecule as described herein above, fused to its N-terminus
  • the single-chain Fc polypeptide can be used as a chimeric receptor protein.
  • Such single-chain Fc polypeptides have the advantageous property that they do not dimerise on the surface of the cell and accordingly avoid inappropriate signaling in the absence of bound ligand.
  • the present invention also provides an isolated DNA sequence encoding any one of the single chain Fc polypeptides of the present invention.
  • the DNA sequences of the present invention may comprise synthetic DNA, for instance produced by chemical processing, cDNA, genomic DNA or any combination thereof.
  • DNA sequences which encode a single chain Fc polypeptide of the present invention can be obtained by methods well known to those skilled in the art.
  • DNA sequences coding for part or all of the antibody Fc domains may be synthesised as desired from the determined DNA sequences or on the basis of the corresponding amino acid sequences.
  • DNA coding for antibody Fc constant domains is widely available to those skilled in the art and can be readily synthesised on the basis of their known amino acid sequences.
  • Standard techniques of molecular biology may be used to prepare DNA sequences coding for the single chain Fc polypeptide of the present invention. Desired DNA sequences may be synthesised completely or in part using oligonucleotide synthesis techniques. Site-directed mutagenesis and polymerase chain reaction (PCR) techniques may be used as appropriate.
  • PCR polymerase chain reaction
  • the present invention also relates to a cloning or expression vector comprising one or more DNA sequences of the present invention.
  • a cloning or expression vector comprising one or more DNA sequences encoding a single chain Fc polypeptide of the present invention.
  • the cloning or expression vector comprises a single DNA sequence, encoding the entire single chain Fc polypeptide and optionally all or part of the biologically active molecule e.g. a scFv or V HH -
  • the cloning or expression vector comprises two DNA sequences, for example the first DNA sequence encoding the single chain Fc polypeptide and one chain of biologically active molecule, eg. VH-CHl and the second DNA sequence encoding a second chain of the biologically active molecule domain e.g. VL-CL.
  • a vector according to the present invention comprises an appropriate leader sequence, such as an antibody leader sequence.
  • a host cell comprising one or more cloning or expression vectors comprising one or more DNA sequences encoding a single chain Fc polypeptide of the present invention.
  • Any suitable host cell/vector system may be used for expression of the DNA sequences encoding the single chain Fc polypeptide of the present invention.
  • Bacterial for example E. coli, and other microbial systems may be used or eukaryotic, for example mammalian, host cell expression systems may also be used.
  • Suitable mammalian host cells include NSO, CHO, myeloma or hybridoma cells.
  • the present invention also provides a process for the production of a single chain Fc polypeptide according to the present invention comprising culturing a host cell containing a vector of the present invention under conditions suitable for leading to expression of protein from DNA encoding the single chain Fc polypeptide of the present invention, and isolating the single chain Fc polypeptide.
  • the single chain Fc polypeptide may comprise only a single chain and where this is expressed alone or as a genetic fusion to the biologically active molecule only a single polypeptide coding sequence needs to be used to transfect the host cells for example, scFvscFc.
  • the cell line may be transfected with two or more vectors, a first vector encoding the single chain Fc polypeptide fused to a first chain of the biologically active molecule (e.g. VH-CHl) and a second vector encoding a second chain of the biologically active molecule (e.g. VL-CL).
  • a single vector may be used, the vector including sequences encoding both chains of the biologically active molecule where one of the chains is fused to the single chain Fc polypeptide.
  • the single-chain Fc polypeptide of the present invention may be purified where necessary using any suitable method known in the art including, for example chromatography techniques such as ion exchange, size exclusion, protein A or hydrophobic interaction chromatography.
  • the size of the single-chain Fc polypeptide may be confirmed by conventional methods known in the art such as size exclusion chromatography and non-reducing SDS-P AGE. Such techniques can be used to confirm that the scFc has not dimerised. If dimers are detected then the monomelic single-chain Fc polypeptides may be purified away from the dimeric species using conventional chromatography techniques as described above.
  • the functionality of the single-chain Fc polypeptides of the present invention may be determined using any suitable method known in the art depending on the effector functions required, including those methods provided in the Examples. Suitable assays include Fc receptor binding assays, complement fixing assays, co- stimulation assays, cell killing assays, cytotoxicity assays and cytostatis assays. In addition, half-life can be measured using suitable pharmacokinetic methods known in the art.
  • the biologically active molecule binds to a surface protein and targets the single-chain Fc polypeptide to this surface expressed protein
  • other functional assays such as cell killing assays (e.g. complement mediated cytotoxicity assays) may also be used. Accordingly, suitable functional assays may be readily established by one skilled in the art to determine whether the desired function is achieved.
  • the single chain Fc polypeptides of the present invention are useful in the treatment and/or prophylaxis of disease. Accordingly, the present invention also provides a pharmaceutical or diagnostic composition comprising a single chain Fc polypeptide of the present invention in combination with one or more of a pharmaceutically acceptable excipient, diluent or carrier. Accordingly, provided is the use of a single chain Fc polypeptide of the invention for the manufacture of a medicament.
  • the composition will usually be supplied as part of a sterile, pharmaceutical composition that will normally include a pharmaceutically acceptable carrier.
  • a pharmaceutical composition of the present invention may additionally comprise a pharmaceutically-acceptable adjuvant.
  • the present invention also provides a process for preparation of a pharmaceutical or diagnostic or research reagent composition comprising adding and mixing the single chain Fc polypeptide of the present invention together with one or more of a pharmaceutically acceptable excipient, diluent or carrier.
  • the single-chain Fc polypeptide may be the sole active ingredient in the pharmaceutical or diagnostic composition or may be accompanied by other active ingredients including for example other antibody or non-antibody ingredients including for example anti-inflammatories and chemotherapeutic agents.
  • the pharmaceutical compositions preferably comprise a therapeutically effective amount of the single-chain Fc polypeptide of the invention.
  • therapeutically effective amount refers to an amount of a therapeutic agent needed to treat, ameliorate or prevent a targeted disease or condition, or to exhibit a detectable therapeutic or preventative effect.
  • the therapeutically effective amount can be estimated initially either in cell culture assays or in animal models, usually in rodents, rabbits, dogs, pigs or primates. The animal model may also be used to determine the appropriate concentration range and route of administration. Such information can then be used to determine useful doses and routes for administration in humans.
  • the precise therapeutically effective amount for a human subject will depend upon the severity of the disease state, the general health of the subject, the age, weight and gender of the subject, diet, time and frequency of administration, drug combination(s), reaction sensitivities and tolerance/response to therapy. This amount can be determined by routine experimentation and is within the judgement of the clinician. Generally, a therapeutically effective amount will be from 0.01 mg/kg to 50 mg/kg, preferably 0.1 mg/kg to 20 mg/kg. Pharmaceutical compositions maybe conveniently presented in unit dose forms containing a predetermined amount of an active agent of the invention per dose.
  • compositions may be administered individually to a patient or may be administered in combination ⁇ e.g. simultaneously, sequentially or separately) with other agents, drugs or hormones.
  • the dose at which the single-chain Fc polypeptide of the present invention is administered depends on the nature of the condition to be treated, the extent of the inflammation present and on whether the antibody molecule is being used prophylactically or to treat an existing condition.
  • the frequency of dose will depend on the half-life of the single-chain Fc polypeptide and the duration of its effect. If the single-chain Fc polypeptide has a short half-life (e.g. 2 to 10 hours) it may be necessary to give one or more doses per day. Alternatively, if the single-chain Fc polypeptide has a long half life (e.g. 2 to 15 days) it may only be necessary to give a dosage once per day, once per week or even once every 1 or 2 months.
  • the pharmaceutically acceptable carrier should not itself induce the production of antibodies harmful to the individual receiving the composition and should not be toxic. Suitable carriers may be large, slowly metabolised macromolecules such as proteins, polypeptides, liposomes, polysaccharides, polylactic acids, polyglycolic acids, polymeric amino acids, amino acid copolymers and inactive virus particles.
  • salts can be used, for example mineral acid salts, such as hydrochlorides, hydrobromides, phosphates and sulphates, or salts of organic acids, such as acetates, propionates, malonates and benzoates.
  • mineral acid salts such as hydrochlorides, hydrobromides, phosphates and sulphates
  • organic acids such as acetates, propionates, malonates and benzoates.
  • Pharmaceutically acceptable carriers in therapeutic compositions may additionally contain liquids such as water, saline, glycerol and ethanol. Additionally, auxiliary substances, such as wetting or emulsifying agents or pH buffering substances, may be present in such compositions. Such carriers enable the pharmaceutical compositions to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries and suspensions, for ingestion by the patient.
  • Preferred forms for administration include forms suitable for parenteral administration, e.g. by injection or infusion, for example by bolus injection or continuous infusion.
  • the product may take the form of a suspension, solution or emulsion in an oily or aqueous vehicle and it may contain formulatory agents, such as suspending, preservative, stabilising and/or dispersing agents.
  • the single-chain Fc polypeptide may be in dry form, for reconstitution before use with an appropriate sterile liquid.
  • compositions of the invention can be administered directly to the subject.
  • the subjects to be treated can be animals. However, it is preferred that the compositions are adapted for administration to human subjects.
  • compositions of this invention maybe administered by any number of routes including, but not limited to, oral, pulmonary, intravenous, intramuscular, intra-arterial, intramedullary, intrathecal, intraventricular, transdermal, transcutaneous (for example, see WO 98/20734), subcutaneous, intraperitoneal, intranasal, enteral, topical, sublingual, intravaginal or rectal routes. Hyposprays and nebulisers may also be used to administer the pharmaceutical compositions of the invention.
  • the therapeutic compositions may be prepared as injectables, either as liquid solutions or suspensions. Solid forms suitable for solution in, or suspension in, liquid vehicles prior to injection may also be prepared.
  • Direct delivery of the compositions will generally be accomplished by injection, subcutaneously, intraperitoneally, intravenously or intramuscularly, or delivered to the interstitial space of a tissue.
  • the compositions can also be administered into a lesion. Dosage treatment may be a single dose schedule or a multiple dose schedule.
  • the active ingredient in the composition will be a single-chain Fc polypeptide. As such, it will be susceptible to degradation in the gastrointestinal tract. Thus, if the composition is to be administered by a route using the gastrointestinal tract, the composition will need to contain agents which protect the single-chain Fc polypeptide from degradation but which release the single-chain Fc polypeptide once it has been absorbed from the gastrointestinal tract.
  • the single-chain Fc polypeptide of the present invention will be administered by use of gene therapy.
  • DNA sequences encoding the single chain Fc polypeptide under the control of appropriate DNA components are introduced into a patient such that the single chain Fc polypeptide is expressed from the DNA sequence(s) and assembled in situ.
  • the single-chain Fc polypeptide may be transfected ex vivo into appropriate cells, such as T cells. Examples of suitable methods for ex vivo transfections are described in WO2004/039840.
  • the present invention also provides a single-chain Fc polypeptide for use in the treatment or prophylaxis of a pathological disorder that is selected from the group consisting of infections (viral, bacterial, fungal and parasitic), endotoxic shock associtated with infection, arthritis, rheumatoid arthritis, asthma, pelvic inflammatory disease, Alzheimer's Disease, Crohn's disease, Peyronie's Disease, coeliac disease, gallbladder disease, Pilonidal disease, peritonitis, psoriasis, vasculitis, surgical adhesions, stroke, Type I Diabetes, lyme arthritis, meningoencephalitis, immune mediated inflammatory disorders of the central and peripheral nervous system such as multiple sclerosis and Guillain-Barr syndrome, other autoimmune disorders, pancreatitis, trauma (surgery), graft-versus-host disease, transplant rejection, cancer (both solid tumours such as melanomas, hepatoblastomas, sarcomas, s
  • the present invention provides a single-chain Fc polypeptide for use in the control of inflammatory diseases and cancer.
  • the single-chain Fc polypeptide can be used to reduce the inflammatory process or cancer or to prevent the inflammatory process or cancer.
  • Figure 1 (a)-(c) A diagrammatic representation of three examples of single chain Fc polypeptide according to the present invention which comprise an antibody Fab fragment and in which a linker connects the C-terminus of the first CH3 domain and the N-terminus of the second CH2 domain.
  • Figure 2 (a)-(c) A diagrammatic representation of three examples of single chain Fc polypeptide which comprise an antibody Fab fragment and in which a linker connects the C-terminus of the first CH2 domain to the N-terminus of the second
  • CH2 domain and another linker connects the C-terminus of the first CH3 domain to the N-terminus of the second CH3 domain.
  • Figure 3 (a)-(c) A diagrammatic representation of three examples of single chain Fc polypeptide which comprise an antibody Fab fragment and in which:
  • a linker connects the C-terminus of the first CH4 domain to the N-terminus of the second CH2 domain.
  • a first linker connects the C-terminus of the first CH2 domain to the N-terminus of the second CH2 domain
  • a second linker connects the C-terminus of the second CH2 domain to the N-terminus of the first CH3 domain
  • a third linker connects the C-terminus of the first CH4 domain to the N-terminus of the second CH3 domain.
  • a first linker connects the C-terminus of the first CH3 domain to the N-terminus of the second CH2 domain, a second linker connects the C-terminus of the second CH3 domain to the N-terminus of the first CH4 domain and a third linker connects the C-tenninus of the first CH4 domain to the N-terminus of the second CH4 domain.
  • Figures 4a and 4b demonstrate that the single chain Fc polypeptides bind to antigen recombinantly expressed on the surface of NSO cells ( Figure 4a) and naturally expressed on the surface of activated T cells ( Figure 4b).
  • Figures 5 a and b show the ability of the single chain Fc polypeptides to induce cytotoxicity of NSO cells in the presence of complement (a) but not in the absence of complement (b).
  • Figures 5c and 5d show the ability of the single chain Fc polypeptides to induce complement-dependent cytotoxicity of activated T cells.
  • Figure 6 Example sequences of single chain Fc polypeptides derived from IgGl.
  • Hinge sequences are in italics and linkers are underlined.
  • Three murine single chain Fc polypeptides comprising a biologically active molecule at the N-terminus were designed in which the biologically active molecule was an antibody Fab fragment.
  • the variable regions of the Fab fragment were derived from murine antibody, Mox46, that binds to a cell surface protein antigen.
  • the Fc domains were derived from murine IgG2a and the three different versions of these domains are shown below.
  • the linker sequences are underlined. Hinge sequences are in italics and where these constitute part of the linker sequence they are in italics and underlined. Version 1 SEQ ID NO:88 (format as illustrated in Figure 2b).
  • the cysteines in the hinge between CHl and CH2 have been substituted for serines.
  • Linker comprises a truncated hinge 'PCP'.
  • each of the single chain Fc polypeptides was synthesized with the same biologically active molecule at the N-terminus i.e. the VHCHl domain from antibody MOX46.
  • the single chain Fc polypeptides comprising versions 1, 2 and 3 above were expressed in a pVAX vector (Invitrogen) in HEK293 cells (a human embryonic kidney epithelial cell line) using an antibody leader sequence (from mouse antibody B72.3 (Whittle et ah, 1987, Protein Eng. 1(6) 499-505)).
  • the VLCL chain of the MOX46 Fab fragment was produced in the same cell but in a separate vector.
  • the resulting single chain Fc polypeptides were purified using protein A.
  • the ability of the single-chain Fc polypeptides (versions 1 and 3) to bind to antigen was compared to the same antibody variable regions (from the MOX46 antibody) expressed in a murine IgGl framework and an irrelevant IgG.
  • Recombinant NSO cells expressing the antigen on their surface (5x10 5 ) were incubated with lOO ⁇ l of the single chain Fc polypeptides for 30 minutes at 4°C.
  • the control was MOPC21 which was titrated from 5 ⁇ g/ml down in 1/3 dilutions.
  • the MOX46 IgG and the single-chain Fc constructs were titrated in 1/3 dilutions.
  • the cells were washed twice in Dulbecco's PBS containing 5% FCS and 0.1% sodium azide and then lOO ⁇ l of anti-mouse heavy and light chain PE labelled (Jackson) antibody diluted 1/250 was added for 30 minutes at 4 0 C. The cells were washed once more as before and analysed by flow cytometry.
  • the ability of the single chain Fc polypeptides (versions 1, 2 and 3) to bind antigen was also tested using the methods described above however primary activated T cells which naturally express the antigen on their surface were used instead of recombinant NSO cells.
  • the primary activated T cells were produced as follows: IxIO 6 DOl 1 splenocytes were cultured with 200ng/ml ovalbumin peptide 323-329 for 3 days, washed and resuspended in twice the volume of medium for a further 2 days. The activated cells were then purified by negatively isolating the CD4 T cells.
  • CD64 and CD32 were immobilised (approx IOOORU each) by amine coupling chemistry on flow cells 2 and 3 (respectively) of a standard CM5 Biacore chip.
  • Flow cell 1 was set up as a reference flow cell to check for background binding.
  • the single-chain Fc proteins were then injected in sequence over the chip to look for binding activity. All samples were run undiluted and at 1 :2 and 1 :5 dilutions. Background binding was insignificant with all samples.
  • Single-chain Fc versions 1 and 3 were tested for their ability to cause complement- mediated cytotoxicity of cells to which they were bound.
  • a recombinant NSO line expressing the relevant antigen on its surface (5x10 6 ) cells/ml media were mixed with 50 ⁇ l/ml baby rabbit complement (Serotec C12CA). Prior to use, the complement was reconstituted with 2ml ice cold tissue culture grade distilled water. It was used within one hour of reconstitution and was maintained on ice until use. The agents tested were at a concentration of 2 ⁇ g/ml and plated onto a 96 well plate (Costar) in lOO ⁇ l volumes in duplicate. lOO ⁇ l of the cell/complement mix was added per well and the plate incubated at 37°C for 4 hours. Cytotoxicity was assessed by uptake of the vital stain, propidium iodide (PI) by FACS.
  • PI propidium iodide
  • a stock solution of 20mg/ml PI (Molecular Probes P- 1304MP) was prepared in distilled water and then diluted in RPMI 1640 to give a final concentration in the well of 3 ⁇ g/ml. The cells were incubated for 10 min at RT in the dark, before being analysed by flow cytometry.
  • Figure 5 a shows that the both versions 1 and 3 of the single-chain Fc polypeptides (1.1 and 3.1) induce complement-dependent cytotoxicity.
  • Example 5 Complement-dependent cytotoxicity assay using activated T cells
  • Another single chain Fc polypeptide (version 3) comprising an antibody Fab fragment derived from murine antibody, 495, that binds to a different cell surface protein antigen was produced as both an IgGl and IgG2a format using murine Fc regions.
  • the ability of these scFc proteins to cause complement-mediated cytotoxicity of activated T cells which express the antigen bound by the 495 antibody on their surface was tested.
  • the methods used were as described in Example 4 except primary activated T cells were used which were produced as described in Example 2.
  • Figure 5d clearly illustrates that as expected only the IgG2a format of the single chain Fc and the IgG2b format of the whole antibody 495 were able to induce complement-dependent cytotoxicity.
  • the IgGl formats were unable to induce complement-dependent cytotoxicity.
  • the human gpl30 receptor domain 1 was cloned as a single chain Fc (mouse gamma 1) fusion protein using the single chain Fc format illustrated in Figure Ia.
  • the sequence of the fusion protein is shown below.
  • GpI 30 domain 1 scFc fusion protein (SEQ ID NO:91)
  • VQFSWFVDDV EVHTAQTQPR EEQFNSTFRS VSELPIMHQD WLNGKEFKCR VNSAAFPAPI
  • the sequence in bold represents the gpl30 receptor domain 1 (amino acids 1 to 125 of SEQ ID NO:91).
  • the linker sequence is underlined. Hinge sequences are in italics and where they constitute part of the linker they are in italics and underlined.
  • the sequence 'SSA' between the C-terminus of the gpl30 domain 1 and the first hinge sequence are amino acids required to introduce the necessary Xhol restriction site for cloning purposes.
  • the constructs were transiently expressed in a mammalian cell system (CHO L761) using a pVAX vector and the B72.3 mouse signal sequence.
  • a western blot was prepared using the resulting scFc protein and the blot was probed with an anti-mouse Fc HRP (Jackson 115-035-071) and also with the biotinylated polyclonal anti gpl30 (R&D BAF228), revealed with a strep-HRP.
  • a protein corresponding to the predicted size of the gpl30 domain 1 fusion protein was detected on the western blot. Previous attempts to express the gp 130 domain 1 on its own or with a his tag attached had been unsuccessful. Fusion of the gpl30 domain 1 to the single chain Fc polypeptide enabled the gpl30 domain 1 to be expressed.

