WO2006131013A2 - STABLE AND SOLUBLE ANTIBODIES INHIBITING TNFα - Google Patents

STABLE AND SOLUBLE ANTIBODIES INHIBITING TNFα Download PDF

Info

Publication number
WO2006131013A2
WO2006131013A2 PCT/CH2006/000300 CH2006000300W WO2006131013A2 WO 2006131013 A2 WO2006131013 A2 WO 2006131013A2 CH 2006000300 W CH2006000300 W CH 2006000300W WO 2006131013 A2 WO2006131013 A2 WO 2006131013A2
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
seq
formulation
tnfα
anyone
Prior art date
Application number
PCT/CH2006/000300
Other languages
French (fr)
Other versions
WO2006131013A3 (en
Inventor
Stefan Ewert
Alcide Barberis
David M. Urech
Adrian Auf Der Maur
Peter Lichtlen
Original Assignee
Esbatech Ag
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to CN200680028415.3A priority Critical patent/CN101233154B/en
Priority to EP06741625A priority patent/EP1891110A2/en
Priority to NZ563580A priority patent/NZ563580A/en
Priority to MX2015006717A priority patent/MX342638B/en
Priority to MX2007015280A priority patent/MX2007015280A/en
Priority to KR1020087000199A priority patent/KR101457223B1/en
Priority to KR1020147011105A priority patent/KR101539545B1/en
Priority to JP2008515021A priority patent/JP5089582B2/en
Priority to US11/916,793 priority patent/US8067547B2/en
Priority to AU2006255415A priority patent/AU2006255415B2/en
Application filed by Esbatech Ag filed Critical Esbatech Ag
Priority to BRPI0611765-1A priority patent/BRPI0611765B1/en
Priority to CA2609999A priority patent/CA2609999C/en
Publication of WO2006131013A2 publication Critical patent/WO2006131013A2/en
Publication of WO2006131013A3 publication Critical patent/WO2006131013A3/en
Priority to IL187503A priority patent/IL187503A0/en
Priority to US13/245,420 priority patent/US8389693B2/en
Priority to PH12012500758A priority patent/PH12012500758A1/en
Priority to US13/708,500 priority patent/US8691228B2/en
Priority to US14/185,783 priority patent/US9315572B2/en
Priority to IL243270A priority patent/IL243270B/en
Priority to US15/064,067 priority patent/US9683034B2/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/24Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against cytokines, lymphokines or interferons
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/24Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against cytokines, lymphokines or interferons
    • C07K16/241Tumor Necrosis Factors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • 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
    • 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
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • 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
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/04Inotropic agents, i.e. stimulants of cardiac contraction; Drugs for heart failure
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/10Immunoglobulins specific features characterized by their source of isolation or production
    • C07K2317/14Specific host cells or culture conditions, e.g. components, pH or temperature
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/21Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/52Constant or Fc region; Isotype
    • C07K2317/522CH1 domain
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/55Fab or Fab'
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/567Framework region [FR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/622Single chain antibody (scFv)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/94Stability, e.g. half-life, pH, temperature or enzyme-resistance