Abstract

The present invention relates to single chain polypeptides comprising one or more immunoglobulin Fc domains. In particular the present invention relates to single-chain Fc polypeptides in which at least one functional Fc domain is formed within the polypeptide chain.

Description

SINGLE CHAIN FC POLYPEPTIDES
The present invention relates to single chain polypeptides comprising one or more immunoglobulin Fc domains. In particular the present invention relates to single- chain polypeptides in which at least one functional Fc domain is formed within the chain.
Immunoglobulins are bivalent Y-shaped molecules comprising two identical heavy chains and two identical light chains. Disulfide bonds link together the heavy and light chain pairs as well as the two heavy chains. Each chain consists of one variable domain that varies in sequence and is responsible for antigen binding, these are known as the VH and VL domains for the heavy and light chains respectively. Each chain also consists of at least one constant domain. In the light chain there is a single constant domain (CL) and in the heavy chain there are at least three (CHI, CH2 and CH3), sometimes four (CH4) depending on the isotype. In humans there are five different classes or isotypes of antibodies including IgA (which includes IgAl and IgA2), IgD, IgE, IgG (which includes subclasses IgGl, IgG2, IgG3 and IgG4) and IgM.
The Fc domain of an antibody typically comprises at least the last two heavy chain constant region domains of each chain which dimerise to form the Fc domain. The Fc domain is responsible for providing antibody effector functions, including determining antibody half-life and distribution throughout the body, ability to fix complement and binding to cell surface Fc receptors. The properties of Fc domains have made them useful therapeutic agents and Fc domains have been fused to other non-antibody proteins, such as receptor proteins e.g etanercept. Fc domain fusions have also been used as research reagents, 'Fc tags', which facilitate fusion protein detection and purification. In addition, a number of alternative antibody structures comprising Fc domains have also been described see for example Dumont et al, 2006, Biodrugs, 20(3) 151-160, WO2005001025, WO2005077981, WO2005017148 and Hayden et al, 1994, Therapeutic Immunology, 1, 3-15. WO2005077981 describes antibodies in which each chain comprises two Fc domains i.e. each antibody chain comprises in linear sequence CH2 CH3 CH2 CH3 and these domains dimerise to form two functional Fc domains to provide enhanced effector functions. WO2005017148 and Hayden et al. supra describe single chain polypeptides comprising a single-chain Fv fused to half of an Fc domain i.e. sc-Fv-CH2 CH3. These polypeptides can exist as both monomers or dimers. The binding specificity of antibodies has made these useful therapeutic agents, however, bivalent molecules such as antibodies are often inappropriate targeting agents for certain cell surface antigens. Bivalent binding can cause the target cell to undergo co-stimulation, activation and/or antigenic modulation, thereby offering the cell a means of evading complement and the various effector cells recruited by the Fc domain of the antibody. Instead, in order to target such cell surface antigens antibodies have typically been conjugated to drugs or toxins that kill the cells upon internalization.
In contrast, univalent antibody fragments do not cause antigenic modulation as no redistribution of surface antigen occurs and hence no co-stimulation and no internalization. It would be desirable therefore to retain the natural effector functions of an antibody in such fragments and thus avoid the need for costly and time consuming attachment of drugs or toxins. An example of one such antibody fragment was produced by proteolytic cleavage of a rabbit IgG by Glennie and Stevenson, 1982, Nature, 295, 712-713. The fragment comprised only a single Fab binding site but retained the entire Fc domain. The fragment was produced by papain digestion of a rabbit IgG antibody Al 2 allotypic variant which is glycosylated on one chain, making that chain resistant to papain digestion, thus allowing one Fab arm to be retained. The fragment produced was demonstrated to be more effective in invoking complement mediated lysis of cells than the whole IgG. Similar fragments have been produced from human IgG by proteolytic digestion (Michaelsen and Natvig, Scand. J. Immunology, 1973, 2, 299-312; Michaelsen and Natvig, Scand. J. Immunology, 1972, 1, 255-268) and by chemical cleavage (Wines and Easterbrook- Smith, Molecular Immunology, 1991, 28, 8, 855-863). These fragments are not practical to produce on a commercial scale as the use of proteolysis requires long preparation times and results in low yields and mixed products.
WO20050010125 describes hybrid proteins comprising two polypeptide chains, the first polypeptide chain comprising an Fc region and a biologically active molecule and the second polypeptide chain comprising an Fc region without the biologically active molecule of the first chain. The two chains are produced separately and are either allowed to dimerise or are chemically conjugated together. Although this achieves the desired functional molecule, the preparation is complex and involves low yielding chromatographic procedures. Surprisingly we have now found that it possible to produce a functional Fc domain as a single-chain polypeptide. The polypeptides of the present invention therefore have the advantage that they can be produced recombinantly in large amounts and can be linked by any suitable means to any other molecule, such as a binding domain. Further, as the antibody constant domains form an Fc domain within the chain, the polypeptides of the present invention are not prone to dimerisation thus, where desired, bi-valent binding domains can be avoided.
Accordingly the present invention provides a single chain polypeptide comprising two CH2 domains and two CH3 domains characterized in that said CH2 and CH3 domains form a functional Fc domain within the chain. The functional Fc domain in the single-chain polypeptides of the present invention is not formed by dimerisation of two chains i.e. the two CH2 domains and two CH3 domains are present in a single chain and form a functional Fc domain within the single chain. Accordingly the present invention provides a single chain polypeptide comprising two CH2 domains and two CH3 domains characterized in that said CH2 and CH3 domains form a functional Fc domain within the chain and not by dimerisation with another polypeptide chain. Accordingly, in the single chain polypeptide of the present invention a first CH2 domain is dimerised with a second CH2 domain and a first CH3 domain is dimerised with a second CH3 domain within the polypeptide chain.
The term 'functional' as used herein refers to the ability of the Fc domain formed within the single chain polypeptide to provide one or more effector functions usually associated with Fc domains although it will be appreciated that other functions maybe engineered into such domains. Examples of effector functions include determining the half-life and/or distribution of the Fc polypeptide throughout the body, the ability of the Fc polypeptide to fix complement and the ability of the Fc polypeptide to bind to cell surface Fc receptors. Examples of such effector functions include but are not limited to, antibody-dependent cellular cytotoxicity (ADCC), antibody-dependent cellular phagocytosis (ADCP) and complement-dependent cytotoxicity (CDC).
The Fc domain of the present invention comprises four or more constant domains which may be derived from any suitable species and/or class of antibody. Preferably the constant domains are human. In humans there are five different classes or isotypes of antibodies including IgA (which includes IgAl and IgA2), IgD, IgE, IgG (which includes subclasses IgGl, IgG2, IgG3 and IgG4) and IgM. Any suitable Fc domain may be used depending on the effector functions required. Typically the term Fc domain as used herein refers to the last two constant region immunoglobulin domains of IgA, IgD and IgG (CH2 and CH3) and the last three constant region domains of IgE and IgM (CH2, CH3 and CH4), although it will be appreciated that in certain circumstances not all the domains may be required, for example in the case of IgE or IgM only CH2 and CH3 domains may be sufficient. It will also be appreciated that more than one Fc domain may be formed within the single chain Fc polypeptide and that these Fc domains may be derived from the same or different isotypes.
The residues in antibody domains are conventionally numbered according to a system devised by Kabat et al. This system is set forth in Kabat et al, 1987, in Sequences of Proteins of Immunological Interest, US Department of Health and Human Services, NIH, USA (hereafter "Kabat et al. {supra)"). This numbering system is used in the present specification except where otherwise indicated.
In one embodiment the Fc domain is derived from IgA and the single-chain Fc polypeptide comprises two Cα2 domains and two Cα3 domains.
In one embodiment the Fc domain is derived from IgM and the single-chain Fc polypeptide comprises two Cμ2 domains, two Cμ3 domains and two Cμ4 domains.
In one embodiment the Fc domain is derived from IgD and the single-chain Fc polypeptide comprises two Cδ2 domains and two Cδ3 domains.
In one embodiment the Fc domain is derived from IgE and the single-chain Fc polypeptide comprises two Cε2 domains, two Cε3 domains and two Cε4 domains.
Preferably the Fc domain of the present invention is derived from an IgG and the single-chain Fc polypeptide comprises two Cγ2 and two Cγ3 domains. The preferred sequences for the Cγ2 domain of IgGl, IgG2, IgG3 and IgG4 for use in the present invention are provided in SEQ ID NOS: 2, 15, 28 and 41 respectively and the preferred sequences for the Cγ3 domain of IgGl, IgG2, IgG3 and IgG4 for use in the invention are provided in SEQ ID NOS: 3, 16, 29 and 42 respectively.
Accordingly, in one embodiment the present invention provides a single chain polypeptide comprising two Cγ2 domains and two Cγ3 domains characterized in that said Cγ2 and Cγ3 domains form a functional Fc domain within the chain i.e. a first Cγ2 domain dimerises with a second Cγ2 domain and a first Cγ3 domain dimerises with a second Cγ3 domain within the polypeptide chain
It will be appreciated that the constant region domains for use in producing the Fc domain of the present invention may include variants of the naturally occurring constant domains described herein above. Such variants may comprise one or more amino acid variations compared to wild type constant domains. In one example the Fc domain of the present invention comprises at least one constant domain which varies in sequence from the wild type constant domain. It will be appreciated that the variant constant domains may be longer or shorter than the wild type constant domain. Preferably the variant constant domains are at least 50% identical or similar to a wild type constant domain. The term "Identity", as used herein, indicates that at any particular position in the aligned sequences, the amino acid residue is identical between the sequences. The term "Similarity", as used herein, indicates that, at any particular position in the aligned sequences, the amino acid residue is of a similar type between the sequences. For example, leucine may be substituted for isoleucine or valine. Other amino acids which can often be substituted for one another include but are not limited to:
- phenylalanine, tyrosine and tryptophan (amino acids having aromatic side chains);
- lysine, arginine and histidine (amino acids having basic side chains);
- aspartate and glutamate (amino acids having acidic side chains);
- asparagine and glutamine (amino acids having amide side chains); and
- cysteine and methionine (amino acids having sulphur-containing side chains). Degrees of identity and similarity can be readily calculated (Computational Molecular Biology, Lesk, A.M., ed., Oxford University Press, New York, 1988; Biocomputing. Informatics and Genome Projects, Smith, D.W., ed., Academic Press, New York, 1993; Computer Analysis of Sequence Data, Part 1, Griffin, A.M., and Griffin, H. G., eds., Humana Press, New Jersey, 1994; Sequence Analysis in Molecular Biology, von Heinje, G., Academic Press, 1987; and Sequence Analysis Primer, Gribskov, M. and Devereux, J., eds., M Stockton Press, New York, 1991). In one example the variant constant domains are at least 60% identical or similar to a wild type constant domain. In another example the variant constant domains are at least 70% identical or similar. In another example the variant constant domains are at least 80% identical or similar. In another example the variant constant domains are at least 90% identical or similar. In another example the variant constant domains are at least 95% identical or similar.
In one embodiment the variant constant domains provide equivalent Fc effector functions compared to the wild type Fc domain. In one embodiment the variant constant domains provide improved effector functions. In one embodiment the variant constant domains provide altered effector functions. In one example the Fc domain provides no effector functions other than extended half-life. In one example the Fc domain provides FcR binding but not CIq binding. In one example the Fc domain provides CIq binding but not FcR binding.
A number of Fc variant polypeptides are known in the art, see for example Idusogie et ah, Journal of Immunology, 2000, 164, 4178-4184 and Shields et ah, Journal of Biological Chemistry, 2001, 276, 9, 6591-6604. A comprehensive list of Fc variants is provided by WO2005077981 (see in particular, paragraph number 80) and these are incorporated herein by reference.
Examples of Fc variants include in IgGl N314Q (or N297Q), T318A (T299A), A349S (A330S) with P350A (P331A), L247A (L234A) with L248A (L235A) or P348A (P329A) (number in brackets is EU numbering). Where the IgG4 Fc domain is used the S241P (S228P) mutation may be used (Angal et al., Molecular Immunology, 1993, 30 (1), 105-108). It will be appreciated that any suitable variant may be produced and tested using routine methods known in the art.
The CH2, CH3 and where present CH4 domains of the single-chain Fc polypeptide of the present invention are linked in the single polypeptide chain such that they can still form a functional Fc domain within the chain. Accordingly any suitable amino acid linkers may be used to link these constant domains provided they allow a functional Fc domain to form within the single-chain polypeptide. Suitable amino acids for use in linkers of the present invention include, but are not limited to, small flexible amino acids such as GIy, Ser, Ala and Thr. In one embodiment the linker comprises or consists of glycine residues. In one embodiment the linker comprises or consists of serine residues. In one embodiment the linker comprises or consists of alanine residues. In one embodiment the linker comprises or consists of threonine residues. In one embodiment the linker comprises or consists of glycine and serine residues. In one embodiment the linker comprises or consists of glycine, serine and alanine residues. In one embodiment the linker comprises or consists of glycine, serine, alanine and threonine residues. For the avoidance of doubt, it is understood that all permutations of glyine and/or serine and/or alanine and/or threonine residues are included. In one example the linker comprises or consists of 30-80% glycine residues and 20-70% serine residues. In one example the linker comprises or consists of 35-50% glycine residues; 30-40% serine residues; 5-15% threonine residues and 10-20% alanine residues. In one example the amino acid residues are randomly distributed within the linker. Specific examples of suitable linkers include glycine-serine polymers comprising for example repeats of sequences such as GS, GSGGS, GGGGS and GGGS.
In one embodiment the Fc domain of the single chain polypeptide of the present invention comprises two CH2 domains and two CH3 domains.
In one embodiment the present invention provides a single chain Fc polypeptide comprising two CH2 domains and two CH3 domains wherein in N- to C-terminal sequence, a first CH2 domain is linked at its C-terminus to the N- terminus of a first CH3 domain and said first CH3 domain is linked at its C-terminus via a linker to the N-terminus of a second CH2 domain which is linked at its C- terminus to the N-terminus a second CH3 domain (as shown in Figure 1).
In one embodiment the first CH2 domain is directly linked i.e. genetically fused at its C-terminus to the N-terminus of the first CH3 domain.
In one embodiment the second CH2 domain is directly linked i.e. genetically fused at its C-terminus to the N-terminus of the second CH3 domain.
Examples of suitable CH2 domains genetically fused to CH3 domain(s) are given in SEQ ID NOS: 5, 18, 31 and 44.
In one embodiment a linker is employed to link the C-terminus of the first CH2 domain to the N-terminus of the first CH3 domain.
In one embodiment a linker is employed to link the C-terminus of the second CH2 domain to the N-terminus of the second CH3 domain.
Where a linker is used to link (i) the C-terminus of the first CH2 domain to the N-terminus of the first CH3 domain and/or (ii) the C-terminus of the second CH2 domain to the N-terminus of the second CH3 domain the linker will be of sufficient length to allow a functional Fc domain to form within the chain. Typically the linker will only be a few amino acids in length, preferably less than 10 amino acids in length. Where a linker is used in both (i) and (ii) above the two linkers may be the same or different. Preferably the linkers will be approximately the same length.
The linker used to link the C-terminus of the first CH3 domain to the N- terminus of the second CH2 domain will be sufficiently long to allow a functional Fc domain to form within the chain i.e. it will be sufficiently long to allow a first CH2 domain to dimerise with a second CH2 domain and a first CH3 domain to dimerise with a second CH3 domain within the polypeptide chain. In one embodiment the linker is around 30-100 amino acids in length, in another embodiment the linker is around 40 to 100 amino acids in length. In one embodiment the linker is around 40 to 90 amino acids in length. In one embodiment the linker is around 40 to 80 amino acids in length, preferably 40 to 70 amino acids in length. Suitable amino acids for use in these linkers are described herein above. In one example the linker comprises or consists of 35-50% glycine residues; 30-40% serine residues; 5-15% threonine residues and 10-20% alanine residues. In one example the amino acid residues are randomly distributed within the linker. An example of a suitable linker is provided in SEQ ID NO:62. In one embodiment the linker may comprise, preferably towards its C-terminus one or more cysteine residues. In one embodiment the linker may comprise at its C-terminus the sequence of all or part of the hinge region of an antibody or variant thereof comprising one or more cysteine residues. Examples of suitable hinge sequences for use in the linkers of the present invention are provided in US5,677,425, WO9915549, WO9825971 and WO2005003171 and these are incorporated herein by reference. Other examples of suitable hinges are provided in SEQ ID NOs:53-57. Accordingly, in one example the linker of SEQ ID NO:62 further comprises at its C-terminus any one of the sequences provided in SEQ ID NOs: 53-57. In this embodiment, the linker is around 30 to 130 amino acids in length. In one embodiment the linker is around 50 to 100 amino acids in length. In one embodiment the linker is around 50 to 80 amino acids in length.
In another embodiment, the present invention provides a single chain Fc polypeptide comprising two CH2 domains and two CH3 domains wherein in N- to C-terminal sequence a first CH2 domain is linked at its C-terminus via a linker to the N-terminus of a second CH2 domain and said second CH2 domain is linked at its C- terminus to the N-terminus of a first CH3 domain and said first CH3 domain is linked at its C-terminus to the N-terminus of a second CH3 domain via a linker (as shown in Figure 2).
In one embodiment the second CH2 domain is genetically fused at its C- terminus to the N-terminus of the first CH3 domain.
In one embodiment the C-terminus of the second CH2 domain is connected by a linker to the N-terminus of the first CH3 domain.
Where a linker is used to link the C-terminus of the second CH2 domain to the N-terminus of the first CH3 domain the linker will be of sufficient length to allow a functional Fc domain to form within the chain. Typically the linker will only be a few amino acids in length, preferably less than 10 amino acids in length.
The linker used to link the C-terminus of the first CH2 domain to the N- terminus of the second CH2 domain and the linker used to link the C-terminus of the first CH3 domain to the N-terminus of the second CH3 domain will be sufficiently long to allow a functional Fc domain to form within the chain. It will be appreciated that these two linkers may be the same or different in both composition and/or length. In one embodiment one or both of the linkers is between 15 and 50 amino acids in length. In one embodiment one or both of the linkers is between 15 and 40 amino acids in length. In one embodiment one or both of the linkers is between 20 and 40 amino acids in length. In another embodiment one or both of the linkers is between 20 and 35 amino acids in length. Suitable amino acids for use in these linkers are described herein above. In one example one or both of the linkers comprises or consists of 50-80% glycine residues and 10-30% serine residues. In one example the amino acid residues are randomly distributed within the linker. In one example the linker comprises the sequence (GGGGS)n where n=3 to 8. In one embodiment the linker between the C-terminus of the first CH2 domain and the N- terminus of the second CH2 domain comprises the sequence (GGGGS)n where n=5 (SEQ ID NO:63). In one embodiment the linker between the C-terminus of the first CH3 domain and the N-terminus of the second CH3 domain comprises the sequence (GGGGS)n where n=5 (SEQ ID NO:63).