Definitions

  • the present invention relates to optimised antibodies and antibody derivatives that bind to and block the function of tumour necrosis factor alpha (TNF ⁇ ) and are useful for the diagnosis and/or treatment, prevention or amelioration of TNF ⁇ -associated diseases; their coding sequences, production, and use in pharmacologically suitable compositions.
  • TNF ⁇ tumour necrosis factor alpha
  • Tumour necrosis factor alpha (TNF ⁇ , also known as cachectin) , is a naturally occurring mammalian cytokine produced by numerous cell types, including monocytes and macrophages in response to endotoxin or other stimuli. TNFoc is a major mediator of inflammatory, immunological, and pathophysiological reactions (Grell, M. , et al. (1995) Cell, 83: 793-802).
  • Soluble TNF ⁇ is formed by the. cleavage of a precursor transmembrane protein (Kriegler, et al . (1988) Cell 53: 45-53), and the secreted 17 kDa polypeptides assemble to soluble homotrimer complexes (Smith, et al.(1987), J. Biol. Chem. 262: 6951-6954; for reviews of TNF, see Butler, et al . (1986), Nature 320:584; Old (1986), Science 230: 630). These complexes then bind to receptors found on a variety of cells.
  • Binding produces an array of pro-inflammatory effects, including (i) release of other pro-inflammatory cytokines such as interleukin (IL) -6, IL-8, and IL-I, (ii) release of matrix metalloproteinases and (iii) up regulation of the expression of endothelial adhesion molecules, further amplifying the inflammatory and immune cascade by attracting leukocytes into extravascular tissues.
  • pro-inflammatory cytokines such as interleukin (IL) -6, IL-8, and IL-I
  • TNF ⁇ has been shown to be up- regulated in a number of human diseases, including chronic diseases such as rheumatoid arthritis (RA) , inflammatory bowel disorders including Crohn's disease and ulcerative colitis, sepsis, congestive heart failure, asthma bronchiale and multiple sclerosis .
  • RA rheumatoid arthritis
  • Mice transgenic for human TNF ⁇ produce high levels of TNF ⁇ constitutively and develop a spontaneous, destructive polyarthritis resembling RA (Keffer et al . 1991, EMBO J., 10,4025- 4031) .
  • TNF ⁇ is therefore referred to as a pro- inflammatory cytokine.
  • TNFa is now well established as key in the pathogenesis of RA, which is a chronic, progressive and debilitating disease characterised by polyarticular joint inflammation and destruction, with systemic symptoms of fever and malaise and fatigue.
  • RA also leads to chronic synovial inflammation, with frequent progression to articular cartilage and bone destruction.
  • Increased levels of TNF ⁇ are found in both the synovial fluid and peripheral blood of patients suffering from RA.
  • TNF ⁇ blocking agents are administered to patients suffering from RA, they reduce inflammation, improve symptoms and retard joint damage (McKown et al . (1999), Arthritis Rheum. 42:1204-1208) .
  • TNF ⁇ is also associated with protection from particular infections (Cerami. et al. (1988), Immunol. Today 9:28). TNF ⁇ is released by macrophages that have been activated by lipopolysaccharides of Gram-negative bacteria. As such, TNF ⁇ appears to be an endogenous mediator of central importance involved in the development and pathogenesis of endotoxic shock associated with bacterial sepsis (Michie, et al . (1989), Br. J. Surg.76 : 670-671.; Debets . et al. (1989), Second Vienna Shock Forum, p. 463-466; Simpson, et al . (1989) Crit . Care Clin.
  • TNF ⁇ has also been shown to play a key. role in the central nervous system, in particular in inflammatory and autoimmune disorders of the nervous system, including multiple sclerosis, Guillain-Barre syndrome and myasthenia gravis, and in degenerative disorders of the nervous system, including Alzheimer's disease, Parkinson's disease and • Huntington's disease. TNF ⁇ is also involved in disorders of related systems of the retina and of muscle, including optic neuritis, macular degeneration, diabetic retinopathy, dermatomyositis, amyotrophic lateral sclerosis, and muscular dystrophy, as well as in injuries to the nervous system, including traumatic brain injury, acute spinal cord injury, and stroke.
  • Hepatitis is another TNF ⁇ -related inflammatory disorder which among other triggers can be caused by viral infections, including Epstein-Barr, cytomegalo- virus, and hepatitis A-E viruses. Hepatitis causes acute liver inflammation in the portal and lobular region, followed by fibrosis and tumor progression.
  • TNF ⁇ can mediate cachexia in cancer, which causes most cancer morbidity and mortality (Tisdale M.J. (2004), Langenbecks Arch Surg. 389:299-305).
  • TNF ⁇ is an important cytokine whose systemic blockade carries the risk for increased frequency and severity of clinically manifested infections, in particular re-activation of latent tuberculosis and possibly other risks including induction of lymphomas, demyelinating diseases and heart failure.
  • TNF ⁇ antagonists designed for the treatment of TNF ⁇ -mediated diseases are antibodies or antibody fragments that specifically bind TNF ⁇ and thereby block its function.
  • the use of anti-TNF ⁇ antibodies has shown that a blockade of TNF ⁇ can reverse effects attributed to TNF ⁇ including decreases in IL-I, GM-CSF, IL-6, IL-8, adhesion molecules and tissue destruction (Feldmann et al. (1997), Adv. Immunol. • 1997:283-350) .
  • Antibodies directed against TNF ⁇ have been proposed for the prophylaxis and treatment of endotoxic shock (Beutler et al . (1985) Science :234, 470-474) .
  • the use of anti-TNF ⁇ antibodies in the treatment of septic shock is discussed by Bodmer et al . , 1993, (Critical Care Medicine, 21:441-446,1993), Wherry et al.,1993, (Critical Care Medicine, 21:436-440) and Kirschenbaum et al . ,1998, (Critical Care Medicine, 26:1625-1626).
  • a method for treating a neurodegenerative disease in a human by administering an anti-TNF ⁇ monoclonal antibody or a TNF ⁇ binding fragment thereof has been disclosed in US2003147891.
  • WO0149321 teaches the use of TNF ⁇ blockers including anti TNF ⁇ antibodies to treat neurologic and related disorders caused by TNF ⁇ . It provides a method for treating said disorders by administering a TNF ⁇ antagonist .
  • WO03047510 discloses various kinds of monoclonal and engineered antibodies directed against
  • TNF ⁇ TNF ⁇ , their production, compounds comprising them and use in medicine.
  • Antibodies useful for therapies of TNF ⁇ mediated diseases are usually either monoclonal antibodies (mAB) produced by hybridoma technology from a natural source, usually a mouse, or engineered antibodies.
  • mAB monoclonal antibodies
  • the latter either correspond to naturally occurring antibodies in that they comprise full-length heavy and light chains, or to the Fab fragments that can also be generated from natural antibodies by proteolytic cleavage, or to single chain scFv antibodies wherein fragments of the variable heavy and light chain regions are linked by a peptide linker.
  • Both, heavy and light chains of an antibody comprise constant and variable domains.
  • non-human antibodies are immunogenic, the amount of human-like sequences in an antibody is often increased in a so-called “hybrid” antibody, which comprises constant regions of a human IgG, and variable regions matching the sequences of an animal antibody, in most cases murine antibodies with the desired specificity. These variable regions can then be further adapted to become more similar to a typical human antibody by mutagenesis, leading to a "humanised” antibody.
  • CDRs complementary determining regions
  • anti-TNF ⁇ antibodies used in the treatment of RA and Crohn's disease is discussed in Feldman et ali(1998), (Transplantation Proceedings 30:4126-4127), Adorini et al . , 1997, (Trends in Immunology Today 18:209-211) and in Feldmann et al . , 1997, (Advanced Immunology 64:283-350).
  • the antibodies to TNF ⁇ used in such treatments are generally chimeric antibodies, such as those described in US5919452.
  • US20003187231 discloses humanised anti-TNF ⁇ antibodies with at least one non-human CDR region that have improved binding characteristics. Furthermore, in the International Patent Application WO 92/11383, recombinant antibodies, including CDR-grafted antibodies, specific for TNF ⁇ are disclosed. Rankin et al . (1995), (British J. Rheumatology 34:334-342) describe the use of such CDR-grafted antibodies in the treatment of RA.
  • WO9211383 discloses a recombinant, humanised CDR-grafted antibody specific for TNF ⁇ that is derived from the murine monoclonal antibody 61El, hTNFI, hTNF3 or 101.4, and it teaches the production and use of said antibodies in diagnosis and/or therapy of TNF ⁇ -associated , disorders .
  • US22002037934 discloses the treatment of hepatitis by administration of an anti-TNF ⁇ antibody such as infliximab.
  • US6428787 teaches the treatment of neurologic and TNF ⁇ -associated diseases with anti-TNF ⁇ antibodies including infliximab, CDP571 and D2E7.
  • D2E7 Adalimumab
  • a human anti-TNF ⁇ monoclonal antibody (Abbott) has been developed to treat RA and Crohn's disease (WO9729131) .
  • Celltech is developing CDP571 (EP0626389) , a humanised monoclonal anti-TNF ⁇ IgG4 antibody to treat Crohn' s disease and CDP870, a humanised monoclonal anti TNF ⁇ antibody- fragment to treat RA.
  • the local administration of said antibodies for treatment of localised disorders is disclosed in US2003185826.
  • scFvs single chain antibodies
  • scFv antibodies can be produced in microbial systems which are associated with fewer costs compared to the production of therapeutic full-length antibodies.
  • scFvs have also been successfully used for intracellular applications (Worn et al . 2000, JBC, 28;275 (4) :2795-2803; Auf der Maur et al . 2002, JBC, 22;277 (47) : 45075-45085; Stocks MR, 2004, Drug Discov Today.
  • the present invention provides antibodies, compositions and methods for effective and continuous treatment of inflammatory processes of arthritis and other TNF ⁇ - mediated disorders or pathophysiological mechanisms, in particular various forms of pain.
  • said antibody derivative is an scFv antibody or Fab fragment.
  • said antibody or antibody 'derivative is manifested by the features that it comprises a light chain variable domain being or derived from the sequence SEQ ID NO:1 that is combined with a heavy chain variable domain being or derived from the sequence SEQ ID NO: 2, wherein in the case of a derived sequence said sequence has at maximum up to 5 changes within the framework of said VL domain and/or at maximum up to 9 changes within the framework of said VH domain.
  • a preferred embodiment of the present invention is said antibody or antibody derivative, wherein one or more amino acid changes are introduced at any of the positions in the framework, preferably at one or more positions selected from the group of the positions 4, 46, 65, 67 70, and 83 of the VL domain, and/or at one or more of the positions selected from the group of the positions 11, 16, 28, 43, 48, 68, 70, 71, 72, 73, 76, 77, 79, 93 and 112 of the VH domain. More preferably, at least one of the conversions leads to an amino acid present in SEQ ID NO: 3 for VL and/or SEQ ID NO: 4 for VH, and even more preferably at most 13 conversions in total are present.
  • said antibody or antibody derivatives comprises a VL domain of the sequence SEQ ID N0:l and/or a VH domain of the sequence or derived from the sequence SEQ ID NO: 2, or a VL domain of the sequence SEQ ID NO: 11 and a VH domain of the sequence SEQ ID NO: 4.
  • the VH domain of the antibody of the present invention comprises a VL domain of SEQ ID NO:1
  • the VH sequence is derived from SEQ ID NO: 2 such that the phenylalanine at position 68 is changed to either alanine, leucine, isoleucine or valine. Additional changes within VH are optional.
  • scFv antibodies of this kind are given in SEQ ID NO: 31, SEQ ID NO: 33, SEQ ID NO:34, SEQ ID NO:35, SEQ ID NO:36, and SEQ ID NO:37.
  • said antibody or antibody derivative is derived from the antibody with the VL sequence SEQ ID N0:l and the VH sequence SEQ ID NO: 2 and comprises at least one amino acid residue that is converted in at least one of the CDRs to a residue present in the corresponding CDR of the VL sequence SEQ ID NO: 5 and/or the VH sequence SEQ ID No: 6 or SEQ ID NO: 25.
  • At least one of the CDRs of the group VL CDR2, VL CDR3, VH CDR2 or VH CDR3 is converted to the corresponding CDR of the VL sequence SEQ ID NO: 5 and/or the VH sequence SEQ ID No:25 or SEQ ID N0:6.
  • said antibody or antibody derivative comprises the following VL/VH sequence combinations :
  • VL SEQ ID NO: 7/VH SEQ ID NO: 2
  • VL SEQ ID NO: 8/VH SEQ ID NO: 2
  • VL SEQ ID NO:1/VH SEQ ID NO: 9 VL SEQ ID NO:1/VH SEQ ID NO: 25
  • VL SEQ ID NO: 26/VH SEQ ID NO: 30,
  • the antibody or antibody derivative of the present invention has specificity to human TNF ⁇ .
  • antigen binding is characterized by a K ⁇ of * 100 nM or less. More preferred is an antibody with a K ⁇ of 10 nM or less, and most preferred of 1 nM and less.
  • Antibody derivatives according to the present invention are for example Fc fusions, toxin fusions, fusions to enzymatic activities, different formats such as minibodies, diabodies, linear antibodies, single chain antibodies, bispecific antibody fragments, in particular scFv and Fab fragments.
  • Another preferred object of the present invention is a scFv antibody whose VL and VH domains are connected by a linker, preferably in a VL-linker-VH sequence arrangement. More preferably said linker has the sequence SEQ ID NO: 10.
  • Another preferred object of the present invention is a scFv antibody derived from SEQ ID NO: 40 (TB-A) . Such an antibody can be obtained by mutagenesis, and comprises three or less mutations in either framework, CDR and/or linker sequences.
  • the scFv antibody has the sequence SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, SEQ ID NO: 24, SEQ ID NO:31, SEQ ID NO: 32, SEQ ID NO:33, SEQ ID NO:34, SEQ ID NO:35, SEQ ID NO:36, SEQ ID NO:37, or SEQ ID NO:38.
  • Another preferred object of the present invention is the Fab fragment comprising a VL domain that is fused to the constant region of a human Ig kappa chain, and a VH domain that is fused to the CHl domain of a human IgG, whereby the two fusion polypeptides are connected by an inter-chain disulfide bridge.
  • the antibody or antibody derivative, e.g. the antibody fragment, of the present invention is labelled or chemically modified.
  • the present invention also provides a DNA sequence encoding any of the antibodies or antibody derivatives of .the present invention, as well as a cloning or expression vector containing said DNA sequence.
  • a suitable host cell transformed with said DNA sequence is provided, which preferentially is E. coli, a yeast or a mammalian cell.
  • a method for the production of the antibodies or antibody derivatives of the present invention comprising culturing of the host cell transformed with the DNA encoding any of said antibodies or antibody derivatives under conditions that allow the synthesis of said antibody or antibody derivative, and recovering said molecule from said culture.
  • said method provides an scFv antibody or Fab fragment purified from E. coli.
  • Another aspect of the present invention is the use of the antibodies or antibody derivatives provided by the present invention as a diagnostic tool for in vitro diagnostics, and/or as a pharmaceutical. This use is particularly preferred in the context of any TNF ⁇ related condition.
  • the present invention also encompasses a composition comprising an antibody or antibody derivative of the present invention in combination with a pharmaceutically acceptable carrier, diluent or excipient, said composition to be used as a medicament for the treatment of TNFa associated diseases.
  • a combination preparation comprising an antibody or antibody derivative of the present invention, preferably with a second compound that is not an antibody or antibody derivative specific for TNF ⁇ .
  • the vector comprising the DNA sequence encoding an scFv antibody of the present invention is used for gene therapy.
  • TNF ⁇ associated diseases is achieved by blocking of TNF ⁇ due to a strong interaction of TNF ⁇ with the antibody or the antibody derivative.
  • a treatment of autoimmune, acute or chronic inflammation conditions, cancer-related diseases, pain, neurological and neurodegenerative disorders, infectious diseases and cardiovascular diseases is envisaged.
  • Figure 1 shows a scheme of the scFv antibodies with the sequences of TB-A and TB-B delimiting the range of the most frequent variations.
  • Asterisks designate positions at which amino acid changes in the framework of the antibodies of the present invention are tolerated.
  • Amino acids indicated below the CDRs (the CDRs being emphasized with a gray background) can be used in the respective CDRs.
  • Figure 2 shows an exemplary scheme for the expression of a Fab fragment.
  • Figure 3 shows the production yield of scFvs when expressed in E. coll.
  • A SDS-polyacryalmide gel electrophoresis of expressed proteins.
  • B Analytical gel filtration of TB-A and TB-wt showing superior solubility of TB-A.
  • Figure 4 shows a comparison of affinity of different scFv antibodies towards TNF ⁇ determined by ELISA.
  • Figure 5 shows the inhibition of human TNF ⁇ - induced cytotoxicity in mouse L929 fibroblasts.
  • A Concentration dependent inhibition of TB-A in comparison to TB-wt and infliximab with IC 5 0 values.
  • B Comparison of TB-A derivatives to block TNF ⁇ induced cytotoxicity.
  • C Comparison of scFv and Fab formats of TB-A.
  • Figure 6 shows the effect of antibody treatment of human TNF ⁇ -induced joint swelling in rat (Experiment: 5.3., Experiment 1).
  • Figure 7 shows the scoring scheme for histopathological inflammation scoring.
  • Figure 8 shows the effect of antibody treatment on human TNF ⁇ -induced joint inflammation in rat (Experiment: 5.3., Experiment 1).
  • Figure 9 shows the effect of antibody treatment of human TNF ⁇ -induced joint swelling in rat (Experiment: 5.3., Experiment 2).
  • Figure 10 shows the effect of antibody treatment on human TNF ⁇ -induced joint inflammation in rat (Experiment: 5.3., Experiment 2).
  • Figure 11 shows the stability of TB-A in different body fluids .
  • antibodies or antibody derivatives comprising the frameworks identified in the so called “quality control” screen (WO0148017) are characterised by a generally high stability and/or solubility and thus may also be useful in the context of extracellular applications such as neutralizing TNF ⁇ .
  • the present invention provides antibodies or antibody derivatives characterized by enhanced stability and solubility that specifically recognize and bind TNF ⁇ and thus are suitable to block the function of TNF ⁇ in vivo.
  • Said antibodies or antibody derivatives are characterized by a special framework derived from the "quality control" screen for antibodies with particularly stable and soluble frameworks independent of their antigen binding site that has been disclosed in EP1479694.
  • Di62 inhibits human TNF ⁇ -induced cytotoxicity in mouse L929 cells.
  • TB-B said scFv antibody being called TB-B.
  • TB-B gave good yields in protein expression (Fig. 3a) , but was unable to specifically bind TNF ⁇ (Fig. 4a) .
  • an antibody or antibody derivative that is (i) sufficiently specific for binding TNF ⁇ , (ii) sufficiently soluble to allow efficient production and purification and to block TNF ⁇ in vivo, (iii) sufficiently stable to be useful as a pharmaceutical without suffering a rapid degradation and (iv) sufficiently non-immunogenic, a compromise between best solubility and best antigen binding characteristics was sought by varying the framework and the CDRs .
  • the present invention provides a sequence for VL and VH that is optimized for the combination of the criteria (i-iv) .
  • An scFv antibody comprising said VL (SEQ ID. NO:1 linked by a (GGGGS) 4 linker to said VH (SEQ ID NO: 2) is called TB- A.
  • TB-A The sequence of TB-A is given by SEQ ID NO: 40.
  • This antibody is still reasonably stable and soluble to give satisfactory yields when expressed and purified from E. coli (Fig. 3A) , and it does not aggregate (Fig. 3B) . Its binding characteristics towards TNF ⁇ are excellent, with a Kd of 0.8 nM .
  • the present invention also discloses VL and VH sequences derived from the sequences present in TB-A in various ways.
  • point mutations at up to five positions in the framework of VL and/or at up to nine positions in the framework of VH have been found acceptable, especially point mutations that render the frameworks more TB-B-like, i.e. more like SEQ ID NO: 3 for VL or SEQ ID NO: 4 for VH.
  • VL and/or VH frameworks of TB-A only single or double point mutations are introduced into VL and/or VH frameworks of TB-A.
  • the preferred framework residues for mutations are at positions 4, 46, 65, 67, 70 and 83 for VL, and at positions 11, 16, 28, 43, 48, 68, 70, 71, 72, 73, 76, 77, 79, 93 and 112 for VH.
  • the positions are numbered according to the numbering in the sequence listings.
  • the amino acids substitutions are preferably either "conservative", or such that the replacing amino acids are more similar or preferably even identical to the corresponding amino acids present in the TB-B sequence.
  • A76 of VH in TB-A can be changed to 176 as it is present in TB-B, but it may also be changed to another amino acid with similar, i.e. a non-polar side chain such as V or L. This is an example of a "conservative" amino acid substitution.
  • Families of amino acid residues having similar side chains suitable for "conservative" substitutions as used herein have been defined in the art, including basic side chains (K, R, H) , acidic side chains (D, E) , uncharged polar side chains (Q, N, S, T, Y, C) , non-polar side chains (G, A, V, L, I, P, F, M, W) , beta-branched side chains (T, V, I) and aromatic side chains (Y, F, W, H) .
  • a preferred conservative change is that of VL at position 83, in that V is changed to either F (SEQ ID NO: 26) or to A (SEQ ID NO: 27).
  • SEQ ID NO: 32 a non-conservative change in VL is V83E, which is combined with a change in CDRl, i.e. N31D, and in VH with V79A.
  • Another extraordinary TB-A variant is that of SEQ ID NO: 33, with a conservative ' F68L exchange in VH connected to VL by a linker carrying an R at position 2, replacing G.
  • Much preferred single amino acid exchanges are R65S or Y67S in VL and K43Q or F68 to V, L, or A in VH.
  • Much preferred double changes are F70L/L72R or A76I/S77G in VH.
  • ScFv antibodies comprising TB-A sequences with said alterations show inhibition of TNF ⁇ induced cytotoxicity in L929 cells. The results of some of them are shown in Fig. 5B. Their sequences are as follows :
  • SEQ ID NO: 18 TB-A H_K43Q (TB-A H43)
  • SEQ ID NO: 19 TB-A H_F68V (TB-A H68)
  • SEQ ID NO:20 TB-A H_F70L/L72R (TB-A H70/72)
  • SEQ ID NO:21 TB-A H_A76I/S77G (TB-A H76/77)
  • SEQ ID NO: 22 TB-A L_L46R (TB-A L46)
  • SEQ ID NO: 23 TB-A L_R65S (TB-A L65)
  • SEQ ID NO: 24 TB-A L_Y67S (TB-A L67)
  • any of the above mentioned VH domains may be combined with any of the above mentioned VL domains .
  • the VL and VH domains of TB-A and TB-B are shuffled such that , the VL domain of TB-A (SEQ ID. NO:1 is combined with the VH domain of TB-B (SEQ ID NO:4), or the VL domain of TB-B (SEQ ID. NO:4) is combined with the VH domain of TB-A (SEQ ID NO: 2) .
  • the shuffled versions obtained in an scFv are connected with the (GGGGS) 4 linker of the sequence SEQ ID. NO: 10, resulting in the scFv antibodies TB-AB (SEQ ID. NO: 12 ⁇ or TB-BA (SEQ ID. NO:13), respectively.
  • Said (GGGGS) 4 linker can have an amino acid exchange of a glycine to a more hydrophilic, i.e. polar, or even charged amino • acid, which ' may render the antibody more soluble.
  • the one with the sequence GRGGS- (GGGGS) 3 (SEQ ID NO: 39) is preferred.
  • one or more amino acids are changed in the CDR regions of the TB-A VL and/or VH sequences to match the corresponding amino acids present in the selected sequences SEQ ID NO: 5 of VL and/or SEQ ID. NO: 6 or SEQ ID NO: 25 of VH.
  • Much preferred are changes in one of the VL CDRs (VL CDR2 or VL CDR3) and/or VH CDRs (CDR2 or CDR3) , with the most preferred changes leading to the VL sequences SEQ ID N0:7or SEQ ID NO: 8 and/or the VH sequences SEQ ID NO:25 or SEQ ID NO: 9, respectively.
  • VL sequences SEQ ID NO: 26 or SEQ ID NO:27 is ' combined with the VH sequence SEQ ID NO:30.
  • VL sequence SEQ ID N0:l is combined with the VH sequences Seq ID NO:28 or SEQ ID NO:29.
  • any of the disclosed VL sequences may be combined with any of the disclosed VH sequences.
  • Objects of the present invention are antibodies and antibody fragments , in particular VL or VH polypeptides, single-chain antibodies (scFv) or Fab fragments.
  • a selected VL domain can be linked to a selected VH domain in either orientation by a flexible linker.
  • a suitable state of the art linker consists of repeated GGGGS amino acid sequences or variants thereof.
  • a (GGGGS) 4 linker of the sequence ID NO: 10 or its derivative SEQ ID NO: 39 is used, but variants of 1-3 repeats are also possible (Holliger et al . (1993), Proc. Natl. Acad. Sci. USA 90:6444-6448).
  • linkers that can be used for the present invention are described by Alfthan et al . (1995.), Protein Eng. 8:725-731, Choi et al. (2001), Eur. J. Immunol. 31:94-106, Hu et al . (1996), Cancer Res. 56:3055-3061, Klpriyanov et al. (1999), J. MoI. Biol. 293:41-56 and Roovers et al. (2001), Cancer Immunol. Immunother . 50:51-59.
  • the arrangement can be either VL- linker-VH or VH-linker-VL, with the former orientation being the preferred one.
  • Fab fragments selected light chain variable domains VL are fused to the constant region of a human Ig kappa chain, while the suitable heavy chain variable domains VH are fused to the first (N-terminal) constant domain CHl of a human IgG.
  • the human CK domain has the sequence SEQ ID NO: 14 and the CHl domain used to construct the Fab fragments has the sequence SEQ ID NO: 15.
  • Fig. 2 shows an example of a Fab fragment wherein the VL and VH domains of TB-A are used such that the VL domain is directly linked to the human kappa constant domain, resulting in the sequence
  • an inter-chain disulfide bridge is formed between the two constant domains.
  • the antibodies or antibody derivatives of the present invention can have affinities to human TNF ⁇ with dissociation constants K ⁇ in a range of 0.8-10'0OO nM.
  • the K ⁇ is ⁇ IO nM.
  • the affinity of an antibody for an antigen can be determined experimentally using a suitable method (Berzofsky et al . "Antibody-Antigen Interactions", in Fundamental Immunology, Paul, W.E., Ed, Raven Press: New York, NY (1992); Kuby, J. Immunology, W. H. Freeman and Company: New York, NY ) and methods described therein.
  • the antibodies or antibody derivatives, especially the scFv or Fab fragments are labeled.
  • Detectable labeling of a TNF ⁇ -specific antibody or antibody derivative can be accomplished by linking it to an enzyme for use in an enzyme immunoassay (EIA) or enzyme-linked immunosorbent assay (ELISA) , which are methods well known to the person skilled in the art (for example Current Protocols in Immunology, Coligan et al . Eds, John Wiley & Sons, 2005) .
  • EIA enzyme immunoassay
  • ELISA enzyme-linked immunosorbent assay
  • RIA radioimmuneassay
  • the radioisotope can be detected by the use of a gamma counter or a scintillation counter or by autoradiography.
  • Particularly useful isotopes are 3 H, 131 I, 35 S, 14 C, and preferably 125 I.
  • the antibodies or antibody derivatives of the present invention can also be labeled with fluorescent labeling compounds such as fluorescein, isothiocyanate, rhodamine, phycoerythrin, phycocyanin, allophycocyanin, o-phthaldehyde and fluorescamine, or with chemilumines- cent compounds such as luminol, isoluminol, theromatic acridinium ester, imidazol acridinium salt and oxalate ester.
  • fluorescent labeling compounds such as fluorescein, isothiocyanate, rhodamine, phycoerythrin, phycocyanin, allophycocyanin, o-phthaldehyde and fluorescamine
  • chemilumines- cent compounds such as luminol, isoluminol, theromatic acridinium ester, imidazol acridinium salt and oxalate ester.
  • Labeling and detection protocols are well known to the person skilled in the art. For example, they are available from Using Antibodies : A Laboratory Manual: Portable Protocol NO. I (Harlow, E. and Lane, D., 1998). Labeled antibodies or antibody derivatives of the present invention are useful for diagnostic purposes, in particular detection of TNF ⁇ in a biological sample removed from a patient. Any sample containing TNF ⁇ can be used, e.g. biological fluids like blood, serum, lymph, urine, inflammatory exudate, cerebrospinal fluid, amniotic fluid, a tissue extract or homogenate, and the like, or histological specimens for in situ detection.
  • biological fluids like blood, serum, lymph, urine, inflammatory exudate, cerebrospinal fluid, amniotic fluid, a tissue extract or homogenate, and the like, or histological specimens for in situ detection.
  • pharmaceutical formulation refers to preparations which are in such form as to permit the biological actvity of the antibody or antibody derivative to be unequivocally effective, and which contain no additional ' components which are toxic to the subjects to which the formulation would be administered.
  • “Pharmaceutically acceptable” excipients are those which can reasonably be administered to a subject mammal to provide an effective dose of the active ingredient employed.
  • a “stable” formulation is one in which the antibody or antibody derivative therein essentially retains its physical stability and/or chemical stability and/or biological activity upon storage.
  • Various analytical techniques for measuring protein stability are available in the art and are reviewed in Peptide and Protein Drug Delivery, 247-301, Vincent Lee Ed., Marcel Dekker, Inc., New York, N. Y., Pubs. (1991) and Jones, .A. Adv. Drug Delivery Rev. 10: 29-90 (1993), for example, Stability ⁇ can be measured at a selected temperature for a selected time period.
  • the formulation is stable at room temperature (about 30° C) or at 40° C for at least 1 month and/or stable at about 2-8° C for at least 1 year for at least 2 years.
  • the formulation is preferably stable following freezing (to, e.g., -70° C) and thawing of the formulation.
  • An antibody or antibody derivative "retains its physical stability" 'in a pharmaceutical formulation if it shows no signs of aggregation, precipitation and/or denaturation upon visual examination of color and/or clarity, or as measured by UV light scattering or by size exclusion chromatography.
  • An antibody or antibody derivative "retains its chemical stability" in a pharmaceutical formulation, if the chemical stability at a given time is such that the protein is considered to still retain its biological activity as defined below.
  • Chemical stability can be assessed by detecting and quantifying chemically altered forms of the protein.
  • Chemical alteration may involve size modification (e.g. clipping) which can be evaluated using size exclusion chromatography, SDS-PAGE and/or matrix-assisted laser desorption ionization/time-of- flight mass spectrometry (MALDI/TOP MS), for example.
  • Other types of chemical alteration include charge alteration (e.g. occurring as a result of deamidation) which can be evaluated by ion-exchange chromatography, for example.
  • An antibody or antibody derivative "retains its biological activity" in a pharmaceutical formulation, if the biological activity of the antibody at a given time is within about 10% (within the errors of the assay) of the biological activity exhibited at the time the pharmaceutical formulation was prepared as determined in an antigen binding assay, for example.
  • Other "biological activity” assays for antibodies are elaborated herein below.
  • isotonic is meant that the formulation of interest has essentially the same osmotic pressure as human blood. Isotonic formulations will generally have an osmotic pressure from about 250 to 350 mOsm. Isotonicity can be measured using a vapor pressure or ice-freezing type osmometer, for example.
  • a "polyol” is a substance with multiple hydroxyl groups, and includes sugars (reducing and non- reducing sugars), sugar alcohols and sugar acids. ' Preferred polyols herein have a molecular weight which is less than about 600 kD (e.g. in the range from about 120 to about 400 kD) .
  • a "reducing sugar” is one which contains a hemiacetal group that can reduce metal ions or react covalently with lysine and other amino groups in proteins and a "non-reducing sugar” is one which does not have these properties of a reducing sugar.
  • reducing sugars are fructose, mannose, maltose, lactose, arabinose, xylose, ribose, rhamnose, galactose and glucose.
  • Non-reducing sugars include sucrose, trehalose, sorbose, melezitose and raffinose .
  • Mannitol, xylitol, erythritol, threitol, sorbitol and glycerol are examples of sugar alcohols.
  • sugar acids these include L- gluconate and metallic salts thereof.
  • the polyol is preferably one which does not crystallize at freezing temperatures (e.g. -20° C) such that it destabilizes the antibody in the formulation.
  • Non-reducing sugars such as sucrose and trehalose are the preferred polyols herein, with trehalose being preferred over sucrose, because of the superior solution stability of trehalose.
  • buffer refers to a buffered solution that resists changes in pH by the action of its acid-base conjugate components.
  • the buffer of this invention has a pH in the range from about 4.5 to about • 6.0; preferably from about 4.8 to about 5.5; and most preferably has a pH of about 5.0.
  • buffers that will control the pH in this range include acetate (e.g. sodium acetate) , succinate (such as sodium succinate) , gluconate, histidine, citrate and other organic acid buffers: Where a freeze-thaw stable formulation is desired, the buffer is preferably not phosphate .
  • a “therapeutically effective amount” of an antibody or antibody derivative refers to an amount effective in the prevention or treatment of a disorder for the treatment of which the antibody or antibody derivative is effective.
  • a “disease/disorder” is any condition that would benefit from treatment with the antibody or antibody derivative. This includes chronic and acute disorders or diseases including those pathological conditions which predispose the mammal to the disorder in question.
  • a "preservative" is a compound which can be included in the formulation to essentially reduce bacterial action therein, thus facilitating the production of a multi-use formulation, for example.
  • potential preservatives include octadecyldimethylbenzyl ammonium chloride, hexamethonium chloride, benzalkonium chloride (a mixture of alkylbenzyldimethylammonium chlorides in which the alkyl groups are long-chain compounds) , and benzethonium chloride.
  • preservatives include aromatic alcohols such as phenol, butyl and benzyl alcohol, alkyl parabens such as methyl or propyl paraben, catechol, resorcinol, cyclohexanol, 3-pentanol, and m-cresol.
  • aromatic alcohols such as phenol, butyl and benzyl alcohol
  • alkyl parabens such as methyl or propyl paraben
  • catechol resorcinol
  • cyclohexanol 3-pentanol
  • m-cresol m-cresol
  • compositions comprising one or more antibodies or antibody derivative compounds, together with at least one physiologically acceptable carrier or excipient.
  • Pharmaceutical compositions may comprise, for example, one or more of water, buffers [e.g., neutral buffered saline or phosphate buffered saline) , ethanol, mineral oil, vegetable oil, dimethylsulfoxide, carbohydrates (e.g., glucose, mannose, sucrose or dextrans) , mannitol, proteins, adjuvants, polypeptides or amino acids such as glycine, antioxidants, chelating agents such as EDTA or glutathione and/or preservatives.
  • buffers e.g., neutral buffered saline or phosphate buffered saline
  • carbohydrates e.g., glucose, mannose, sucrose or dextrans
  • mannitol e.g., mannitol
  • proteins e.g., glucose, mannose, sucrose or dextrans
  • a carrier is a substance that may be associated with an antibody or antibody derivative prior to administration to a patient, often for the purpose of controlling stability or bioavailability of the compound.
  • Carriers for use within such formulations are generally biocompatible, and may also be biodegradable.
  • Carriers include, for example, monovalent or multivalent molecules such as serum albumin (e.g., human or bovine), egg albumin, peptides, polylysine and polysaccharides such as aminodextran and polyamidoamines .
  • Carriers also include solid support materials such as beads and microparticles comprising, for example, polylactate polyglycolate, poly (lactide-co-glycolide) , polyacrylate, latex, starch, cellulose or dextran.
  • a carrier may bear the compounds in a variety of ways, including covalent bonding (either directly or via a linker group) , noncovalent interaction or admixture.
  • compositions may be formulated for any appropriate manner of administration, including, for example, topical, oral, nasal, rectal or parenteral administration.
  • compositions in a form suitable for oral use are preferred. Such, forms include, for example, pills, tablets, troches, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsion, hard or soft capsules, or syrups or elixirs.
  • compositions provided herein may be formulated as a lyophilizate.
  • parenteral as used herein includes subcutaneous, intradermal, intravascular (e.g., intravenous), intramuscular, spinal, -intracranial, intrathecal and intraperitoneal injection, as well as any similar injection or infusion technique.
  • compositions intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and may contain one or more agents, such as sweetening agents, flavoring agents, coloring agent, and preserving agents in order to provide appealing and palatable preparations.
  • Tablets contain the active ingredient in admixture with physiologically acceptable excipients that are suitable for the manufacture of tablets.
  • excipients include, for example, inert diluents (e.g., calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate), granulating and disintegrating agents ⁇ e.g., corn starch or alginic acid), binding agents ⁇ e.g., starch, gelatin or acacia) and lubricating agents (e.g., magnesium stearate, stearic acid or talc) .
  • the tablets may be uncoated or they may be coated by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period.
  • a time delay material such as glyceryl monosterate or glyceryl distearate may be employed.
  • Formulations for oral use may also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent (e.g., calcium carbonate, calcium phosphate or kaolin) , or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium ⁇ e.g., peanut oil, liquid paraffin or olive oil) .
  • Aqueous suspensions contain the antibody or antibody derivative in admixture ⁇ with excipients suitable for the manufacture of aqueous suspensions .
  • excipients include suspending agents (e.g., sodium carboxymethylcellulose, methylcellulose, hydropropylmethylcellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia) ; and- dispersing or wetting agents (e.g., naturally-occurring phosphatides such as lecithin, condensation products of an alkylene 'oxide with fatty acids such as .
  • suspending agents e.g., sodium carboxymethylcellulose, methylcellulose, hydropropylmethylcellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia
  • dispersing or wetting agents e.g., naturally-occurring phosphatides such as lecithin, condensation products of an alkylene 'oxide with fatty acids such as .
  • polyoxyethylene stearate condensation products of ⁇ ethylene oxide with long chain aliphatic alcohols such as heptadecaethyleneoxycetanol, condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol such as polyoxyethylene sorbitol monooleate, or condensation products of ethylene oxide . 5. with partial esters derived- from fatty acids and hexitol anhydrides such as polyethylene sorbitan monooleate) .
  • Aqueous suspensions may also comprise one or more preservatives-, for example ethyl or n-propyl p- hydroxybenzoate, one or more coloring agents, one or more 0 flavoring agents, and one or more sweetening agents, such as sucrose or saccharin.
  • preservatives- for example ethyl or n-propyl p- hydroxybenzoate
  • coloring agents such as glycerol, propylene glycol, sorbitol, or sucrose.
  • sweetening agents such as sucrose or saccharin.
  • Syrups and elixirs may be formulated with sweetening agents, such as glycerol, propylene glycol, sorbitol, or sucrose.
  • Such formulations may also comprise one or more demulcents, preservatives, 5 flavoring agents, and/or coloring agents.
  • Oily suspensions may be formulated by suspending the active ingredients in a vegetable oil (e.g., arachis oil, olive oil, sesame oil, or coconut oil) or in a mineral oil such as liquid paraffin.
  • the 0 oily suspensions may contain a thickening agent such as beeswax, hard paraffin, or cetyl alcohol.
  • Sweetening agents, such as those set forth above, and/or flavoring agents may be added to provide palatable oral preparations.
  • Such suspensions may be preserved by the 5 addition of an anti-oxidant such as ascorbic acid.
  • Dispersible powders and granules suitable for preparation of an • aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent,- suspending agent and one or 0 more. preservatives .
  • Suitable dispersing or wetting agents and suspending agents are exemplified by those already mentioned above. Additional excipients, for example sweetening, flavoring 'and coloring agents, may also be present.
  • Pharmaceutical compositions may also be in the form of oil-in-water. emulsions.
  • the oily phase may be a vegetable oil ⁇ e.g., olive oil " or arachis oil), a mineral oil ⁇ e.g., liquid paraffin), or a mixture thereof.
  • Suitable emulsifying agents include naturally- occurring gums ⁇ e.g., gum acacia or gum tragacanth) , naturalIy-occurring phosphatides ⁇ e.g. , soy bean, lecithin, and esters or partial esters derived from fatty- acids and hexitol), anhydrides ⁇ e.g., sorbitan monoleate) , and condensation products of partial esters derived from fatty acids and hexitol with ethylene oxide (e.g., polyoxyethylene sorbitan monoleate) .
  • An emulsion may also comprise one or more sweetening and/or flavoring agents .
  • the pharmaceutical composition may be prepared as a sterile injectible aqueous or oleaginous suspension in which the modulator, depending on the vehicle and concentration used, is either suspended or ⁇ • dissolved in the vehicle.
  • a composition may be formulated according to the known art using suitable dispersing, wetting agents and/or suspending agents such as those mentioned above.
  • suitable dispersing, wetting agents and/or suspending agents such as those mentioned above.
  • the acceptable vehicles and solvents that may be employed are water, 1,3- butanedi-ol, Ringer's solution and isotonic sodium chloride solution.
  • sterile, fixed oils may be employed as a solvent or suspending medium.
  • any bland fixed oil may be employed, including synthetic mono- or diglycerides .
  • compositions may be formulated as sustained release formulations (i.e., a formulation such as a capsule that effects a slow release of modulator following administration) .
  • sustained release formulations i.e., a formulation such as a capsule that effects a slow release of modulator following administration
  • Such formulations may generally be prepared using well known technology and administered by/ for example, oral, rectal, or subcutaneous implantation, or by implantation at the desired target site.
  • Carriers for use within such formulations are biocompatible, and may also be biodegradable; preferably the formulation provides a relatively constant level of modulator release.
  • the amount of an antibody or antibody derivative contained within a sustained release formulation depends upon, for example, the site of implantation, the rate and expected duration of release and the nature of the disease/disorder to be treated or prevented.
  • Antibody or antibody derivatives provided herein are generally administered in an amount that achieves a concentration in a body fluid (e.g., blood, plasma, serum, CSF, synovial fluid, lymph, cellular interstitial fluid, tears or urine) that is sufficient to detectably bind to TWF ⁇ and prevent or inhibit TNF ⁇ associated diseases/disorders.
  • a dose is considered to be effective if it results in a discernible patient benefit as described herein.
  • Preferred systemic doses range from about 0.1 mg to about 140 mg per kilogram of body weight per day (about 0.5 mg to about 7 g per patient per day), with oral doses generally being about 5-20 fold higher than intravenous doses.
  • the amount of antibody or antibody derivative that may be combined with the carrier materials to produce a single dosage form will vary depending upon the host treated and the particular mode of administration. Dosage unit forms will generally contain between from about 1 mg to about 500 mg of an active ingredient.
  • compositions may be packaged for treating conditions responsive to an antibody or antibody derivative directed to TNF- ⁇ .
  • Packaged pharmaceutical compositions may include a container holding a effective amount of at least one antibody or antibody derivative as described herein and instructions (e.g., labeling) indicating that the contained composition is to be used for treating a disease/disorder responsive to one antibody or antibody derivative following administration in the patient.
  • the antibodies or antibody derivatives of the present invention can also be chemically modified.
  • Preferred modifying groups are polymers, for example an optionally substituted straight or branched chain polyalkene, polyalkenylene, or polyoxyalkylene polymer or a branched or unbranched polysaccharide. Such effector group may increase the half-live of the antibody in vivo.
  • Particular examples of synthetic polymers include optionally substituted straight or branched chain poly (ethyleneglycol) (PEG), poly (propyleneglycol) , poly (vinylalcohol) or derivatives thereof.
  • Particular naturally occurring polymers include lactose, amylose, dextran, glycogen or derivatives thereof.
  • the size of the polymer may be varied as desired, but will generally be in an average molecular weight range from 500Da to 50000Da. For local application where the antibody is designed to penetrate tissue, a preferred molecular weight of the polymer is around 5000Da.
  • the polymer molecule can be attached to the antibody, in particular to the C-terminal end of the Fab fragment heavy chain via a covalently linked hinge peptide as described in WO0194585.
  • PEG moieties reference is made to "Poly (ethyleneglycol) Chemistry, Biotechnological and Biomedical Applications", 1992, J. Milton Harris (ed) , Plenum Press, New York and
  • the pharmaceutical formulation comprising it is prepared.
  • the antibody to be formulated has not been subjected to prior lyophilization and the formulation of interest herein is an aqueous formulation.
  • the antibody or antibody derivative in the formulation is an antibody fragment, such as an scFv.
  • the therapeutically effective amount of antibody present in the formulation is determined by taking into account the desired dose volumes and mode(s) of administration, for example. From about 0.1 mg/ml to about 50 mg/ml, preferably from about 0.5 mg/ml to about 25 mg/ml and most preferably from about 2 mg/ml to about 10 mg/ml is an exemplary antibody concentration in the formulation.
  • An aqueous formulation comprising the antibody or antibody derivative in a pH-buffered solution
  • the buffer of this invention has a pH in the range from about 4.5 to about 6.0, preferably from about 4.8 to about 5.5, and most preferably has a pH of about 5.0.
  • buffers that will control the pH within this range include acetate (e.g. sodium acetate), succinate (such as sodium succinate) , gluconate, histidine, citrate and other organic acid buffers.
  • the buffer concentration can be from about 1 mM to about 50 ⁇ iM, preferably from about 5 mM to about 30 mM, depending, for example, on the buffer and the desired isotonicity of the formulation.
  • the preferred buffer is sodium acetate (about 10 mM) , pH 5.0.
  • a polyol which acts as a tonicifier and may stabilize the antibody, is included in the formulation.
  • the formulation does not contain a tonicifying amount of a salt such as sodium chloride, as this may cause the antibody or antibody derivative to precipitate and/or may result in oxidation at low pH.
  • the polyol is a non- reducing sugar, such as sucrose or trehalose. The polyol is added to the formulation in an amount which may vary ⁇ with respect to the desired isotonicity of the formulation.
  • the aqueous formulation is isotonic, in which case suitable concentrations of the • polyol in the formulation are in the range from about 1% to about 15% w/v, preferably in the range from about 2% to about 10% whv, for example.
  • suitable concentrations of the • polyol in the formulation are in the range from about 1% to about 15% w/v, preferably in the range from about 2% to about 10% whv, for example.
  • hypertonic or hypotonic formulations may also be suitable.
  • the amount of polyol added may also alter with respect to the molecular weight of the polyol. For example, a lower amount of a monosaccharide (e.g. mannitol) may be added, compared to a disaccharide (such as trehalose) .
  • a surfactant is also added to the antibody or antibody derivative formulation.
  • Exemplary surfactants include nonionic surfactants such as polysorbates (e.g.
  • the amount of surfactant added is such that it reduces aggregation of the formulated antibody/antibody derivative and/or minimizes the formation of particulates in the formulation and/or reduces adsorption.
  • the surfactant may be present in the formulation in an amount from about 0.001% to about 0.5%, preferably from about 0.005% to about 0.2% and most preferably from about 0.01% to about 0.1%.
  • the formulation contains the above-identified agents (i.e. antibody or antibody derivative, buffer, polyol and surfactant) and is essentially free of one or more preservatives, such as benzyl alcohol, phenol, m-cresol, chlorobutanol and benzethonium Cl.
  • a preservative may be included in the formulation, particularly where the formulation is a multidose formulation.
  • the concentration of preservative may be in the range from about 0.1% to about 2%, most preferably from about 0.5% to about 1%.
  • One or more other pharmaceutically acceptable carriers, excipients or stabilizers such as those described in Remington's Pharmaceutical Sciences 21st edition, Osol, A. Ed.
  • Acceptable carriers, excipients or stabilizers are nontoxic to recipients at the dosages and concentrations employed and include,- additional buffering agents; co- solvents, • antioxidants including ascorbic acid and methionine; chelating agents such- as EDTA; metal complexes (e.g. Zn-protein complexes); biodegradable polymers such as polyesters; and/or salt-forming counterions such as sodium.
  • the formulations to be used for in vivo administration must be sterile. This is readily accomplished by filtration through sterile filtration membranes, prior to, or following, preparation of the formulation.
  • the formulation is administered to a mammal in need of treatment with the antibody, preferably a human, in accord with known methods, such as intravenous administration as a bolus or by continuous infusion over a period of time, by intramuscular, intraperitoneal, intracerobrospinal , subcutaneous, intra-articular, intrasynovial , intrathecal, oral, topical, or inhalation routes.
  • the formulation is administered to the mammal by intravenous administration.
  • the formulation may be injected using a syringe or via an IV line, for example.
  • the appropriate dosage (therapeutically effective amount) of the antibody will depend, for example, on the condition to be treated, the severity and course of the condition, whether the antibody is administered for preventive or therapeutic purposes, previous therapy, the patient's clinical history and response to the antibody, the type of antibody used, and the discretion of the attending physician.
  • the antibody or antibody derivative is suitably administered to the patent at one time or over a series of treatments and may be administered to the patent at any time from diagnosis onwards.
  • the antibody or antibody derivative may be administered as the. sole treatment or in conjunction with other, drugs or therapies useful in treating the condition in question.
  • the therapeutically effective amount of the antibody or antibody derivative administered will be in the range of about 0.1 to about 50 mg/kg of patent body weight whether by one or more administrations, with the typical range of antibody used being about 0.3 to about 20 mg/kg, more preferably about 0.3 to about 15 mg/kg, administered daily, for example.
  • the typical range of antibody used being about 0.3 to about 20 mg/kg, more preferably about 0.3 to about 15 mg/kg, administered daily, for example.
  • other dosage regimens may be useful. The progress of this therapy is easily monitored by conventional techniques .
  • an article of manufacture comprising a container which holds the aqueous pharmaceutical formulation of the present invention and optionally provides instructions for its use.
  • Suitable containers include, for example, bottles, vials and syringes.
  • the container may be formed from a variety of materials such as glass or plastic.
  • An exemplary container is a 3-20 cc single use glass vial.
  • the container may be 3-100 cc glass vial.
  • the container holds the formulation and the label on, or associated with, the container may indicate directions for use.
  • the article of manufacture may further include other materials desirable from a commercial and user standpoint , including other buffers, diluents, filters, needles, syringes, and package inserts with instructions for use.
  • the antibodies or antibody derivatives of the present invention may be generated using routine techniques in the field of recombinant genetics. Knowing the sequences of the polypeptides, the cDNAs encoding them can be generated by gene synthesis (www . genscript . com) . These cDNAs can be cloned into suitable vector plasmids. Once the DNA encoding a VL and/or a VH domain are obtained, site directed mutagenesis, for example by PCR using mutagenic primers, can be performed to obtain various derivatives. The best "starting" sequence can be chosen depending on the number of alterations desired in the VL and/or VH sequences. A preferred sequence is the TB-A sequences and its derivatives, e.g. scFv sequences or Fab fusion peptide sequences, may be chosen as templates for PCR driven mutagenesis and/or cloning.
  • the antibodies of the present invention comprise the disclosed sequences rather than they consist of them.
  • cloning strategies may require that a construct is made from which an antibody with one or a few additional residues at the N-terminal end are present.
  • the methionine derived from the start codon may be present in the final protein in cases where it has not been cleaved posttranslationally .
  • Most of the constructs for scFv antibodies give rise to an additional alanine at the N-terminal end.
  • an expression vector for periplasmic expression in E. coli is chosen (Krebber, 1997) .
  • Said vector comprises a promoter in front of a cleavable signal sequence.
  • the coding sequence for the antibody peptide is then fused in frame to the cleavable signal sequence. This allows the targeting of the expressed polypeptide to the bacterial periplasm where the signal sequence is cleaved.
  • the antibody is then folded.
  • both the VL-CK and the VH-CHl fusions peptides must be linked to an export signal.
  • the covalent S-S bond is formed at the C- terminal cysteines after the peptides have reached the periplasm.
  • cytoplasmic expression of antibodies is preferred, said antibodies usually can be obtained at high yields from inclusion bodies, which can be easily separated from other cellular fragments and protein.
  • inclusion bodies are solubilized in a denaturing agent such as e.g. guaridine hydrochloride (GndHCl) and then refolded by renaturation procedures well known to those skilled in the art.
  • a denaturing agent such as e.g. guaridine hydrochloride (GndHCl)
  • Plasmids expressing the scFv or Fab polypeptides are introduced into a suitable host, preferably a bacterial, yeast or mammalian cell, most preferably a suitable E. coli strain as for example JM83 for periplasmic expression or BL21 for expression in inclusion bodies.
  • the polypeptide can be harvested either from the periplasm ox form inclusion bodies and purified using standard technique ' s such as ion exchange chromatography, reversed phase chromatography, affinity chromatography and/or gel filtration known to the person skilled in the art.
  • the antibodies or antibody derivatives of the present invention can be characterized with respect to yield, solubility and stability in vitro. Binding capacities towards TNF ⁇ , preferably towards human TNFa 7 can be tested in vitro by ELISA or surface plasmon resonance (BIACore) , using recombinant human TNF ⁇ as described in WO9729131, the latter method also allowing to determine the k Off rate constant, which should preferably be less than 10 "3 S "1 . Kd values of, ⁇ IO nM are preferred. In vivo neutralizing activity of an antibody or antibody derivative of the present invention can be estimated using the L929 cytotoxicity assay.
  • TNF ⁇ Human recombinant TNFot exerts a cytotoxic effect towards cultured mouse L929 fibroblast cells in a concentration- dependent manner. This TNF ⁇ -induced cytotoxicity can be . inhibited by TNF ⁇ neutralizing antibodies (D ⁇ ring, 1994) . A preferred IC50 value corresponding to a half-maximal inhibitor concentration is ' ⁇ IOO ng ml "1 .
  • TNF ⁇ has a proven pathophysiological role in various human diseases, in particular inflammatory disorders, immune and immune-regulated disorders, infections causing septic, endotoxic and cardiovascular shock, neurodegenerative diseases, and malignant diseases.
  • the antibodies or antibody derivatives of the present invention can be applied to treat the diseases listed in the following catalogue, which is not to be considered as a complete or exclusive list.
  • Other diseases not mentioned specifically, which directly or indirectly are influenced by TNF ⁇ , are also included.
  • Autoimmune or chronic inflammation Chronic and/or autoimmune states of inflammation in general, immune mediated inflammatory disorders in general, inflammatory CNS disease, inflammatory diseases affecting the eye, joint, skin, mucuous membranes, central nervous system, gastrointestinal tract, urinary tract or lung, states of .
  • uveitis in general, retinitis, HLA-B27+ ' uveitis, Behcet's disease, dry eye syndrome, glaucoma, Sjogren syndrome, diabetes mellitus (incl.
  • diabetic neuropathy insulin resistance
  • states of arthritis in general rheumatoid arthritis, • osteoarthritis, reactive arthritis and Reiter' s syndrome, juvenile arthritis, ankylosing spondylitis, multiple sclerosis, Guillain-Barre syndrome, myasthenia gravis, amyotrophic lateral sclerosis, sarcoidosis, glomerulonephritis, chronic kidney disease, cystitis, Psoriasis (incl.
  • extraintestinal manifestastations ulcerative colitis
  • asthma bronchiale hypersensitivity pneumonitis
  • general allergies allergic rhinitis, allergic sinusitis, chronic obstructive pulmonary disease (COPD)
  • COPD chronic obstructive pulmonary disease
  • lung fibrosis ' " Wegener's granulomatosis, Kawasaki syndrome, Giant cell arteritis, Churg-Strauss vasculitis, polyarteritis nodosa, burns, graft versus host disease, host versus graft reactions, rejection episodes following organ or bone marrow -transplantation, sytemic and local states of vasculitis in general, systemic and discoid lupus erythematodes, polymyositis and dermatomyositis, sclerodermia, pre-eclampsia, acute and chronic pancreatitis, viral hepatitis, alcoholic hepatitis.
  • eye surgery e.g. cataract (eye lens replacement) or glaucoma surgery
  • joint surgery incl. arthroscopic surgery
  • surgery at. joint-related structures e.g. ligaments
  • oral and/or dental surgery minimally invasive cardiovascular procedures (e.g. PTCA, atherectomy, stent placement)
  • laparoscopic and/or endoscopic intra-abdominal and gynecological procedures ⁇ endoscopic urological procedures (e.g. prostate surgery, ureteroscopy, cystoscopy, interstitial cystitis)
  • perioperative inflammation prevention in general.
  • Neurological and neurodegenarative diseases Alzheimer disease, Parkinson's disease, Huntington' s disease, Bell' palsy, Creutzfeld-Jakob disease.
  • Cancer-related osteolysis Cancer-related inflammation, cancer-related pain, cancer-related cachexia, bone metastases.
  • Acute and chronic forms of pain irrespective whether these are caused by central or peripheral effects of TNF ⁇ and whether they are classified as inflammatory , nociceptive or neuropathic forms of pain, sciatica, low back pain, carpal tunnel syndrome, complex regional pain syndrome (CRPS) , gout, postherpetic neuralgia, fibromyalgia, local pain states, chronic pain syndroms due to metastatic tumor, dismenorrhea.
  • CRPS complex regional pain syndrome
  • Atherosclerosis coronary artery disease, hypertension, dyslipidemia, heart insufficiency and chronic heart failure.
  • treatment of osteoarthritis or uveitis or inflammatory bowel disease can be achieved with the antibodies, or antibody derivatives of the present invention.
  • the present invention also provides a pharmaceutical composition
  • a pharmaceutical composition comprising an antibody or antibody derivative molecule of the present invention in combination with a pharmaceutically acceptable excipient, diluent ' or carrier.
  • the pharmaceutical composition should preferably comprise a therapeutically effective amount .of the antibody of the present invention, i.e. an amount of said antibody that is needed to treat, ameliorate or prevent the TNF ⁇ -related disease or condition, or to exhibit a detectable therapeutic or preventive effect.
  • the therapeutically effective dose can be estimated 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.
  • a suitable animal model to observe an effect of the antibody or antibody derivative of the present invention is a rat model for acute monoarthritis (Bolon et al . (2004), Vet. Pathol .41 : 235-243.
  • Human TNF ⁇ is injected intraarticularly into the knee joint of a rat, leading to an acute, self-limiting monoarthritis in the injected joint.
  • Bioactivity of an anti-TNF ⁇ antibody can be quantified by reduction of TNF ⁇ - induced knee joint swelling and or reduction of histological parameters of inflammation.
  • antibodies or antibody derivatives of the present invention are highly soluble, high antibody concentrations (60 mg ml "1 or more) enable the use of small application volumes.
  • the antibody or antibody derivative of the present invention may be utilised in any therapy where it is desired to reduce the level of biological active TNF ⁇ present in the human or animal body.
  • the TNF ⁇ may be circulating in the body or be present at an undesirably high level localised at a particular site in the body.
  • the present invention provides modes for systemic as well as local applications in general, which include, but are not limited to the following ones: peroral application, intravenous, subcutaneous, intramuscular, intraarticular, intravitreal, intradermal, or intraparenchymal injection, aerosol inhalation, topical application to the skin, to mucous membranes or to eye, systemic or local release via implantable minipump or local release via implantable formulation/device allowing for retarded release, topical application to serosal surfaces, intrathecal or intra- ventricular application, oral application in formulations allowing for controlled intralumenal release in selected parts of the gastrointestinal tract, localized intravasal release from adequate formulation/devices (e.g.
  • a preferred application is a local one such as intraarticular injection or topic application e.g. into the eye.
  • the antibody of the present invention needs to-be in solution.
  • the present invention also reveals the use of the antibody or antibody derivative of the present invention for the production of a medicament for the treatment of TNF ⁇ associated diseases.
  • the antibody or antibody derivative is comprised in a therapeutic composition.
  • Said composition is used as a medicament, most preferably for the prevention or therapy of TNF ⁇ related diseases .
  • the scFv antibodies of the present invention are used in gene therapy, in particular in adoptive cellular gene therapy.
  • Autoimmune disorders represent inappropriate immune responses directed at self-tissue.
  • Antigen-specific CD4+ T cells and antigen- presenting dendritic cells (DCs) are important mediators ' in the pathogenesis of auto-immune disease and thus are ideal candidates for adoptive cellular gene therapy, an ex vivo approach to therapeutic gene transfer.
  • DCs dendritic cells
  • T cells T cell hybridomas, and DCs rapidly and preferentially , home to the sites of inflammation in animal models of multiple sclerosis, arthritis, and diabetes.
  • These cells transduced with retroviral vectors that drive expression of various "regulatory proteins" such as interleukins and anti-TNF scFv, deliver these immunoregulatory proteins to the inflamed lesions, providing therapy for experimental autoimmune encephalitis, collagen-induced arthritis, and nonobese diabetic mice.
  • the stable and soluble frameworks of the antibodies or antibody derivatives of the present inventions are particularly suitable for intracellular • delivery of the antigen, for example when the antibody or- antibody derivative is expressed from a transgene carried by a suitable retroviral vector.
  • scFvs derived from a TNF ⁇ neutralizing monoclonal antibody can neutralize TNF ⁇ in vitro and (ii) change the cytokine expression pattern in mice suffering from collagen-induced arthritis locally in the joints, but not systemically .
  • suitable vectors e.g. viruses
  • sequences of the present invention are the following ones:
  • SEQ ID NOrI8 TB-A H_K43Q also named TB-A H43 DIVMTQSPSSLSASVGDRVTLTCTASQSVSNDWWYQQRPGKAPK LLIYSAFNRYTGVPSRFSGRGYGTDFTLTISSLQPEDVAVYYCQQDYNSPRTFGQGT KLEVKRGGGGSGGGGSGGGGSGGGGSQVQLVQSGAEVKKPGASVKVSCTASGYTFTH YGMNWVRQAPGQGLEWMGWINTYTGEPTYADKFKDRFTFSLETSASTVYMELTSLTS DDTAVYYCARERGDAMDYWGQGTLVTVSS
  • DDTAVYYCARERGDAMDYWGQGTLVTVSS SEQ ID NO.-20 TB-A H_F70L/L72R, also named TB-A H70/72
  • the starting material for the generation of humanised anti-human TNF ⁇ antibodies or antibody ⁇ derivatives, such as single-chain fragments (scFv) or Fab fragments was the murine monoclonal antibody Di62.
  • Di62 inhibits human TNF ⁇ - induced cytotoxicity in mouse L929 cells.
  • Di62 Based on its published sequence Di62 was constructed in the form of a single-chain antibody derivative (scFv) in the orientation VL ⁇ linker-VH, in which the linker sequence is composed of four repeats of four glycine and one serine residue (Gly 4 Ser) 4 .
  • this scFv is referred to as TB-wt, with a VL of SEQ ID NO: 16 and a VH of SEQ ID NO: 17.
  • this antibody derivative for the purpose of a) render it more similar to human sequences . in order to minimise potential immunogenicity, and b) render it more stable and soluble, TB-wt CDR sequences were grafted on stable and soluble human frameworks (Au-f der Maur et al . (2001), FEBS Lett. 508:407-412; Auf der Maur et al . (2004), Methods 34:215-224).
  • the human VL- kappa subgroup I and VH subgroup I were identified as nearest human subfamily.
  • acceptor framework was chosen from a pool of human VL and VH sequences, selected for advantageous biochemical and biophysical properties, such as for example stability, solubility, and expression properties (Auf der Maur, et al. (2001), FEBS Lett. 508:407-412; Auf der Maur, et al . (2004), Methods 34:215-224).
  • the isolation and properties of these antibody frameworks are described in WO03097697/EP1506236. From this pool, a single-chain antibody framework with undefined antigen binding properties was identified as suitable acceptor.
  • This acceptor consists of a human VL-kappa I domain (SEQ ID NO: 14) in combination with a human VHI domain (SEQ ID NO: 15) .
  • this acceptor framework is referred to as FW2.3.
  • FW2.3 this acceptor framework
  • TB-wt and FW2.3 share 55 identical amino acid residues, which amounts to 67% identity.
  • TB-wt and FW2.3 have 55 identical residues, corresponding to 63% identity.
  • Both single-chain antibody derivatives have identical CDR lengths, apart from the VH-CDR3, which is longer in FW2.3.
  • the minimal approach namely the conservative transfer of all mouse CDR loops from TB-wt onto FW2.3 was carried out first.
  • the resul- ting scFv is- referred to as TB-B and has the VL sequence of SEQ ID NO: 3 and the VH sequence of SEQ ID NO: 4.
  • the CDR-loops in TB-wt were defined according to the Rabat' ⁇ numbering scheme (Rabat et al . (1991), Sequences of Proteins of Immunological Interest, 5 th Ed , Natl. Inst. Health, Bethesda, MD) and confine the following residues (see Figure 1) : .VL
  • CDRl L24-L34 (same Kabat numbering)
  • CDR2 L50-L56 (same Kabat numbering)
  • CDR3 L89-L97 (same Kabat numbering)
  • VH CDRl: H31-H35 (same Kabat numbering)
  • CDR2 H50-H66 (Kabat numbering H50-H65)
  • CDR3 H99-H106 (Kabat numbering H95-H102) .
  • This antibody was unable to bind TNF ⁇ efficiently (Fig. 4A) .
  • the next step was to determine which residue or residues from these components- should be substituted to optimise the properties of the resulting humanised antibody.
  • TB-B L46 VL SEQ ID NO: 11; VH SEQ ID NO: 4
  • This variant is based on TB-B and contains one single amino acid change at position 46 in VL, namely R- ⁇ L. This amino acid is located within the upper core of the light chain and takes part in the dimer interface.
  • the first variant is composed of the VL domain from TB-A connected via a glycine serine linker (SEQ ID NO: 10) with the VH domain from TB-B, resulting in TB-AB (SEQ ID NO: 12) .
  • the second variant, TB-BA is the reverse of the first variant, namely, the VL domain from TB-B in combination with the VH domain of TB-A (SEQ ID NO: 13) .
  • FIG. 1 shows a sequence comparison of the VL and VH sequences of TB-A and TB-B.
  • a total of 14 framework residues differ between TB-A and TB-B (asterisks) . Only five of them, VL residues 4 and 70, and VH residues 28, 71, and 73 show only minor differences in size and property and therefore were not considered for mutagenesis at this point.
  • the following framework positions- were replaced with the corresponding amino acid from TB-B.
  • TB-A H43 K->Q interface (SEQ ID NO: 18) TB-A H68 F->V outer loop VH (SEQ ID NO: 19) TB-A H70/72 F->L,L->R outer loop VH (SEQ ID NO: 20) TB-A H76/77 A->I,S->G outer loop VH (SEQ ID NO:21) TB-A L46 L->R interface (SEQ ID NO:22) TB-A L65 R->S outer loop VL (SEQ ID NO: 23) TB-A L67 Y->S outer loop VL (SEQ ID NO: 24)
  • TB_L2 SEQ ID N0:7
  • TB_L3 SEQ ID NO:8
  • TB_H2 SEQ ID NO:9
  • the cDNAs encoding the murine single-chain version of the monoclonal antibody Di62, and the two humanised versions TB-B and TB-A were generated by gene synthesis (www.genscript.com). All the point mutations in the other variants (TB-B L46, TB-A H43, TB-A H67, TB-A H69/71, TB-A H75/76, TB-A L46, TB-A L65, TB-A L67, TB-A V83F, TB-A V83A, TB-A D66G) were introduced by PCR-driven site-directed mutagenesis following standard cloning procedures.
  • TB-A Some further preferred TB-A are disclosed in SEQ ID NO: 31 to SEQ ID NO: 38. These antibodies were found to be particularly stable and soluble, as shown below in Table I. All scFv fragments were cloned into an expression vector for periplasmic production in E. coli (Krebber et al . (1997), J. Immunol. Methods 201:35-55).
  • the corresponding Fab fragments were generated as follows .
  • Selected light chain variable domains (VL) were fused to the constant region of a human Ig kappa chain, while the suitable heavy chain variable domains (VH) were fused to the first (N-terminal) constant domain (CHl) of a human IgG.
  • Both human constant domains were amplified by PCR from a human spleen cDNA library and resulted in the sequence SEQ ID NO: 14 for CK- and SEQ ID NO: 15 for CHl.
  • Plasmids encoding TB-wt, its humanised derivatives, or Fab fragments were introduced into a suitable E. coli strain (e.g. JM83) for periplasmic expression.
  • the scFv variants were also expressed as inclusion bodies, for example in the BL21 E. coli strain.
  • Functional single-chain antibodies were obtained by refolding of inclusion bodies and subsequent purification by, for example, gel filtration.
  • the expression yields upon periplasmic expression of the scFvs ranged between 0.5 mg up to 12 mg per litre culture under standard laboratory cultivation conditions (dYT medium, with approx. 3 hours induction- time at 30°C, shaking at 200 rpm) with conventional ' . shaking flasks.
  • 3B shows the superiority of the framework TB-A over the donor framework (TB-wt) in terms of solubility in phosphate buffered saline.
  • TB-A m ⁇ igrates predominantly in a monomer state (peak at 70ml) whereas TB-wt shows a strong tendency to form aggregates (peak at 50ml) .
  • maximal solubility of TB-A and certain derivatives thereof was assessed by PEG precipitation (Athat .DH.. et al . JBC. 1981, 256;23. 12108-12117).
  • test proteins were measured in the presence of polyethylene glycol 3000 (PEG3000) .
  • Solubility curves were determined by measuring protein concentration in the supernatant of centrifuged protein-PEG300 mixtures. All curves showed a linear dependence of log S (mg/ml) on PEG3000 concentration (%, w/v) . Maximal solubility (S ma ⁇ ) ⁇ of a test protein was estimated by extrapolation of the linear regression towards 0% PEG (Tab. I) . For TB-A was S max calculated to be about 70 mg/ml. All test proteins showed exceptionally good solubility.
  • SIC self-interaction -chromatography
  • the protein osmotic second virion coefficient B 22 which is a measure for intermolecular attraction/repulsion, was calculated according to Tessier, PM et al .. Biophys . J. 2002, 82: 1620-1632 (Tab. I) .
  • Protein stability of TB-A, TB-A H_M48L/F68I (SEQ ID NO: 31), TB-A Linker_G2R H_F68L (SEQ ID N0:33), TB-A H_K43R/F68I (SEQ ID NO:34) and TB-A H_F68L (SEQ ID NO: 35) was assessed by determining the temperature for onset of unfolding by circular dichroism and light scattering at both 218 and 292 nm (Tab. II) .
  • TB-A started to unfold at a temperature of 5-3 0 C whereas its derivatives TB-A H_M48L/F68I (SEQ ID NO:31), TB-A Linker G2R H F68L (SEQ ID NO:33), TB-A H__K43R/F68I (SEQ ID NO:34) and TB-A H_F68L (SEQ ID NO:35) showed increased thermal stability (56 and 58°C) . All test proteins showed irreversible denaturation and precipitated upon unfolding, making it impossible to ⁇ determine the melting temperature.
  • TB-A The stability of TB-A in human serum, human urine, pig vitreous body fluid and pig anterior chamber fluid was assayed by measuring TNF ⁇ binding activity of TB-A after incubation for 3 days at 37 0 C in the respective body fluid or in assay buffer (TBSTM) as a positive control.
  • TBSTM assay buffer
  • TB-A was diluted in body fluids to a final concentration of 10 ⁇ M. After the incubation period dilution series of the samples were assayed by ELISA in order to determine the binding constant Kd of TB-A (Fig. 11) .
  • a shift of the K d towards higher concentrations would indicate a decrease of active protein during the incubation period. In our experiments, however, no such shift was detectable, indicating that the amount of fully active TB-A remained constant in every body fluid assay due to a high stability of the antibody. _ . .. . ..
  • TB-A represents a good compromise between the apparent trade-off of expression yield and affinity. In terms of affinity, no significant difference between the single-chain and the Fab fragment format of TB-A was detectable (data not shown) .
  • the function ' of antibodies or antibody derivatives in neutralising TNF ⁇ in vivo can be tested by measuring inhibition of cytotoxicity of TNF ⁇ towards cultured mouse L929 fibroblasts or alternatively towards human KYM-I myelosarcoma cells (Tab. Ill) .
  • the humanised scFv derivatives of Di62 display different efficacies in the L929 assay as shown in Fig. 5B. Some scFv derivatives show IC50 (inhibitory concentration to achieve 50% inhibition) values in the range of 5ng/ml, whereas others had no effect in the L929 assays. ELISA data and results from the L929 assay do not always correlate.
  • KYM-I data and L929 results correlated nicely with the only difference that much higher concentrations of recombinant human TNF and consequently also of the antagonist where required to see an effect.
  • KYM-I was, therefore, mainly used to confirm L929 results.
  • potency is expressed as a relative value normalized to TB-A (EC 50 X / EC 50 TB-A) .
  • IC50 values become again higher the closer the sequence of a binder is to the human acceptor framework (FW2.3).
  • Fig. 5 compares the potency of different derivatives of TB-A to block TNF ⁇ induced cytotoxicity towards mouse L929 fibroblast cells. Absorption at 450nm correlates with cell survival.
  • TB-A and the anti-hTNF ⁇ IgG Infliximab® show a similar IC 50 value in the L929 assay, whereas the potency of TB-wt to block human TNF ⁇ induced cytotoxicity is significantly lower (Fig. 5A).
  • TB-A derivatives are compared with TB-A for their potential to block TNF ⁇ induced cytotoxicity, most of these derivatives except TB-H43 have a .reduced efficacy in the L929 (Fig. 5B) .
  • Table III Functional properties of TB-A derivatives
  • Cytokine and antibody instillation Anaesthesia and cytokine injections were performed as described by Bolon and colleagues .
  • cytokines and antibody derivatives were instilled in two separate intra-articular injections whereby the 10 micorgrams of recombinant human TNF ⁇ was injected in 10 microliter of filter-sterilised phosphate- buffered saline (PBS) and the respective dose of the respective antibody was injected in 40 microliter of filter-sterilised phosphate-buffered saline.
  • Animals treated with intraperitoneally applied Infliximab/ Remicade® were i.p.
  • Recombinant human TNF ⁇ was purchased from PeproTech EC Ltd. Measurement of joint diameter
  • the diameter of the knee joint to be injected was determined by means of a standard calliper. 48 hours after injection of TNF ⁇ (or PBS in control animals) and immediately before euthanisation of the animals, the diameter of the injected knee joint was determined again and joint swelling was calculated by subtracting the value of the second diameter measurement from the value of the first diameter measurement (Fig. 6 and 9) . Tissue processing ⁇ 48 hours after injection of TNF ⁇ (or PBS in case of control animals) animals were euthanised.
  • injected knees were separated from the foot and thigh, fixed intact by immersion in 70% ethanol and proceeded for standard hematoxylin and eosin (HE) staining, as described by Bolon and colleagues.
  • HE hematoxylin and eosin
  • Histological scoring analysis for measurement of joint inflammation was performed as described by Bolon and colleagues. Histopathological scoring criteria for assessment of joint inflammation were applied according to Bolon and colleagues (Fig. 7, 8 and 10) .
  • Score 2 Thickening of synovial lining and mild inflammation of the sublining
  • Score 3 Thickening of synovial lining and moderate inflammation of the sublining
  • both the representative ESBATech anti-TNF ⁇ scFv and the representative ESBATech anti-TNF ⁇ Fab antibody were highly efficient in blocking human TNF ⁇ -induced monoarthritis upon local (intraarticular) administration .