In one embodiment the linker between the first CH2 domain and the second CH2 domain comprises, preferably towards its C-terminus, one or more cysteine residues. In one embodiment the linker comprises all or part of an antibody hinge sequence or variant thereof as described herein above. Accordingly, in one embodiment the linker between the C-terminus of the first CH2 domain and the N- terminus of the second CH2 domain having the sequence given in SEQ ID NO: 63 further comprises at its C-terminus any one of the hinge sequences provided in SEQ ID NOs 53-57. In this embodiment the linker between the C-terminus of the first CH2 domain and the N-terminus of the second CH2 domain is between 25 and 90 amino acids in length. In one embodiment this linker is between 25 and 80 amino acids in length. In one embodiment this linker is between 25 and 70 amino acids in length. In one embodiment this linker is between 25 and 60 amino acids in length. In one embodiment this linker is between 25 and 50 amino acids in length. In one embodiment this linker is between 25 and 40 amino acids in length.
In one embodiment the Fc domain of the single chain Fc polypeptide of the present invention comprises two CH2 domains, two CH3 domains and one or two CH4 domains, preferably two.
In one embodiment the present invention provides a single chain Fc polypeptide comprising two CH2 domains, two CH3 domains and two CH4 domains wherein in N- to C-terminal sequence, a first CH2 domain is linked at its C-terminus to the N-terminus of a first CH3 domain, said first CH3 domain is linked at its C- terminus to the N-terminus of a first CH4 domain and said first CH4 domain is linked at its C-terminus via a linker to the N-terminus of a second CH2 domain which is linked at its C-terminus to the N-terminus of a second CH3 domain which is linked at its C-terminus to the N-terminus of a second CH4 domain (see for example Figure 3 a).
In one embodiment one or more of the following domains are directly linked i.e. genetically fused (i) the C-terminus of the first CH2 domain to the N-terminus of the first CH3 domain (ii) the C-terminus of the second CH2 domain to the N- terminus of the second CH3 domain (iii) the C-terminus of the first CH3 domain to the N-terminus of the first CH4 domain (iv) the C-terminus of the second CH3 domain to the N-terminus of the second CH4 domain.
In one embodiment one or more of the following domains are connected by a linker (v) the C-terminus of the first CH2 domain to the N-terminus of the first CH3 domain (vi) the C-terminus of the second CH2 domain to the N-terminus of the second CH3 domain (vii) the C-terminus of the first CH3 domain to the N-terminus of the first CH4 domain (viii) the C-terminus of the second CH3 domain to the N- terminus of the second CH4 domain. Where a linker is present between any one of (v, vi, vii or viii) this linker will be of sufficient length to allow a functional Fc domain to form within the chain. Typically the linker will only be a few amino acids in length. Where there is more than one linker it will be appreciated that these may be the same or different. Preferably the linkers will be approximately the same length.
The linker used to link the C-terminus of the first CH4 domain to the N- terminus of the second CH2 domain will be sufficiently long to allow a functional Fc domain to form within the chain. In one embodiment the linker is around 50-100 amino acids in length, in another embodiment the linker is around 60 to 100 amino acids in length. In one embodiment the linker is around 70 to 100 amino acids in length, preferably 80 to 100 amino acids in length. Suitable amino acids for use in these linkers have been described herein above. In one embodiment the linker may comprise, preferably towards its C-terminus, one or more cysteine residues. In one embodiment the linker comprises all or part of an antibody hinge sequence or variant thereof as described herein above.
In another embodiment, the present invention provides a single chain Fc polypeptide comprising two CH2 domains, two CH3 domains and two CH4 domains wherein in N- to C-terminal sequence a first CH2 domain is linked at its C-terminus via a linker to the N-terminus of a second CH2 domain and said second CH2 domain is linked at its C-terminus to the N-terminus of a first CH3 domain and said first CH3 domain is linked at its C-terminus to the N-terminus of a first CH4 domain which is linked at its C-terminus, via a linker, to the N-terminus of a second CH3 domain which is linked at its C-terminus to the N-terminus of a second CH4 domain (see for example Figure 3b).
In one embodiment one or more of the following domains are directly linked i.e. genetically fused (i) the C-terminus of the second CH2 domain to the N-terminus of the first CH3 domain (ii) the C-terminus of the first CH3 domain to the N- terminus of the first CH4 domain (iii) the C-terminus of the second CH3 domain to the N-terminus of the second CH4 domain.
In one embodiment one or more of the following domains are connected by a linker (i) the C-terminus of the second CH2 domain to the N-terminus of the first CH3 domain (ii) the C-terminus of the first CH3 domain to the N-terminus of the first CH4 domain (iii) the C-terminus of the second CH3 domain to the N-terminus of the second CH4 domain. Where a linker is present between one or more of (i) the C-terminus of the second CH2 domain and the N-terminus of the first CH3 domain (ii) the C-terminus of the first CH3 domain and the N-terminus of the first CH4 domain (iii) the C- terminus of the second CH3 domain and the N-terminus of the second CH4 domain the linker will be of sufficient length to allow a functional Fc domain to form within the chain. Typically the linker will only be a few amino acids in length. Where there is more than one linker it will be appreciated that these may be the same or different. Preferably the linkers will be approximately the same length.
The linker between the C-terminus of the first CH2 domain and the N- terminus of the second CH2 domain and the linker between the C-terminus of the first CH4 domain and the N-terminus of the second CH3 domain will be sufficiently long to allow a functional Fc domain to form within the chain. Suitable amino acids for these linkers have been described herein above.
The linker between the C-terminus of the first CH2 domain and the N- terminus of the second CH2 domain is typically between 15 and 40 amino acids in length. In another embodiment the linker is between 20 and 35 amino acids in length. In one embodiment the linker between the C-terminus of the first CH2 domain and the N-terminus of the second CH2 domain comprises the sequence (GGGGS)n where n-5 (SEQ ID NO:63).
In one embodiment the linker between the first CH2 domain and the second CH2 domain comprises, preferably towards its C-terminus, one or more cysteine residues. In one embodiment the linker comprises all or part of an antibody hinge sequence or variant thereof as described herein above which may comprise one or more cysteine residues. Suitable hinge sequences include those provided in SEQ ID NOs 53-57.
The linker between the C-terminus of the first CH4 domain and the N- terrninus of the second CH3 domain is typically around 30-100 amino acids in length, in another embodiment the linker is around 40 to 100 amino acids in length. In one embodiment the linker is around 40 to 80 amino acids in length, preferably 40 to 70 amino acids in length. An example of a suitable linker is provided in SEQ ID NO:62.
In another embodiment, the present invention provides a single chain Fc polypeptide comprising two CH2 domains, two CH3 domains and two CH4 domains wherein in N- to C-terminal sequence a first CH2 domain is linked at its C-terminus to the N-terminus of a first CH3 domain and said first CH3 domain is linked at its C- terminus via a linker to the N-terminus of a second CH2 domain and said second CH2 domain is linked at its C-terminus to the N-terminus of a second CH3 domain which is linked at its C-terminus to the N-terminus of a first CH4 domain which is linked at its C-terminus, via a linker, to the N-terminus of a second CH4 domain (see for example Figure Ic).
In one embodiment one or more of the following domains are directly linked
1 i.e. genetically fused (i) the C-terminus of the first CH2 domain and the N-terminus of the first CH3 domain (ii) the C-terminus of the second CH2 domain and the N- terminus of the second CH3 domain (iii) the C-terminus of the second CH3 domain and the N-terminus of the first CH4 domain.
In one embodiment one or more of the following domains are connected by a linker (i) the C-terminus of the first CH2 domain and the N-terminus of the first CH3 domain (ii) the C-terminus of the second CH2 domain and the N-terminus of the second CH3 domain (iii) the C-terminus of the second CH3 domain and the N- terminus of the first CH4 domain.
Where a linker is present between one or more of (i) the C-terminus of the first CH2 domain and the N-terminus of the first CH3 domain (ii) the C-terminus of the second CH2 domain and the N-terminus of the second CH3 domain (iii) the C- terminus of the second CH3 domain and the N-terminus of the first CH4 domain the linker will be of sufficient length to allow a functional Fc domain to form within the chain. Typically the linker will only be a few amino acids in length. Where there is more than one linker it will be appreciated that these may be the same or different. Preferably the linkers will be approximately the same length.
The linker between the C-terminus of the first CH3 domain and the N- terminus of the second CH2 domain and the linker between the C-terminus of the first CH4 domain and the N-terminus of the second CH4 domain will be of sufficient length to allow a functional Fc domain to form within the chain.
The linker between the C-terminus of the first CH4 domain and the N- terminus of the second CH4 domain is typically between 15 and 40 amino acids in length. In another embodiment the linker is between 20 and 35 amino acids in length. In one embodiment the linker between the C-terminus of the first CH4 domain and the N-terminus of the second CH4 domain comprises the sequence (GGGGS)n where n=5 (SEQ ID NO:63). The linker between the C-terminus of the first CH3 domain and the N- terminus of the second CH2 domain is typically around 30-100 amino acids in length, in another embodiment the linker is around 40 to 100 amino acids in length. In one embodiment the linker is around 40 to 80 amino acids in length, preferably 40 to 70 amino acids in length. An example of a suitable linker is provided in SEQ ID NO:62.
In one embodiment the linker between the first CH3 domain and the second CH2 domain comprises, preferably towards its C-terminus, one or more cysteine residues. In one embodiment this linker comprises all or part of an antibody hinge sequence or variant thereof as described herein above which may comprise one or more cysteine residues. Suitable hinge sequences include SEQ ID NOs 53-57.
In one embodiment the single chain Fc polypeptide of the present invention further comprises an amino acid linker genetically fused to the N-terminus of the first CH2 domain. The linker may comprise any suitable amino acids and be of any suitable length. In one embodiment the linker comprises one or more cysteine residues. In one embodiment the linker genetically fused to the N-terminus of the first CH2 domain comprises all or part of an antibody hinge sequence or variant thereof as described herein above. In one embodiment the linker comprises the sequence given in any one of SEQ ID NOs:53-57. In one embodiment one or more of the cysteine residues present in the linker are disulphide linked to one or more cysteine residues present in any one of the following linkers where present (i) the linker which connects the C-terminus of the first CH3 domain and the N-terminus of the second CH2 domain (see for example Figure 1 a) (ii) the linker which connects the C-terminus of the first CH2 domain to the N-terminus of the second CH2 domain (see for example Figure 2a) or (iii) the linker which connects the C-terminus of the first CH4 domain to the N-terminus of the second CH2 domain.
In another embodiment the linker fused to the N-terminus may comprise all or part of the hinge region of an antibody or variant thereof in which one or more cysteines have been substituted for another amino acid, preferably serine. Examples of suitable linkers of this type are provided in SEQ ID NOs: 58-61.
Examples of single chain Fc polypeptides according to the present invention are provided for IgGl, 2, 3 and 4 in SEQ ID NOS: 8-13, 21-26, 34-39 and 47-52 respectively. See also Figure 6 for examples of IgGl sequences. The invention also extends to variants of these sequences as set out herein above. In one example the present invention provides a single chain Fc polypeptide comprising a sequence having at least 70% identity or similarity to any one of the sequences given in SEQ ID NOS: 8-13, 21-26, 34-39 and 47-52. In another example a single chain Fc polypeptide of the present invention comprises a sequence having at least 80% identity or similarity to any one of the sequences given in SEQ ID NOS: 8-13, 21-26, 34-39 and 47-52. In another example a single chain Fc polypeptide of the present invention comprises a sequence having at least 90% identity or similarity to any one of the sequences given in SEQ ID NOS: 8-13, 21-26, 34-39 and 47-52. In another example a single chain Fc polypeptide of the present invention comprises a sequence having at least 95% or 98% identity or similarity to any one of the sequences given in SEQ ID NOS: 8-13, 21-26, 34-39 and 47-52.
In one embodiment the single chain Fc polypeptide of the present invention further comprises a CHl domain fused optionally via a hinge to the N-terminus of the first CH2 domain. Examples of suitable CHl domains are provided in SEQ ID NOs I, 14, 27 and 40.
The single chain Fc polypeptides of the present invention can be used in a number of applications, including therapeutic, diagnostic and research applications. Preferably the single chain Fc polypeptides of the present invention further comprise one or more other molecules which may be fused or otherwise linked at the N and/or C-terminus and/or elsewhere on the polypeptide. Such molecules include, but are not limited to, nucleic acids, small molecules, carbohydrates, proteins and peptides, including for example receptor proteins, antibodies and antibody fragments. The single-chain Fc polypeptide may be linked to another molecule, optionally via a linker (amino acid or chemical), by any suitable means known in the art, including for example, chemical conjugation, chemical cross-linking or genetic fusion. In one embodiment the single chain Fc polypeptide comprises a cysteine containing linker, such as an antibody hinge, at its N-terminus and one of these free cysteines is used as a site of attachment for another molecule, preferably a biologically active molecule as described below.
In one embodiment the single-chain Fc polypeptides of the present invention are used as an Fc tag, for example to aid protein purification and/or protein detection. Accordingly in one embodiment the single-chain Fc polypeptide further comprises at its N-terminus all or part of another protein. Such Fc fusions advantageously do not dimerise unlike currently available Fc fusions thus ensuring that the fusion protein remains monomelic. In certain applications, where it is desirable to be able to remove the Fc domain, for example after purification, the single-chain Fc polypeptide may be linked to another protein via a cleavable linker.
In one embodiment the single-chain Fc polypeptide of the present invention is linked at its N and/or C-terminus to a biologically active molecule. The biologically active molecule may be any protein or other suitable molecule, including nucleic acids, small molecules, carbohydrates, receptor proteins or immunoglobulins. Some examples of biologically active molecules include enzymes, antibody fragments, domain antibodies, single chain antibodies, aptamers, Microbodies™, binding agents based on protein scaffolds (see for example Nygren and Uhlen, 1997, Current Opinion in Structural Biology, 7, 463-469) versabodies, avimers, adnectins, anticalins, phylomers, aptamers, cyclic peptides, peptides, antiviral agents, hemostatic agents and cytokines and growth factors such as EPO, RANTES, interleukins such as IL-I, IL-2, IL-3, IL-4, IL-5, IL-6, IL-8, IL-12, IL-13, IL-16 or IL-17, interferons such as interferon α, interferon β or interferon γ, tumour necrosis factor-α, tumor necrosis factor-β, colony stimulating factors such as G-CSF or GM- CSF.
In one embodiment the biologically active molecule brings the single-chain Fc polypeptide of the present invention into contact with a desired target, for example a target protein. In one embodiment the biologically active molecule binds to a desired target protein. In one example the target protein is a cell-associated protein, for example a cell surface protein on cells such as bacterial cells, yeast cells, T-cells, endothelial cells or tumour cells, or it may be a soluble protein. Target proteins may also be any medically relevant protein such as those proteins upregulated during disease or infection, for example receptors and/or their corresponding ligands. Particular examples of cell surface proteins include adhesion molecules, for example integrins such as βl integrins e.g. VLA-4, E-selectin, P selectin or L-selectin, CD2, CD3, CD4, CD5, CD7, CD8, CDl Ia, CDl Ib, CD18, CD19, CD20, CD23, CD25, CD33, CD38, CD40, CD45, CDW52, CD69, CD134 (OX40), ICOS, BCMP7, CD137, CD27L, CD28, CD40L, CTLA-4, CD22, CDCPl, DPCRl, DPCRl, dudulin2, FLJ20584, FLJ40787, HEK2, KIAA0634, KIAA0659, KIAA1246, KIAA1455, LTBP2, LTK, MAL2, MRP2, nectin-like2, NKCCl, PTK7, RAIGl, TCAMl, SC6, BCMPlOl, BCMP84, BCMPI l, DTD, carcinoembryonic antigen (CEA), human milk fat globulin (HMFGl and 2), MHC Class I and MHC Class II antigens, and VEGF, and where appropriate, receptors thereof.
In one embodiment the target protein is a soluble protein. Soluble proteins include interleukins such as IL-I, IL-2, IL-3, IL-4, IL-5, IL-6, IL-8, IL-12, IL-13, IL- 16 or IL- 17, viral proteins for example respiratory syncytial virus or cytomegalovirus proteins, immunoglobulins, such as IgE, interferons such as interferon α, interferon β or interferon γ, tumour necrosis factor-α, tumor necrosis factor-β, colony stimulating factors such as G-CSF or GM-CSF, and platelet derived growth factors such as PDGF-α, and PDGF-β and where appropriate receptors thereof.
In one embodiment the single-chain Fc polypeptides of the present invention may be used to functionally alter the activity of a particular protein to which the biologically active molecule binds. For example the single-chain Fc polypeptide may neutralize, antagonize or agonise the activity of a protein. In one embodiment the binding of the single-chain Fc polypeptide to a cell via the biologically active molecule results in cell killing e.g via complement mediated cytotoxicity.
In one embodiment the biologically active molecule is a monovalent binding domain, in particular, a monovalent protein such as a receptor or fragment thereof or an immunoglobulin or fragment thereof.
In one embodiment the biologically active molecule is a receptor that may be naturally expressed on a cell surface or inside the cell. Examples of suitable receptors include, but are not limited to, viral receptors, cytokine receptors, growth factor receptors, hormone receptors and bacterial receptors. It will be appreciated that the term 'receptor' as used herein also includes suitable fragments of such receptors, an example of which includes the extracellular domain of a receptor. In one example the receptor is the human gpl30 receptor or a cytokine binding fragment thereof, such as domain 1, 2 and/or 3. In one example the biologically active molecule comprises domain 1 of the gpl30 receptor or a fragment thereof. In one example the biologically active molecule comprises amino acids 1 to 125 of SEQ ID NO:91. In one example the biologically active molecule comprises domain 2 and domain 3 of the gpl30 receptor. In one example the biologically active molecule comprises domain 1, domain 2 and domain 3 of the gp 130 receptor. It will also be appreciated that the term 'receptor' as used herein includes modified forms of naturally occurring receptors, including for example amino acid substitutions, additions or deletions. In one example a receptor comprising two chains may be produced as a single chain and linked to a single chain Fc polypeptide of the present invention. In one example the receptor may comprise all or part of the extracellular domains of the alpha and beta chains of the T cell receptor (TCR). Preferably these alpha and beta extracellular domains are linked in a single chain by a suitable linker which is in turn linked to a single chain Fc polypeptide of the present invention.
Preferably the monovalent binding protein is an antibody fragment. Examples of suitable antibody fragments include but are not limited to, scFv, Fab, Fab', VHH, FV, VK, VH, Vλ, epitope-binding fragments of any of the above. Examples of suitable antibody fragments include those described in Adair and Lawson, 2005. Therapeutic antibodies. Drug Design Reviews - Online 2(3):209-217, WO2005003169, WO2005003170 and WO2005003171.
An antibody fragment for use in the present invention can be derived from any class (e.g. IgG, IgE, IgM, IgD or IgA) or subclass of immunoglobulin molecule and may be obtained from any species including for example mouse, rat, shark, rabbit, pig, hamster, camel, llama, goat or human.
In one embodiment the antibody fragment is a monoclonal, humanized and/or chimeric antibody fragment.
Humanized antibodies are antibody molecules having one or more complementarity determining regions (CDRs) from a non-human species and a framework region from a human immunoglobulin molecule which optionally comprises one or more donor residues from the non-human species (see, for example, US 5,585,089).