Abstract

The present invention relates to particularly stable and soluble scFv antibodies and Fab fragments specific for TNFα, which comprise specific light chain and heavy chain sequences that are optimized for stability, solubility, in vitro and in vivo binding of TNFα, and low immunogenicity. Said antibodies are designed for the diagnosis and/or treatment of TNFα-related disorders. The nucleic acids, vectors and host cells for expression of the recombinant antibodies of the invention, methods for isolating them and the use of said antibodies in medicine are also disclosed.

Description

Stable and soluble antibodies inhibiting TNFoc
Technical Field
The present invention relates to optimised antibodies and antibody derivatives that bind to and block the function of tumour necrosis factor alpha (TNFα) and are useful for the diagnosis and/or treatment, prevention or amelioration of TNFα-associated diseases; their coding sequences, production, and use in pharmacologically suitable compositions.
Background Art
• Tumour necrosis factor alpha (TNFα, also known as cachectin) , is a naturally occurring mammalian cytokine produced by numerous cell types, including monocytes and macrophages in response to endotoxin or other stimuli. TNFoc is a major mediator of inflammatory, immunological, and pathophysiological reactions (Grell, M. , et al. (1995) Cell, 83: 793-802).
Soluble TNFα is formed by the. cleavage of a precursor transmembrane protein (Kriegler, et al . (1988) Cell 53: 45-53), and the secreted 17 kDa polypeptides assemble to soluble homotrimer complexes (Smith, et al.(1987), J. Biol. Chem. 262: 6951-6954; for reviews of TNF, see Butler, et al . (1986), Nature 320:584; Old (1986), Science 230: 630). These complexes then bind to receptors found on a variety of cells. Binding produces an array of pro-inflammatory effects, including (i) release of other pro-inflammatory cytokines such as interleukin (IL) -6, IL-8, and IL-I, (ii) release of matrix metalloproteinases and (iii) up regulation of the expression of endothelial adhesion molecules, further amplifying the inflammatory and immune cascade by attracting leukocytes into extravascular tissues.
A large number of disorders are associated with elevated levels of TNFα, many of them of significant medical importance. TNFα has been shown to be up- regulated in a number of human diseases, including chronic diseases such as rheumatoid arthritis (RA) , inflammatory bowel disorders including Crohn's disease and ulcerative colitis, sepsis, congestive heart failure, asthma bronchiale and multiple sclerosis . Mice transgenic for human TNFα produce high levels of TNFα constitutively and develop a spontaneous, destructive polyarthritis resembling RA (Keffer et al . 1991, EMBO J., 10,4025- 4031) . TNFα is therefore referred to as a pro- inflammatory cytokine.
TNFa is now well established as key in the pathogenesis of RA, which is a chronic, progressive and debilitating disease characterised by polyarticular joint inflammation and destruction, with systemic symptoms of fever and malaise and fatigue. RA also leads to chronic synovial inflammation, with frequent progression to articular cartilage and bone destruction. Increased levels of TNFα are found in both the synovial fluid and peripheral blood of patients suffering from RA. When TNFα blocking agents are administered to patients suffering from RA, they reduce inflammation, improve symptoms and retard joint damage (McKown et al . (1999), Arthritis Rheum. 42:1204-1208) .
Physiologically, TNFα is also associated with protection from particular infections (Cerami. et al. (1988), Immunol. Today 9:28). TNFα is released by macrophages that have been activated by lipopolysaccharides of Gram-negative bacteria. As such, TNFα appears to be an endogenous mediator of central importance involved in the development and pathogenesis of endotoxic shock associated with bacterial sepsis (Michie, et al . (1989), Br. J. Surg.76 : 670-671.; Debets . et al. (1989), Second Vienna Shock Forum, p. 463-466; Simpson, et al . (1989) Crit . Care Clin. 5: 27-47; Waage et al. (1987). Lancet 1: 355-357; Hammerle. et al. (1989) Second Vienna Shock Forum p. 715-718; Debets. et al . (1989), Crit. Care Med. 17:489-497; Calandra. et al . (1990), J. Infect. Dis. 161:982-987; Revhaug et al . (1988), Arch. Surg. 123:162-170).
As with other organ systems, TNFα has also been shown to play a key. role in the central nervous system, in particular in inflammatory and autoimmune disorders of the nervous system, including multiple sclerosis, Guillain-Barre syndrome and myasthenia gravis, and in degenerative disorders of the nervous system, including Alzheimer's disease, Parkinson's disease and • Huntington's disease. TNFα is also involved in disorders of related systems of the retina and of muscle, including optic neuritis, macular degeneration, diabetic retinopathy, dermatomyositis, amyotrophic lateral sclerosis, and muscular dystrophy, as well as in injuries to the nervous system, including traumatic brain injury, acute spinal cord injury, and stroke.
Hepatitis is another TNFα-related inflammatory disorder which among other triggers can be caused by viral infections, including Epstein-Barr, cytomegalo- virus, and hepatitis A-E viruses. Hepatitis causes acute liver inflammation in the portal and lobular region, followed by fibrosis and tumor progression.
TNFα can mediate cachexia in cancer, which causes most cancer morbidity and mortality (Tisdale M.J. (2004), Langenbecks Arch Surg. 389:299-305).
The key role played by TNFα in inflammation, cellular immune responses and the pathology of many diseases has led to the search for antagonists of TNFα. TNFα is an important cytokine whose systemic blockade carries the risk for increased frequency and severity of clinically manifested infections, in particular re-activation of latent tuberculosis and possibly other risks including induction of lymphomas, demyelinating diseases and heart failure.
One class of TNFα antagonists designed for the treatment of TNFα-mediated diseases are antibodies or antibody fragments that specifically bind TNFα and thereby block its function. The use of anti-TNFα antibodies has shown that a blockade of TNFα can reverse effects attributed to TNFα including decreases in IL-I, GM-CSF, IL-6, IL-8, adhesion molecules and tissue destruction (Feldmann et al. (1997), Adv. Immunol. • 1997:283-350) .
Antibodies directed against TNFα have been proposed for the prophylaxis and treatment of endotoxic shock (Beutler et al . (1985) Science :234, 470-474) . The use of anti-TNFα antibodies in the treatment of septic shock is discussed by Bodmer et al . , 1993, (Critical Care Medicine, 21:441-446,1993), Wherry et al.,1993, (Critical Care Medicine, 21:436-440) and Kirschenbaum et al . ,1998, (Critical Care Medicine, 26:1625-1626). A method for treating a neurodegenerative disease in a human by administering an anti-TNFα monoclonal antibody or a TNFα binding fragment thereof has been disclosed in US2003147891.
WO0149321 teaches the use of TNFα blockers including anti TNFα antibodies to treat neurologic and related disorders caused by TNFα. It provides a method for treating said disorders by administering a TNFα antagonist .
WO03047510 discloses various kinds of monoclonal and engineered antibodies directed against
TNFα, their production, compounds comprising them and use in medicine.
Antibodies useful for therapies of TNFα mediated diseases are usually either monoclonal antibodies (mAB) produced by hybridoma technology from a natural source, usually a mouse, or engineered antibodies. The latter either correspond to naturally occurring antibodies in that they comprise full-length heavy and light chains, or to the Fab fragments that can also be generated from natural antibodies by proteolytic cleavage, or to single chain scFv antibodies wherein fragments of the variable heavy and light chain regions are linked by a peptide linker.
Both, heavy and light chains of an antibody comprise constant and variable domains. As non-human antibodies are immunogenic, the amount of human-like sequences in an antibody is often increased in a so- called "hybrid" antibody, which comprises constant regions of a human IgG, and variable regions matching the sequences of an animal antibody, in most cases murine antibodies with the desired specificity. These variable regions can then be further adapted to become more similar to a typical human antibody by mutagenesis, leading to a "humanised" antibody. In yet an alternative approach, only the antigen binding portions, i.e. the complementary determining regions (CDRs) of the variable regions of a mouse antibody are combined with a framework of a human antibody, resulting in a "CDR-grafted" antibody.
Monoclonal antibodies against TNFα have been described in the prior art. Meager et al., 1087 (Hybridoma 6:305-311) describe murine monoclonal antibodies against recombinant TNFα. Shimamoto et al. , 1988, (Immunology Letters 17:311-318) describe the use of murine monoclonal antibodies against TNFα in preventing endotoxic shock in mice. US5919452 discloses anti-TNFα chimeric antibodies and their use in treating pathologies associated with the presence of TNFα.
The use of anti-TNFα antibodies in the treatment of RA and Crohn's disease is discussed in Feldman et ali(1998), (Transplantation Proceedings 30:4126-4127), Adorini et al . , 1997, (Trends in Immunology Today 18:209-211) and in Feldmann et al . , 1997, (Advanced Immunology 64:283-350). The antibodies to TNFα used in such treatments are generally chimeric antibodies, such as those described in US5919452.
US20003187231 discloses humanised anti-TNFα antibodies with at least one non-human CDR region that have improved binding characteristics. Furthermore, in the International Patent Application WO 92/11383, recombinant antibodies, including CDR-grafted antibodies, specific for TNFα are disclosed. Rankin et al . (1995), (British J. Rheumatology 34:334-342) describe the use of such CDR-grafted antibodies in the treatment of RA.
WO9211383 discloses a recombinant, humanised CDR-grafted antibody specific for TNFα that is derived from the murine monoclonal antibody 61El, hTNFI, hTNF3 or 101.4, and it teaches the production and use of said antibodies in diagnosis and/or therapy of TNFα-associated , disorders .
Among the specific inhibitors of TNFα that have become commercially available only recently, a monoclonal, chimeric mouse-human antibody directed against TNFα (infliximab, Remicade™; Centocor Corporation/Johnson & Johnson) has demonstrated clinical efficacy in the treatment of RA (Elliott et. al.1994, Lancet 344:1105-1110; Mani et al . (1998), Arthritis &. Rheumatism 41: 1552-1563) . Infliximab has also demonstrated clinical efficacy in the treatment of the inflammatory bowel disorder Crohn's disease (Baert et al . 1999, Gastroenterology 116: 22-28.)
US22002037934 discloses the treatment of hepatitis by administration of an anti-TNFα antibody such as infliximab.
US6428787 teaches the treatment of neurologic and TNFα-associated diseases with anti-TNFα antibodies including infliximab, CDP571 and D2E7. • D2E7 (Adalimumab) , a human anti-TNFα monoclonal antibody (Abbott) has been developed to treat RA and Crohn's disease (WO9729131) . Celltech is developing CDP571 (EP0626389) , a humanised monoclonal anti-TNFα IgG4 antibody to treat Crohn' s disease and CDP870, a humanised monoclonal anti TNFα antibody- fragment to treat RA. The local administration of said antibodies for treatment of localised disorders is disclosed in US2003185826.
Many single chain antibodies (scFvs) were generated against a multitude of different antigens, in particular because they can be easily selected for high binding capacity using techniques such as for example phage display or ribosome display. Moreover, scFv antibodies can be produced in microbial systems which are associated with fewer costs compared to the production of therapeutic full-length antibodies. In addition to conventional extracellular and in vitro applications, scFvs have also been successfully used for intracellular applications (Worn et al . 2000, JBC, 28;275 (4) :2795-2803; Auf der Maur et al . 2002, JBC, 22;277 (47) : 45075-45085; Stocks MR, 2004, Drug Discov Today. 15; 9 (22) : 960-966) ; hence, scFvs directed against intracellular antigens have been developed. In general, intracellular expression of functional scFvs is limited by their instability, insolubility, and tendency to form aggregates. For this -reason, in vivo screening systems for scFv antibodies, which are particularly soluble and stable under reducing conditions typical for the intracellular environment (e.g. nucleus, cytoplasm) have been successfully developed using a so called "Quality Control" screen (WO0148017; Auf der Maur et al . (2001), FEBS Lett. 508:407-412; Auf der Maur et al . (2004),
Methods 34:215-224) and have led to the identification of particularly stable and soluble scFv framework sequences for such purposes (WO03097697) . Furthermore, these frameworks show exceptional expression levels and enhanced stability and solubility properties also under natural, oxidizing conditions in the extracellular environment. Hence, these favourable biophysical and biochemical properties translate into favourable high production yields and enable these antibody fragments, once directed against specific antigens, to be applied locally and/or systemically as protein therapeutics in particular therapeutic areas. As both scFv antibodies and Fab fragments, in contrast to full-length antibodies, lack the Fc part that is recognized by the Fc~receptor of monocytes, such as e.g. natural killer cells, they do not evoke antibody-dependent cell-mediated cytotoxicity (ADCC) and thus do not provoke unspecific toxicity due to binding to Fc-receptors on non-target cells.
. Hence, there is a need for new, effective forms of antibodies for the treatment for TNFα-associated disorders such as RA, particularly treatments that can provide sustained, controlled therapy by local administration with a low degree of side effects. The present invention provides antibodies, compositions and methods for effective and continuous treatment of inflammatory processes of arthritis and other TNFα- mediated disorders or pathophysiological mechanisms, in particular various forms of pain.
All publications and references cited herein are hereby, incorporated by reference in their entirety.
Disclosure of the Invention
Hence, it is a general object of the invention to provide a stable and soluble antibody or antibody derivative, which specifically binds TNFα in vitro and in vivo. In a preferred embodiment said antibody derivative is an scFv antibody or Fab fragment.
Now, in order to implement these and still further objects of the invention, which will become more readily apparent as 'the description proceeds, said antibody or antibody 'derivative is manifested by the features that it comprises a light chain variable domain being or derived from the sequence SEQ ID NO:1 that is combined with a heavy chain variable domain being or derived from the sequence SEQ ID NO: 2, wherein in the case of a derived sequence said sequence has at maximum up to 5 changes within the framework of said VL domain and/or at maximum up to 9 changes within the framework of said VH domain.
A preferred embodiment of the present invention is said antibody or antibody derivative, wherein one or more amino acid changes are introduced at any of the positions in the framework, preferably at one or more positions selected from the group of the positions 4, 46, 65, 67 70, and 83 of the VL domain, and/or at one or more of the positions selected from the group of the positions 11, 16, 28, 43, 48, 68, 70, 71, 72, 73, 76, 77, 79, 93 and 112 of the VH domain. More preferably, at least one of the conversions leads to an amino acid present in SEQ ID NO: 3 for VL and/or SEQ ID NO: 4 for VH, and even more preferably at most 13 conversions in total are present.
Most preferably, said antibody or antibody derivatives comprises a VL domain of the sequence SEQ ID N0:l and/or a VH domain of the sequence or derived from the sequence SEQ ID NO: 2, or a VL domain of the sequence SEQ ID NO: 11 and a VH domain of the sequence SEQ ID NO: 4. If the VH domain of the antibody of the present invention comprises a VL domain of SEQ ID NO:1, in a preferred embodiment the VH sequence is derived from SEQ ID NO: 2 such that the phenylalanine at position 68 is changed to either alanine, leucine, isoleucine or valine. Additional changes within VH are optional. scFv antibodies of this kind are given in SEQ ID NO: 31, SEQ ID NO: 33, SEQ ID NO:34, SEQ ID NO:35, SEQ ID NO:36, and SEQ ID NO:37.
In another preferred embodiment of the present invention said antibody or antibody derivative is derived from the antibody with the VL sequence SEQ ID N0:l and the VH sequence SEQ ID NO: 2 and comprises at least one amino acid residue that is converted in at least one of the CDRs to a residue present in the corresponding CDR of the VL sequence SEQ ID NO: 5 and/or the VH sequence SEQ ID No: 6 or SEQ ID NO: 25.
In a much preferred embodiment of the present invention in said antibody or antibody derivative at least one of the CDRs of the group VL CDR2, VL CDR3, VH CDR2 or VH CDR3 is converted to the corresponding CDR of the VL sequence SEQ ID NO: 5 and/or the VH sequence SEQ ID No:25 or SEQ ID N0:6.
Most preferably, said antibody or antibody derivative comprises the following VL/VH sequence combinations :
VL SEQ ID NO: 7/VH SEQ ID NO: 2,
VL SEQ ID NO: 8/VH SEQ ID NO: 2,
VL SEQ ID NO:1/VH SEQ ID NO: 9, VL SEQ ID NO:1/VH SEQ ID NO: 25,
VL SEQ ID NO:1/VH SEQ ID NO: 28,
VL SEQ ID NO:1/VH SEQ ID NO: 29,
VL SEQ ID NO: 26/VH SEQ ID NO: 30,
VL SEQ ID NO:27/VH SEQ ID NO:30. In another preferred embodiment the antibody or antibody derivative of the present invention has specificity to human TNFα. Preferably, antigen binding is characterized by a K^ of * 100 nM or less. More preferred is an antibody with a K^ of 10 nM or less, and most preferred of 1 nM and less.
Antibody derivatives according to the present invention are for example Fc fusions, toxin fusions, fusions to enzymatic activities, different formats such as minibodies, diabodies, linear antibodies, single chain antibodies, bispecific antibody fragments, in particular scFv and Fab fragments.
Another preferred object of the present invention is a scFv antibody whose VL and VH domains are connected by a linker, preferably in a VL-linker-VH sequence arrangement. More preferably said linker has the sequence SEQ ID NO: 10. Another preferred object of the present invention is a scFv antibody derived from SEQ ID NO: 40 (TB-A) . Such an antibody can be obtained by mutagenesis, and comprises three or less mutations in either framework, CDR and/or linker sequences. Preferably, the scFv antibody has the sequence SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, SEQ ID NO: 24, SEQ ID NO:31, SEQ ID NO: 32, SEQ ID NO:33, SEQ ID NO:34, SEQ ID NO:35, SEQ ID NO:36, SEQ ID NO:37, or SEQ ID NO:38. Another preferred object of the present invention is the Fab fragment comprising a VL domain that is fused to the constant region of a human Ig kappa chain, and a VH domain that is fused to the CHl domain of a human IgG, whereby the two fusion polypeptides are connected by an inter-chain disulfide bridge.
Still in another aspect the antibody or antibody derivative, e.g. the antibody fragment, of the present invention is labelled or chemically modified. The present invention also provides a DNA sequence encoding any of the antibodies or antibody derivatives of .the present invention, as well as a cloning or expression vector containing said DNA sequence. In addition, a suitable host cell transformed with said DNA sequence is provided, which preferentially is E. coli, a yeast or a mammalian cell.
Furthermore, a method for the production of the antibodies or antibody derivatives of the present invention is provided, comprising culturing of the host cell transformed with the DNA encoding any of said antibodies or antibody derivatives under conditions that allow the synthesis of said antibody or antibody derivative, and recovering said molecule from said culture. Preferably, said method provides an scFv antibody or Fab fragment purified from E. coli. . Another aspect of the present invention is the use of the antibodies or antibody derivatives provided by the present invention as a diagnostic tool for in vitro diagnostics, and/or as a pharmaceutical. This use is particularly preferred in the context of any TNFα related condition.
The present invention also encompasses a composition comprising an antibody or antibody derivative of the present invention in combination with a pharmaceutically acceptable carrier, diluent or excipient, said composition to be used as a medicament for the treatment of TNFa associated diseases. In a further aspect the present invention provides a combination preparation comprising an antibody or antibody derivative of the present invention, preferably with a second compound that is not an antibody or antibody derivative specific for TNFα. In yet another aspect of the present invention the vector comprising the DNA sequence encoding an scFv antibody of the present invention is used for gene therapy.
The treatment of TNFα associated diseases is achieved by blocking of TNFα due to a strong interaction of TNFα with the antibody or the antibody derivative. Preferably, a treatment of autoimmune, acute or chronic inflammation conditions, cancer-related diseases, pain, neurological and neurodegenerative disorders, infectious diseases and cardiovascular diseases is envisaged.
Brief Description of the Drawings
The invention will be better understood and objects other than those set forth above will become apparent when consideration is given to the following detailed description thereof. Such description makes reference to the annexed drawings, wherein: Figure 1 shows a scheme of the scFv antibodies with the sequences of TB-A and TB-B delimiting the range of the most frequent variations. Asterisks designate positions at which amino acid changes in the framework of the antibodies of the present invention are tolerated. Amino acids indicated below the CDRs (the CDRs being emphasized with a gray background) can be used in the respective CDRs.
Figure 2 shows an exemplary scheme for the expression of a Fab fragment.
Figure 3 shows the production yield of scFvs when expressed in E. coll. A. SDS-polyacryalmide gel electrophoresis of expressed proteins. B. Analytical gel filtration of TB-A and TB-wt showing superior solubility of TB-A.
Figure 4 shows a comparison of affinity of different scFv antibodies towards TNFα determined by ELISA.
Figure 5 shows the inhibition of human TNFα- induced cytotoxicity in mouse L929 fibroblasts. A. Concentration dependent inhibition of TB-A in comparison to TB-wt and infliximab with IC50 values. B. Comparison of TB-A derivatives to block TNFα induced cytotoxicity. C. Comparison of scFv and Fab formats of TB-A.
Figure 6 shows the effect of antibody treatment of human TNFα-induced joint swelling in rat (Experiment: 5.3., Experiment 1). Figure 7 shows the scoring scheme for histopathological inflammation scoring.
Figure 8 shows the effect of antibody treatment on human TNFα-induced joint inflammation in rat (Experiment: 5.3., Experiment 1). Figure 9 shows the effect of antibody treatment of human TNFα-induced joint swelling in rat (Experiment: 5.3., Experiment 2).
Figure 10 shows the effect of antibody treatment on human TNFα-induced joint inflammation in rat (Experiment: 5.3., Experiment 2).
Figure 11 shows the stability of TB-A in different body fluids . Modes for Carrying Out the Invention
It has been found that antibodies or antibody derivatives comprising the frameworks identified in the so called "quality control" screen (WO0148017) are characterised by a generally high stability and/or solubility and thus may also be useful in the context of extracellular applications such as neutralizing TNFα. The present invention provides antibodies or antibody derivatives characterized by enhanced stability and solubility that specifically recognize and bind TNFα and thus are suitable to block the function of TNFα in vivo. Said antibodies or antibody derivatives are characterized by a special framework derived from the "quality control" screen for antibodies with particularly stable and soluble frameworks independent of their antigen binding site that has been disclosed in EP1479694. If the frameworks used in the screening are human antibody frameworks, they can be considered as non-immunogenic frameworks for human applications. The CDRs of the antibodies of the present invention are identical to or derived from the CDRs of the murine monoclonal antibody Di62 (Dδring et al . , 1994) that specifically binds to human TNFα with a high affinity (Kd = 0.4 nM) and can block TNFα binding to its receptor. In addition, Di62 inhibits human TNFα-induced cytotoxicity in mouse L929 cells. The obvious step of grafting the CDRs from the mouse antibody onto the apparently best suitable human acceptor framework with undefined antigen binding properties, said framework having the VL sequence SEQ ID . NO: 5 and the VH sequence SEQ ID NO: 6, said sequences being linked by a (GGGGS)4 linker (SEQ ID NOrIO), resulted in an scFv antibody of the sequence
DIVLTQS PS SLSAS VGDRVTLTCTASQS VSNDWWYQQRPGKAPKRLIYSAFNRYTG VPSRFSGSGSGTEFTLTISSLQPEDVAVYYCQQDYNSPRTFGQGTKLEVKRGGGGSG GGGSGGGGSGGGGSQVQLVQSGAEVKKPGASVKVSCTASGYSFTHYGMNWVRQAPGQ GLEWMGWINTYTGEPTYADKFKDRVTLTRDTSIGTVYMELTSLTSDDTAVYYCARER GDAMDYWGQGTLVTVSS (SEQ ID NO: 3+SEQ ID NO: 10+ SEQ ID NO: 4)
said scFv antibody being called TB-B. TB-B gave good yields in protein expression (Fig. 3a) , but was unable to specifically bind TNFα (Fig. 4a) .
Hence, to obtain an antibody or antibody derivative that is (i) sufficiently specific for binding TNFα, (ii) sufficiently soluble to allow efficient production and purification and to block TNFα in vivo, (iii) sufficiently stable to be useful as a pharmaceutical without suffering a rapid degradation and (iv) sufficiently non-immunogenic, a compromise between best solubility and best antigen binding characteristics was sought by varying the framework and the CDRs . The present invention provides a sequence for VL and VH that is optimized for the combination of the criteria (i-iv) . An scFv antibody comprising said VL (SEQ ID. NO:1 linked by a (GGGGS)4 linker to said VH (SEQ ID NO: 2) is called TB- A. The sequence of TB-A is given by SEQ ID NO: 40. This antibody is still reasonably stable and soluble to give satisfactory yields when expressed and purified from E. coli (Fig. 3A) , and it does not aggregate (Fig. 3B) . Its binding characteristics towards TNFα are excellent, with a Kd of 0.8 nM .
The present invention also discloses VL and VH sequences derived from the sequences present in TB-A in various ways. First, point mutations at up to five positions in the framework of VL and/or at up to nine positions in the framework of VH have been found acceptable, especially point mutations that render the frameworks more TB-B-like, i.e. more like SEQ ID NO: 3 for VL or SEQ ID NO: 4 for VH. An scFv antibody comprising a VL domain of the sequence SEQ ID NO: 11 and a VH domain of the sequence SEQ ID NO: 4, linked by the (GGGGS) 4 linker, is called TB-B R46L because it differs only at position 46 of VL from TB-B. In contrast to TB-B this antibody still has good binding properties towards TNFα (Kd = 100 nM) . This suggests that the number of changes in TB-B R46L relative to TB-A represents approximately the upper limit for changes in the variable domain framework.
In a preferred embodiment of the present invention only single or double point mutations are introduced into VL and/or VH frameworks of TB-A. The preferred framework residues for mutations are at positions 4, 46, 65, 67, 70 and 83 for VL, and at positions 11, 16, 28, 43, 48, 68, 70, 71, 72, 73, 76, 77, 79, 93 and 112 for VH. The positions are numbered according to the numbering in the sequence listings. The amino acids substitutions are preferably either "conservative", or such that the replacing amino acids are more similar or preferably even identical to the corresponding amino acids present in the TB-B sequence. For example, A76 of VH in TB-A can be changed to 176 as it is present in TB-B, but it may also be changed to another amino acid with similar, i.e. a non-polar side chain such as V or L. This is an example of a "conservative" amino acid substitution. Families of amino acid residues having similar side chains suitable for "conservative" substitutions as used herein have been defined in the art, including basic side chains (K, R, H) , acidic side chains (D, E) , uncharged polar side chains (Q, N, S, T, Y, C) , non-polar side chains (G, A, V, L, I, P, F, M, W) , beta-branched side chains (T, V, I) and aromatic side chains (Y, F, W, H) . A preferred conservative change is that of VL at position 83, in that V is changed to either F (SEQ ID NO: 26) or to A (SEQ ID NO: 27). However, in SEQ ID NO: 32 a non-conservative change in VL is V83E, which is combined with a change in CDRl, i.e. N31D, and in VH with V79A. Another extraordinary TB-A variant is that of SEQ ID NO: 33, with a conservative' F68L exchange in VH connected to VL by a linker carrying an R at position 2, replacing G. Much preferred single amino acid exchanges are R65S or Y67S in VL and K43Q or F68 to V, L, or A in VH. Much preferred double changes are F70L/L72R or A76I/S77G in VH. ScFv antibodies comprising TB-A sequences with said alterations show inhibition of TNFα induced cytotoxicity in L929 cells. The results of some of them are shown in Fig. 5B. Their sequences are as follows :
SEQ ID NO: 18 = TB-A H_K43Q (TB-A H43) SEQ ID NO: 19 = TB-A H_F68V (TB-A H68)
SEQ ID NO:20 = TB-A H_F70L/L72R (TB-A H70/72) SEQ ID NO:21 = TB-A H_A76I/S77G (TB-A H76/77) SEQ ID NO: 22 = TB-A L_L46R (TB-A L46) SEQ ID NO: 23 = TB-A L_R65S (TB-A L65) SEQ ID NO: 24 = TB-A L_Y67S (TB-A L67)
In a preferred embodiment any of the above mentioned VH domains may be combined with any of the above mentioned VL domains .
In another preferred embodiment of the present invention the VL and VH domains of TB-A and TB-B are shuffled such that , the VL domain of TB-A (SEQ ID. NO:1 is combined with the VH domain of TB-B (SEQ ID NO:4), or the VL domain of TB-B (SEQ ID. NO:4) is combined with the VH domain of TB-A (SEQ ID NO: 2) . In a much preferred embodiment the shuffled versions obtained in an scFv are connected with the (GGGGS) 4 linker of the sequence SEQ ID. NO: 10, resulting in the scFv antibodies TB-AB (SEQ ID. NO: 12} or TB-BA (SEQ ID. NO:13), respectively. Said (GGGGS) 4 linker can have an amino acid exchange of a glycine to a more hydrophilic, i.e. polar, or even charged amino acid, which 'may render the antibody more soluble. Among these variations, the one with the sequence GRGGS- (GGGGS) 3 (SEQ ID NO: 39) is preferred.
It is also within the scope of the present invention to combine VL or VH domains of TB-B-like sequences with VH or VL domains of. TB-A-like sequences, whereby TB-B/TB-A-like means that the sequences are closer to the one than to the other.
In yet another preferred embodiment of the present invention one or more amino acids are changed in the CDR regions of the TB-A VL and/or VH sequences to match the corresponding amino acids present in the selected sequences SEQ ID NO: 5 of VL and/or SEQ ID. NO: 6 or SEQ ID NO: 25 of VH. Much preferred are changes in one of the VL CDRs (VL CDR2 or VL CDR3) and/or VH CDRs (CDR2 or CDR3) , with the most preferred changes leading to the VL sequences SEQ ID N0:7or SEQ ID NO: 8 and/or the VH sequences SEQ ID NO:25 or SEQ ID NO: 9, respectively.
In another preferred embodiment of the present invention the VL sequences SEQ ID NO: 26 or SEQ ID NO:27 is' combined with the VH sequence SEQ ID NO:30. In yet another preferred embodiment of the present invention the VL sequence SEQ ID N0:l is combined with the VH sequences Seq ID NO:28 or SEQ ID NO:29.
Generally, any of the disclosed VL sequences may be combined with any of the disclosed VH sequences.
Objects of the present invention are antibodies and antibody fragments , in particular VL or VH polypeptides, single-chain antibodies (scFv) or Fab fragments. In the case of scFv antibodies, a selected VL domain can be linked to a selected VH domain in either orientation by a flexible linker. A suitable state of the art linker consists of repeated GGGGS amino acid sequences or variants thereof. In a preferred embodiment of the present invention a (GGGGS) 4 linker of the sequence ID NO: 10 or its derivative SEQ ID NO: 39 is used, but variants of 1-3 repeats are also possible (Holliger et al . (1993), Proc. Natl. Acad. Sci. USA 90:6444-6448). Other linkers that can be used for the present invention are described by Alfthan et al . (1995.), Protein Eng. 8:725-731, Choi et al. (2001), Eur. J. Immunol. 31:94-106, Hu et al . (1996), Cancer Res. 56:3055-3061, Klpriyanov et al. (1999), J. MoI. Biol. 293:41-56 and Roovers et al. (2001), Cancer Immunol. Immunother . 50:51-59. The arrangement can be either VL- linker-VH or VH-linker-VL, with the former orientation being the preferred one. In the case of Fab fragments, selected light chain variable domains VL are fused to the constant region of a human Ig kappa chain, while the suitable heavy chain variable domains VH are fused to the first (N-terminal) constant domain CHl of a human IgG. In an exemplary embodiment of the present invention, the human CK domain has the sequence SEQ ID NO: 14 and the CHl domain used to construct the Fab fragments has the sequence SEQ ID NO: 15. Fig. 2 shows an example of a Fab fragment wherein the VL and VH domains of TB-A are used such that the VL domain is directly linked to the human kappa constant domain, resulting in the sequence
DIVMTQSPSSLSASVGDRVTLTCTASQSVSNDVVWYQQRPGKAPKLLIYSAFNRXTG VPSRFSGRGYGTDFTLTISSLQPEDVAVYYCQQDYNSPRTFGQGTKLEIKRTVAAPS ' VFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWVDNALQSGNSQESVTEQDSKDST YSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKSFNRGEC (SEQ ID NO-.l+SEQ ID NO:14) and the VH domain is fused to the first constant domain (CHl) , resulting in the sequence
QVQLVQSGAEVKKPGASVKVSCTASGYTFTHYGMNWVRQAPGKGLEWMGWINTYTGE PTYADKFKDRFTFSLETSASTVYMELTSLTSDDTAVYYCARERGDAMDYWGQGTLVT VSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFSEPVTVSWNSGALTSGVHTFP AVLQSSGLYSLSSWTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCTS (SEQ ID NO:2+SEQ ID NO:15).
At the C-terminus, an inter-chain disulfide bridge is formed between the two constant domains.
The antibodies or antibody derivatives of the present invention can have affinities to human TNFα with dissociation constants K^ in a range of 0.8-10'0OO nM. In a preferred embodiment 'of the present invention the K^ is ≤IO nM. The affinity of an antibody for an antigen can be determined experimentally using a suitable method (Berzofsky et al . "Antibody-Antigen Interactions", in Fundamental Immunology, Paul, W.E., Ed, Raven Press: New York, NY (1992); Kuby, J. Immunology, W. H. Freeman and Company: New York, NY ) and methods described therein. In one aspect of the present invention the antibodies or antibody derivatives, especially the scFv or Fab fragments, are labeled. Detectable labeling of a TNFα-specific antibody or antibody derivative can be accomplished by linking it to an enzyme for use in an enzyme immunoassay (EIA) or enzyme-linked immunosorbent assay (ELISA) , which are methods well known to the person skilled in the art (for example Current Protocols in Immunology, Coligan et al . Eds, John Wiley & Sons, 2005) .
By radioactively labeling the TNFα-specific antibodies or antibody derivatives, it is possible to detect TNF-α through the use of radioimmuneassay (RIA) (see for example, Work et al . , Laboratory Techniques and Biochemistry in Molecular Biology, North Holland
Publishing Company, N.Y. (1978) . The radioisotope can be detected by the use of a gamma counter or a scintillation counter or by autoradiography. Particularly useful isotopes are 3H, 131I, 35S, 14C, and preferably 125I. The antibodies or antibody derivatives of the present invention can also be labeled with fluorescent labeling compounds such as fluorescein, isothiocyanate, rhodamine, phycoerythrin, phycocyanin, allophycocyanin, o-phthaldehyde and fluorescamine, or with chemilumines- cent compounds such as luminol, isoluminol, theromatic acridinium ester, imidazol acridinium salt and oxalate ester.
Labeling and detection protocols are well known to the person skilled in the art. For example, they are available from Using Antibodies : A Laboratory Manual: Portable Protocol NO. I (Harlow, E. and Lane, D., 1998). Labeled antibodies or antibody derivatives of the present invention are useful for diagnostic purposes, in particular detection of TNFα in a biological sample removed from a patient. Any sample containing TNFα can be used, e.g. biological fluids like blood, serum, lymph, urine, inflammatory exudate, cerebrospinal fluid, amniotic fluid, a tissue extract or homogenate, and the like, or histological specimens for in situ detection.
PHARMACEUTICAL PREPARATIONS
Definitions : The term "pharmaceutical formulation" refers to preparations which are in such form as to permit the biological actvity of the antibody or antibody derivative to be unequivocally effective, and which contain no additional ' components which are toxic to the subjects to which the formulation would be administered. "Pharmaceutically acceptable" excipients (vehicles, additives) are those which can reasonably be administered to a subject mammal to provide an effective dose of the active ingredient employed.
A "stable" formulation is one in which the antibody or antibody derivative therein essentially retains its physical stability and/or chemical stability and/or biological activity upon storage. Various analytical techniques for measuring protein stability are available in the art and are reviewed in Peptide and Protein Drug Delivery, 247-301, Vincent Lee Ed., Marcel Dekker, Inc., New York, N. Y., Pubs. (1991) and Jones, .A. Adv. Drug Delivery Rev. 10: 29-90 (1993), for example, Stability ■ can be measured at a selected temperature for a selected time period. Preferably, the formulation is stable at room temperature (about 30° C) or at 40° C for at least 1 month and/or stable at about 2-8° C for at least 1 year for at least 2 years. Furthermore, the formulation is preferably stable following freezing (to, e.g., -70° C) and thawing of the formulation. An antibody or antibody derivative "retains its physical stability" 'in a pharmaceutical formulation if it shows no signs of aggregation, precipitation and/or denaturation upon visual examination of color and/or clarity, or as measured by UV light scattering or by size exclusion chromatography.
An antibody or antibody derivative "retains its chemical stability" in a pharmaceutical formulation, if the chemical stability at a given time is such that the protein is considered to still retain its biological activity as defined below. Chemical stability can be assessed by detecting and quantifying chemically altered forms of the protein. Chemical alteration may involve size modification (e.g. clipping) which can be evaluated using size exclusion chromatography, SDS-PAGE and/or matrix-assisted laser desorption ionization/time-of- flight mass spectrometry (MALDI/TOP MS), for example. Other types of chemical alteration include charge alteration (e.g. occurring as a result of deamidation) which can be evaluated by ion-exchange chromatography, for example.
An antibody or antibody derivative "retains its biological activity" in a pharmaceutical formulation, if the biological activity of the antibody at a given time is within about 10% (within the errors of the assay) of the biological activity exhibited at the time the pharmaceutical formulation was prepared as determined in an antigen binding assay, for example. Other "biological activity" assays for antibodies are elaborated herein below.
By "isotonic" is meant that the formulation of interest has essentially the same osmotic pressure as human blood. Isotonic formulations will generally have an osmotic pressure from about 250 to 350 mOsm. Isotonicity can be measured using a vapor pressure or ice-freezing type osmometer, for example. A "polyol" is a substance with multiple hydroxyl groups, and includes sugars (reducing and non- reducing sugars), sugar alcohols and sugar acids. ' Preferred polyols herein have a molecular weight which is less than about 600 kD (e.g. in the range from about 120 to about 400 kD) . A "reducing sugar" is one which contains a hemiacetal group that can reduce metal ions or react covalently with lysine and other amino groups in proteins and a "non-reducing sugar" is one which does not have these properties of a reducing sugar. Examples of reducing sugars are fructose, mannose, maltose, lactose, arabinose, xylose, ribose, rhamnose, galactose and glucose. Non-reducing sugars include sucrose, trehalose, sorbose, melezitose and raffinose . Mannitol, xylitol, erythritol, threitol, sorbitol and glycerol are examples of sugar alcohols. As to sugar acids, these include L- gluconate and metallic salts thereof. Where it is desired that the formulation is freeze-thaw stable, the polyol is preferably one which does not crystallize at freezing temperatures (e.g. -20° C) such that it destabilizes the antibody in the formulation. Non-reducing sugars such as sucrose and trehalose are the preferred polyols herein, with trehalose being preferred over sucrose, because of the superior solution stability of trehalose. As used herein, "buffer" refers to a buffered solution that resists changes in pH by the action of its acid-base conjugate components. The buffer of this invention has a pH in the range from about 4.5 to about 6.0; preferably from about 4.8 to about 5.5; and most preferably has a pH of about 5.0. Examples of buffers that will control the pH in this range include acetate (e.g. sodium acetate) , succinate (such as sodium succinate) , gluconate, histidine, citrate and other organic acid buffers: Where a freeze-thaw stable formulation is desired, the buffer is preferably not phosphate . In a pharmacological sense, in the context of the present invention, a "therapeutically effective amount" of an antibody or antibody derivative refers to an amount effective in the prevention or treatment of a disorder for the treatment of which the antibody or antibody derivative is effective. A "disease/disorder" is any condition that would benefit from treatment with the antibody or antibody derivative. This includes chronic and acute disorders or diseases including those pathological conditions which predispose the mammal to the disorder in question.
A "preservative" is a compound which can be included in the formulation to essentially reduce bacterial action therein, thus facilitating the production of a multi-use formulation, for example. Examples of potential preservatives include octadecyldimethylbenzyl ammonium chloride, hexamethonium chloride, benzalkonium chloride (a mixture of alkylbenzyldimethylammonium chlorides in which the alkyl groups are long-chain compounds) , and benzethonium chloride. Other types of preservatives include aromatic alcohols such as phenol, butyl and benzyl alcohol, alkyl parabens such as methyl or propyl paraben, catechol, resorcinol, cyclohexanol, 3-pentanol, and m-cresol. The most preferred preservative herein is benzyl alcohol.
The present invention also provides pharmaceutical compositions comprising one or more antibodies or antibody derivative compounds, together with at least one physiologically acceptable carrier or excipient. Pharmaceutical compositions may comprise, for example, one or more of water, buffers [e.g., neutral buffered saline or phosphate buffered saline) , ethanol, mineral oil, vegetable oil, dimethylsulfoxide, carbohydrates (e.g., glucose, mannose, sucrose or dextrans) , mannitol, proteins, adjuvants, polypeptides or amino acids such as glycine, antioxidants, chelating agents such as EDTA or glutathione and/or preservatives. As noted above, other active ingredients may (but need not) be included in the pharmaceutical compositions provided herein.
A carrier is a substance that may be associated with an antibody or antibody derivative prior to administration to a patient, often for the purpose of controlling stability or bioavailability of the compound. Carriers for use within such formulations are generally biocompatible, and may also be biodegradable. Carriers include, for example, monovalent or multivalent molecules such as serum albumin (e.g., human or bovine), egg albumin, peptides, polylysine and polysaccharides such as aminodextran and polyamidoamines . Carriers also include solid support materials such as beads and microparticles comprising, for example, polylactate polyglycolate, poly (lactide-co-glycolide) , polyacrylate, latex, starch, cellulose or dextran. A carrier may bear the compounds in a variety of ways, including covalent bonding (either directly or via a linker group) , noncovalent interaction or admixture.
Pharmaceutical compositions may be formulated for any appropriate manner of administration, including, for example, topical, oral, nasal, rectal or parenteral administration. In certain embodiments, compositions in a form suitable for oral use are preferred. Such, forms include, for example, pills, tablets, troches, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsion, hard or soft capsules, or syrups or elixirs. Within yet other embodiments, compositions provided herein may be formulated as a lyophilizate. The term parenteral as used herein includes subcutaneous, intradermal, intravascular (e.g., intravenous), intramuscular, spinal, -intracranial, intrathecal and intraperitoneal injection, as well as any similar injection or infusion technique.
Compositions intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and may contain one or more agents, such as sweetening agents, flavoring agents, coloring agent, and preserving agents in order to provide appealing and palatable preparations. Tablets contain the active ingredient in admixture with physiologically acceptable excipients that are suitable for the manufacture of tablets. Such excipients include, for example, inert diluents (e.g., calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate), granulating and disintegrating agents {e.g., corn starch or alginic acid), binding agents {e.g., starch, gelatin or acacia) and lubricating agents (e.g., magnesium stearate, stearic acid or talc) . The tablets may be uncoated or they may be coated by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period. For example, a time delay material such as glyceryl monosterate or glyceryl distearate may be employed. Formulations for oral use may also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent (e.g., calcium carbonate, calcium phosphate or kaolin) , or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium {e.g., peanut oil, liquid paraffin or olive oil) . Aqueous suspensions contain the antibody or antibody derivative in admixture with excipients suitable for the manufacture of aqueous suspensions . Such excipients include suspending agents (e.g., sodium carboxymethylcellulose, methylcellulose, hydropropylmethylcellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia) ; and- dispersing or wetting agents (e.g., naturally-occurring phosphatides such as lecithin, condensation products of an alkylene 'oxide with fatty acids such as . ' polyoxyethylene stearate, condensation products of ethylene oxide with long chain aliphatic alcohols such as heptadecaethyleneoxycetanol, condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol such as polyoxyethylene sorbitol monooleate, or condensation products of ethylene oxide . 5. with partial esters derived- from fatty acids and hexitol anhydrides such as polyethylene sorbitan monooleate) . Aqueous suspensions may also comprise one or more preservatives-, for example ethyl or n-propyl p- hydroxybenzoate, one or more coloring agents, one or more 0 flavoring agents, and one or more sweetening agents, such as sucrose or saccharin. Syrups and elixirs may be formulated with sweetening agents, such as glycerol, propylene glycol, sorbitol, or sucrose. Such formulations may also comprise one or more demulcents, preservatives, 5 flavoring agents, and/or coloring agents.
Oily suspensions may be formulated by suspending the active ingredients in a vegetable oil (e.g., arachis oil, olive oil, sesame oil, or coconut oil) or in a mineral oil such as liquid paraffin. The 0 oily suspensions may contain a thickening agent such as beeswax, hard paraffin, or cetyl alcohol. Sweetening agents, such as those set forth above, and/or flavoring agents may be added to provide palatable oral preparations. Such suspensions may be preserved by the 5 addition of an anti-oxidant such as ascorbic acid.
Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent,- suspending agent and one or 0 more. preservatives . Suitable dispersing or wetting agents and suspending agents are exemplified by those already mentioned above. Additional excipients, for example sweetening, flavoring 'and coloring agents, may also be present. 5 Pharmaceutical compositions may also be in the form of oil-in-water. emulsions. The oily phase may be a vegetable oil {e.g., olive oil "or arachis oil), a mineral oil {e.g., liquid paraffin), or a mixture thereof. Suitable emulsifying agents include naturally- occurring gums {e.g., gum acacia or gum tragacanth) , naturalIy-occurring phosphatides {e.g. , soy bean, lecithin, and esters or partial esters derived from fatty- acids and hexitol), anhydrides {e.g., sorbitan monoleate) , and condensation products of partial esters derived from fatty acids and hexitol with ethylene oxide (e.g., polyoxyethylene sorbitan monoleate) . An emulsion may also comprise one or more sweetening and/or flavoring agents .
The pharmaceutical composition may be prepared as a sterile injectible aqueous or oleaginous suspension in which the modulator, depending on the vehicle and concentration used, is either suspended or ■ dissolved in the vehicle. Such a composition may be formulated according to the known art using suitable dispersing, wetting agents and/or suspending agents such as those mentioned above. Among the acceptable vehicles and solvents that may be employed are water, 1,3- butanedi-ol, Ringer's solution and isotonic sodium chloride solution. In addition, sterile, fixed oils may be employed as a solvent or suspending medium. For this purpose any bland fixed oil may be employed, including synthetic mono- or diglycerides . In addition, fatty acids such as oleic acid may be used in the preparation of injectible compositions, and adjuvants such as local anesthetics, preservatives and/or buffering agents can be dissolved in the vehicle.. Pharmaceutical compositions may be formulated as sustained release formulations (i.e., a formulation such as a capsule that effects a slow release of modulator following administration) . Such formulations may generally be prepared using well known technology and administered by/ for example, oral, rectal, or subcutaneous implantation, or by implantation at the desired target site. Carriers for use within such formulations are biocompatible, and may also be biodegradable; preferably the formulation provides a relatively constant level of modulator release. The amount of an antibody or antibody derivative contained within a sustained release formulation depends upon, for example, the site of implantation, the rate and expected duration of release and the nature of the disease/disorder to be treated or prevented.
Antibody or antibody derivatives provided herein are generally administered in an amount that achieves a concentration in a body fluid (e.g., blood, plasma, serum, CSF, synovial fluid, lymph, cellular interstitial fluid, tears or urine) that is sufficient to detectably bind to TWFα and prevent or inhibit TNFα associated diseases/disorders. A dose is considered to be effective if it results in a discernible patient benefit as described herein. Preferred systemic doses range from about 0.1 mg to about 140 mg per kilogram of body weight per day (about 0.5 mg to about 7 g per patient per day), with oral doses generally being about 5-20 fold higher than intravenous doses. The amount of antibody or antibody derivative that may be combined with the carrier materials to produce a single dosage form will vary depending upon the host treated and the particular mode of administration. Dosage unit forms will generally contain between from about 1 mg to about 500 mg of an active ingredient.
Pharmaceutical compositions may be packaged for treating conditions responsive to an antibody or antibody derivative directed to TNF-α. Packaged pharmaceutical compositions may include a container holding a effective amount of at least one antibody or antibody derivative as described herein and instructions (e.g., labeling) indicating that the contained composition is to be used for treating a disease/disorder responsive to one antibody or antibody derivative following administration in the patient. The antibodies or antibody derivatives of the present invention can also be chemically modified. Preferred modifying groups are polymers, for example an optionally substituted straight or branched chain polyalkene, polyalkenylene, or polyoxyalkylene polymer or a branched or unbranched polysaccharide. Such effector group may increase the half-live of the antibody in vivo. Particular examples of synthetic polymers include optionally substituted straight or branched chain poly (ethyleneglycol) (PEG), poly (propyleneglycol) , poly (vinylalcohol) or derivatives thereof. Particular naturally occurring polymers include lactose, amylose, dextran, glycogen or derivatives thereof. The size of the polymer may be varied as desired, but will generally be in an average molecular weight range from 500Da to 50000Da. For local application where the antibody is designed to penetrate tissue, a preferred molecular weight of the polymer is around 5000Da. The polymer molecule can be attached to the antibody, in particular to the C-terminal end of the Fab fragment heavy chain via a covalently linked hinge peptide as described in WO0194585. Regarding the attachment of PEG moieties, reference is made to "Poly (ethyleneglycol) Chemistry, Biotechnological and Biomedical Applications", 1992, J. Milton Harris (ed) , Plenum Press, New York and
"Bioconjugation Protein Coupling Techniques for the Biomedical Sciences", 1998, M. Aslam and A. Dent, Grove Publishers, New York.
Preparation of the Formulation After preparation of the antibody or antibody- derivative of interest as described above, the pharmaceutical formulation comprising it is prepared. The antibody to be formulated has not been subjected to prior lyophilization and the formulation of interest herein is an aqueous formulation. Preferably the antibody or antibody derivative in the formulation is an antibody fragment, such as an scFv. The therapeutically effective amount of antibody present in the formulation is determined by taking into account the desired dose volumes and mode(s) of administration, for example. From about 0.1 mg/ml to about 50 mg/ml, preferably from about 0.5 mg/ml to about 25 mg/ml and most preferably from about 2 mg/ml to about 10 mg/ml is an exemplary antibody concentration in the formulation.
An aqueous formulation is prepared comprising the antibody or antibody derivative in a pH-buffered solution The buffer of this invention has a pH in the range from about 4.5 to about 6.0, preferably from about 4.8 to about 5.5, and most preferably has a pH of about 5.0. Examples of buffers that will control the pH within this range include acetate (e.g. sodium acetate), succinate (such as sodium succinate) , gluconate, histidine, citrate and other organic acid buffers. The buffer concentration can be from about 1 mM to about 50 πiM, preferably from about 5 mM to about 30 mM, depending, for example, on the buffer and the desired isotonicity of the formulation. The preferred buffer is sodium acetate (about 10 mM) , pH 5.0.
A polyol , which acts as a tonicifier and may stabilize the antibody, is included in the formulation. In preferred embodiments, the formulation does not contain a tonicifying amount of a salt such as sodium chloride, as this may cause the antibody or antibody derivative to precipitate and/or may result in oxidation at low pH. In preferred embodiments, the polyol is a non- reducing sugar, such as sucrose or trehalose. The polyol is added to the formulation in an amount which may vary with respect to the desired isotonicity of the formulation. Preferably the aqueous formulation is isotonic, in which case suitable concentrations of the polyol in the formulation are in the range from about 1% to about 15% w/v, preferably in the range from about 2% to about 10% whv, for example. However, hypertonic or hypotonic formulations may also be suitable. The amount of polyol added may also alter with respect to the molecular weight of the polyol. For example, a lower amount of a monosaccharide (e.g. mannitol) may be added, compared to a disaccharide (such as trehalose) . A surfactant is also added to the antibody or antibody derivative formulation. Exemplary surfactants include nonionic surfactants such as polysorbates (e.g. polysorbates 20, 80 etc) or poloxamers (e.g. poloxamer 188) . The amount of surfactant added is such that it reduces aggregation of the formulated antibody/antibody derivative and/or minimizes the formation of particulates in the formulation and/or reduces adsorption. For example, the surfactant may be present in the formulation in an amount from about 0.001% to about 0.5%, preferably from about 0.005% to about 0.2% and most preferably from about 0.01% to about 0.1%.
In one embodiment, the formulation contains the above-identified agents (i.e. antibody or antibody derivative, buffer, polyol and surfactant) and is essentially free of one or more preservatives, such as benzyl alcohol, phenol, m-cresol, chlorobutanol and benzethonium Cl. In another embodiment, a preservative may be included in the formulation, particularly where the formulation is a multidose formulation. The concentration of preservative may be in the range from about 0.1% to about 2%, most preferably from about 0.5% to about 1%. One or more other pharmaceutically acceptable carriers, excipients or stabilizers such as those described in Remington's Pharmaceutical Sciences 21st edition, Osol, A. Ed. (2006) may be included in the formulation provided that they do not adversely affect the desired characteristics of the formulation. Acceptable carriers, excipients or stabilizers are nontoxic to recipients at the dosages and concentrations employed and include,- additional buffering agents; co- solvents, antioxidants including ascorbic acid and methionine; chelating agents such- as EDTA; metal complexes (e.g. Zn-protein complexes); biodegradable polymers such as polyesters; and/or salt-forming counterions such as sodium.
The formulations to be used for in vivo administration must be sterile. This is readily accomplished by filtration through sterile filtration membranes, prior to, or following, preparation of the formulation.
Administration of the formulation The formulation is administered to a mammal in need of treatment with the antibody, preferably a human, in accord with known methods, such as intravenous administration as a bolus or by continuous infusion over a period of time, by intramuscular, intraperitoneal, intracerobrospinal , subcutaneous, intra-articular, intrasynovial , intrathecal, oral, topical, or inhalation routes. In preferred embodiments, the formulation is administered to the mammal by intravenous administration. For such purposes, the formulation may be injected using a syringe or via an IV line, for example.
The appropriate dosage ("therapeutically effective amount") of the antibody will depend, for example, on the condition to be treated, the severity and course of the condition, whether the antibody is administered for preventive or therapeutic purposes, previous therapy, the patient's clinical history and response to the antibody, the type of antibody used, and the discretion of the attending physician. The antibody or antibody derivative is suitably administered to the patent at one time or over a series of treatments and may be administered to the patent at any time from diagnosis onwards. The antibody or antibody derivative may be administered as the. sole treatment or in conjunction with other, drugs or therapies useful in treating the condition in question.
As a general proposition, the therapeutically effective amount of the antibody or antibody derivative administered will be in the range of about 0.1 to about 50 mg/kg of patent body weight whether by one or more administrations, with the typical range of antibody used being about 0.3 to about 20 mg/kg, more preferably about 0.3 to about 15 mg/kg, administered daily, for example. However, other dosage regimens may be useful. The progress of this therapy is easily monitored by conventional techniques .
Articles of Manufacture In another embodiment of the invention, an article of manufacture is provided comprising a container which holds the aqueous pharmaceutical formulation of the present invention and optionally provides instructions for its use. Suitable containers include, for example, bottles, vials and syringes. The container may be formed from a variety of materials such as glass or plastic. An exemplary container is a 3-20 cc single use glass vial. Alternatively, for a multidose formulation, the container may be 3-100 cc glass vial. The container holds the formulation and the label on, or associated with, the container may indicate directions for use. The article of manufacture may further include other materials desirable from a commercial and user standpoint , including other buffers, diluents, filters, needles, syringes, and package inserts with instructions for use.
Generating the antibodies of the present invention
The antibodies or antibody derivatives of the present invention may be generated using routine techniques in the field of recombinant genetics. Knowing the sequences of the polypeptides, the cDNAs encoding them can be generated by gene synthesis (www . genscript . com) . These cDNAs can be cloned into suitable vector plasmids. Once the DNA encoding a VL and/or a VH domain are obtained, site directed mutagenesis, for example by PCR using mutagenic primers, can be performed to obtain various derivatives. The best "starting" sequence can be chosen depending on the number of alterations desired in the VL and/or VH sequences. A preferred sequence is the TB-A sequences and its derivatives, e.g. scFv sequences or Fab fusion peptide sequences, may be chosen as templates for PCR driven mutagenesis and/or cloning.
Standard cloning and mutagenesis techniques well known to the person skilled in the art can be used to attach linkers, shuffle domains or construct fusions for the production of Fab fragments. Basic protocols disclosing the general methods of this invention are described in Molecular Cloning, A Laboratory Manual (Sambrook & Russell, 3rd ed. 2001) and in Current Protocols in Molecular Biology (Ausubel et al . , 1999). The DNA sequence harboring a gene encoding a scFv polypeptide, or in the case of Fab fragments, encoding either two separate genes or a bi-cistronic operon comprising the two genes for the VL-CK and the VH- CHl fusions (Fig. 2) are cloned in a suitable expression vector, preferably one with an inducible promoter. Care must be taken that in front of each gene an appropriate ribosome binding site (RBS in Fig. 2) is present that ensures translation. It is to be understood that the antibodies of the present invention comprise the disclosed sequences rather than they consist of them. For example, cloning strategies may require that a construct is made from which an antibody with one or a few additional residues at the N-terminal end are present. Specifically, the methionine derived from the start codon may be present in the final protein in cases where it has not been cleaved posttranslationally . Most of the constructs for scFv antibodies give rise to an additional alanine at the N-terminal end. In a preferred embodiment of the present invention, an expression vector for periplasmic expression in E. coli is chosen (Krebber, 1997) . Said vector comprises a promoter in front of a cleavable signal sequence. The coding sequence for the antibody peptide is then fused in frame to the cleavable signal sequence. This allows the targeting of the expressed polypeptide to the bacterial periplasm where the signal sequence is cleaved. The antibody is then folded. In the case of the Fab fragments, both the VL-CK and the VH-CHl fusions peptides must be linked to an export signal. The covalent S-S bond is formed at the C- terminal cysteines after the peptides have reached the periplasm. If cytoplasmic expression of antibodies is preferred, said antibodies usually can be obtained at high yields from inclusion bodies, which can be easily separated from other cellular fragments and protein. In this case the inclusion bodies are solubilized in a denaturing agent such as e.g. guaridine hydrochloride (GndHCl) and then refolded by renaturation procedures well known to those skilled in the art.
Plasmids expressing the scFv or Fab polypeptides are introduced into a suitable host, preferably a bacterial, yeast or mammalian cell, most preferably a suitable E. coli strain as for example JM83 for periplasmic expression or BL21 for expression in inclusion bodies. The polypeptide can be harvested either from the periplasm ox form inclusion bodies and purified using standard technique's such as ion exchange chromatography, reversed phase chromatography, affinity chromatography and/or gel filtration known to the person skilled in the art.
The antibodies or antibody derivatives of the present invention can be characterized with respect to yield, solubility and stability in vitro. Binding capacities towards TNFα, preferably towards human TNFa7 can be tested in vitro by ELISA or surface plasmon resonance (BIACore) , using recombinant human TNFα as described in WO9729131, the latter method also allowing to determine the kOff rate constant, which should preferably be less than 10"3S"1. Kd values of, ≤IO nM are preferred. In vivo neutralizing activity of an antibody or antibody derivative of the present invention can be estimated using the L929 cytotoxicity assay. Human recombinant TNFot exerts a cytotoxic effect towards cultured mouse L929 fibroblast cells in a concentration- dependent manner. This TNFα-induced cytotoxicity can be . inhibited by TNFα neutralizing antibodies (Dδring, 1994) . A preferred IC50 value corresponding to a half-maximal inhibitor concentration is '≤IOO ng ml"1. As TNFα has a proven pathophysiological role in various human diseases, in particular inflammatory disorders, immune and immune-regulated disorders, infections causing septic, endotoxic and cardiovascular shock, neurodegenerative diseases, and malignant diseases. As TNFα is suspected to play a disease-relevant role in a steadily growing number of additional human diseases, it is difficult to give a comprehensive list of indications that also ensures a complete representation of the spectrum of clinical applications for TNFα inhibitors in the future. Therefore, the antibodies or antibody derivatives of the present invention can be applied to treat the diseases listed in the following catalogue, which is not to be considered as a complete or exclusive list. Other diseases not mentioned specifically, which directly or indirectly are influenced by TNFα, are also included.
Autoimmune or chronic inflammation: Chronic and/or autoimmune states of inflammation in general, immune mediated inflammatory disorders in general, inflammatory CNS disease, inflammatory diseases affecting the eye, joint, skin, mucuous membranes, central nervous system, gastrointestinal tract, urinary tract or lung, states of . uveitis in general, retinitis, HLA-B27+' uveitis, Behcet's disease, dry eye syndrome, glaucoma, Sjogren syndrome, diabetes mellitus (incl. diabetic neuropathy), insulin resistance, states of arthritis in general, rheumatoid arthritis, • osteoarthritis, reactive arthritis and Reiter' s syndrome, juvenile arthritis, ankylosing spondylitis, multiple sclerosis, Guillain-Barre syndrome, myasthenia gravis, amyotrophic lateral sclerosis, sarcoidosis, glomerulonephritis, chronic kidney disease, cystitis, Psoriasis (incl. psoriatic arthritis), hidradenitis suppurativa, panniculitis, pyoderma gangrenosum, SAPHO syndrome (synovitis, acne, pustulosis, hyperostosis and osteitis), acne, Sweet's sydrome, pemphigus, Crohn's disease (incl. extraintestinal manifestastations) , ulcerative colitis, asthma bronchiale, hypersensitivity pneumonitis, general allergies, allergic rhinitis, allergic sinusitis, chronic obstructive pulmonary disease (COPD) , lung fibrosis, ' "Wegener's granulomatosis, Kawasaki syndrome, Giant cell arteritis, Churg-Strauss vasculitis, polyarteritis nodosa, burns, graft versus host disease, host versus graft reactions, rejection episodes following organ or bone marrow -transplantation, sytemic and local states of vasculitis in general, systemic and discoid lupus erythematodes, polymyositis and dermatomyositis, sclerodermia, pre-eclampsia, acute and chronic pancreatitis, viral hepatitis, alcoholic hepatitis. Acute ' inflammation and/or prevention of postsurgical or ■ posttraumatic inflammation and pain:
Prevention of postsurgical inflammation in general, eye surgery' (e.g. cataract (eye lens replacement) or glaucoma surgery), joint surgery (incl. arthroscopic surgery) , surgery at. joint-related structures (e.g. ligaments), oral and/or dental surgery, minimally invasive cardiovascular procedures (e.g. PTCA, atherectomy, stent placement) , laparoscopic and/or endoscopic intra-abdominal and gynecological procedures, endoscopic urological procedures (e.g. prostate surgery, ureteroscopy, cystoscopy, interstitial cystitis) , perioperative inflammation (prevention) in general.
Neurological and neurodegenarative diseases: Alzheimer disease, Parkinson's disease, Huntington' s disease, Bell' palsy, Creutzfeld-Jakob disease.
Cancer: Cancer-related osteolysis, cancer-related inflammation, cancer-related pain, cancer-related cachexia, bone metastases.
Pain:
Acute and chronic forms of pain, irrespective whether these are caused by central or peripheral effects of TNFα and whether they are classified as inflammatory , nociceptive or neuropathic forms of pain, sciatica, low back pain, carpal tunnel syndrome, complex regional pain syndrome (CRPS) , gout, postherpetic neuralgia, fibromyalgia, local pain states, chronic pain syndroms due to metastatic tumor, dismenorrhea.
Infection:
Bacterial, viral or fungal sepsis, tuberculosis, AIDS. Cardiovascular Disease:
Atherosclerosis, coronary artery disease, hypertension, dyslipidemia, heart insufficiency and chronic heart failure.
In a preferred embodiment of the present invention, treatment of osteoarthritis or uveitis or inflammatory bowel disease can be achieved with the antibodies, or antibody derivatives of the present invention.
The present invention also provides a pharmaceutical composition comprising an antibody or antibody derivative molecule of the present invention in combination with a pharmaceutically acceptable excipient, diluent' or carrier.
The pharmaceutical composition should preferably comprise a therapeutically effective amount .of the antibody of the present invention, i.e. an amount of said antibody that is needed to treat, ameliorate or prevent the TNFα-related disease or condition, or to exhibit a detectable therapeutic or preventive effect. The therapeutically effective dose can be estimated 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. A suitable animal model to observe an effect of the antibody or antibody derivative of the present invention is a rat model for acute monoarthritis (Bolon et al . (2004), Vet. Pathol .41 : 235-243. Human TNFα is injected intraarticularly into the knee joint of a rat, leading to an acute, self-limiting monoarthritis in the injected joint. Bioactivity of an anti-TNFα antibody (derivative) can be quantified by reduction of TNFα- induced knee joint swelling and or reduction of histological parameters of inflammation.
As the antibodies or antibody derivatives of the present invention are highly soluble, high antibody concentrations (60 mg ml"1 or more) enable the use of small application volumes.
The antibody or antibody derivative of the present invention may be utilised in any therapy where it is desired to reduce the level of biological active TNFα present in the human or animal body. The TNFα may be circulating in the body or be present at an undesirably high level localised at a particular site in the body. The present invention provides modes for systemic as well as local applications in general, which include, but are not limited to the following ones: peroral application, intravenous, subcutaneous, intramuscular, intraarticular, intravitreal, intradermal, or intraparenchymal injection, aerosol inhalation, topical application to the skin, to mucous membranes or to eye, systemic or local release via implantable minipump or local release via implantable formulation/device allowing for retarded release, topical application to serosal surfaces, intrathecal or intra- ventricular application, oral application in formulations allowing for controlled intralumenal release in selected parts of the gastrointestinal tract, localized intravasal release from adequate formulation/devices (e.g. stents), local delivery to urinary cyst, localized intralumenal release (e.g. to biliary tract, ureter), or local delivery through endoscopic devices, or release from contact lenses (contacts) . A preferred application is a local one such as intraarticular injection or topic application e.g. into the eye. For both preferred applications, the antibody of the present invention needs to-be in solution.
The present invention also reveals the use of the antibody or antibody derivative of the present invention for the production of a medicament for the treatment of TNFα associated diseases. In this case, the antibody or antibody derivative is comprised in a therapeutic composition. Said composition is used as a medicament, most preferably for the prevention or therapy of TNFα related diseases .