Chimeric antibodies have been genetically engineered so that the light and heavy chain genes are composed of immunoglobulin gene segments belonging to different species. Preferably the heavy and light chain constant regions are human and the variable regions are derived from another species.
Monoclonal antibodies may be prepared by any method known in the art such as the hybridoma technique (Kohler & Milstein, Nature, 1975, 256, 495-497), the trioma technique, the human B-cell hybridoma technique (Kozbor et al, Immunology Today, 1983, 4, 72) and the EBV-hybridoma technique (Cole et al, "Monoclonal Antibodies and Cancer Therapy", pp. 77-96, Alan R. Liss, Inc., 1985). Antibodies may also be obtained by any other suitable method such as those described in Babcook, J. et al, Proc. Natl. Acad. Sd. USA, 1996, 93 (15), 7843- 7848, WO 92/02551, WO2004/051268 and WO2004/106377.
An antibody fragment for use in the present invention may be obtained from any whole antibody, especially a whole monoclonal antibody, using any suitable enzymatic cleavage and/or digestion techniques, for example by treatment with pepsin. Alternatively antibody fragments may be prepared by the use of recombinant DNA techniques involving the manipulation and re-expression of DNA encoding antibody variable and/or constant regions. Standard molecular biology techniques may be used to modify, add or delete amino acids or domains as desired. Any alterations to the variable or constant regions are still encompassed by the terms 'variable' and 'constant' regions as used herein.
The methods for creating and manufacturing antibodies and antibody fragments are well known in the art (see for example, Boss et al., US 4,816,397; Cabilly et al., US 6,331,415; Shrader et al., WO 92/02551; Ward et al., 1989, Nature, 341, 544; Orlandi et al., 1989, Proc.Natl.Acad.Sci. USA, 86, 3833; Riechmann et al., 1988, Nature, 322, 323; Bird et al, 1988, Science, 242, 423; Queen et al., US 5,585,089; Adair, WO91/09967; Mountain and Adair, 1992, Biotechnol. Genet. Eng. Rev, 10, 1-142; Verma et al., 1998, Journal of Immunological Methods, 216, 165- 181).
Antibody fragments for use in the present invention may possess a native or a modified hinge region comprising one or more cysteines. The native hinge region is the hinge region normally associated with the CHI domain of the antibody molecule. A modified hinge region is any hinge that differs in length and/or composition from the native hinge region. Such hinges can include hinge regions from other species, such as human, mouse, rat, rabbit, shark, pig, hamster, camel, llama or goat hinge regions. Other modified hinge regions may comprise a complete hinge region derived from an antibody of a different class or subclass from that of the CHI domain. Thus, for instance, a CHI domain of class γl may be attached to a hinge region of class γ4. Alternatively, the modified hinge region may comprise part of a natural hinge or a repeating unit in which each unit in the repeat is derived from a natural hinge region. In a further alternative, the natural hinge region may be altered by converting one or more cysteine or other residues into neutral residues, such as serine or alanine, or by converting suitably placed residues into cysteine residues. By such means the number of cysteine residues in the hinge region may be increased or decreased. Other modified hinge regions may be entirely synthetic and may be designed to possess desired properties such as length, cysteine composition and flexibility.
A number of modified hinge regions have already been described for example, in US5,677,425, WO9915549, WO9825971 and WO2005003171 and these are incorporated herein by reference. In one example the protein for use in the present invention is a Fab' fragment with a native or a modified hinge region.
In one example, one or more cysteines may be engineered into the antibody fragments of the present invention, for example to create surface exposed cysteine(s) (US 5,219,996). Thus by using suitable engineering techniques the number of cysteines in an antibody fragment may be modified in order to provide a specific number of sites for example for effector molecule attachment.
In one embodiment the single chain-Fc polypeptide of the present invention further comprises an antibody fragment.
In one embodiment the antibody fragment is a single chain-Fv polypeptide. In one embodiment the single chain-Fc polypeptide of the present invention further comprises a single-chain Fv polypeptide. In one embodiment the C-terminus of the VH domain of the sc-Fv is genetically fused to the N-terminus of the first CH2 domain, optionally via one of the linkers described herein above. In one embodiment the C-terminus of the VL domain of the sc-Fv is genetically fused to the N-terminus of the first CH2 domain, optionally via one of the linkers described herein above.
In one embodiment the biologically active molecule is a Fab or Fab'(See for example Figure 1). In one embodiment the single chain-Fc polypeptide of the present invention further comprises an antibody Fab or Fab' fragment. In one embodiment the C-terminus of the VH-CHl chain of the Fab or Fab' is genetically fused to the N-terminus of a single-chain Fc polypeptide of the present invention. In this embodiment the VL-CL chain of the Fab or Fab' is linked to the VH-CHl chain by a disulphide bond, preferably the native interchain disulphide bond. In one embodiment the C-terminus of the VL-CL chain of the Fab or Fab' is genetically fused to the N-terminus of a single-chain Fc polypeptide of the present invention. In this embodiment the VH-CHl chain of the Fab or Fab' is linked to the VL-CL by a disulphide bond, preferably the native interchain disulphide bond. The single chain Fc polypeptide of the present invention may have one or more effector molecules attached. Effector molecules may be attached by any suitable method, for example by chemical conjugation or genetic fusion.
The term 'effector molecule' as used herein includes, for example, antineoplastic agents, drugs, toxins (such as enzymatically active toxins of bacterial or plant origin and fragments thereof e.g. ricin and fragments thereof) biologically active proteins, for example enzymes, other antibody or antibody fragments, synthetic or naturally occurring polymers, nucleic acids and fragments thereof e.g. DNA, RNA and fragments thereof, radionuclides, particularly radioiodide, radioisotopes, chelated metals, nanoparticles and reporter groups such as fluorescent compounds or compounds which may be detected by NMR or ESR spectroscopy. It will be appreciated that an effector molecule may comprise a single effector molecule or two or more such molecules so linked as to form a single moiety that can be attached to a protein using the process of the present invention.
Particular antineoplastic agents include cytotoxic and cytostatic agents for example alkylating agents, such as nitrogen mustards (e.g. chlorambucil, melphalan, mechlorethamine, cyclosphophamide, or uracil mustard) and derivatives thereof, triethylenephosphoramide , triethylenethiophosphor-amide, busulphan, or cisplatin; antimetabolites, such as methotrexate, fluorouracil, floxuridine, cytarabine, mercaptopurine, thioguanine, fluoroacetic acid, or fluorocitric acid, antibiotics, such as bleomycins (e.g. bleomycin sulphate), doxorubicin, daunorubicin, mitomycins (e.g. mitomycin C), actionmycins (e.g. dactinomycin) plicamyin, calicheamicin and derivatives thereof, or esperamicin and derivatives thereof; mitotic inhibitors, such as etoposide, vincristine or vinblastine and derivatives thereof; alkaloids such as ellipticine; polyols such as taxicin-I or taxicin-II; hormones, such as androgens (e.g. dromostanolone or testolactone), progestins (e.g. megestrol acetate or medroxyprogesterone acetate), estrogens (e.g. dimethylstilbestrol diphosphate, polyestradiol phosphate or estramustine phosphate) or antiestrogens (e.g. tamoxifen); anthraquinones, such as mitoxantrone, ureas, such as hydroxyurea; hydrazines, such as procarbazine; or imidazoles, such as dacarbazine.
Chelated metals include chelates of di- or tripositive metals having a coordination number from 2 to 8 inclusive. Particular examples of such metals include technetium (Tc), rhenium (Re), cobalt (Co), copper (Cu), gold (Au), silver (Ag), lead (Pb), bismuth (Bi), indium (In), gallium (Ga), yttrium (Y), terbium (Tb), gadolinium (Gd), and scandium (Sc). In general the metal is preferably a radionuclide. Particular radionuclides include 99mTc, 186Re, 188Re, 58Co, 60Co, 67Cu, 195Au, 199Au, πo Ag, 203Pb, 206Bi, 207Bi, 111In, 67Ga, 68Ga, 88Y, 90Y, 160Tb, 153Gd and 47Sc.
The chelated metal may be for example one of the above types of metal chelated with any suitable polyadentate chelating agent, for example acyclic or cyclic polyamines, polyethers, (e.g. crown ethers and derivatives thereof); polyamides; porphyrins; and carbocyclic derivatives.
In general, the type of chelating agent will depend on the metal in use. One particularly useful group of chelating agents in conjugates according to the invention, however, are acyclic and cyclic polyamines, especially polyaminocarboxylic acids, for example diethylenetriaminepentaacetic acid and derivatives thereof, and macrocyclic amines, e.g. cyclic tri-aza and tetra-aza derivatives (for example as described in International Patent Specification No. WO 92/22583); and polyamides, especially desferriox-amine and derivatives thereof.
Other effector molecules include other proteins, peptides and enzymes. Enzymes of interest include, but are not limited to, proteolytic enzymes, hydrolases, lyases, isomerases, transferases. Proteins, polypeptides and peptides of interest include, but are not limited to, immunoglobulins, albumin, toxins such as abrin, ricin A, pseudomonas exotoxin, or diphtheria toxin, a protein such as insulin, tumour necrosis factor, α-interferon, β-interferon, nerve growth factor, platelet derived growth factor or tissue plasminogen activator, a thrombotic agent or an anti- angiogenic agent, e.g. angiostatin or endostatin, or, a biological response modifier such as a lymphokine, interleukin-1 (IL-I), interleukin-2 (IL-2), interleukin-6 (IL-6), granulocyte macrophage colony stimulating factor (GM-CSF), granulocyte colony stimulating factor (G-CSF), nerve growth factor (NGF) or other growth factors.
Other effector molecules may include detectable substances useful for example in diagnosis. Examples of detectable substances include various enzymes, prosthetic groups, fluorescent materials, luminescent materials, bioluminescent materials, radioactive nuclides, positron emitting metals (for use in positron emission tomography), and nonradioactive paramagnetic metal ions. See generally U.S. Patent No. 4,741,900 for metal ions which can be conjugated to antibodies for use as diagnostics. Suitable enzymes include horseradish peroxidase, alkaline phosphatase, beta-galactosidase, or acetylcholinesterase; suitable prosthetic groups include streptavidin, avidin and biotin; suitable fluorescent materials include umbelliferone, fluorescein, rhodamine red, rhodamine green, B-phycoerythrin, R-phycoerythrin, allophycosyanin, Texas red, Pacific blue, Marina blue, Oregon green and the Alexa Fluor series 350, 405, 430, 488, 500, 514, 532, 546, 555, 568, 594, 610, 633, 647, 660, 680, 700 and 750; suitable luminescent materials include luminol; suitable bioluminescent materials include luciferase, luciferin, and aequorin; and suitable radioactive nuclides include 125I, 1311, 111In and 99Tc.
Synthetic or naturally occurring polymers for use as effector molecules include, for example optionally substituted straight or branched chain polyalkylene, polyalkenylene, or polyoxyalkylene polymers or branched or unbranched polysaccharides, e.g. a homo- or hetero- polysaccharide such as lactose, amylose, dextran, starch or glycogen. Particular optional substituents which may be present on the above-mentioned synthetic polymers include one or more hydroxy, methyl or methoxy groups. Particular examples of synthetic polymers include optionally substituted straight or branched chain poly(ethyleneglycol), poly(propyleneglycol), poly(vinylalcohol) or derivatives thereof, especially optionally substituted poly(ethyleneglycol) such as methoxypoly(ethyleneglycol) or derivatives thereof.
"Derivatives" as used herein is intended to include reactive derivatives, for example thiol-selective reactive groups such as an α-halocaraboxylic acid or ester, e.g. iodoacetamide, an imide, e.g. maleimide, a vinyl sulphone or disulphide malemides and the like. The reactive group may be linked directly or through a linker segment to the polymer. It will be appreciated that the residue of such a group will in some instances form part of the product as the linking group between the protein and the polymer.
It will be appreciated that one or more other domains or biologically active molecules maybe genetically fused or otherwise conjugated to the C-terminus of the single-chain polypeptide.
In one embodiment the single-chain Fc polypeptide further comprises a transmembrane domain fused to the C-terminus of the single chain Fc polypeptide. The transmembrane domain allows the single-chain Fc polypeptides to be expressed on the surface of a cell. Accordingly, appropriate transmembrane domains may be used depending on the cell type of interest. A number of different transmembrane domains have been described, see for example WO97/23613, WO99/00494, WO99/57268, WO00/63374 and WO00/63373. Other examples of suitable transmembrane domains include the natural transmembrane domains with which immunoglobulins are expressed on the surface of B cells, see for example the sequences given in SEQ ID NOs: 65, 68, 71, 74, 77, 80, 83 and 86. In one embodiment the transmembrane domains are connected to the C-terminus of the single chain Fc polypeptide via a linker. In one embodiment this is the natural linker with which immunoglobulins are expressed on the surface of B cells, see for example the sequences given in SEQ ID NOs: 64, 67, 70, 73, 76, 79, 82 and 85.
In one embodiment the present invention provides a single-chain Fc polypeptide which further comprises a transmembrane domain and one or more signaling domains. In one embodiment the present invention provides a single-chain Fc polypeptide further comprising a transmembrane domain fused to the C-terminus, optionally via a linker, which is in turn fused at its C-terminus to one or more signaling domains. Suitable signaling domains are well known in the art and appropriate signaling and transmembrane domains may be chosen in order to obtain the desired expression and/or signaling in the cell in which the single-chain Fc is expressed.
In one example the intracellular domains are the natural intracellular domains with which immunoglobulins are expressed on the surface of B cells, see for example the sequences given in SEQ ID NOs: 66, 69, 72, 75, 78, 81, 84 and 87.
Examples of suitable signaling domains have also been described in WO97/23613, WO99/00494, WO99/57268, WO00/63372, WO00/63374, WOOO/63373, WO01/32709, WO01/32866, WO01/32867, WO02/33101 and WO2004/039840.
In one embodiment where the single chain Fc polypeptide also comprises a biological molecule as described herein above, fused to its N-terminus, the single- chain Fc polypeptide can be used as a chimeric receptor protein. Such single-chain Fc polypeptides have the advantageous property that they do not dimerise on the surface of the cell and accordingly avoid inappropriate signaling in the absence of bound ligand.
The present invention also provides an isolated DNA sequence encoding any one of the single chain Fc polypeptides of the present invention. The DNA sequences of the present invention may comprise synthetic DNA, for instance produced by chemical processing, cDNA, genomic DNA or any combination thereof. DNA sequences which encode a single chain Fc polypeptide of the present invention can be obtained by methods well known to those skilled in the art. For example, DNA sequences coding for part or all of the antibody Fc domains may be synthesised as desired from the determined DNA sequences or on the basis of the corresponding amino acid sequences. DNA coding for antibody Fc constant domains is widely available to those skilled in the art and can be readily synthesised on the basis of their known amino acid sequences.
Standard techniques of molecular biology may be used to prepare DNA sequences coding for the single chain Fc polypeptide of the present invention. Desired DNA sequences may be synthesised completely or in part using oligonucleotide synthesis techniques. Site-directed mutagenesis and polymerase chain reaction (PCR) techniques may be used as appropriate.
The present invention also relates to a cloning or expression vector comprising one or more DNA sequences of the present invention. Accordingly, provided is a cloning or expression vector comprising one or more DNA sequences encoding a single chain Fc polypeptide of the present invention. In one embodiment the cloning or expression vector comprises a single DNA sequence, encoding the entire single chain Fc polypeptide and optionally all or part of the biologically active molecule e.g. a scFv or VHH- In another embodiment the cloning or expression vector comprises two DNA sequences, for example the first DNA sequence encoding the single chain Fc polypeptide and one chain of biologically active molecule, eg. VH-CHl and the second DNA sequence encoding a second chain of the biologically active molecule domain e.g. VL-CL. Preferably, a vector according to the present invention comprises an appropriate leader sequence, such as an antibody leader sequence.
General methods by which the vectors may be constructed, transfection methods and culture methods are well known to those skilled in the art. In this respect, reference is made to "Current Protocols in Molecular Biology", 1999, F. M. Ausubel (ed), Wiley Interscience, New York and the Maniatis Manual produced by Cold Spring Harbor Publishing.
Also provided is a host cell comprising one or more cloning or expression vectors comprising one or more DNA sequences encoding a single chain Fc polypeptide of the present invention. Any suitable host cell/vector system may be used for expression of the DNA sequences encoding the single chain Fc polypeptide of the present invention. Bacterial, for example E. coli, and other microbial systems may be used or eukaryotic, for example mammalian, host cell expression systems may also be used. Suitable mammalian host cells include NSO, CHO, myeloma or hybridoma cells.
The present invention also provides a process for the production of a single chain Fc polypeptide according to the present invention comprising culturing a host cell containing a vector of the present invention under conditions suitable for leading to expression of protein from DNA encoding the single chain Fc polypeptide of the present invention, and isolating the single chain Fc polypeptide.
The single chain Fc polypeptide may comprise only a single chain and where this is expressed alone or as a genetic fusion to the biologically active molecule only a single polypeptide coding sequence needs to be used to transfect the host cells for example, scFvscFc. For production of single-chain Fc polypeptides comprising a biologically active molecule which comprises two or more chains the cell line may be transfected with two or more vectors, a first vector encoding the single chain Fc polypeptide fused to a first chain of the biologically active molecule (e.g. VH-CHl) and a second vector encoding a second chain of the biologically active molecule (e.g. VL-CL). Alternatively, a single vector may be used, the vector including sequences encoding both chains of the biologically active molecule where one of the chains is fused to the single chain Fc polypeptide.
Once produced the single-chain Fc polypeptide of the present invention may be purified where necessary using any suitable method known in the art including, for example chromatography techniques such as ion exchange, size exclusion, protein A or hydrophobic interaction chromatography.
The size of the single-chain Fc polypeptide may be confirmed by conventional methods known in the art such as size exclusion chromatography and non-reducing SDS-P AGE. Such techniques can be used to confirm that the scFc has not dimerised. If dimers are detected then the monomelic single-chain Fc polypeptides may be purified away from the dimeric species using conventional chromatography techniques as described above. The functionality of the single-chain Fc polypeptides of the present invention may be determined using any suitable method known in the art depending on the effector functions required, including those methods provided in the Examples. Suitable assays include Fc receptor binding assays, complement fixing assays, co- stimulation assays, cell killing assays, cytotoxicity assays and cytostatis assays. In addition, half-life can be measured using suitable pharmacokinetic methods known in the art.
Further, where the biologically active molecule binds to a surface protein and targets the single-chain Fc polypeptide to this surface expressed protein, other functional assays, such as cell killing assays (e.g. complement mediated cytotoxicity assays) may also be used. Accordingly, suitable functional assays may be readily established by one skilled in the art to determine whether the desired function is achieved.
The single chain Fc polypeptides of the present invention are useful in the treatment and/or prophylaxis of disease. Accordingly, the present invention also provides a pharmaceutical or diagnostic composition comprising a single chain Fc polypeptide of the present invention in combination with one or more of a pharmaceutically acceptable excipient, diluent or carrier. Accordingly, provided is the use of a single chain Fc polypeptide of the invention for the manufacture of a medicament. The composition will usually be supplied as part of a sterile, pharmaceutical composition that will normally include a pharmaceutically acceptable carrier. A pharmaceutical composition of the present invention may additionally comprise a pharmaceutically-acceptable adjuvant.