In another aspect the scFv antibodies of the present invention are used in gene therapy, in particular in adoptive cellular gene therapy. Autoimmune disorders represent inappropriate immune responses directed at self-tissue. Antigen-specific CD4+ T cells and antigen- presenting dendritic cells (DCs) are important mediators ' in the pathogenesis of auto-immune disease and thus are ideal candidates for adoptive cellular gene therapy, an ex vivo approach to therapeutic gene transfer. Using retrovirally transduced cells and luciferase bioluminescence, ' Tarner et al . (2003, Ann. N. Y. Acad. Sci. 998:512-519) have demonstrated that primary T cells, T cell hybridomas, and DCs rapidly and preferentially, home to the sites of inflammation in animal models of multiple sclerosis, arthritis, and diabetes. These cells, transduced with retroviral vectors that drive expression of various "regulatory proteins" such as interleukins and anti-TNF scFv, deliver these immunoregulatory proteins to the inflamed lesions, providing therapy for experimental autoimmune encephalitis, collagen-induced arthritis, and nonobese diabetic mice. The stable and soluble frameworks of the antibodies or antibody derivatives of the present inventions are particularly suitable for intracellular • delivery of the antigen, for example when the antibody or- antibody derivative is expressed from a transgene carried by a suitable retroviral vector. Adoptive cellular gene therapy leads to localized expression and secretion of the anti-TNFα scFv. Smith et al . (2003) have demonstrated that scFvs derived from a TNFα neutralizing monoclonal antibody (i) can neutralize TNFα in vitro and (ii) change the cytokine expression pattern in mice suffering from collagen-induced arthritis locally in the joints, but not systemically . Alternatively, direct systemic or local injection of suitable vectors (e.g. viruses) allowing for continuous expression of an anti-TNFα scFv antibody, is considered as another possible gene therapy approach.
The sequences of the present invention are the following ones:
SEQ ID NO:1 VL of TB-A
DIVMTQSPSSLSASVGDRVTLTCTASQSVSNDWWYQQRPGKAPK LLIYSAFNRYTGVPSRFSGRGYGTDFTLTISSLQPEDVAVYYCQQDYNSPRTFGQGT KLEVKR
SEQ ID NO : 2 VH of TB-A
QVQLVQSGAEVKKPGASVKVSCTASGYTFTHYGMNWVRQAPGKGL EWMGWINTYTGEPTYADKFKDRFTFSLETSASTVYMELTSLTSDDTAVYYCARERGD AMDYWGQGTLVTVSS
SEQ ID NO: 3 VL of TB-B
DIVLTQSPSSLSASVGDRVTLTCTASQSVSNDVVWYQQRPGKAPK RLIYSAFNRYTGVPSRFSGSGSGTEFTLTISSLQPEDVAVYYCQQDYNSPRTFGQGT KLEVKR SEQ ID NO : 4 VH of TB-B
QVQLVQSGAEVKKPGASVKVSCTASGYSFTHYGMNWVRQAPGQGL EWMGWINTYTGEPTYADKFKDRVTLTRDTSIGTVYMELTSLTSDDTAVYYCARERGD AMDYWGQGTLVTVSS . SEQ ID NO : 5 VL of FW2 . 3
DIVLTQSPSSLSASVGDRVTLTCRASQGIRNELAWYQQRPGKAPK RLIYAGSILQSGVPSRFSGSGSGTE-FTLTISSLQPEDVAVYYCQQYYSLPYMFGQGT KLEVKR
SEQ ID NO : 6 VH of FW2 . 3 QVQLVQSGAEVKKPGASVKVSCTASGYSFTGYFLHWVRQAPGQGL
EWMGRINPDSGDTIYAQKFQDRVTLTRDTSIGTVYMELTSLTSDDTAVYYCARVPRG TYLDPWDYFDYWGQGTLVTVSS
SEQ ID NO: 7 VL of TB_L2
DIVMTQSPSSLSASVGDRVTLTCTASQSVSNDWWYQQRPGKAPK LLIYAGSILQSGVPSRFSGRGYGTDFTLTISSLQPEDVAVYYCQQDYNSPRTFGQGT KLEVKR
SEQ ID NO: 8 VL of TB_L3
DIVMTQSPSSLSASVGDRVTLTCTASQSVSNDWWYQQRPGKAPK LLIYSAFNRYTGVPSRFSGRGYGTDFTLTISSLQPEDVAVYYCQQYYSLPYMFGQGT KLEVKR
SEQ ID NO : 9 VH of TB_H2
QVQLVQSGAEVKKPGASVKVSCTASGYTFTHYGMNWVRQAPGKGL EWMGRINPDSGDTIYAQKFQDRFTFSLETSASTVYMELTSLTSDDTAVYYCARERGD AMDYWGQGTLVTVSS SEQ ID NO: 10 Linker
GGGGSGGGGSGGGGSGGGGS SEQ ID NO: 11 VL of TB-B R46L
DIVLTQSPSSLSASVGDRVTLTCTASQSVSNDWWYQQRPGKAPK LLIYSAFNRYTGVPSRFSGSGSGTEFTLTISSLQPEDVAVYYCQQDYNSPRTFGQGT KLEVKR
SEQ ID NO: 12 TB-AB
DIVMTQSPSSLSASVGDRVTLTCTASQSVSNDVVWYQQRPGKAPK
LLIYSAFNRYTGVPSRFSGRGYGTDFTLTISSLQPEDVΆVYYCQQDYNSPRTFGQGT
KLEVKRGGGGSGGGGSGGGGSGGGGSQVQLVQSGAEVKKPGASVKVSCTASGYSFTH YGMNWVRQAPGQGLEWMGWINTYTGEPTYADKFKDRVTLTRDTSIGTVYMELTSLTS
DDTAVYYCARERGDAMDYWGQGTLVTVSS SEQ ID NO : 13 TB-BA
DIVLTQSPS SLSASVGDRVTLTCTASQSVSNDVVWYQQRPGKAPK
RLIYSAFNRYTG.VPSRFSGSGSGTEFTLTISSLQPEDVAVYYCQQDYNSPRTFGQGT
KLEVKRGGGGSGGGGSGGGGSGGGGSQVQLVQSGAEVKKPGASVKVSCTASGYTFTH YGMNWVRQAPGKGLEWMGWINTYTGEPTYADKFKDRFTFSLETSASTVYMELTSLTS
DDTAVYYCARERGDAMDYWGQGTLVTVSS
SEQ ID NO : 14 CK of Fab
TVAAPSVFIFPPSDEQLKSGTASWCLLNNFYPREAKVQWKVDNA LQSGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKSFN RGEC
SEQ ID NO: 15 CHl of Fab
ASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFSEPVTVSWNSGA .LTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKS CTS SEQ ID" NO-:1-6. VL' of TB-wt
DIVMTQTPKFLLVSAGDRVTITCTASQSVSNDWWYQQKPGQSPK MLMYSAFNRYTGVPDRFTGRGYGTDFTFTISSVQAEDLAVYFCQQDYNSPRTFGGGT KLEIKR
SEQ ID NO: 17 VH of TB-wt QIQLVQSGPELKKPGETVKISCKASGYTFTHYGMNWVKQAPGKGL
KWMGWINTYTGEPTYADDFKEHFAFSLETSASTVFLQINNLKNEDTATYFCARERGD AMDYWGQGTSVTVSS
SEQ ID NOrI8 TB-A H_K43Q, also named TB-A H43 DIVMTQSPSSLSASVGDRVTLTCTASQSVSNDWWYQQRPGKAPK LLIYSAFNRYTGVPSRFSGRGYGTDFTLTISSLQPEDVAVYYCQQDYNSPRTFGQGT KLEVKRGGGGSGGGGSGGGGSGGGGSQVQLVQSGAEVKKPGASVKVSCTASGYTFTH YGMNWVRQAPGQGLEWMGWINTYTGEPTYADKFKDRFTFSLETSASTVYMELTSLTS DDTAVYYCARERGDAMDYWGQGTLVTVSS
SEQ ID NO: 19 TB-A H_F68V, also named TB-A H68 DIVMTQSPSSLSASVGDRVTLTCTASQSVSNDWWYQQRPGKAPK
• LLIYSAFNRYTGVPSRFSGRGYGTDFTLTISSLQPEDVAVYYCQQDYNSPRTFGQGT
KLEVKRGGGGSGGGGSGGGGSGGGGSQVQLVQSGAEVKKPGASVKVSCTASGYTFTH
YGMNWVRQAPGKGLEWMGWINTYTGEPTYADKFKDRVTFSLETSASTVYMELTSLTS
. DDTAVYYCARERGDAMDYWGQGTLVTVSS SEQ ID NO.-20 TB-A H_F70L/L72R, also named TB-A H70/72
DIVMTQSPSSLSASVGDRVTLTCTASQSVSNDVVWYQQRPGKAPK
LLIYSAFNRYTGVPSRFSGRGYGTDFTLTISSLQPEDVAVYYCQQDYNSPRTFGQGT KLEVKRGGGGSGGGGSGGGGSGGGGSQVQLVQSGAEVKKPGASVKVSCTASGYTFTH
YGMNWVRQAPGKGLEWMGWINTYTGEPTYADKFKDRFTLSRETSASTVYMELTSLTS
DDTAVYYCARERGDAMDYWGQGTLVTVSS
SEQ ID NO:21, TB-A H_A76I/S77G, also named TB-A H76/77 . DIVMTQSPSSLSASVGDRVTLTCTASQSVSNDVVWYQQRPGKAPK LLIYSAFNRYTGVPSRFSGRGYGTDFTLTISSLQPEDVAVYYCQQDYNSPRTFGQGT KLEVKRGGGGSGGGGSGGGGSGGGGSQVQLVQSGAEVKKPGASVKVSCTASGYTFTH YGMNWVRQAPGKGLEWMGWINTYTGEPTYADKFKDRFTFSLETSIGTVYMELTSLTS DDTAVYYCARERGDAMDYWGQGTLVTVSS SEQ ID NO:22 TB-A L_L46R, also named TB-A L46
DIVMTQSPSSLSASVGDRVTLTCTASQSVSNDVVWYQQRPGKAPK
RLIYSAFNRYTGVPSRFSGRGYGTDFTLTISSLQPEDVAVYYCQQDYNSPRTFGQGT
KLEVKRGGGGSGGGGSGGGGSGGGGSQVQLVQSGAEVKKPGASVKVSCTASGYTFTH
YGMNWVRQAPGKGLEWMGWINTYTGEPTYADKFKDRFTFSLETSASTVYMELTSLTS DDTAVYYCARERGDAMDYWGQGTLVTVSS
SEQ ID NO: 23 TB-A L_R65S, also named TB-A L65
DIVMTQSPSSLSASVGDRVTLTCTASQSVSNDVVWYQQRPGKAPK LLIYSAFNRYTGVPSRFSGSGYGTDFTLTISSLQPEDVAVYYCQQDYNSPRTFGQGT KLEVKRGGGGSGGGGSGGGGSGGGGSQVQLVQSGAEVKKPGASVKVSCTASGYTFTH YGMNWVRQAPGKGLEWMGWINTYTGEPTYADKFKDRFTFSLETSASTVYMELTSLTS DDTAVYYCARERGDAMDYWGQGTLVTVSS
SEQ ID NO: 24 TB-A L_Y67S, also named TB-A L67
DIVMTQSPSSLSASVGDRVTLTCTASQSVSNDVVWYQQRPGKAPK
LLIYSAFNRYTGVPSRFSGRGSGTDFTLTISSLQPEDVAVYYCQQDYNSPRTFGQGT KLEVKRGGGGSGGGGSGGGGSGGGGSQVQLVQSGAEVKKPGASVKVSCTASGYTFTH
YGMNWVRQAPGKGLEWMGWINTYTGEPTYADKFKDRFTFSLETSASTVYMELTSLTS
DDTAVYYCARERGDAMDYWGQGTLVTVSS
SEQ ID NO: 25 VH of TB-A with D66G
QVQLVQSGAEVKKPGASVKVSCTASGYTFTHYGMNWVRQAPGKGL EWMGWINTYTGEPTYADKFKGRFTFSLETSASTVYMELTSLTSDDTAVYYCARERGD AMDYWGQGTLVTVSS
SEQ ID NO : 26 VL of TB-A with V83F DIVMTQSPSSLSASVGDRVTLTCTASQSVSNDWWYQQRPGKAPK LLIYSAFNRYTGVPSRFSGRGYGTDFTLTISSLQPEDFAVYYCQQDYNSPRTFGQGT KLEVKR
SEQ ID NO: 27 VL of TB-A with V83A DIVMTQSPSΞLSASVGDRVTLTCTASQSVSNDVVWYQQRPGKAPK
LLIYSAFNRYTGVPSRFSGRGYGTDFTLTISSLQPEDAAVYYCQQDYNSPRTFGQGT KLEVKR
SEQ ID NO:28 VH of TB-A H43/70/71/73/77
QVQLVQSGAEVKKPGASVKVSCTASGYTFTHYGMNWVRQAPGQGL EWMGWINTYTGEPTYADKFKDRFTLTLDTSAGTVYMELTSLTSDDTAVYYCARERGD AMDYWGQGTLVTVSS
SEQ ID NO: 29 VH of TB-A H43/70/71 QVQLVQSGAEVKKPGASVKVSCTASGYTFTHYGMNWVRQAPGQGL
EWMGWINTYTGEPTYADKFKDRFTLTLETSASTVYMELTSLTSDDTAVYYCARERGD AMDYWGQGTLVTVSS
SEQ ID NO: 30 VH of TB-A
Hll/16/43/66/70/71/73/77 /93/112
QVQLVQSGAEDKKPGGSVKVSCTASGYTFTHYGMNWVRQAPGQGL EWMGWINTYTGEPTYADKFKGRFTLTLDTSAGTVYMELTSLTSDDTATYYCARERGD AMDYWGQGTSVTVSS
SEQ ID NO: 31 TB-A H_M48L/F68I
DIVMTQSPSSLSASVGDRVTLTCTASQSVSNDVVWYQQRPGKAPK
LLIYSAFNRYTGVPSRFSGRGYGTDFTLTISSLQPEDVAVYYCQQDYNSPRTFGQGT
KLEVKRGGGGSGGGGSGGGGSGGGGSQVQLVQSGAEVKKP.GASVKVSCTASGYTFTH YGMNWVRQAPGKGLEWLGWINTYTGEPTYADKFKDRITFSLETSASTVYMELTSLTS
DDTAVYYCARERGDAMDYWGQGTLVTVSS
SEQ ID NO: 32 TB-A L_V83E H_V79A
DIVMTQSPSSLSASVGDRVTLTCTASQSVSNDWWYQQRPGKAPK
LLIYSAFNRYTGVPSRFSGRGYGTDFTLTISSLQPEDEAVYYCQQDYNSPRTFGQGT KLEVKRGGGGSGGGGSGGGGSGGGGSQVQLVQSGAEVKKPGASVKVSCTASGYTFTH
YGMNWVRQAPGKGLEWMGWINTYTGEPTYADKFKDRFTFSLETSASTAYMELTSLTS
DDTAVYYCARERGDAMDYWGQGTLVTVSS
SEQ ID NO: 33 TB-A Linker_G2R H_F68L DIVMTQSPSSLSASVGDRVTLTCTASQSVSNDVVWYQQRPGKAPK LLIYSAFNRYTGVPSRFSGRGYGTDFTLTISSLQPEDVAVYYCQQDYNSPRTFGQGT KLEVKRGRGGSGGGGSGGGGSGGGGSQVQLVQSGAEVKKPGASVKVSCTASGYTFTH YGMNWVRQAPGKGLEWMGWINTYTGEPTYADKFKDRLTFSLETSASTVYMELTSLTS DDTAVYYCARERGDAMDYWGQGTLVTVSS
SEQ ID NO: 34 TB-A H_K43R/F68I
DIVMTQSPSSLSASVGDRVTLTCTASQSVSNDVVWYQQRPGKAPK LLIYSAFNRYTGVPSRFSGRGYGTDFTLTISSLQPEDVAVYYCQQDYNSPRTFGQGT
KLEVKRGGGGSGGGGSGGGGSGGGGSQVQLVQSGAEVKKPGASVKVSCTASGYTFTH
YGMNWVRQAPGRGLEWMGWINTYTGEPTYADKFKDRITFSLETSASTVYMELTSLTS
DDTAVYYCARERGDAMDYWGQGTLVTVSS
SEQ ID NO: 35 TB-A H_F68L DIVMTQSPSSLSASVGDRVTLTCTASQSVSNDVVWYQQRPGKAPK
LLIYSAFNRYTGVPSRFSGRGYGTDFTLTISSLQPEDVAVYYCQQDYNSPRTFGQGT KLEVKRGGGGSGGGGSGGGGSGGGGSQVQLVQSGAEVKKPGASVKVSCTASGYTFTH YGMNWVRQAPGKGLEWMGWINTYTGEPTYADKFKDRLTFSLETSASTVYMELTSLTS DDTAVYYCARERGDAMDYWGQGTLVTVSS SEQ ID NO: 36 TB-A H__F68A
DIVMTQSPSSLSASVGDRVTLTCTASQSVSNDWWYQQRPGKAPK
LLIYSAFNRYTGVPSRFSGRGYGTDFTLTISSLQPEDVAVYYCQQDYNSPRTFGQGT
KLEVKRGGGGSGGGGSGGGGSGGGGSQVQLVQSGΆEVKKPGASVKVSCTASGYTFTH
YGMNWVRQAPGKGLEWMGWINTYTGEPTYADKFKDRATFSLETSΆSTVYMELTSLTS DDTAVYYCARERGDAMDYWGQGTLVTVSS
SEQ ID NO: 37 TB-A H_F68V/F70L
DIVMTQSPSSLSASVGDRVTLTCTASQSVSNDWWYQQRPGKAPK
LLIYSAFNRYTGVPSRFSGRGYGTDFTLTISSLQPEDVAVYYCQQDYNSPRTFGQGT
KLEVKRGGGGSGGGGSGGGGSGGGGSQVQLVQSGAEVKKPGASVKVSCTASGYTFTH YGMNWVRQAPGKGLEWMGWINTYTGEPTYADKFKDRVTLSLETSASTVYMELTSLTS
DDTAVYYCARERGDAMDYWGQGTLVTVSS
SEQ ID NO: 38 TB-A H_F70L
DIVMTQSPSSLSASVGDRVTLTCTASQSVSNDVVWYQQRPGKAPK
LLIYSAFNRYTGVPSRFSGRGYGTDFTLTISSLQPEDVAVYYCQQDYNSPRTFGQGT KLEVKRGGGGSGGGGSGGGGSGGGGSQVQLVQSGAEVKKPGASVKVSCTASGYTFTH
YGMNWVRQAPGKGLEWMGWINTYTGEPTYADKFKDRFTLSLETSASTVYMELTSLTS
DDTAVYYCARERGDAMDYWGQGTLVTVSS
SEQ ID NO: 39 Linker G2R • GRGGSGGGGSGGGGSGGGGS SEQ ID NO: 40 TB-A
DIVMTQSPSSLSASVGDRVTLTCTASQSVSNDVVWYQQRPGKAPK LLIYSAFNRYTGVPSRFSGRGYGTDFTLTISSLQPEDVAVYYCQQDYNSPRTFGQGT KLEVKRGGGGSGGGGSGGGGSGGGGSQVQLVQSGAEVKKPGASVKVSCTASGYTFTH YGMNWVRQAPGKGLEWMGWINTYTGEPTYADKFKDRFTFSLETSASTVYMELTSLTS DDTAVYYCARERGDAMDYWGQGTLVTVSS
The invention will be more fully understood by reference to the following examples. They should not, however, be construed as limiting the scope of the invention. All literature and patent citations are incorporated herein by reference.
Experiment 1 : Construction of scFv antibodies
The starting material for the generation of humanised anti-human TNFα antibodies or antibody derivatives, such as single-chain fragments (scFv) or Fab fragments, was the murine monoclonal antibody Di62. The sequences of the variable region of the light chain and the heavy chain are disclosed in During et al . (1994, MoI. Immunol. 31:1059-1067). The properties of this monoclonal antibody are also discussed in the same publication. Briefly, Di62 specifically binds to human TNFα in a concentration-dependent manner. It is a high affinity antibody (Kd = 0.4nM) and can block TNFα binding to its receptor. In addition, Di62 inhibits human TNFα- induced cytotoxicity in mouse L929 cells.
Based on its published sequence Di62 was constructed in the form of a single-chain antibody derivative (scFv) in the orientation VL~linker-VH, in which the linker sequence is composed of four repeats of four glycine and one serine residue (Gly4Ser)4. Herein, this scFv is referred to as TB-wt, with a VL of SEQ ID NO: 16 and a VH of SEQ ID NO: 17.
To humanise this antibody derivative for the purpose of a) render it more similar to human sequences . in order to minimise potential immunogenicity, and b) render it more stable and soluble, TB-wt CDR sequences were grafted on stable and soluble human frameworks (Au-f der Maur et al . (2001), FEBS Lett. 508:407-412; Auf der Maur et al . (2004), Methods 34:215-224). The human VL- kappa subgroup I and VH subgroup I were identified as nearest human subfamily. The appropriate acceptor framework was chosen from a pool of human VL and VH sequences, selected for advantageous biochemical and biophysical properties, such as for example stability, solubility, and expression properties (Auf der Maur, et al. (2001), FEBS Lett. 508:407-412; Auf der Maur, et al . (2004), Methods 34:215-224). The isolation and properties of these antibody frameworks are described in WO03097697/EP1506236. From this pool, a single-chain antibody framework with undefined antigen binding properties was identified as suitable acceptor. This acceptor consists of a human VL-kappa I domain (SEQ ID NO: 14) in combination with a human VHI domain (SEQ ID NO: 15) . Herein, this acceptor framework is referred to as FW2.3. Among 81 VL framework residues, TB-wt and FW2.3 share 55 identical amino acid residues, which amounts to 67% identity. Among the 87 VH framework residues, TB-wt and FW2.3 have 55 identical residues, corresponding to 63% identity. Both single-chain antibody derivatives have identical CDR lengths, apart from the VH-CDR3, which is longer in FW2.3. The amino acid composition within the
CDR residues is different for both scFvs. Various methods for the humanisation of antibody variable domains are . described (Riechmann et al . (1998), Nature 332:323-327; Padlan, E. A. (1991). MoI. Immunol. 28:489-498; Roguska et al. (1994), Proc. Natl. Acad. Sci. USA 91:969-973;
Gonzales et al . (2005), Tumor Biol. 26:31-43; Ewert, S., et al. (2004), Methods 34:184-199). The minimal approach, namely the conservative transfer of all mouse CDR loops from TB-wt onto FW2.3 was carried out first. The resul- ting scFv is- referred to as TB-B and has the VL sequence of SEQ ID NO: 3 and the VH sequence of SEQ ID NO: 4. The CDR-loops in TB-wt were defined according to the Rabat' ■ numbering scheme (Rabat et al . (1991), Sequences of Proteins of Immunological Interest, 5th Ed , Natl. Inst. Health, Bethesda, MD) and confine the following residues (see Figure 1) : .VL
CDRl: L24-L34 (same Kabat numbering) CDR2 : L50-L56 (same Kabat numbering) CDR3: L89-L97 (same Kabat numbering) VH CDRl: H31-H35 (same Kabat numbering)
CDR2: H50-H66 (Kabat numbering H50-H65) CDR3: H99-H106 (Kabat numbering H95-H102) . This antibody was unable to bind TNFα efficiently (Fig. 4A) . The next step was to determine which residue or residues from these components- should be substituted to optimise the properties of the resulting humanised antibody.
Since substitution of human amino acid residues with other amino acids should be minimised because introduction of foreign amino acid sequences increases the risk of immunogenicity of the antibody or antibody derivative in humans (Gonzales et al . (2005), Tumor Biol. 26:31-43), several variants were constructed. One of said variants - herein referred to as TB-B L46 (VL SEQ ID NO: 11; VH SEQ ID NO: 4) was constructed with the aim to minimize the risk of being immunogenic but still showing sufficient binding activity. This variant is based on TB-B and contains one single amino acid change at position 46 in VL, namely R-^L. This amino acid is located within the upper core of the light chain and takes part in the dimer interface. It is involved in defining the conformation of L-CDRl and has an influence on VH/VL packing.. It was reported that a leucine residue is favoured at this particular position (PCT/US03/19333) . In contrast to TB-B, the scFv TB-B L46 retains some TNFα- specific binding. (Fig. 4A; Kd ? 100 nM)' ) . In order to further improve the TNFα-binding activity, one or more further exchanges were made at one or more of the .VL residues 4, 46, 65, 67, 70, and/or VH residues 28, 43, 68, 70, 71, 72, 73, 76, 77. Herein, the variant with exchanges in all positions is referred to as TB-A (VL SEQ ID NO:1; VH SEQ ID NO:2).
Furthermore, regarding particular anti-TNFα antibodies of the present invention, competition assays with peptides derived from L-CDRl and L-CDR2 showed that both CDR loops are important for the binding of the scFv to- the antigen (Dδring et al . (1994), MoI. Immunol. 31:1059-1067) .
In further experiments aimed at minimising the number of non-human residues required to retain binding .and at optimising biophysical properties
(stability and solubility) , systematic mutagenesis and domain shuffling was applied allowing to elucidate the functional differences between mouse and humanised VL and VH domains . Two variants were obtained by domain shuffling. The first variant is composed of the VL domain from TB-A connected via a glycine serine linker (SEQ ID NO: 10) with the VH domain from TB-B, resulting in TB-AB (SEQ ID NO: 12) . The second variant, TB-BA, is the reverse of the first variant, namely, the VL domain from TB-B in combination with the VH domain of TB-A (SEQ ID NO: 13) .
Additional variants were generated by systematic mutagenesis of TB-A. Fig. 1 shows a sequence comparison of the VL and VH sequences of TB-A and TB-B. A total of 14 framework residues differ between TB-A and TB-B (asterisks) . Only five of them, VL residues 4 and 70, and VH residues 28, 71, and 73 show only minor differences in size and property and therefore were not considered for mutagenesis at this point. The following framework positions- were replaced with the corresponding amino acid from TB-B. These single or double mutants of TB-A are as follows: .. . TB-A H43 K->Q interface (SEQ ID NO: 18) TB-A H68 F->V outer loop VH (SEQ ID NO: 19) TB-A H70/72 F->L,L->R outer loop VH (SEQ ID NO: 20) TB-A H76/77 A->I,S->G outer loop VH (SEQ ID NO:21) TB-A L46 L->R interface (SEQ ID NO:22) TB-A L65 R->S outer loop VL (SEQ ID NO: 23) TB-A L67 Y->S outer loop VL (SEQ ID NO: 24)
Many factors can influence the immunogenicity of an antibody or antibody derivative (Gonzales et al . (2005), Tumor Biol. 26:31-43). To further reduce the non- human content of the variable regions of the humanised TB-A scFv, the murine CDR2 and CDR3 loops of VL and murine CDR2 loop of VH were exchanged with the corresponding human CDR loops from FW2.3. The resulting constructs herein are referred to as TB_L2 (SEQ ID N0:7), TB_L3 (SEQ ID NO:8), and TB_H2 (SEQ ID NO:9), respectively.
The cDNAs encoding the murine single-chain version of the monoclonal antibody Di62, and the two humanised versions TB-B and TB-A were generated by gene synthesis (www.genscript.com). All the point mutations in the other variants (TB-B L46, TB-A H43, TB-A H67, TB-A H69/71, TB-A H75/76, TB-A L46, TB-A L65, TB-A L67, TB-A V83F, TB-A V83A, TB-A D66G) were introduced by PCR-driven site-directed mutagenesis following standard cloning procedures. Exchange of the murine CDR loops of TB-A with the human CDR loops from FW2.3 was accomplished using PCR and state of the art cloning procedures . The cDNA encoding all the VH TB-A variations as disclosed in SEQ ID NO.: 28, SEQ ID NO: 29, and SEQ ID NO: 30 were realized by complete gene synthesis.
Some further preferred TB-A are disclosed in SEQ ID NO: 31 to SEQ ID NO: 38. These antibodies were found to be particularly stable and soluble, as shown below in Table I. All scFv fragments were cloned into an expression vector for periplasmic production in E. coli (Krebber et al . (1997), J. Immunol. Methods 201:35-55).
In addition to the single-chain antibody derivatives (scFvs) described above, the corresponding Fab fragments were generated as follows . Selected light chain variable domains (VL) were fused to the constant region of a human Ig kappa chain, while the suitable heavy chain variable domains (VH) were fused to the first (N-terminal) constant domain (CHl) of a human IgG. Both human constant domains were amplified by PCR from a human spleen cDNA library and resulted in the sequence SEQ ID NO: 14 for CK- and SEQ ID NO: 15 for CHl.
Experiment 2: Expression, production and stability of humanised scFv or Fab antibodies
Plasmids encoding TB-wt, its humanised derivatives, or Fab fragments were introduced into a suitable E. coli strain (e.g. JM83) for periplasmic expression. The scFv variants were also expressed as inclusion bodies, for example in the BL21 E. coli strain. Functional single-chain antibodies were obtained by refolding of inclusion bodies and subsequent purification by, for example, gel filtration. The expression yields upon periplasmic expression of the scFvs ranged between 0.5 mg up to 12 mg per litre culture under standard laboratory cultivation conditions (dYT medium, with approx. 3 hours induction- time at 30°C, shaking at 200 rpm) with conventional ' . shaking flasks. In general, we observed that, as expected from our previous analysis of frameworks selected for stability and solubility (Auf der Maur, et al . (2004), Methods 34:215-224), the closer the sequence of a humanised derivative is to the sequence of the acceptor framework (FW2.3), the higher is the yield obtained upon expression in bacteria. For example, the yield obtained from expression of TB-B is far better than that obtained from expression of TB-A. In accordance with these findings, reducing the number of differing amino acid . residues present in TB-A had a positive effect toward the expression yields (Fig. 3A) . Another important characteristic of the antibodies or antibody derivatives of the present invention is their solubility. Fig. 3B shows the superiority of the framework TB-A over the donor framework (TB-wt) in terms of solubility in phosphate buffered saline. In analytical gel filtration, TB-A m§igrates predominantly in a monomer state (peak at 70ml) whereas TB-wt shows a strong tendency to form aggregates (peak at 50ml) . In addition to that, maximal solubility of TB-A and certain derivatives thereof was assessed by PEG precipitation (Athat .DH.. et al . JBC. 1981, 256;23. 12108-12117).
Briefly, the apparent solubility of test proteins was measured in the presence of polyethylene glycol 3000 (PEG3000) . Solubility curves were determined by measuring protein concentration in the supernatant of centrifuged protein-PEG300 mixtures. All curves showed a linear dependence of log S (mg/ml) on PEG3000 concentration (%, w/v) . Maximal solubility (Smaχ) ■ of a test protein was estimated by extrapolation of the linear regression towards 0% PEG (Tab. I) . For TB-A was Smax calculated to be about 70 mg/ml. All test proteins showed exceptionally good solubility. In a second approach a method called self-interaction -chromatography (SIC) was applied to assess intermolecular attraction / repulsion of TB-A (SEQ ID NO:40), TB-A Linker_G2R H_F68L (SEQ ID NO:33), TB-A " H_K43R/F68I (SEQ ID NO:34) and TB-A H_F68L (SEQ ID NO: 35) at a concentration of 1 mg/ml in PBS (50 mM phosphate pH 6.5, 150 mM NaCl) . In this method the protein of interest is immobilized onto a porous stationary phase and packed into a column. Interactions between the free (mobile phase) and immobilized protein are detected as shifts in the retention volume. The protein osmotic second virion coefficient B22, which is a measure for intermolecular attraction/repulsion, was calculated according to Tessier, PM et al .. Biophys . J. 2002, 82: 1620-1632 (Tab. I) . The more positive B22 / the lower is the intermolecular attraction of the test protein and, therefore, the higher is its solubility. Due to the high, similarity of test protein sequences it was assumed that B22 values of the different proteins can be directly compared to each other.
Table I : Solubility features of TB-A derivatives
Sequence pi Log B22 value (SIC)
TB-A 7.8 1.84 ± 0.13 -24.5xlO~4 + 3.8xlO~4
TB-A _M48L/F68I 7.8 nd nd
TB-A L__ V83E H_V79A 6.58 nd nd
TB-A Linker G2R 8.2 1.91 ± 0.09 1.59xlO~3 + 5 .9xlO~5 H_F68L
TB-A H_ JK43R/F68I 7.8 1.86 ± 0.02 1.28xlO"3 ± 3 .OxIO"4
TB-A H_ F68L 7.8 1.88 ± 0.07 1.06xl0"4 ± 2 .9xlO~5
TB-A H_ F68A 7.8 nd nd
TB-A H__ _F68V/F70L 7.8 nd nd
TB-A H_ _F70L 7..8 nd nd
Yet another relevant feature of the antibodies or antibody derivatives of the present invention is their high stability. Protein stability of TB-A, TB-A H_M48L/F68I (SEQ ID NO: 31), TB-A Linker_G2R H_F68L (SEQ ID N0:33), TB-A H_K43R/F68I (SEQ ID NO:34) and TB-A H_F68L (SEQ ID NO: 35) was assessed by determining the temperature for onset of unfolding by circular dichroism and light scattering at both 218 and 292 nm (Tab. II) . In this 'experiment TB-A started to unfold at a temperature of 5-30C whereas its derivatives TB-A H_M48L/F68I (SEQ ID NO:31), TB-A Linker G2R H F68L (SEQ ID NO:33), TB-A H__K43R/F68I (SEQ ID NO:34) and TB-A H_F68L (SEQ ID NO:35) showed increased thermal stability (56 and 58°C) . All test proteins showed irreversible denaturation and precipitated upon unfolding, making it impossible to determine the melting temperature. In order to determine midpoint of transition in a reversible process, unfolding was 'induced with guanidine hydrochloride (GdnHCl) , to keep the unfolded proteins in solution. In this approach fluorescence emission maxima where determined in by fluorimetry to follow unfolding. In this set-up, TB-A showed again good stability with a midpoint of transition at 2.07 M GdnHCl. In line with the results from thermal unfolding, the derivatives TB-A Linker_G2R H_F68L (SEQ ID NO: 33) and TB-A H_K43R/F68I (SEQ ID NO: 34) showed increased stability as displayed with higher midpoints of transition, 2.33 and 2.3 M GdnHCl, respectively.
Table II: Stability features of TB-A derivatives
Figure imgf000057_0001
The stability of TB-A in human serum, human urine, pig vitreous body fluid and pig anterior chamber fluid was assayed by measuring TNFα binding activity of TB-A after incubation for 3 days at 370C in the respective body fluid or in assay buffer (TBSTM) as a positive control. TB-A was diluted in body fluids to a final concentration of 10 μM. After the incubation period dilution series of the samples were assayed by ELISA in order to determine the binding constant Kd of TB-A (Fig. 11) . When comparing body fluid samples with the positive control TBSTM samples, a shift of the Kd towards higher concentrations would indicate a decrease of active protein during the incubation period. In our experiments, however, no such shift was detectable, indicating that the amount of fully active TB-A remained constant in every body fluid assay due to a high stability of the antibody. _ . .. . . ..
Experiment 3 : Binding, features of humanised antibody derivatives
The binding properties of all humanised scFv variants were tested in ELISA on recombinant human TNFα. The dissociation constants (Kd) for all variants lay within a range of 0.8 to more than 10' 00OnM. There seems to exist a reverse correlation between the grade of homology to the human acceptor framework and the affinity of the respective binder (Fig. 4A) . Nevertheless, some TB-A variants containing mutations towards the TB-B sequence show affinity levels towards human TNFα that are comparable to that of TB-A. Fig. 4B shows two expression yield-improved derivatives of TB-A (compare Fig. 3A) that exhibit similar affinities as TB-A when compared in ELISA.
TB-A represents a good compromise between the apparent trade-off of expression yield and affinity. In terms of affinity, no significant difference between the single-chain and the Fab fragment format of TB-A was detectable (data not shown) .
The affinity for TNFα and binding kinetics were also determined for TB-A by surface plasmon resonance (BIACore) , resulting in a dissociation constant Ka = 0.8 nM, an off rate of kOff = 4.4 x 10"4S"1 and a on rate of kon = 5 x 105S-1M"1.
Experiment 4: L929 cytotoxicity assay
The function' of antibodies or antibody derivatives in neutralising TNFα in vivo can be tested by measuring inhibition of cytotoxicity of TNFα towards cultured mouse L929 fibroblasts or alternatively towards human KYM-I myelosarcoma cells (Tab. Ill) . The humanised scFv derivatives of Di62 display different efficacies in the L929 assay as shown in Fig. 5B. Some scFv derivatives show IC50 (inhibitory concentration to achieve 50% inhibition) values in the range of 5ng/ml, whereas others had no effect in the L929 assays. ELISA data and results from the L929 assay do not always correlate. KYM-I data and L929 results, however, correlated nicely with the only difference that much higher concentrations of recombinant human TNF and consequently also of the antagonist where required to see an effect. KYM-I was, therefore, mainly used to confirm L929 results. For direct comparison of test proteins, potency is expressed as a relative value normalized to TB-A (EC50X / EC50TB-A) . In general, however, IC50 values become again higher the closer the sequence of a binder is to the human acceptor framework (FW2.3). Fig. 5 compares the potency of different derivatives of TB-A to block TNFα induced cytotoxicity towards mouse L929 fibroblast cells. Absorption at 450nm correlates with cell survival. TB-A and the anti-hTNFα IgG Infliximab® show a similar IC50 value in the L929 assay, whereas the potency of TB-wt to block human TNFα induced cytotoxicity is significantly lower (Fig. 5A). When TB-A derivatives are compared with TB-A for their potential to block TNFα induced cytotoxicity, most of these derivatives except TB-H43 have a .reduced efficacy in the L929 (Fig. 5B) . Table III: Functional properties of TB-A derivatives
Figure imgf000060_0001
In line with the ELISA data, there is no significant difference in the ability to block TNFα- induced cytotoxicity between the scFv and the Fab format of TB-A (Fig. 5C) . The IC50 value of the TB-A Fab format lies about a factor of two above the ICs0 value of the TB- A scFv format (Fig. 5C), most probably as a result of the higher molecular mass of the Fab fragment.
Experiment 5. Animal experiments with anti- TNFα antibody derivatives
5.1. Description of experiment
In order to test for the efficacy of
ESBATech' s anti-TNFα antibody derivatives (scFv and Fab) in functionally neutralising human TNFα bioactivity in an in vivo situation, a recently published rat model for acute monoarthritis was used. This model has extensively been described by Bolon and colleagues (see Bolon et al . (2004), Vet. Pathol. 41:235-243). Briefly, in this animal arthritis model, human TNFα is injected intra-articularly into the knee joint of male Lewis rats. Injection of human TNFα leads to an acute, self-limiting monoarthritis in the injected joint. Arthritis can be quantified by measurement of joint swelling and histological scoring. Consequently, bioactivity of respective TNFα antagonists can be quantified by reduction of TNFα-induced joint swelling and/or reduction of histological parameters of inflammation .
5.2. Materials and Methods
Experimental Design
The current studies were designed to examine the respective potential of a representative scFv antibody and a representative Fab antibody of the series described above with the marketed antibody Infliximab (Remicade®) to inhibit bioactivity of human TNFα in an appropriate ■ animal arthritis model. Bolon and colleagues had shown before that intra-articular application of 10 microgram of recombinant human TNFα into the rat knee joint provokes an acute, self-limiting monoarthritis that can be quantified by standard macroscopic and microscopic analysis. Thus this animal model served as ideal system to assess the therapeutic effect of locally applied antibody derivatives. Two experiments were completed in series (Tab. IV and V) . 1) A basic efficacy study assessing the overall potential of the antibodies to block human TNFα-induced monoarthritis; and 2) A dose response study assessing the relative efficacy of the antibody derivatives among each other. Both, cytokines and antibody derivatives were given once by separate injections, as described below. The cytokine dose used was based on -the publication by Bolon and colleagues, whereas the range of doses- of the antibody derivatives was based on available cell culture data and educated guessing. The experiments were conducted in accordance with general animal care guidelines . Animals and husbandry
Young, adult male Lewis rats (6-7 weeks and 175-20Og) were randomly assigned to treatment groups (n=3/cohort) and housed according to Bolon et al . (2004), Vet. Pathol. 41:235-243.
Cytokine and antibody instillation Anaesthesia and cytokine injections were performed as described by Bolon and colleagues . In order not to exceed a total intraarticularly injected volume of 50 microliter, cytokines and antibody derivatives were instilled in two separate intra-articular injections whereby the 10 micorgrams of recombinant human TNFα was injected in 10 microliter of filter-sterilised phosphate- buffered saline (PBS) and the respective dose of the respective antibody was injected in 40 microliter of filter-sterilised phosphate-buffered saline. Animals treated with intraperitoneally applied Infliximab/ Remicade® were i.p. injected with the antibody derivatives 3 hours prior to intra articular injection of the human TNFα. In all animals treated with intraarticularly applied antibody treatments, the respective antibody dose was injected 5 minutes prior to injection of the human TNFα. Control animals were injected with 10 microliter of PBS without human TNFα. Infliximab/Remicade® used in the experiments was purchased at an official Swiss pharmacy. Anti-human TNFα specific scFv and Fab (TB-A) antibodies as well as a naϊve scFv antibody framework used as unspecific control antibody in the dose response experiment were expressed- in E. coli and purified by standard methods. Endotoxin contamination was held below 10 EU per milligram protein- in all preparations because the lipopolysaccharide component is a potent inducer of TNFα.
Recombinant human TNFα was purchased from PeproTech EC Ltd. Measurement of joint diameter
Immediately before the injection of the respective, intraperitoneally or intraarticularly applied antibody derivative, or, in case of the control animals, before the injection of PBS or TNFα, the diameter of the knee joint to be injected was determined by means of a standard calliper. 48 hours after injection of TNFα (or PBS in control animals) and immediately before euthanisation of the animals, the diameter of the injected knee joint was determined again and joint swelling was calculated by subtracting the value of the second diameter measurement from the value of the first diameter measurement (Fig. 6 and 9) . Tissue processing 48 hours after injection of TNFα (or PBS in case of control animals) animals were euthanised. At necropsy, injected knees were separated from the foot and thigh, fixed intact by immersion in 70% ethanol and proceeded for standard hematoxylin and eosin (HE) staining, as described by Bolon and colleagues. Morphologic analysis
Histological scoring analysis for measurement of joint inflammation was performed as described by Bolon and colleagues. Histopathological scoring criteria for assessment of joint inflammation were applied according to Bolon and colleagues (Fig. 7, 8 and 10) .
5.3. Results
In a first set of experiments a representative intraarticularly applied ESBATech scFv antibody, TB-A, and the corresponding intraarticularly applied ESBATech Fab antibody were compared for their ability to block induction of the acute monoarthritis with intraarticularly and intraperitoneally applied Infliximab/Remicade® according to Table IV: Table IV: Injection scheme experiment 1
Figure imgf000064_0001
The results obtained regarding treatment effects on change of knee diameter (as an indicator of ' effects on TNFα-induced joint swelling) are represented in Fig. 6. All antibodies completely blocked TNFα-induced joint swelling.
For evaluation of treatment effects on joint inflammation, histological scoring of HE-stained tissue slides was performed. Joint inflammation was scored by the following criteria (see Fig. 7 for representative scoring examples) :
Score 0 : normal
Score 1: Mild thickening of synovial lining
Score 2: Thickening of synovial lining and mild inflammation of the sublining Score 3: Thickening of synovial lining and moderate inflammation of the sublining
The results obtained regarding treatment effects on histopathological inflammation scores are shown in Fig . 8. Comparable effects of all treatment on histopathological inflammation scores were observed.
In a second set of experiments, the relative dose response to the assessed antibody derivatives was compared. The representative intraarticularly applied ESBATech scFv antibody TB-A and the corresponding intraarticularly applied Fab antibody of experiment 1 were compared with intraarticularly and intraperitoneally applied Infliximab/Remicade® and an unrelated scFv antibody lacking any binding activity to human TNFα over a broader and different dose range as compared with experiment 1, according to Table V. Table V: Injection scheme experiment 2
Figure imgf000066_0001
The results obtained regarding treatment effects on change of knee diameter (as an indicator of effects on TNFα-induced joint swelling) are shown in Fig.. 9. The results obtained regarding treatment effects on histopathological inflammation scores are presented in Fig. 10.
In summary, both the representative ESBATech anti-TNFα scFv and the representative ESBATech anti-TNFα Fab antibody were highly efficient in blocking human TNFα-induced monoarthritis upon local (intraarticular) administration .
While there are shown and described presently preferred embodiments of the invention, it is to be distinctly understood that the invention is not limited thereto but may be otherwise variously embodied and prac- ticed within the scope of the following claims.

Claims

Claims
1. A stable and soluble antibody or antibody derivative which specifically binds TNFα, said antibody or antibody derivative comprising a light chain variable domain (VL) -being or derived from the sequence SEQ ID N0:l that is combined with a heavy chain variable domain
(VH) being or derived from the sequence SEQ ID NO: 2, wherein in the case of a derived sequence said sequence has at maximum up to 5 changes within the framework of said VL domain and/or at maximum up to nine changes within the framework of said VH domain.
2. The antibody or antibody derivative of claim 1, wherein the changes of VL are at one or more of the positions 4, 46, 65, 67, 70, and/or 83 and the changes of VH are at one of the positions 11, 16, 28, 43, 68, 70, 71, 72, 73, 76, 77, 93 and/or 112.
3. The antibody or antibody derivative of claim 1, wherein the changes of VL are at one or more of- the positions 4, 46, 65, 67, 70, and/or 83 and the changes of VH are at one or more of the positions. 11, 16, 28, 43, 48, 68, 70, 71, 72, 73, 76, 77, 79, 93 and/or 112.
4. The antibody or antibody derivative of anyone of the preceding claims, in which at least one of the conversions leads to an amino acid present in SEQ ID NO: 3 for VL and/or SEQ ID NO: 4 for VH.
5. The antibody or antibody derivative of anyone of the preceding claims, wherein the VL domain comprises the sequence SEQ ID NO: 1.
6. The antibody or antibody derivative of claim 5 comprising the VH domain of the sequence SEQ ID NO: 2.
7. The antibody or antibody derivative of claim 5 comprising a VH domain derived from the sequence SEQ ID NO: 2, wherein F68 is changed to A, L, I, or V.
8. The antibody or antibody derivative of claim 1 comprising the VL domain of the sequence SEQ ID NO: 11, and the VH domain of the sequence SEQ ID NO: 4.
9. The antibody or antibody derivative of anyone of the preceding claims, wherein at least one amino acid residue is converted in at least one of the CDRs to a residue present in the corresponding CDR of the VL sequence SEQ ID NO: 5 and/or the VH sequence SEQ ID
No: 6 or SEQ ID No: 25.
10. The antibody or antibody derivative of claim 9, wherein at least one of the CDRs of the group VL CDR2, VL CDR3, VH CDR2 or VH CDR3 is converted to the corresponding CDR of the VL sequence SEQ ID NO: 5 and/or the VH sequence SEQ ID No: 25 or SEQ ID No: 6.
11. The antibody or antibody derivative of claim 10 with the VL sequence SEQ ID NO: 7 or SEQ ID NO: 8 and the VH sequence SEQ ID NO: 2.
12. The antibody or antibody derivative of claim 10 with the VL sequence SEQ ID NO:1 and the VH sequence SEQ ID NO: 4, or SEQ ID NO: 9, or SEQ ID NO: 28 or SEQ ID NO:29.
13. The antibody or antibody derivative of claim 10 with the VL sequence SEQ ID NO: 26 or SEQ ID NO: 27 and the VH sequence SEQ ID NO: 30.
14. The antibody or antibody derivative of anyone of the preceding claims having a specificity to human TNFα.
15. The antibody derivative according to anyone of the preceding claims, which is an scFv antibody, wherein the VL and VH domains are connected by a linker.
16.- The scFv antibody according to claim 15 comprising a VL-linker-VH sequence arrangement.
17. The scFv antibody according to claim 15 or 16, wherein ' the linker has the sequence SEQ ID NO: 10 or is -derived -from said- -sequence-.
18. The scFv antibody of claim 17, wherein at least one G of said linker is changed to a more polar or charged amino acid.
19. The scFv antibody of claim 18, wherein the linker has the sequence SEQ ID NO: 39.
20. The antibody derivative according to anyone of claims 1-14, which is a Fab fragment wherein the VL domain is fused to the constant region of a human Ig kappa chain, the VH domain is fused to the CHl domain of a human IgG, and the two fusion polypeptides are connected by an inter-chain disulfide bridge.
21. The -scFv antibody or Fab fragment according to any one of claims "1-20, which is labeled or . chemically modified.
1 22. A DNA sequence encoding the antibody or antibody derivative according to any one of claims 1-2.0.
23. A cloning or expression vector containing the DNA sequence according to claim 22.
24. A suitable host cell transformed with an expression vector according to claim 23.
25. The host cell of claim 24, being a prokaryotic or eukaryotic cell, in particular an E. colir yeast, plant, insect or a mammalian cell.
26. A method for the production of an antibody or antibody derivative molecule according to any¬ one of claims 1-20 comprising culturing of the host cell of claims 24 or 25 under conditions that allow the synthesis of said antibody molecule and recovering it from said culture.
27. The antibody or antibody derivative of any of claims 1-21 as a pharmaceutical. .
28. The use of an antibody or antibody derivative of. any of claims 1-21 for the production of a medicament for the treatment of or as an in vitro diagnostic for detection of TNFα-associated diseases, in particular osteoarthritis, uveitis, inflammatory bowel disease .
29. A diagnostic or therapeutic composition comprising • an antibody of any of claims 1-21 in combination with a pharmaceutically acceptable carrier, diluent or excipient. '
30. A combination preparation comprising the antibody of anyone of claims 1-21 together with at least a second compound.
31. The combination preparation of claim 30 wherein the second compound is not an antibody specific for TNFα.
32. The combination preparation of claim 30 or 31 for the use as a medicament.
33. The vector of claim 23 as a pharmaceutical for gene therapeutical applications in humans and/or in animals .
34. A method for the treatment of a TNFα related disease wherein an antibody or antibody derivative of anyone of claims 1-21 is locally or topically administered.
35. Use of any of the VL and/or VH sequences specified in anyone of claims 1-20 in an expression vector.
36. A stable aqueous pharmaceutical formulation comprising a therapeutically effective amount of an antibody or antibody derivative of any of claims 1- 21 not subjected to prior lyophilization, an acetate buffer from about pH 4.8 to about 5.5, a. surfactant and a polyol, wherein the formulation lacks a tonicifying amount of sodium chloride .
37. The formulation of claim 36 which is isotonic.
38. The formulation of claim 36 or 37 which is stable at a temperature of about 2-8° C for at least one year.
39. The formulation of anyone of claims 36 to 38 which is stable following freezing and thawing of the formulation.
40. The formulation of anyone of claims 36 to
39 which is stable at about 30° C for at least one month.
41. The formulation of anyone of claims 36 to
40 wherein the polyol is a non-reducing sugar.
42. The formulation of anyone of claims 36 to
41 wherein the non-reducing sugar is trehalose.
43. The formulation of anyone of claims 36 to
42 wherein the non-reducing sugar is sucrose.
44. The formulation of anyone of claims 36 to
43 wherein the antibody is an antibody fragment.
45. The formulation of claim 44 wherein the antibody fragment is a scFv.
46. The formulation of anyone of claims 36 to 45 wherein the antibody binds TNFα.
47. The formulation of anyone of claims 36 to 46 which is stable at a temperature of about 2-8° C for at least two years.
48. The formulation of anyone of claims 36 to
47 wherein the antibody concentration in the formulation is from about 0.1 to about 50 mg/ml.
49. The formulation of anyone of claims 36 to
48 wherein the surfactant is a polysorbate.
50. The formulation of claim 45 wherein the scFv binds TNFα.
51. The formulation of anyone of claims 36 to 50 wherein the acetate is present in an amount of about 5-30 mM.
52. The formulation of claim 51 wherein the acetate is present in an amount of 10-30 mM.
53. The formulation of anyone of claims 50 to 52 further comprising a preservative.
54. The formulation of claim 53 wherein the preservative is benzyl alcohol.
55. The formulation of anyone of claims 50 to 54 wherein the antibody is present in an amount of about - 30-50 mg/ml.
56. The formulation of claim 55 wherein the buffer is 10-30 mM sodium acetate at pH 5, the polyol is trehalose in an amount of about 2-10% w/v, the surfactant is polysorbate in an amount of about 0.01-0.1% v/v, wherein the formulation further comprises benzyl alcohol as a preservative and wherein the formulation is stable at a temperature of about 2-8° C for at least two years.
57. An article of manufacture comprising a container holding- a stable aqueous pharmaceutical formulation comprising a therapeutically effective amount of an antibody, not subjected to prior lyophilization, ' an acetate buffer from about pH 4.8 to about 5.5, a surfactant and a polyol, wherein the formulation lacks a tonicifying amount of sodium chloride.
58. The article of claim 57 wherein the antibody is an antibody or antibody derivative of anyone of claims 1-21. • ' "
PCT/CH2006/000300 2005-06-07 2006-06-06 STABLE AND SOLUBLE ANTIBODIES INHIBITING TNFα WO2006131013A2 (en)