The present invention also provides a process for preparation of a pharmaceutical or diagnostic or research reagent composition comprising adding and mixing the single chain Fc polypeptide of the present invention together with one or more of a pharmaceutically acceptable excipient, diluent or carrier.
The single-chain Fc polypeptide may be the sole active ingredient in the pharmaceutical or diagnostic composition or may be accompanied by other active ingredients including for example other antibody or non-antibody ingredients including for example anti-inflammatories and chemotherapeutic agents. The pharmaceutical compositions preferably comprise a therapeutically effective amount of the single-chain Fc polypeptide of the invention. The term "therapeutically effective amount" as used herein refers to an amount of a therapeutic agent needed to treat, ameliorate or prevent a targeted disease or condition, or to exhibit a detectable therapeutic or preventative effect. For any single-chain Fc polypeptide, the therapeutically effective amount can be estimated initially either in cell culture assays or in animal models, usually in rodents, rabbits, dogs, pigs or primates. The animal model may also be used to determine the appropriate concentration range and route of administration. Such information can then be used to determine useful doses and routes for administration in humans.
The precise therapeutically effective amount for a human subject will depend upon the severity of the disease state, the general health of the subject, the age, weight and gender of the subject, diet, time and frequency of administration, drug combination(s), reaction sensitivities and tolerance/response to therapy. This amount can be determined by routine experimentation and is within the judgement of the clinician. Generally, a therapeutically effective amount will be from 0.01 mg/kg to 50 mg/kg, preferably 0.1 mg/kg to 20 mg/kg. Pharmaceutical compositions maybe conveniently presented in unit dose forms containing a predetermined amount of an active agent of the invention per dose.
Compositions may be administered individually to a patient or may be administered in combination {e.g. simultaneously, sequentially or separately) with other agents, drugs or hormones.
The dose at which the single-chain Fc polypeptide of the present invention is administered depends on the nature of the condition to be treated, the extent of the inflammation present and on whether the antibody molecule is being used prophylactically or to treat an existing condition.
The frequency of dose will depend on the half-life of the single-chain Fc polypeptide and the duration of its effect. If the single-chain Fc polypeptide has a short half-life (e.g. 2 to 10 hours) it may be necessary to give one or more doses per day. Alternatively, if the single-chain Fc polypeptide has a long half life (e.g. 2 to 15 days) it may only be necessary to give a dosage once per day, once per week or even once every 1 or 2 months. The pharmaceutically acceptable carrier should not itself induce the production of antibodies harmful to the individual receiving the composition and should not be toxic. Suitable carriers may be large, slowly metabolised macromolecules such as proteins, polypeptides, liposomes, polysaccharides, polylactic acids, polyglycolic acids, polymeric amino acids, amino acid copolymers and inactive virus particles.
Pharmaceutically acceptable salts can be used, for example mineral acid salts, such as hydrochlorides, hydrobromides, phosphates and sulphates, or salts of organic acids, such as acetates, propionates, malonates and benzoates.
Pharmaceutically acceptable carriers in therapeutic compositions may additionally contain liquids such as water, saline, glycerol and ethanol. Additionally, auxiliary substances, such as wetting or emulsifying agents or pH buffering substances, may be present in such compositions. Such carriers enable the pharmaceutical compositions to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries and suspensions, for ingestion by the patient.
Preferred forms for administration include forms suitable for parenteral administration, e.g. by injection or infusion, for example by bolus injection or continuous infusion. Where the product is for injection or infusion, it may take the form of a suspension, solution or emulsion in an oily or aqueous vehicle and it may contain formulatory agents, such as suspending, preservative, stabilising and/or dispersing agents. Alternatively, the single-chain Fc polypeptide may be in dry form, for reconstitution before use with an appropriate sterile liquid.
Once formulated, the compositions of the invention can be administered directly to the subject. The subjects to be treated can be animals. However, it is preferred that the compositions are adapted for administration to human subjects.
The pharmaceutical compositions of this invention maybe administered by any number of routes including, but not limited to, oral, pulmonary, intravenous, intramuscular, intra-arterial, intramedullary, intrathecal, intraventricular, transdermal, transcutaneous (for example, see WO 98/20734), subcutaneous, intraperitoneal, intranasal, enteral, topical, sublingual, intravaginal or rectal routes. Hyposprays and nebulisers may also be used to administer the pharmaceutical compositions of the invention. Typically, the therapeutic compositions may be prepared as injectables, either as liquid solutions or suspensions. Solid forms suitable for solution in, or suspension in, liquid vehicles prior to injection may also be prepared.
Direct delivery of the compositions will generally be accomplished by injection, subcutaneously, intraperitoneally, intravenously or intramuscularly, or delivered to the interstitial space of a tissue. The compositions can also be administered into a lesion. Dosage treatment may be a single dose schedule or a multiple dose schedule.
It will be appreciated that the active ingredient in the composition will be a single-chain Fc polypeptide. As such, it will be susceptible to degradation in the gastrointestinal tract. Thus, if the composition is to be administered by a route using the gastrointestinal tract, the composition will need to contain agents which protect the single-chain Fc polypeptide from degradation but which release the single-chain Fc polypeptide once it has been absorbed from the gastrointestinal tract.
A thorough discussion of pharmaceutically acceptable carriers is available in Remington's Pharmaceutical Sciences (Mack Publishing Company, NJ. 1991).
It is also .envisaged that the single-chain Fc polypeptide of the present invention will be administered by use of gene therapy. In order to achieve this, DNA sequences encoding the single chain Fc polypeptide under the control of appropriate DNA components are introduced into a patient such that the single chain Fc polypeptide is expressed from the DNA sequence(s) and assembled in situ. Alternatively the single-chain Fc polypeptide may be transfected ex vivo into appropriate cells, such as T cells. Examples of suitable methods for ex vivo transfections are described in WO2004/039840.
The present invention also provides a single-chain Fc polypeptide for use in the treatment or prophylaxis of a pathological disorder that is selected from the group consisting of infections (viral, bacterial, fungal and parasitic), endotoxic shock associtated with infection, arthritis, rheumatoid arthritis, asthma, pelvic inflammatory disease, Alzheimer's Disease, Crohn's disease, Peyronie's Disease, coeliac disease, gallbladder disease, Pilonidal disease, peritonitis, psoriasis, vasculitis, surgical adhesions, stroke, Type I Diabetes, lyme arthritis, meningoencephalitis, immune mediated inflammatory disorders of the central and peripheral nervous system such as multiple sclerosis and Guillain-Barr syndrome, other autoimmune disorders, pancreatitis, trauma (surgery), graft-versus-host disease, transplant rejection, cancer (both solid tumours such as melanomas, hepatoblastomas, sarcomas, squamous cell carcinomas, transitional cell cancers, ovarian cancers and hematologic malignancies and in particular acute myelogenous leukaemia, chronic myelogenous leukemia, gastric cancer and colon cancer), heart disease including ischaemic diseases such as myocardial infarction as well as atherosclerosis, intravascular coagulation, bone resorption, osteoporosis, periodontitis and hypochlorhydia.
Preferably the present invention provides a single-chain Fc polypeptide for use in the control of inflammatory diseases and cancer. Preferably, the single-chain Fc polypeptide can be used to reduce the inflammatory process or cancer or to prevent the inflammatory process or cancer.
EXAMPLES
The present invention will now be described by way of example, in which reference is made to:
Figure 1 (a)-(c): A diagrammatic representation of three examples of single chain Fc polypeptide according to the present invention which comprise an antibody Fab fragment and in which a linker connects the C-terminus of the first CH3 domain and the N-terminus of the second CH2 domain.
Figure 2 (a)-(c): A diagrammatic representation of three examples of single chain Fc polypeptide which comprise an antibody Fab fragment and in which a linker connects the C-terminus of the first CH2 domain to the N-terminus of the second
CH2 domain and another linker connects the C-terminus of the first CH3 domain to the N-terminus of the second CH3 domain.
Figure 3 (a)-(c): A diagrammatic representation of three examples of single chain Fc polypeptide which comprise an antibody Fab fragment and in which:
(a) a linker connects the C-terminus of the first CH4 domain to the N-terminus of the second CH2 domain.
(b) a first linker connects the C-terminus of the first CH2 domain to the N-terminus of the second CH2 domain, a second linker connects the C-terminus of the second CH2 domain to the N-terminus of the first CH3 domain and a third linker connects the C-terminus of the first CH4 domain to the N-terminus of the second CH3 domain. (c) a first linker connects the C-terminus of the first CH3 domain to the N-terminus of the second CH2 domain, a second linker connects the C-terminus of the second CH3 domain to the N-terminus of the first CH4 domain and a third linker connects the C-tenninus of the first CH4 domain to the N-terminus of the second CH4 domain.
Figures 4a and 4b: demonstrate that the single chain Fc polypeptides bind to antigen recombinantly expressed on the surface of NSO cells (Figure 4a) and naturally expressed on the surface of activated T cells (Figure 4b).
Figures 5 a and b: show the ability of the single chain Fc polypeptides to induce cytotoxicity of NSO cells in the presence of complement (a) but not in the absence of complement (b).
Figures 5c and 5d: show the ability of the single chain Fc polypeptides to induce complement-dependent cytotoxicity of activated T cells.
Figure 6: Example sequences of single chain Fc polypeptides derived from IgGl.
Hinge sequences are in italics and linkers are underlined.
(a) format as shown in Figure l(a)
(b) format as shown in Figure l(b)
(c) format as shown in Figure l(c)
(d) format as shown in Figure 2(a)
(e) format as shown in Figure 2(b)
(f) format as shown in Figure 2(c)
Example 1
Three murine single chain Fc polypeptides comprising a biologically active molecule at the N-terminus were designed in which the biologically active molecule was an antibody Fab fragment. The variable regions of the Fab fragment were derived from murine antibody, Mox46, that binds to a cell surface protein antigen. The Fc domains were derived from murine IgG2a and the three different versions of these domains are shown below. The linker sequences are underlined. Hinge sequences are in italics and where these constitute part of the linker sequence they are in italics and underlined. Version 1 SEQ ID NO:88 (format as illustrated in Figure 2b). The cysteines in the hinge between CHl and CH2 have been substituted for serines.
EPRGPWZPSPPSKSPAPMJJGGPSVFWPTKLKD VLMISLSPIVTCWVDVSED
DPDVQISWFVNNVEVHTAQTQTHREDYNSTLRVVSALPIQHQDWMSGKEFK
CKVNNKDLPAPIERTISKPKGGGGSGGGGSGGGGSGGGGSGGGGSAPNLLG
GPSVFIFPPKIKDVLMISLSPIVTCVVVDVSEDDPDVQISWFVNNVEVHTAQT
QTHREDYNSTLRVVSALPIQHQDWMSGKEFKCKVNNKDLPAPIERTISKPKG
SVRAPQVYVLPPPEEEMTKKQVTLTCMVTDFMPEDIYVEWTNNGKTELNYK
NTEPVLDSDGSYFMYSKLRVEKKNWVERNSYSCSVVHEGLHNHHTTKSFSR
TPGKGGGGSGGGGSGGGGSGGGGSGGGGSGSVRAPOVYVLPPPEEEMTKK
QVTLTCMVTDFMPEDIYVEWTNNGKTELNYKNTEPVLDSDGSYFMYSKLR
VEKKNWVERNSYSCSVVHEGLHNHHTTKSFSRTPGK
Version 2 SEQ ID NO: 89 (format as illustrated in Figure 2a)
EPRGPr/^CPAPNLLGGPSVFIFPPKIKDVLMISLSPIVTCVWDVSEDDPDV
QISWFVNNVEVHTAQTQTHREDYNSTLRWSALPIQHQDWMSGKEFKCKV
NNKDLP APIERTISKPKGGGGSGGGGSGGGGSGGGGSGGGGSEPRGPTYKPC
PAPNLLGGPSVFIFPPKIKD VLMISLSPIVTCVVVDVSΕDDPDVQISWFVNNVΕ
VHTAQTQTHRΕDYNSTLRWSALPIQHQDWMSGKΕFKCKVNNKDLPAPIΕR
TISKPKGSVRAPQVYVLPPPΕΕΕMTKKQVTLTCMVTDFMPΕDIYVΕWTNNG
KTΕLNYKNTΕPVLDSDGSYFMYSKLRVΕKKNWVΕRNSYSCSWHΕGLHNH
HTTKSFSRTPGKGGGGSGGGGSGGGGSGGGGSGGGGSGSVRAPOVYVLPPP
ΕΕΕMTKKQVTLTCMVTDFMPΕDIYVΕWTNNGKTΕLNYKNTΕPVLDSDGSYF
MYSKLRVΕKKNWVΕRNSYSCSVVHΕGLHNHHTTKSFSRTPGK
Version 3 SΕQ ID NO: 90 (format as illustrated in Figure Ia). Linker comprises a truncated hinge 'PCP'.
EPRGPTYZPCPAPNLLGGPSVFIFPPKIKDVLMISLSPIVTCVWDVSΕDDPDV QISWFVNNVΕVHTAQTQTHRΕDYNSTLRVVSALPIQHQDWMSGKΕFKCKV NNKX)LPAPIERTISK-PKGSVRAPQVYVLPPPEEEMTKKQVTLTCMVTDFMPE
DIYVEWTNNGKTELNYKNTEPVLDSDGSYFMYSKLRVEKKNWVERNSYSC SVVHEGLHNHHTTKSFSRTPGKGGSSTASGSGSGGSGTAGSSGGAGSSGGST
TAGGSASGSGSTGSGTGGASSGGASGASGPCPAPNT1LGGPSVFTFPPKTKDVL MISLSPIVTCVVVDVSEDDPDVQISWFVNNVEVHTAQTQTHREDYNSTLRW SALPIQHQDWMSGKEFKCKVNNKDLPAPIERTISKPKGSVRAPQVYVLPPPE EEMTKKQVTLTCMVTDFMPEDΓYVEWTNNGKTELNYKNTEPVLDSDGSYF MYSKLRVEKKNWVERNSYSCSWHEGLHNHHTTKSFSRTPGK
The DNA encoding each of the single chain Fc polypeptides was synthesized with the same biologically active molecule at the N-terminus i.e. the VHCHl domain from antibody MOX46.
The single chain Fc polypeptides comprising versions 1, 2 and 3 above were expressed in a pVAX vector (Invitrogen) in HEK293 cells (a human embryonic kidney epithelial cell line) using an antibody leader sequence (from mouse antibody B72.3 (Whittle et ah, 1987, Protein Eng. 1(6) 499-505)). The VLCL chain of the MOX46 Fab fragment was produced in the same cell but in a separate vector. The resulting single chain Fc polypeptides were purified using protein A.
Example 2: Antigen binding
The ability of the single-chain Fc polypeptides (versions 1 and 3) to bind to antigen was compared to the same antibody variable regions (from the MOX46 antibody) expressed in a murine IgGl framework and an irrelevant IgG. Recombinant NSO cells expressing the antigen on their surface (5x105) were incubated with lOOμl of the single chain Fc polypeptides for 30 minutes at 4°C. The control was MOPC21 which was titrated from 5μg/ml down in 1/3 dilutions. The MOX46 IgG and the single-chain Fc constructs were titrated in 1/3 dilutions. The cells were washed twice in Dulbecco's PBS containing 5% FCS and 0.1% sodium azide and then lOOμl of anti-mouse heavy and light chain PE labelled (Jackson) antibody diluted 1/250 was added for 30 minutes at 40C. The cells were washed once more as before and analysed by flow cytometry.
Both of the single-chain Fc constructs tested bound antigen (1.1 and 3.1), as did the MOX46 IgG. The irrelevant control did not bind to the antigen. See Figure 4a.
The ability of the single chain Fc polypeptides (versions 1, 2 and 3) to bind antigen was also tested using the methods described above however primary activated T cells which naturally express the antigen on their surface were used instead of recombinant NSO cells. The primary activated T cells were produced as follows: IxIO6 DOl 1 splenocytes were cultured with 200ng/ml ovalbumin peptide 323-329 for 3 days, washed and resuspended in twice the volume of medium for a further 2 days. The activated cells were then purified by negatively isolating the CD4 T cells.
All three single-chain Fc constructs bound antigen, as did the MOX46 IgG. The irrelevant control did not bind to the antigen. See Figure 4b.
Example 3: Fc receptor binding
The ability of the single-chain Fc versions 1 and 3 to bind to Fc receptors CD64 (FcγRII) and CD32 (FcγRI) was determined by BIAcore. CD64 and CD32 were immobilised (approx IOOORU each) by amine coupling chemistry on flow cells 2 and 3 (respectively) of a standard CM5 Biacore chip. Flow cell 1 was set up as a reference flow cell to check for background binding. The single-chain Fc proteins were then injected in sequence over the chip to look for binding activity. All samples were run undiluted and at 1 :2 and 1 :5 dilutions. Background binding was insignificant with all samples.
Both versions 1 and 3 of the single-chain Fc were found to bind to both CD64 and CD32.
Example 4: Complement-dependent cytotoxicity assay using recombinant NSO cells
Single-chain Fc versions 1 and 3 were tested for their ability to cause complement- mediated cytotoxicity of cells to which they were bound.
A recombinant NSO line expressing the relevant antigen on its surface (5x106) cells/ml media were mixed with 50μl/ml baby rabbit complement (Serotec C12CA). Prior to use, the complement was reconstituted with 2ml ice cold tissue culture grade distilled water. It was used within one hour of reconstitution and was maintained on ice until use. The agents tested were at a concentration of 2μg/ml and plated onto a 96 well plate (Costar) in lOOμl volumes in duplicate. lOOμl of the cell/complement mix was added per well and the plate incubated at 37°C for 4 hours. Cytotoxicity was assessed by uptake of the vital stain, propidium iodide (PI) by FACS. A stock solution of 20mg/ml PI (Molecular Probes P- 1304MP) was prepared in distilled water and then diluted in RPMI 1640 to give a final concentration in the well of 3μg/ml. The cells were incubated for 10 min at RT in the dark, before being analysed by flow cytometry.
Figure 5 a (with complement) and Figure 5b (without complement) show that the both versions 1 and 3 of the single-chain Fc polypeptides (1.1 and 3.1) induce complement-dependent cytotoxicity.
Example 5: Complement-dependent cytotoxicity assay using activated T cells
(i) Single chain Fc versions 1 , 2 and 3 were tested for their ability to cause complement-mediated cytotoxicity of activated T cells which express the antigen bound by the MOX46 antibody on their surface. The methods used were as described in Example 4 except primary activated T cells were used instead of NSO cells and these were produced as described in Example 2.
% specific lysis of cells was calculated as follows:
% PI positive cells in experimental condition - background % PI positive cells maximum % PI positive cells (lysis buffer) - background % PI positive cells
All three versions of the single chain Fc polypeptides were found to induce complement-dependent cytotoxicity (Figure 5 c).
(ii) Another single chain Fc polypeptide (version 3) comprising an antibody Fab fragment derived from murine antibody, 495, that binds to a different cell surface protein antigen was produced as both an IgGl and IgG2a format using murine Fc regions. The ability of these scFc proteins to cause complement-mediated cytotoxicity of activated T cells which express the antigen bound by the 495 antibody on their surface was tested. The methods used were as described in Example 4 except primary activated T cells were used which were produced as described in Example 2.
Figure 5d clearly illustrates that as expected only the IgG2a format of the single chain Fc and the IgG2b format of the whole antibody 495 were able to induce complement-dependent cytotoxicity. The IgGl formats were unable to induce complement-dependent cytotoxicity.
Example 6: Receptor-scFc fusion
The human gpl30 receptor domain 1 was cloned as a single chain Fc (mouse gamma 1) fusion protein using the single chain Fc format illustrated in Figure Ia. The sequence of the fusion protein is shown below.
GpI 30 domain 1 scFc fusion protein (SEQ ID NO:91)
KLATMSVPTQ VLGLLLLWLT DARCELLDPC GYISPESPW QLHSNFTAVC VLKEKCMDYF HVNANYIVWK TNHFTIPKEQ YTIINRTASS VTFTDIASLN IQLTCNILTF GQLEQNVYGI
TIISGSSAVP RDGGSKPGIC TVPEVSSVFI FPPKPKDVLT ITLTPKVTCV VVDISKDDPE
VQFSWFVDDV EVHTAQTQPR EEQFNSTFRS VSELPIMHQD WLNGKEFKCR VNSAAFPAPI
EKTISKTKGR PKAPQVYTIP PPKEQMAKDK VSLTCMITDF FPEDITVEWQ WNGQPAENYK NTQPIMDTDG SYFVYSKLNV QKSNWEAGNT FTCSVLHEGL HNHHTEKSLS HSPGKGGSST ASGSGSGGSG TAGSSGGAGS SGGSTTAGGS ASGSGSTGSG TGGASSGGAS GASGVPRDGG SKPGICTVPE VSSVFIFPPK PKDVLTITLT PKVTCVVVDI SKDDPEVQFS WFVDDVEVHT AQTQPREEQF NSTFRSVSEL PIMHQDWLNG KEFKCRVNSA AFPAPIEKTI SKTKGRPKAP QVYTIPPPKE QMAKDKVSLT CMITDFFPED ITVEWQWNGQ PAENYKNTQP IMDTDGSYFV YSKLNVQKSN WEAGNTFTCS VLHEGLHNHH TEKSLSHSPG K*
The sequence in bold represents the gpl30 receptor domain 1 (amino acids 1 to 125 of SEQ ID NO:91). The linker sequence is underlined. Hinge sequences are in italics and where they constitute part of the linker they are in italics and underlined. The sequence 'SSA' between the C-terminus of the gpl30 domain 1 and the first hinge sequence are amino acids required to introduce the necessary Xhol restriction site for cloning purposes.
The constructs were transiently expressed in a mammalian cell system (CHO L761) using a pVAX vector and the B72.3 mouse signal sequence. A western blot was prepared using the resulting scFc protein and the blot was probed with an anti-mouse Fc HRP (Jackson 115-035-071) and also with the biotinylated polyclonal anti gpl30 (R&D BAF228), revealed with a strep-HRP. A protein corresponding to the predicted size of the gpl30 domain 1 fusion protein was detected on the western blot. Previous attempts to express the gp 130 domain 1 on its own or with a his tag attached had been unsuccessful. Fusion of the gpl30 domain 1 to the single chain Fc polypeptide enabled the gpl30 domain 1 to be expressed.
It will of course be understood that the present invention has been described by way of example only, is in no way meant to be limiting, and that modifications of detail can be made within the scope of the claims hereinafter. Preferred features of each embodiment of the invention are as for each of the other embodiments mutatis mutandis. All publications, including but not limited to patents and patent applications, cited in this specification are herein incorporated by reference as if each individual publication were specifically and individually indicated to be incorporated by reference herein as though fully set forth.

Claims

1. A single chain Fc polypeptide comprising two CH2 domains and two CH3 domains characterized in that said CH2 and CH3 domains form a functional Fc domain within the polypeptide chain.
2. A single chain Fc polypeptide according to claim 1 in which a first CH2 domain is dimerised with a second CH2 domain and a first CH3 domain is dimerised with a second CH3 domain within the polypeptide chain.
3. A polypeptide according to claim 2 in which, in N- to C-terminal sequence a first CH2 domain is linked at its C-terminus to the N-terminus of a first CH3 domain, optionally via a linker, and said first CH3 domain is linked at its C- terminus via a linker to the N-terminus of a second CH2 domain which is linked at its C-terminus to the N-terminus of said second CH3 domain, optionally via a linker.
4. A polypeptide according to claim 2 in which, in N- to C-terminal sequence, a first CH2 domain is linked at its C-terminus via a linker to the N-terminus of a second CH2 domain and said second CH2 domain is linked at its C- terminus to the N-terminus of a first CH3 domain, optionally via a linker, and said first CH3 domain is linked at its C-terminus to the N-terminus of a second CH3 domain via a linker.
5. A polypeptide according to claim 2 which further comprises two CH4 domains which are dimerised to each other.
6. A polypeptide according to claim 3 in which the linker between the C- terminus of the first CH3 domain and the N-terminus of the second CH2 domain is between 30 and 130 amino acids in length.
7. A polypeptide according to claim 4 in which the linker between the C- terminus of the first CH2 domain and the N-terminus of the second CH2 domain is between 15 and 90 amino acids in length.
8. A polypeptide according to claim 4 in which the linker between the C- terminus of the first CH3 domain and the N-terminus of the second CH3 domain is between 15 and 50 amino acids in length.
9. A polypeptide according to any one of claims 6-9 in which the linker comprises one or more amino acids selected from glycine, serine, alanine and threonine.
10. A polypeptide according to claim 6 in which the linker comprises the sequence given in SEQ ID NO:62.
11. A polypeptide according to claim 7 in which the linker comprises the sequence given in SEQ ID NO: 63.
12. A polypeptide according to claim 8 in which the linker comprises the sequence given in SEQ ID NO:63.
13. A polypeptide according to any one of claims 6 to 12 in which the linker comprises one or more cysteine residues.
14. A polypeptide according to any one of claims 6 to 13 in which the linker comprises all or part of an antibody hinge sequence or a modified antibody hinge sequence.
15. A polypeptide according to claim 14 in which the linker comprises an antibody hinge sequence selected from the sequence given in SEQ ID NO:53, SEQ ID NO:54, SEQ ID NO:55, SEQ ID NO:56, SEQ ID NO:57, SEQ ID NO:58, SEQ ID NO:59, SEQ ID NO:60 or SEQ ID NO:61.
16. A polypeptide according to claim 6 wherein the linker comprises the sequences given in SEQ ID NO:62 and SEQ ID NO:53.
17. A polypeptide according to claim 7 wherein the linker comprises the sequence given in SEQ ID NO:63 and SEQ ID NO:53.
18. A polypeptide according to any one of claims 1-17 in which each CH2 domain comprises the sequence given in SEQ ID NO:2 or SEQ ID NO: 15 or SEQ ID NO:28 or SEQ ID NO:41.
19. A polypeptide according to any one of claims 1-18 in which each CH3 domain comprises the sequence given in SEQ ID NO:3 or SEQ ID NO: 16 or SEQ ID NO:29 or SEQ ID NO:42.
20. A polypeptide according to any one of claims 1-17 in which each CH2 domain has at least 80% identity or similarity to a sequence given in Claim 18 and each CH3 domain has at least 80% identity or similarity to a sequence given in Claim 19.
21. A polypeptide according to any one of claims 1-20 in which the C-terminus of the second CH3 domain or CH4 domain, where present, is linked, optionally via a linker, to a transmembrane domain.
22. The polypeptide according to claim 21 wherein the C-terminus of the transmembrane domain is linked to the N-terminus of a signalling domain, optionally via a linker.
23. A single chain Fc polypeptide according to any one of claims 1 to 22 which is linked to one or more biologically active molecules.
24. A single chain Fc polypeptide according to claim 23 wherein the biologically active molecules are selected from a drug, a carbohydrate, a receptor or an antibody fragment.
25. A single chain Fc polypeptide according to claim 23 or claim 24 wherein a biologically active molecule is linked to the N-terminus of the first CH2 domain of the single chain Fc polypeptide.
26. A single chain Fc polypeptide according to claim 25 in which the biologically active molecule and the single chain Fc polypeptide are linked by a peptide linker of between 1 and 100 amino acids in length.
27. A single chain Fc polypeptide according to claim 26 wherein the linker comprises a cysteine residue.
28. A single chain Fc polypeptide according to claim 26 or claim 27 wherein the linker comprises an antibody hinge sequence selected from the sequence given in SEQ ID NO:53, SEQ ID NO:54, SEQ ID NO:55, SEQ ID NO:56, SEQ ID NO:57, SEQ ID NO:58, SEQ ID NO:59, SEQ ID NO:60 or SEQ ID NO:61.
29. A single chain Fc polypeptide according to any one of claims 1 to 28 comprising the sequence given in SEQ ID NO:8, SEQ ID NO:9, SEQ ID NO:10, SEQ ID NO:11, SEQ ID NO:12, SEQ ID NO:13, SEQ ID NO:21, SEQ ID NO:22, SEQ ID NO:23, SEQ ID NO:24, SEQ ID NO:25, SEQ ID NO:26, SEQ ID NO:34, SEQ ID NO:35, SEQ ID NO:36, SEQ ID NO:37, SEQ ID NO:38, SEQ ID NO:39, SEQ ID NO:47, SEQ ID NO:48, SEQ ID NO:49, SEQ ID NO:50, SEQ ID NO:51 or SEQ ID NO:52.
30. A single chain Fc polypeptide according to any one of claims 1 to 28 comprising a sequence having at least 80% identity or similarity to any one of the sequences given in Claim 29.
31. A single chain Fc polypeptide according to any one of claims 23 to 30 wherein the biologically active molecule comprises an antibody fragment.
32. A single chain Fc polypeptide according to claim 31 wherein the antibody fragment is selected from VHH, VH, VL, VH-CHl, VL-CL, Fab, Fab' or a scFv.
33. A single chain Fc polypeptide according to claim 32 wherein the antibody fragment is a Fab and the C-terminus of the VH-CHl chain of the Fab is genetically fused to the N-terminus of the single chain Fc polypeptide and the VL-CL chain of the Fab is linked to the VH-CHl chain by a disulphide bond.
34. A single chain Fc polypeptide according to any one of claims 23 to 30 wherein the biologically active molecule comprises one or more domains of the human gpl30 receptor or fragments thereof.
35. A single chain Fc polypeptide according to claim 34 that comprises domain 1 of the human gpl30 receptor or a fragment thereof.
36. A single chain Fc polypeptide according to any one of claims 1 to 35 to which one or more effector molecules is attached.
37. An isolated DNA sequence encoding the single chain Fc polypeptide according to any one of claims 1 to 36.
38. A cloning or expression vector comprising one or more DNA sequences according to claim 37.
39. A host cell comprising one or more cloning or expression vectors according to claim 38.
40. A process for the production of the single chain Fc polypeptide of any one of claims 1 to 36 comprising culturing the host cell of claim 39 and isolating the single chain Fc polypeptide.
41. A pharmaceutical composition comprising a single chain Fc polypeptide according to any one of claims 1 to 36, in combination with one or more of a pharmaceutically acceptable excipient, diluent or carrier.
42. A pharmaceutical composition according to claim 41, additionally comprising other active ingredients.
PCT/GB2007/002842 2006-07-25 2007-07-24 Single chain fc polypeptides WO2008012543A1 (en)

Priority Applications (6)

Application Number Priority Date Filing Date Title
JP2009521344A JP5384338B2 (en) 2006-07-25 2007-07-24 Single chain Fc polypeptide
EP07789062.2A EP2046832B1 (en) 2006-07-25 2007-07-24 Single chain fc polypeptides
ES07789062.2T ES2564389T3 (en) 2006-07-25 2007-07-24 Single chain Fc polypeptides
CA2658542A CA2658542C (en) 2006-07-25 2007-07-24 Single chain fc polypeptides
CN200780027807.2A CN101495510B (en) 2006-07-25 2007-07-24 Single chain FC polypeptides
US12/374,213 US10479824B2 (en) 2006-07-25 2007-07-24 Single chain FC polypeptides

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GB0614780.5 2006-07-25
GBGB0614780.5A GB0614780D0 (en) 2006-07-25 2006-07-25 Biological products

Publications (1)

Publication Number Publication Date
WO2008012543A1 true WO2008012543A1 (en) 2008-01-31

Family

ID=37006136

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2007/002842 WO2008012543A1 (en) 2006-07-25 2007-07-24 Single chain fc polypeptides

Country Status (8)

Country Link
US (1) US10479824B2 (en)
EP (2) EP2046832B1 (en)
JP (1) JP5384338B2 (en)
CN (2) CN104744587B (en)
CA (1) CA2658542C (en)
ES (2) ES2564389T3 (en)
GB (1) GB0614780D0 (en)
WO (1) WO2008012543A1 (en)

Cited By (55)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008131242A1 (en) * 2007-04-18 2008-10-30 Zymogenetics, Inc. Single chain fc, methods of making and methods of treatment
WO2008143954A2 (en) * 2007-05-14 2008-11-27 Biogen Idec Ma Inc. Single-chain fc (scfc) regions, binding polypeptides comprising same, and methods related thereto
WO2010045261A1 (en) * 2008-10-13 2010-04-22 Zymogenetics, Llc Single chain fc type iii interferons and methods of using same
WO2011030107A1 (en) 2009-09-10 2011-03-17 Ucb Pharma S.A. Multivalent antibodies
CN102264390A (en) * 2008-07-02 2011-11-30 新兴产品开发西雅图有限公司 IL6 immunotherapeutics
WO2012006633A1 (en) 2010-07-09 2012-01-12 Biogen Idec Hemophilia Inc. Chimeric clotting factors
US20120142593A1 (en) * 2009-03-24 2012-06-07 Bayer Healthcare Llc Factor VIII Variants and Methods of Use
WO2012142515A2 (en) 2011-04-13 2012-10-18 Bristol-Myers Squibb Company Fc fusion proteins comprising novel linkers or arrangements
WO2013156148A1 (en) * 2012-04-16 2013-10-24 Universität Stuttgart THE IgM AND IgE HEAVY CHAIN DOMAIN 2 AS COVALENTLY LINKED HOMODIMERIZATION MODULES FOR THE GENERATION OF FUSION PROTEINS WITH DUAL SPECIFICITY
WO2013185114A2 (en) 2012-06-08 2013-12-12 Biogen Idec Ma Inc. Chimeric clotting factors
WO2014126884A1 (en) 2013-02-12 2014-08-21 Bristol-Myers Squibb Company High ph protein refolding methods
WO2014127215A1 (en) 2013-02-15 2014-08-21 Biogen Idec Ma Inc. Optimized factor viii gene
WO2014126871A1 (en) 2013-02-12 2014-08-21 Bristol-Myers Squibb Company Tangential flow filtration based protein refolding methods
US9163079B2 (en) 2009-11-27 2015-10-20 Orgentec Diagnostika Gmbh Monospecific polypeptide reagents
WO2016065301A1 (en) 2014-10-23 2016-04-28 Biogen Ma Inc. Anti-gpiib/iiia antibodies and uses thereof
WO2016070050A1 (en) 2014-10-31 2016-05-06 Biogen Ma Inc. ANTI-GLYCOPROTEIN IIb/IIIa ANTIBODIES
WO2016110468A1 (en) * 2015-01-05 2016-07-14 Innate Pharma Monomeric fc domains
US9518132B2 (en) 2010-11-09 2016-12-13 Altimab Therapeutics, Inc. Protein complexes for antigen binding and methods of use
WO2017005734A1 (en) 2015-07-06 2017-01-12 Ucb Biopharma Sprl Tau-binding antibodies
WO2017009473A1 (en) 2015-07-16 2017-01-19 Ucb Biopharma Sprl Antibody molecules which bind cd45
WO2017060242A1 (en) 2015-10-05 2017-04-13 Ucb Biopharma Sprl Molecular signatures for use in diagnosis and response to treatment analysis of autoimmune diseases
WO2017136358A1 (en) 2016-02-01 2017-08-10 Bioverativ Therapeutics Inc. Optimized factor viii genes
WO2018026642A1 (en) * 2016-08-03 2018-02-08 The Feinstein Institute For Medical Research C1q and hmgb1 fusion proteins and uses thereof
WO2018112362A1 (en) 2016-12-16 2018-06-21 Biogen Ma Inc. Stabilized proteolytically activated growth differentiation factor 11
EP3307304A4 (en) * 2015-06-12 2018-12-26 UBI Pharma Inc. Immunoglobulin fusion proteins and uses thereof
WO2019032898A1 (en) 2017-08-09 2019-02-14 Bioverativ Therapeutics Inc. Nucleic acid molecules and uses thereof
US10239944B2 (en) 2014-05-02 2019-03-26 Momenta Pharmaceuticals, Inc. Compositions and methods related to engineered Fc constructs
US10287564B2 (en) 2012-06-08 2019-05-14 Bioverativ Therapeutics Inc. Procoagulant compounds
US10344081B2 (en) 2015-07-06 2019-07-09 Ucb Biopharma Sprl Tau-binding antibodies
WO2019152692A1 (en) 2018-02-01 2019-08-08 Bioverativ Therapeutics, Inc. Use of lentiviral vectors expressing factor viii
WO2020033863A1 (en) 2018-08-09 2020-02-13 Bioverativ Therapeutics Inc. Nucleic acid molecules and uses thereof for non-viral gene therapy
US10633460B2 (en) 2010-12-23 2020-04-28 Roche Diagnostic Operations, Inc. Binding agent
WO2020118069A2 (en) 2018-12-06 2020-06-11 Bioverativ Therapeutics Inc. Use of lentiviral vectors expressing factor ix
WO2020215010A1 (en) 2019-04-17 2020-10-22 Codiak Biosciences, Inc. Compositions of exosomes and aav
US10982007B2 (en) 2010-12-23 2021-04-20 Roche Diagnostics Operations, Inc. Detection of a posttranslationally modified polypeptide by a bivalent binding agent
US11008561B2 (en) 2014-06-30 2021-05-18 Bioverativ Therapeutics Inc. Optimized factor IX gene
WO2021191424A1 (en) 2020-03-27 2021-09-30 UCB Biopharma SRL Autonomous knob domain peptides
US11135301B2 (en) 2016-09-14 2021-10-05 Duke University Triblock polypeptide-based nanoparticles for the delivery of hydrophilic drugs
US11155640B2 (en) 2016-05-23 2021-10-26 Janssen Biotech, Inc. Compositions and methods related to engineered Fc constructs
US11155584B2 (en) 2016-09-23 2021-10-26 Duke University Unstructured non-repetitive polypeptides having LCST behavior
US11168125B2 (en) 2003-05-06 2021-11-09 Bioverativ Therapeutics Inc. Immunoglobulin chimeric monomer-dimer hybrids
US11220531B2 (en) 2017-01-06 2022-01-11 Janssen Biotech, Inc. Engineered Fc constructs
US11352433B2 (en) 2016-02-03 2022-06-07 Amgen Research (Munich) Gmbh BCMA and CD3 bispecific T cell engaging antibody constructs
US11434302B2 (en) 2016-02-03 2022-09-06 Amgen Research (Munich) Gmbh Bispecific T cell engaging antibody constructs
US11458205B2 (en) 2015-08-04 2022-10-04 Duke University Genetically encoded intrinsically disordered stealth polymers for delivery and methods of using same
US11467156B2 (en) 2016-06-01 2022-10-11 Duke University Nonfouling biosensors
US11512314B2 (en) 2019-07-12 2022-11-29 Duke University Amphiphilic polynucleotides
US11554097B2 (en) 2017-05-15 2023-01-17 Duke University Recombinant production of hybrid lipid-biopolymer materials that self-assemble and encapsulate agents
WO2023028440A2 (en) 2021-08-23 2023-03-02 Bioverativ Therapeutics Inc. Baculovirus expression system
US11618790B2 (en) 2010-12-23 2023-04-04 Hoffmann-La Roche Inc. Polypeptide-polynucleotide-complex and its use in targeted effector moiety delivery
WO2023064708A1 (en) * 2021-10-12 2023-04-20 Wang Chang Yi Vaccine compositions against sars-cov-2 variants of concern to prevent infection and treat long-haul covid
US11649275B2 (en) 2018-08-02 2023-05-16 Duke University Dual agonist fusion proteins
US11648200B2 (en) 2017-01-12 2023-05-16 Duke University Genetically encoded lipid-polypeptide hybrid biomaterials that exhibit temperature triggered hierarchical self-assembly
US11680083B2 (en) 2017-06-30 2023-06-20 Duke University Order and disorder as a design principle for stimuli-responsive biopolymer networks
US11752213B2 (en) 2015-12-21 2023-09-12 Duke University Surfaces having reduced non-specific binding and antigenicity

Families Citing this family (34)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8759297B2 (en) 2006-08-18 2014-06-24 Armagen Technologies, Inc. Genetically encoded multifunctional compositions bidirectionally transported between peripheral blood and the cns
MX2009013004A (en) 2007-06-01 2010-01-20 Univ Maryland Immunoglobulin constant region fc receptor binding agents.
CA2694762A1 (en) 2007-07-27 2009-02-05 Armagen Technologies, Inc. Methods and compositions for increasing alpha-l-iduronidase activity in the cns
US20100143353A1 (en) * 2008-12-04 2010-06-10 Mosser David M POLYPEPTIDES COMPRISING Fc FRAGMENTS OF IMMUNOGLOBULIN G (lgG) AND METHODS OF USING THE SAME
EP2485761B1 (en) 2009-10-09 2019-02-27 Armagen, Inc. Methods and compositions for increasing iduronate 2-sulfatase activity in the cns
KR101331101B1 (en) 2010-06-04 2013-11-19 에스케이케미칼주식회사 Fusion protein having factor vii activity
BR112013002074B1 (en) 2010-07-28 2021-09-14 Gliknik Inc HOMODIMERIC COMPOUND, DIMER OR MULTIMER OF A HIGHER ORDER OF HOMODIMERIC COMPOUND, COMPOSITION, COMPOUND, MULTIMER AND USES THEREOF
US8986688B2 (en) * 2011-06-28 2015-03-24 Inhibrx, Llc WAP domain fusion polypeptides and methods of use thereof
US10400029B2 (en) 2011-06-28 2019-09-03 Inhibrx, Lp Serpin fusion polypeptides and methods of use thereof
WO2013003641A2 (en) * 2011-06-28 2013-01-03 Inhibrx Llc Serpin fusion polypeptides and methods of use thereof
KR101963230B1 (en) * 2011-12-26 2019-03-29 삼성전자주식회사 Protein complex comprising multi-specific monoclonal antibodies
EP2847224A4 (en) * 2012-05-10 2016-04-27 Zymeworks Inc Single-arm monovalent antibody constructs and uses thereof
AU2013305885B2 (en) 2012-08-20 2017-12-21 Gliknik Inc. Molecules with antigen binding and polyvalent Fc gamma receptor binding activity
KR101911438B1 (en) * 2012-10-31 2018-10-24 삼성전자주식회사 Bispecific antigen binding protein complex and preparation methods of bispecific antibodies
US10538589B2 (en) * 2015-01-14 2020-01-21 Armagen Inc. Methods and compositions for increasing N-acetylglucosaminidase (NAGLU) activity in the CNS using a fusion antibody comprising an anti-human insulin receptor antibody and NAGLU
JP6828005B2 (en) * 2015-03-31 2021-02-10 ファンダメンタル・ソリューションズ・コーポレイションFundamental Solutions Corporation Biosensor system for rapid detection of substances to be measured
US9752199B2 (en) 2015-03-31 2017-09-05 Fundamental Solutions Corporation Biosensor system for the rapid detection of analytes
TWI829617B (en) 2015-07-31 2024-01-21 德商安美基研究(慕尼黑)公司 Antibody constructs for flt3 and cd3
TWI796283B (en) 2015-07-31 2023-03-21 德商安美基研究(慕尼黑)公司 Antibody constructs for msln and cd3
WO2017096221A1 (en) * 2015-12-02 2017-06-08 The Rockefeller University Bispecific anti-hiv broadly neutralizing antibodies
WO2017134158A1 (en) * 2016-02-03 2017-08-10 Amgen Research (Munich) Gmbh Psma and cd3 bispecific t cell engaging antibody constructs
AU2017245143B2 (en) 2016-03-30 2024-01-11 Ab Biosciences, Inc. Recombinant intravenous immunoglobulin (rIVIG) compositions and methods for their production and use
WO2017214321A1 (en) 2016-06-07 2017-12-14 Gliknik Inc. Cysteine-optimized stradomers
PE20191354A1 (en) 2016-12-09 2019-10-01 Gliknik Inc MANUFACTURING OPTIMIZATION OF GL-2045, A MULTIMERIZING STRADOMER
US10613083B2 (en) 2016-12-22 2020-04-07 Fundamental Solutions Corporation Universal biosensor system for analyte detection
JOP20190189A1 (en) * 2017-02-02 2019-08-01 Amgen Res Munich Gmbh Low ph pharmaceutical composition comprising t cell engaging antibody constructs
KR20240023449A (en) 2017-02-08 2024-02-21 드래곤플라이 쎄라퓨틱스, 인크. Multi-specific binding proteins for activation of natural killer cells and therapeutic uses thereof to treat cancer
CA3054079A1 (en) 2017-02-20 2018-08-23 Dragonfly Therapeutics, Inc. Proteins binding her2, nkg2d and cd16
US11372001B2 (en) * 2017-07-06 2022-06-28 Nitto Boseki Co., Ltd. Anti-human IgG4 monoclonal antibody and methods of making and using same
EP3749346A4 (en) 2018-02-08 2021-12-15 Dragonfly Therapeutics, Inc. Antibody variable domains targeting the nkg2d receptor
CA3135166A1 (en) * 2018-04-26 2019-10-31 The Trustees Of The University Of Pennsylvania Compositions and methods for retrieving tumor-related antibodies and antigens
EA202091977A1 (en) * 2018-05-28 2021-02-09 Драгонфлай Терапьютикс, Инк. MULTI-SPECIFIC BINDING PROTEINS THAT BIND CD33, NKG2D AND CD16 AND METHODS OF APPLICATION
CN113490690A (en) * 2018-12-29 2021-10-08 南通壹宸生物医药科技有限公司 Heterodimeric fusion proteins
CN113406320B (en) * 2021-08-18 2021-11-02 南京立顶医疗科技有限公司 Directional coupling method based on recombinant gene engineering antibody and microsphere and application

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6165745A (en) * 1992-04-24 2000-12-26 Board Of Regents, The University Of Texas System Recombinant production of immunoglobulin-like domains in prokaryotic cells
WO2005001025A2 (en) * 2003-05-06 2005-01-06 Syntonix Pharmaceuticals, Inc. Immunoglobulin chimeric monomer-dimer hybrids
WO2005077981A2 (en) * 2003-12-22 2005-08-25 Xencor, Inc. Fc POLYPEPTIDES WITH NOVEL Fc LIGAND BINDING SITES

Family Cites Families (43)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4741900A (en) 1982-11-16 1988-05-03 Cytogen Corporation Antibody-metal ion complexes
GB8308235D0 (en) 1983-03-25 1983-05-05 Celltech Ltd Polypeptides
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
GB8720833D0 (en) 1987-09-04 1987-10-14 Celltech Ltd Recombinant dna product
US5677425A (en) 1987-09-04 1997-10-14 Celltech Therapeutics Limited Recombinant antibody
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
GB8928874D0 (en) 1989-12-21 1990-02-28 Celltech Ltd Humanised antibodies
WO1992002551A1 (en) 1990-08-02 1992-02-20 B.R. Centre Limited Methods for the production of proteins with a desired function
GB9112536D0 (en) 1991-06-11 1991-07-31 Celltech Ltd Chemical compounds
US6030613A (en) * 1995-01-17 2000-02-29 The Brigham And Women's Hospital, Inc. Receptor specific transepithelial transport of therapeutics
GB9526131D0 (en) 1995-12-21 1996-02-21 Celltech Therapeutics Ltd Recombinant chimeric receptors
US5980898A (en) 1996-11-14 1999-11-09 The United States Of America As Represented By The U.S. Army Medical Research & Material Command Adjuvant for transcutaneous immunization
GB9625640D0 (en) 1996-12-10 1997-01-29 Celltech Therapeutics Ltd Biological products
GB9713473D0 (en) 1997-06-25 1997-09-03 Celltech Therapeutics Ltd Biological products
GB9720054D0 (en) 1997-09-19 1997-11-19 Celltech Therapeutics Ltd Biological products
GB9809658D0 (en) 1998-05-06 1998-07-01 Celltech Therapeutics Ltd Biological products
AU3871099A (en) * 1998-05-06 1999-11-23 Temple University - Of The Commonwealth System Of Higher Education Reversal of proinflammatory response by ligating the macrophage fcgammari receptor
GB2348203B (en) * 1998-11-04 2002-06-19 Imp College Innovations Ltd Solube beta-forms of prion proteins, methods of preparation and use
GB9908807D0 (en) 1999-04-16 1999-06-09 Celltech Therapeutics Ltd Synthetic signalling molecules
GB9908816D0 (en) 1999-04-16 1999-06-09 Celltech Therapeutics Ltd Biological product
US7052906B1 (en) 1999-04-16 2006-05-30 Celltech R & D Limited Synthetic transmembrane components
GB9925854D0 (en) 1999-11-01 1999-12-29 Celltech Therapeutics Ltd Biological products
GB9925853D0 (en) 1999-11-01 1999-12-29 Celltech Therapeutics Ltd Biological products
GB9925848D0 (en) 1999-11-01 1999-12-29 Celltech Therapeutics Ltd Biological products
GB0025307D0 (en) 2000-10-16 2000-11-29 Celltech Chiroscience Ltd Biological products
US7829084B2 (en) 2001-01-17 2010-11-09 Trubion Pharmaceuticals, Inc. Binding constructs and methods for use thereof
US7070995B2 (en) * 2001-04-11 2006-07-04 City Of Hope CE7-specific redirected immune cells
US7332474B2 (en) * 2001-10-11 2008-02-19 Amgen Inc. Peptides and related compounds having thrombopoietic activity
US7053202B2 (en) * 2001-10-19 2006-05-30 Millennium Pharmaceuticals, Inc. Immunoglobulin DNA cassette molecules, monobody constructs, methods of production, and methods of use therefor
CA2493706A1 (en) * 2002-07-23 2004-01-29 Duke University Igg fc/hiv-gp120/c3d fusion protein
GB0225279D0 (en) 2002-10-30 2002-12-11 Celltech R&D Ltd Biological products
EP1570267B1 (en) 2002-12-03 2011-10-12 UCB Pharma, S.A. Assay for identifying antibody producing cells
GB0312481D0 (en) 2003-05-30 2003-07-09 Celltech R&D Ltd Antibodies
JP2007500016A (en) * 2003-06-11 2007-01-11 バイオジェン・アイデック・エムエイ・インコーポレイテッド Method for increasing protein production in culture
GB0315450D0 (en) 2003-07-01 2003-08-06 Celltech R&D Ltd Biological products
GB0315457D0 (en) 2003-07-01 2003-08-06 Celltech R&D Ltd Biological products
ES2551439T5 (en) 2003-07-01 2018-11-08 Ucb Biopharma Sprl Fab fragments of modified antibodies
US6924422B2 (en) 2003-07-23 2005-08-02 Jonathan Wise Drum pedal stabilizer
PT1718677E (en) * 2003-12-19 2012-07-18 Genentech Inc Monovalent antibody fragments useful as therapeutics
US7666622B2 (en) * 2005-10-19 2010-02-23 Regeneron Pharmaceuticals, Inc. Monomeric self-associating fusion polypeptides and therapeutic uses thereof
US7803769B2 (en) * 2006-10-25 2010-09-28 Amgen Inc. OSK1 peptide analogs and pharmaceutical compositions
US20080260738A1 (en) * 2007-04-18 2008-10-23 Moore Margaret D Single chain fc, methods of making and methods of treatment
NZ581395A (en) * 2007-05-14 2012-08-31 Biogen Idec Inc Single-chain fc (scfc) regions, binding polypeptides comprising same, and methods related thereto

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6165745A (en) * 1992-04-24 2000-12-26 Board Of Regents, The University Of Texas System Recombinant production of immunoglobulin-like domains in prokaryotic cells
WO2005001025A2 (en) * 2003-05-06 2005-01-06 Syntonix Pharmaceuticals, Inc. Immunoglobulin chimeric monomer-dimer hybrids
WO2005077981A2 (en) * 2003-12-22 2005-08-25 Xencor, Inc. Fc POLYPEPTIDES WITH NOVEL Fc LIGAND BINDING SITES

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
BITONTI A J ET AL: "Pulmonary delivery of an erythropoietin Fc fusion protein in non-human primates through an immunoglobulin transport pathway", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF USA, NATIONAL ACADEMY OF SCIENCE, WASHINGTON, DC, US, vol. 101, no. 26, 29 June 2004 (2004-06-29), pages 9763 - 9768, XP003008495, ISSN: 0027-8424 *
DUMONT JENNIFER A ET AL: "MONOMERIC FC FUSIONS: IMPACT ON PHARMACOKINETIC AND BIOLOGICAL ACTIVITY OF PROTEIN THERAPEUTICS", BIODRUGS, AUCKLAND, NZ, vol. 20, no. 3, 2006, pages 151 - 160, XP009082835, ISSN: 1173-8804 *
GREENWOOD J ET AL: "ENGINEERING MULTIPLE-DOMAIN FORMS OF THE THERAPEUTIC ANTIBODY CAMPATH-1H: EFFECTS ON COMPLEMENT LYSIS", THERAPEUTIC IMMUNOLOGY, BLACKWELL SCIENTIFIC PUBL. LONDON, GB, vol. 1, no. 5, October 1994 (1994-10-01), pages 247 - 255, XP009059035, ISSN: 0967-0149 *
LOW S C ET AL: "Oral and pulmonary delivery of FSH-Fc fusion proteins via neonatal Fc receptor-mediated transcytosis", HUMAN REPRODUCTION (OXFORD), vol. 20, no. 7, July 2005 (2005-07-01), pages 1805 - 1813, XP002455704, ISSN: 0268-1161 *

Cited By (92)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11168125B2 (en) 2003-05-06 2021-11-09 Bioverativ Therapeutics Inc. Immunoglobulin chimeric monomer-dimer hybrids
WO2008131242A1 (en) * 2007-04-18 2008-10-30 Zymogenetics, Inc. Single chain fc, methods of making and methods of treatment
WO2008143954A2 (en) * 2007-05-14 2008-11-27 Biogen Idec Ma Inc. Single-chain fc (scfc) regions, binding polypeptides comprising same, and methods related thereto
WO2008143954A3 (en) * 2007-05-14 2009-03-19 Biogen Idec Inc Single-chain fc (scfc) regions, binding polypeptides comprising same, and methods related thereto
CN102264390A (en) * 2008-07-02 2011-11-30 新兴产品开发西雅图有限公司 IL6 immunotherapeutics
WO2010045261A1 (en) * 2008-10-13 2010-04-22 Zymogenetics, Llc Single chain fc type iii interferons and methods of using same
US20120142593A1 (en) * 2009-03-24 2012-06-07 Bayer Healthcare Llc Factor VIII Variants and Methods of Use
WO2011030107A1 (en) 2009-09-10 2011-03-17 Ucb Pharma S.A. Multivalent antibodies
US9163079B2 (en) 2009-11-27 2015-10-20 Orgentec Diagnostika Gmbh Monospecific polypeptide reagents
US9815891B2 (en) 2009-11-27 2017-11-14 Robert Poppe Monospecific polypeptide reagents
US10927362B2 (en) 2010-07-09 2021-02-23 Bioverativ Therapeutics Inc. Processable single chain molecules and polypeptides made using same
US20130216513A1 (en) * 2010-07-09 2013-08-22 Biogen Idec Hemophilia Inc. Chimeric Clotting Factors
WO2012006633A1 (en) 2010-07-09 2012-01-12 Biogen Idec Hemophilia Inc. Chimeric clotting factors
US9856468B2 (en) 2010-07-09 2018-01-02 Bioverativ Therapeutics Inc. Processable single chain molecules and polypeptides made using same
US10968442B2 (en) 2010-07-09 2021-04-06 Bioverativ Therapeutics Inc. Chimeric clotting factors
CN103180439A (en) * 2010-07-09 2013-06-26 比奥根艾迪克依蒙菲利亚公司 Chimeric clotting factors
US9518132B2 (en) 2010-11-09 2016-12-13 Altimab Therapeutics, Inc. Protein complexes for antigen binding and methods of use
US10633460B2 (en) 2010-12-23 2020-04-28 Roche Diagnostic Operations, Inc. Binding agent
US10982007B2 (en) 2010-12-23 2021-04-20 Roche Diagnostics Operations, Inc. Detection of a posttranslationally modified polypeptide by a bivalent binding agent
US11618790B2 (en) 2010-12-23 2023-04-04 Hoffmann-La Roche Inc. Polypeptide-polynucleotide-complex and its use in targeted effector moiety delivery
EP3415528A2 (en) 2011-04-13 2018-12-19 Bristol-Myers Squibb Company Fc fusion proteins comprising novel linkers or arrangements
EP3896083A1 (en) 2011-04-13 2021-10-20 Bristol-Myers Squibb Company Fc fusion proteins comprising novel linkers or arrangements
WO2012142515A2 (en) 2011-04-13 2012-10-18 Bristol-Myers Squibb Company Fc fusion proteins comprising novel linkers or arrangements
US9469676B2 (en) 2011-04-13 2016-10-18 Bristol-Myers Squibb Company Fc fusion proteins comprising novel linkers or arrangements
US10214579B2 (en) 2011-04-13 2019-02-26 Bristol-Myers Squibb Company Fc fusion proteins comprising novel linkers or arrangements
EP3144320A1 (en) 2011-04-13 2017-03-22 Bristol-Myers Squibb Company Fc fusion proteins comprising novel linkers or arrangements
WO2013156054A1 (en) * 2012-04-16 2013-10-24 Universität Stuttgart The igm and ige heavy chain domain 2 as covalently linked homodimerization modules for the generation of fusion proteins with dual specificity
US10875928B2 (en) 2012-04-16 2020-12-29 Universität Stuttgart IgM and IgE heavy chain domain 2 as covalently linked homodimerization modules for the generation of fusion proteins with dual specificity
WO2013156148A1 (en) * 2012-04-16 2013-10-24 Universität Stuttgart THE IgM AND IgE HEAVY CHAIN DOMAIN 2 AS COVALENTLY LINKED HOMODIMERIZATION MODULES FOR THE GENERATION OF FUSION PROTEINS WITH DUAL SPECIFICITY
US11261437B2 (en) 2012-06-08 2022-03-01 Bioverativ Therapeutics Inc. Procoagulant compounds
WO2013185114A2 (en) 2012-06-08 2013-12-12 Biogen Idec Ma Inc. Chimeric clotting factors
US11168316B2 (en) 2012-06-08 2021-11-09 Bioverativ Therapeutics, Inc. Chimeric clotting factors
US10202595B2 (en) 2012-06-08 2019-02-12 Bioverativ Therapeutics Inc. Chimeric clotting factors
US10287564B2 (en) 2012-06-08 2019-05-14 Bioverativ Therapeutics Inc. Procoagulant compounds
EP3744728A1 (en) 2013-02-12 2020-12-02 Bristol-Myers Squibb Company Tangential flow filtration based protein refolding methods
EP3299378A1 (en) 2013-02-12 2018-03-28 Bristol-Myers Squibb Company High ph protein refolding methods
WO2014126884A1 (en) 2013-02-12 2014-08-21 Bristol-Myers Squibb Company High ph protein refolding methods
WO2014126871A1 (en) 2013-02-12 2014-08-21 Bristol-Myers Squibb Company Tangential flow filtration based protein refolding methods
EP3617220A1 (en) 2013-02-12 2020-03-04 Bristol-Myers Squibb Company High ph protein refolding methods
EP3889173A1 (en) 2013-02-15 2021-10-06 Bioverativ Therapeutics Inc. Optimized factor viii gene
US10370431B2 (en) 2013-02-15 2019-08-06 Bioverativ Therapeutics Inc. Optimized factor VIII gene
WO2014127215A1 (en) 2013-02-15 2014-08-21 Biogen Idec Ma Inc. Optimized factor viii gene
US11787851B2 (en) 2013-02-15 2023-10-17 Bioverativ Therapeutics Inc. Optimized factor VIII gene
EP4223772A2 (en) 2013-02-15 2023-08-09 Bioverativ Therapeutics Inc. Optimized factor viii gene
US10239944B2 (en) 2014-05-02 2019-03-26 Momenta Pharmaceuticals, Inc. Compositions and methods related to engineered Fc constructs
US11124573B2 (en) 2014-05-02 2021-09-21 Janssen Biotech, Inc. Compositions and methods related to engineered Fc constructs
US11008561B2 (en) 2014-06-30 2021-05-18 Bioverativ Therapeutics Inc. Optimized factor IX gene
WO2016065301A1 (en) 2014-10-23 2016-04-28 Biogen Ma Inc. Anti-gpiib/iiia antibodies and uses thereof
WO2016070050A1 (en) 2014-10-31 2016-05-06 Biogen Ma Inc. ANTI-GLYCOPROTEIN IIb/IIIa ANTIBODIES
AU2016206108B2 (en) * 2015-01-05 2021-09-09 Innate Pharma Monomeric Fc domains
WO2016110468A1 (en) * 2015-01-05 2016-07-14 Innate Pharma Monomeric fc domains
AU2016274890B2 (en) * 2015-06-12 2022-02-10 Ubi Pharma Inc Immunoglobulin fusion proteins and uses thereof
EP3307304A4 (en) * 2015-06-12 2018-12-26 UBI Pharma Inc. Immunoglobulin fusion proteins and uses thereof
US10344081B2 (en) 2015-07-06 2019-07-09 Ucb Biopharma Sprl Tau-binding antibodies
US10906966B2 (en) 2015-07-06 2021-02-02 UCB Biopharma SRL Tau-binding antibodies
WO2017005734A1 (en) 2015-07-06 2017-01-12 Ucb Biopharma Sprl Tau-binding antibodies
US10889640B2 (en) 2015-07-06 2021-01-12 Ucb Biopharma Sprl Tau-binding antibodies
US11732034B2 (en) 2015-07-06 2023-08-22 UCB Biopharma SRL Tau-binding antibodies
US10287343B2 (en) 2015-07-06 2019-05-14 Ucb Biopharma Sprl Tau-binding antibodies
US11746145B2 (en) 2015-07-06 2023-09-05 UCB Biopharma SRL Tau-binding antibodies
WO2017009473A1 (en) 2015-07-16 2017-01-19 Ucb Biopharma Sprl Antibody molecules which bind cd45
US11458205B2 (en) 2015-08-04 2022-10-04 Duke University Genetically encoded intrinsically disordered stealth polymers for delivery and methods of using same
WO2017060242A1 (en) 2015-10-05 2017-04-13 Ucb Biopharma Sprl Molecular signatures for use in diagnosis and response to treatment analysis of autoimmune diseases
US11752213B2 (en) 2015-12-21 2023-09-12 Duke University Surfaces having reduced non-specific binding and antigenicity
WO2017136358A1 (en) 2016-02-01 2017-08-10 Bioverativ Therapeutics Inc. Optimized factor viii genes
EP4137570A1 (en) 2016-02-01 2023-02-22 Bioverativ Therapeutics Inc. Optimized factor viii genes
US11753461B2 (en) 2016-02-01 2023-09-12 Bioverativ Therapeutics Inc. Optimized factor VIII genes
US11352433B2 (en) 2016-02-03 2022-06-07 Amgen Research (Munich) Gmbh BCMA and CD3 bispecific T cell engaging antibody constructs
US11434302B2 (en) 2016-02-03 2022-09-06 Amgen Research (Munich) Gmbh Bispecific T cell engaging antibody constructs
US11155640B2 (en) 2016-05-23 2021-10-26 Janssen Biotech, Inc. Compositions and methods related to engineered Fc constructs
US11623964B2 (en) 2016-05-23 2023-04-11 Momenta Pharmaceuticals, Inc. Compositions and methods related to engineered Fc constructs
US11467156B2 (en) 2016-06-01 2022-10-11 Duke University Nonfouling biosensors
WO2018026642A1 (en) * 2016-08-03 2018-02-08 The Feinstein Institute For Medical Research C1q and hmgb1 fusion proteins and uses thereof
US11135301B2 (en) 2016-09-14 2021-10-05 Duke University Triblock polypeptide-based nanoparticles for the delivery of hydrophilic drugs
US11155584B2 (en) 2016-09-23 2021-10-26 Duke University Unstructured non-repetitive polypeptides having LCST behavior
WO2018112362A1 (en) 2016-12-16 2018-06-21 Biogen Ma Inc. Stabilized proteolytically activated growth differentiation factor 11
US11220531B2 (en) 2017-01-06 2022-01-11 Janssen Biotech, Inc. Engineered Fc constructs
US11827682B2 (en) 2017-01-06 2023-11-28 Momenta Pharmaceuticals, Inc. Engineered Fc constructs
US11648200B2 (en) 2017-01-12 2023-05-16 Duke University Genetically encoded lipid-polypeptide hybrid biomaterials that exhibit temperature triggered hierarchical self-assembly
US11554097B2 (en) 2017-05-15 2023-01-17 Duke University Recombinant production of hybrid lipid-biopolymer materials that self-assemble and encapsulate agents
US11680083B2 (en) 2017-06-30 2023-06-20 Duke University Order and disorder as a design principle for stimuli-responsive biopolymer networks
WO2019032898A1 (en) 2017-08-09 2019-02-14 Bioverativ Therapeutics Inc. Nucleic acid molecules and uses thereof
WO2019152692A1 (en) 2018-02-01 2019-08-08 Bioverativ Therapeutics, Inc. Use of lentiviral vectors expressing factor viii
US11649275B2 (en) 2018-08-02 2023-05-16 Duke University Dual agonist fusion proteins
WO2020033863A1 (en) 2018-08-09 2020-02-13 Bioverativ Therapeutics Inc. Nucleic acid molecules and uses thereof for non-viral gene therapy
WO2020118069A2 (en) 2018-12-06 2020-06-11 Bioverativ Therapeutics Inc. Use of lentiviral vectors expressing factor ix
WO2020215010A1 (en) 2019-04-17 2020-10-22 Codiak Biosciences, Inc. Compositions of exosomes and aav
US11512314B2 (en) 2019-07-12 2022-11-29 Duke University Amphiphilic polynucleotides
US11965164B2 (en) 2019-07-12 2024-04-23 Duke University Amphiphilic polynucleotides
WO2021191424A1 (en) 2020-03-27 2021-09-30 UCB Biopharma SRL Autonomous knob domain peptides
WO2023028440A2 (en) 2021-08-23 2023-03-02 Bioverativ Therapeutics Inc. Baculovirus expression system
WO2023064708A1 (en) * 2021-10-12 2023-04-20 Wang Chang Yi Vaccine compositions against sars-cov-2 variants of concern to prevent infection and treat long-haul covid

Also Published As

Publication number Publication date
JP5384338B2 (en) 2014-01-08
GB0614780D0 (en) 2006-09-06
EP2046832A1 (en) 2009-04-15
CN101495510A (en) 2009-07-29
CN101495510B (en) 2015-02-25
JP2009544300A (en) 2009-12-17
EP3009448B1 (en) 2017-09-13
CA2658542A1 (en) 2008-01-31
ES2651309T3 (en) 2018-01-25
EP3009448A1 (en) 2016-04-20
ES2564389T3 (en) 2016-03-22
US20090304696A1 (en) 2009-12-10
CA2658542C (en) 2017-10-31
CN104744587B (en) 2019-01-11
EP2046832B1 (en) 2015-12-02
CN104744587A (en) 2015-07-01
US10479824B2 (en) 2019-11-19

Similar Documents

Publication Publication Date Title
EP2046832B1 (en) Single chain fc polypeptides
US20230322957A1 (en) Monoclonal antibodies against bcma
US20220135676A1 (en) Multispecific antigen binding proteins
US11845795B2 (en) NKp46 binding proteins
CN110352200B (en) Antibodies against T cell immunoreceptor (TIGIT) with Ig and ITIM domains and uses thereof
US10421816B2 (en) Multivalent antibodies
WO2015063187A1 (en) Multivalent antigen-binding proteins
AU2014227638A1 (en) Tetravalent bispecific antibodies
CN104968683A (en) Method for the selection of antibodies against bcma
US20200325232A1 (en) Multispecific antigen binding proteins
WO2020192709A1 (en) Novel bispecific polypeptide complexes
CN110563842B (en) Antibodies to programmed death ligand (PD-L1) and uses thereof
US20230340142A1 (en) Pd-l1 antibodies, fusion proteins, and uses thereof
US20240141042A1 (en) MULTISPECIFIC NKp46 BINDING PROTEINS
CA3228641A1 (en) Fap/cd40 binding molecule and medicinal use thereof
TW202313699A (en) Novel anti-sirpa antibodies
CN116964091A (en) Human CCR8 binding agents

Legal Events

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

Ref document number: 200780027807.2

Country of ref document: CN

121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 07789062

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 2007789062

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2658542

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2009521344

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

NENP Non-entry into the national phase

Ref country code: RU

WWE Wipo information: entry into national phase

Ref document number: 12374213

Country of ref document: US