Priority Applications (19)

Application Number Priority Date Filing Date Title
BRPI0611765-1A BRPI0611765B1 (en) 2005-06-07 2006-06-06 STABLE AND SOLUBLE ANTIBODY OR FRAGMENT THAT SPECIFICALLY BINDS TO TNF-ALFA ITS USES AND DIAGNOSTIC OR THERAPEUTIC COMPOSITION
EP06741625A EP1891110A2 (en) 2005-06-07 2006-06-06 STABLE AND SOLUBLE ANTIBODIES INHIBITING TNF-alpha
MX2015006717A MX342638B (en) 2005-06-07 2006-06-06 Stable and soluble antibodies inhibiting tnfî±.
MX2007015280A MX2007015280A (en) 2005-06-07 2006-06-06 Stable and soluble antibodies inhibiting tnf??.
KR1020087000199A KR101457223B1 (en) 2005-06-07 2006-06-06 STABLE AND SOLUBLE ANTIBODIES INHIBITING TNFα
KR1020147011105A KR101539545B1 (en) 2005-06-07 2006-06-06 STABLE AND SOLUBLE ANTIBODIES INHIBITING TNFα
JP2008515021A JP5089582B2 (en) 2005-06-07 2006-06-06 Stable soluble antibody that inhibits TNFα
CA2609999A CA2609999C (en) 2005-06-07 2006-06-06 Stable and soluble antibodies inhibiting tnf.alpha.
AU2006255415A AU2006255415B2 (en) 2005-06-07 2006-06-06 Stable and soluble antibodies inhibiting TNFalpha
CN200680028415.3A CN101233154B (en) 2005-06-07 2006-06-06 Suppress the stable of TNF α and soluble antibodies
NZ563580A NZ563580A (en) 2005-06-07 2006-06-06 Stable and soluble antibodies inhibiting TNFaplha
US11/916,793 US8067547B2 (en) 2005-06-07 2006-06-06 Stable and soluble antibodies inhibiting TNFα
IL187503A IL187503A0 (en) 2005-06-07 2007-11-20 Stable and soluble antibodies
US13/245,420 US8389693B2 (en) 2005-06-07 2011-09-26 Stable and soluble antibodies inhibiting TNFα
PH12012500758A PH12012500758A1 (en) 2005-06-07 2012-04-18 STABLE AND SOLUBLE ANTIBODIES INHIBITING TNFa
US13/708,500 US8691228B2 (en) 2005-06-07 2012-12-07 Stable and soluble antibodies inhibiting TNFα
US14/185,783 US9315572B2 (en) 2005-06-07 2014-02-20 Stable and soluble antibodies inhibiting TNF alpha
IL243270A IL243270B (en) 2005-06-07 2015-12-21 Stable and soluble antibodies inhibiting tnfα
US15/064,067 US9683034B2 (en) 2005-06-07 2016-03-08 Stable and soluble antibodies inhibiting TNFα

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US68797105P 2005-06-07 2005-06-07
US60/687,971 2005-06-07
US78535306P 2006-03-23 2006-03-23
US60/785,353 2006-03-23

Related Child Applications (3)

Application Number Title Priority Date Filing Date
US11/916,793 A-371-Of-International US8067547B2 (en) 2005-06-07 2006-06-06 Stable and soluble antibodies inhibiting TNFα
US91679308A A-371-Of-International 2005-06-07 2008-06-06
US13/245,420 Continuation US8389693B2 (en) 2005-06-07 2011-09-26 Stable and soluble antibodies inhibiting TNFα

Publications (2)

Publication Number Publication Date
WO2006131013A2 true WO2006131013A2 (en) 2006-12-14
WO2006131013A3 WO2006131013A3 (en) 2007-01-25

Family

ID=37193351

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CH2006/000300 WO2006131013A2 (en) 2005-06-07 2006-06-06 STABLE AND SOLUBLE ANTIBODIES INHIBITING TNFα

Country Status (21)

Country Link
US (5) US8067547B2 (en)
EP (3) EP3260465A1 (en)
JP (2) JP5089582B2 (en)
KR (2) KR101457223B1 (en)
CN (2) CN105153306B (en)
AU (1) AU2006255415B2 (en)
BR (1) BRPI0611765B1 (en)
CA (1) CA2609999C (en)
DK (1) DK2390267T3 (en)
ES (1) ES2424042T3 (en)
HK (2) HK1164891A1 (en)
HR (1) HRP20130632T1 (en)
IL (2) IL187503A0 (en)
MX (1) MX2007015280A (en)
NZ (1) NZ563580A (en)
PH (1) PH12012500758A1 (en)
PL (1) PL2390267T3 (en)
PT (1) PT2390267E (en)
RS (1) RS52861B (en)
RU (1) RU2415151C2 (en)
WO (1) WO2006131013A2 (en)

Cited By (67)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1532174A2 (en) * 2002-06-14 2005-05-25 Immunogen, Inc. Anti-igf-i receptor antibody
WO2009000099A2 (en) 2007-06-25 2008-12-31 Esbatech Ag Methods of modifying antibodies, and modified antibodies with improved functional properties
WO2009040361A1 (en) * 2007-09-25 2009-04-02 Intervet International B.V. Vaccine for the treatment of osteoarthritis
WO2009062690A1 (en) * 2007-11-12 2009-05-22 U3 Pharma Gmbh Axl antibodies
WO2009097704A1 (en) * 2008-02-05 2009-08-13 Esbatech Ag Antigen-binding polypeptides against cartilage degeneration
US20090311251A1 (en) * 2006-07-10 2009-12-17 Esbatech Ag Scfv antibodies which pass epithelial and/or endothelial layers
WO2009155723A2 (en) * 2008-06-25 2009-12-30 Esbatech, An Alcon Biomedical Research Unit Llc STABLE AND SOLUBLE ANTIBODIES INHIBITING TNFα
WO2009155726A2 (en) * 2008-06-25 2009-12-30 Esbatech, An Alcon Biomedical Research Unit Llc Humanization of rabbit antibodies using a universal antibody framework
EP2243031A1 (en) * 2008-01-21 2010-10-27 Centocor Ortho Biotech Inc. Methods of predicting antibody solubility
WO2010130751A1 (en) * 2009-05-11 2010-11-18 U3 Pharma Gmbh Humanized axl antibodies
EP2270053A1 (en) * 2009-05-11 2011-01-05 U3 Pharma GmbH Humanized AXL antibodies
US8389693B2 (en) 2005-06-07 2013-03-05 ESBATech, an Alcon Boimedical Research Unit, LLC Stable and soluble antibodies inhibiting TNFα
EP2649189A1 (en) * 2010-12-08 2013-10-16 AbbVie Inc. Tnf- binding proteins
US8637022B2 (en) 2008-06-30 2014-01-28 Esbatech, An Alcon Biomedical Research Unit Llc Functionalized polypeptides
US8697074B2 (en) 2008-07-10 2014-04-15 Esbatech, An Alcon Biomedical Research Unit Llc Methods and compositions for enhanced delivery of macromolecules
WO2014068132A1 (en) 2012-11-05 2014-05-08 Delenex Therapeutics Ag Binding members to il-1 beta
JP2015120746A (en) * 2008-06-25 2015-07-02 エスバテック − ア ノバルティスカンパニー エルエルシー Humanization of rabbit antibodies using universal antibody framework
WO2015144852A1 (en) * 2014-03-26 2015-10-01 Delenex Therapeutics Ag Binding members to tnf alpha
US9226983B2 (en) 2010-04-07 2016-01-05 Abbvie Inc. TNF-α binding proteins
US9556265B2 (en) 2008-06-25 2017-01-31 Esbatech, An Alcon Biomedical Research Unit Llc Solubility optimization of immunobinders
EP3165928A1 (en) * 2015-11-06 2017-05-10 Promise Advanced Proteomics A method for quantifying anti-tnf antibodies
EP3202786A2 (en) 2007-03-12 2017-08-09 ESBATech, an Alcon Biomedical Research Unit LLC Sequence based engineering and optimization of single chain antibodies
EP3219727A1 (en) 2016-03-17 2017-09-20 Tillotts Pharma AG Anti-tnf alpha-antibodies and functional fragments thereof
EP3219726A1 (en) 2016-03-17 2017-09-20 Tillotts Pharma AG Anti-tnf alpha-antibodies and functional fragments thereof
WO2017158084A1 (en) 2016-03-17 2017-09-21 Numab Innovation Ag ANTI-TNFα-ANTIBODIES AND FUNCTIONAL FRAGMENTS THEREOF
WO2017158101A1 (en) 2016-03-17 2017-09-21 Numab Innovation Ag ANTI-TNFα-ANTIBODIES AND FUNCTIONAL FRAGMENTS THEREOF
WO2017158079A1 (en) 2016-03-17 2017-09-21 Numab Innovation Ag Anti-tnfalpha-antibodies and functional fragments thereof
AU2016200367B2 (en) * 2006-07-10 2017-10-12 Novartis Ag scFv antibodies which pass epithelial and/or endothelial layers
US9908945B2 (en) 2007-06-25 2018-03-06 Esbatech, An Alcon Biomedical Research Unit Llc Sequence based engineering and optimization of single chain antibodies
EP3145487B1 (en) 2014-05-23 2018-08-22 Fresenius Kabi Deutschland GmbH Liquid pharmaceutical composition
EP3409688A1 (en) 2017-05-31 2018-12-05 Tillotts Pharma Ag Topical treatment of inflammatory bowel disease using anti-tnf-alpha antibodies and fragments thereof
EP3456739A1 (en) 2017-09-19 2019-03-20 Tillotts Pharma Ag Use of anti-tnfalpha antibodies for treating wounds
EP3459529A1 (en) 2017-09-20 2019-03-27 Tillotts Pharma Ag Preparation of sustained release solid dosage forms comprising antibodies by spray drying
EP3459527A1 (en) 2017-09-20 2019-03-27 Tillotts Pharma Ag Method for preparing a solid dosage form comprising antibodies by wet granulation, extrusion and spheronization
EP3459528A1 (en) 2017-09-20 2019-03-27 Tillotts Pharma Ag Preparation of solid dosage forms comprising antibodies by solution/suspension layering
US10370692B2 (en) 2013-12-20 2019-08-06 Hoffmann-La Roche Inc. Recombinant polypeptide production methods
US10426833B2 (en) 2014-05-23 2019-10-01 Fresenius Kabi Deutschland Gmbh Liquid pharmaceutical composition
US10493152B2 (en) 2014-05-23 2019-12-03 Fresenius Kabi Deutschland Gmbh Adalimumab formulations
US10570198B2 (en) 2010-10-22 2020-02-25 Novartis Ag Stable and soluble antibodies
WO2020114616A1 (en) 2018-12-07 2020-06-11 Tillotts Pharma Ag Topical treatment of immune checkpoint inhibitor induced diarrhoea, colitis or enterocolitis using antibodies and fragments thereof
WO2020157305A1 (en) 2019-01-31 2020-08-06 Numab Therapeutics AG Multispecific antibodies having specificity for tnfa and il-17a, antibodies targeting il-17a, and methods of use thereof
AU2018285441B2 (en) * 2008-06-25 2020-12-24 Novartis Ag STABLE AND SOLUBLE ANTIBODIES INHIBITING TNFa
US11229702B1 (en) 2015-10-28 2022-01-25 Coherus Biosciences, Inc. High concentration formulations of adalimumab
US11299554B2 (en) 2013-03-15 2022-04-12 Xencor, Inc. Heterodimeric proteins
US11352442B2 (en) 2014-11-26 2022-06-07 Xencor, Inc. Heterodimeric antibodies that bind CD3 and CD38
US11426566B2 (en) 2016-12-14 2022-08-30 Biora Therapeutics, Inc. Treatment of a disease of the gastrointestinal tract with a TLR modulator
US11472890B2 (en) 2019-03-01 2022-10-18 Xencor, Inc. Heterodimeric antibodies that bind ENPP3 and CD3
US11492407B2 (en) 2016-06-14 2022-11-08 Xencor, Inc. Bispecific checkpoint inhibitor antibodies
US11523772B2 (en) 2016-12-14 2022-12-13 Biora Therapeutics, Inc. Treatment of a disease of the gastrointestinal tract with an immunosuppressant
US11543416B2 (en) 2015-11-06 2023-01-03 Promise Proteomics Method for quantifying therapeutic antibodies
US11584793B2 (en) 2015-06-24 2023-02-21 Hoffmann-La Roche Inc. Anti-transferrin receptor antibodies with tailored affinity
US11591401B2 (en) 2020-08-19 2023-02-28 Xencor, Inc. Anti-CD28 compositions
US11597762B2 (en) 2016-12-14 2023-03-07 Biora Therapeutics, Inc. Treatment of a disease of the gastrointestinal tract with an IL-12/IL-23 inhibitor released using an ingestible device
US11596670B2 (en) 2017-03-30 2023-03-07 Biora Therapeutics, Inc. Treatment of a disease of the gastrointestinal tract with IL-10 or an IL-10 agonist
US11603411B2 (en) 2015-10-02 2023-03-14 Hoffmann-La Roche Inc. Bispecific anti-human CD20/human transferrin receptor antibodies and methods of use
US11623957B2 (en) 2015-12-07 2023-04-11 Xencor, Inc. Heterodimeric antibodies that bind CD3 and PSMA
US11634506B2 (en) 2013-01-14 2023-04-25 Xencor, Inc. Heterodimeric proteins
US11673972B2 (en) 2014-11-26 2023-06-13 Xencor, Inc. Heterodimeric antibodies that bind CD3 and tumor antigens
US11718667B2 (en) 2013-01-14 2023-08-08 Xencor, Inc. Optimized antibody variable regions
US11739144B2 (en) 2021-03-09 2023-08-29 Xencor, Inc. Heterodimeric antibodies that bind CD3 and CLDN6
US11787868B2 (en) 2015-10-02 2023-10-17 Hoffmann-La Roche Inc. Bispecific anti-human A-beta/human transferrin receptor antibodies and methods of use
US11814423B2 (en) 2013-03-15 2023-11-14 Xencor, Inc. Heterodimeric proteins
US11840579B2 (en) 2014-03-28 2023-12-12 Xencor, Inc. Bispecific antibodies that bind to CD38 and CD3
US11857669B2 (en) 2016-12-14 2024-01-02 Biora Therapeutics, Inc. Treatment of a disease of the gastrointestinal tract with a JAK inhibitor and devices
US11859012B2 (en) 2021-03-10 2024-01-02 Xencor, Inc. Heterodimeric antibodies that bind CD3 and GPC3
US11919956B2 (en) 2020-05-14 2024-03-05 Xencor, Inc. Heterodimeric antibodies that bind prostate specific membrane antigen (PSMA) and CD3
US11945880B2 (en) 2014-11-26 2024-04-02 Xencor, Inc. Heterodimeric antibodies that bind CD3 and tumor antigens

Families Citing this family (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008121615A2 (en) * 2007-03-30 2008-10-09 Medimmune, Inc. Antibody formulation
EA037083B1 (en) 2010-11-12 2021-02-03 Нектар Терапьютикс Immunomodulating conjugate comprising il-2 and water-soluble polymers
PE20191242A1 (en) 2011-10-25 2019-09-16 Prothena Biosciences Ltd ANTIBODY FORMULATIONS AND METHODS
SG11201504897YA (en) * 2012-12-21 2015-07-30 Glenmark Pharmaceuticals Sa Anti her2 antibody formulation
CN105263476A (en) 2013-03-13 2016-01-20 库尔制药开发公司 Immune-modifying particles for the treatment of inflammation
LT2951203T (en) * 2013-03-15 2019-09-10 Xencor, Inc. Heterodimeric proteins
EP3052132B1 (en) 2013-09-30 2020-07-29 Beth Israel Deaconess Medical Center, Inc. Antibody therapies for human immunodeficiency virus (hiv)
MX2016004926A (en) 2013-10-16 2017-01-18 Oncobiologics Inc Buffer formulations for enhanced antibody stability.
JP6620094B2 (en) * 2013-11-26 2019-12-11 ザ ブリガム アンド ウィメンズ ホスピタル インコーポレイテッドThe Brigham and Women’s Hospital, Inc. Compositions and methods for modulating immune responses
EP3247718B1 (en) 2015-01-21 2021-09-01 Outlook Therapeutics, Inc. Modulation of charge variants in a monoclonal antibody composition
JP7084308B2 (en) 2016-02-03 2022-06-14 アウトルック セラピューティクス,インコーポレイティド Buffer formulation to increase antibody stability
JOP20190260A1 (en) 2017-05-02 2019-10-31 Merck Sharp & Dohme Stable formulations of programmed death receptor 1 (pd-1) antibodies and methods of use thereof
EP3618871A4 (en) 2017-05-02 2021-01-06 Merck Sharp & Dohme Corp. Formulations of anti-lag3 antibodies and co-formulations of anti-lag3 antibodies and anti-pd-1 antibodies
US11845788B2 (en) 2018-05-22 2023-12-19 Beth Israel Deaconess Medical Center, Inc. Antibody therapies for human immunodeficiency virus (HIV)
EP3883962A4 (en) * 2018-11-21 2023-04-12 Beth Israel Deaconess Medical Center, Inc. Antibody therapies for human immunodeficiency virus (hiv)
AU2020225202B2 (en) 2019-02-18 2023-10-26 Eli Lilly And Company Therapeutic antibody formulation
CN111909267B (en) * 2019-05-07 2022-03-25 北京天成新脉生物技术有限公司 Low-immunogenicity anti-TNF-alpha humanized monoclonal antibody TCX063 and its application

Family Cites Families (32)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2000318A (en) 1933-05-22 1935-05-07 James H Cannon Cord connecter
US2200203A (en) 1938-12-16 1940-05-07 William W Heintz Legend sheet and method of application
US5672347A (en) 1984-07-05 1997-09-30 Genentech, Inc. Tumor necrosis factor antagonists and their use
US5225539A (en) 1986-03-27 1993-07-06 Medical Research Council Recombinant altered antibodies and methods of making altered antibodies
US6548640B1 (en) 1986-03-27 2003-04-15 Btg International Limited Altered antibodies
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
US20030225254A1 (en) 1989-08-07 2003-12-04 Rathjen Deborah Ann Tumour necrosis factor binding ligands
US5644034A (en) 1989-08-07 1997-07-01 Peptide Technology Ltd. Tumour necrosis factor binding ligands
US5859205A (en) 1989-12-21 1999-01-12 Celltech Limited Humanised antibodies
GB8928874D0 (en) * 1989-12-21 1990-02-28 Celltech Ltd Humanised antibodies
GB9109645D0 (en) 1991-05-03 1991-06-26 Celltech Ltd Recombinant antibodies
US5919452A (en) 1991-03-18 1999-07-06 New York University Methods of treating TNFα-mediated disease using chimeric anti-TNF antibodies
US6277969B1 (en) 1991-03-18 2001-08-21 New York University Anti-TNF antibodies and peptides of human tumor necrosis factor
LU91067I2 (en) 1991-06-14 2004-04-02 Genentech Inc Trastuzumab and its variants and immunochemical derivatives including immotoxins
US5198030A (en) 1991-06-18 1993-03-30 E. I. Du Pont De Nemours And Company Bead edge guide for use in slide-bead coating
ATE204299T1 (en) 1993-03-05 2001-09-15 Bayer Ag HUMAN MONOCLONAL ANTI-TNF ALPHA ANTIBODIES
US6090382A (en) 1996-02-09 2000-07-18 Basf Aktiengesellschaft Human antibodies that bind human TNFα
NZ536216A (en) 1996-02-09 2006-08-31 Abbott Biotech Ltd Use of an isolated antibody D2E7 for the treatment of a disorder in which TNF(alpha) activity is detrimental
US6171586B1 (en) * 1997-06-13 2001-01-09 Genentech, Inc. Antibody formulation
WO1998056418A1 (en) * 1997-06-13 1998-12-17 Genentech, Inc. Stabilized antibody formulation
US20030185826A1 (en) 1999-02-24 2003-10-02 Tobinick Edward L. Cytokine antagonists for the treatment of localized disorders
US6177077B1 (en) 1999-02-24 2001-01-23 Edward L. Tobinick TNT inhibitors for the treatment of neurological disorders
DK1242457T3 (en) 1999-12-28 2004-12-20 Esbatech Ag Intracellular antibodies [intracellular antibodies] of defined structure that are stable in a reducing environment and use thereof
GB0013810D0 (en) 2000-06-06 2000-07-26 Celltech Chiroscience Ltd Biological products
US6406863B1 (en) * 2000-06-23 2002-06-18 Genetastix Corporation High throughput generation and screening of fully human antibody repertoire in yeast
SG97908A1 (en) 2000-08-03 2003-08-20 Inst Data Storage A method and apparatus for load/unload testing of disk drives
US6881407B2 (en) 2000-08-11 2005-04-19 Ashok Amin Method for treating hepatitis
EP1585477A4 (en) 2001-11-30 2007-06-27 Centocor Inc Anti-tnf antibodies, compositions, methods and uses
EP2314622B1 (en) 2002-05-22 2017-10-18 ESBATech, an Alcon Biomedical Research Unit LLC Immunoglobulin frameworks which demonstrate enhanced stability in the intracellular environment and methods of identifying same
SI2390267T1 (en) * 2005-06-07 2013-07-31 Esbatech - A Novartis Company Llc Stable and soluble antibodies inhibiting TNF(alpha)
EP3260465A1 (en) 2005-06-07 2017-12-27 ESBATech, an Alcon Biomedical Research Unit LLC Stable and soluble antibodies inhibiting tnf-alpha
WO2009000098A2 (en) * 2007-06-25 2008-12-31 Esbatech Ag Sequence based engineering and optimization of single chain antibodies

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
None

Cited By (152)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1532174A2 (en) * 2002-06-14 2005-05-25 Immunogen, Inc. Anti-igf-i receptor antibody
EP1532174B1 (en) * 2002-06-14 2011-04-27 ImmunoGen, Inc. Anti-igf-i receptor antibody
US8691228B2 (en) 2005-06-07 2014-04-08 Esbatech, An Alcon Biomedical Research Unit Llc Stable and soluble antibodies inhibiting TNFα
US8389693B2 (en) 2005-06-07 2013-03-05 ESBATech, an Alcon Boimedical Research Unit, LLC Stable and soluble antibodies inhibiting TNFα
US9315572B2 (en) 2005-06-07 2016-04-19 ESBATech, an Alcon Biomedical Research, Unit LLC Stable and soluble antibodies inhibiting TNF alpha
US9683034B2 (en) 2005-06-07 2017-06-20 Esbatech, An Alcon Biomedical Research Unit Llc Stable and soluble antibodies inhibiting TNFα
AU2016200367B2 (en) * 2006-07-10 2017-10-12 Novartis Ag scFv antibodies which pass epithelial and/or endothelial layers
US8936785B2 (en) * 2006-07-10 2015-01-20 ESBATech, an Alcon Biomedical Research Unit, LLC scFv antibodies which pass epithelial and/or endothelial layers
US20090311251A1 (en) * 2006-07-10 2009-12-17 Esbatech Ag Scfv antibodies which pass epithelial and/or endothelial layers
US9938336B2 (en) 2007-03-12 2018-04-10 Esbatech, An Alcon Biomedical Research Unit Llc Sequence based engineering and optimization of single chain antibodies
EP3202786A2 (en) 2007-03-12 2017-08-09 ESBATech, an Alcon Biomedical Research Unit LLC Sequence based engineering and optimization of single chain antibodies
CN106366192A (en) * 2007-06-25 2017-02-01 艾斯巴技术-诺华有限责任公司 Methods of modifying antibodies, and modified antibodies with improved functional properties
WO2009000099A2 (en) 2007-06-25 2008-12-31 Esbatech Ag Methods of modifying antibodies, and modified antibodies with improved functional properties
US9908945B2 (en) 2007-06-25 2018-03-06 Esbatech, An Alcon Biomedical Research Unit Llc Sequence based engineering and optimization of single chain antibodies
CN105017427A (en) * 2007-06-25 2015-11-04 艾斯巴技术-诺华有限责任公司 Methods of modifying antibodies, and modified antibodies with improved functional properties
EP2158315B1 (en) 2007-06-25 2016-03-23 ESBATech, an Alcon Biomedical Research Unit LLC Methods of modifying antibodies, and modified antibodies with improved functional properties
CN103435696B (en) * 2007-06-25 2016-10-12 艾斯巴技术-诺华有限责任公司 The method of modified antibodies and there is the modified antibodies of functional character of improvement
WO2009040361A1 (en) * 2007-09-25 2009-04-02 Intervet International B.V. Vaccine for the treatment of osteoarthritis
WO2009062690A1 (en) * 2007-11-12 2009-05-22 U3 Pharma Gmbh Axl antibodies
US8933202B2 (en) 2007-11-12 2015-01-13 U3 Pharma Gmbh AXL antibodies
EP2918605A1 (en) * 2007-11-12 2015-09-16 U3 Pharma GmbH Axl antibodies
US20110189790A1 (en) * 2008-01-21 2011-08-04 Karyn O'neil Methods of Predicting Antibody Solubility
EP2243031A1 (en) * 2008-01-21 2010-10-27 Centocor Ortho Biotech Inc. Methods of predicting antibody solubility
EP2243031A4 (en) * 2008-01-21 2011-01-19 Centocor Ortho Biotech Inc Methods of predicting antibody solubility
AU2009212079B2 (en) * 2008-02-05 2012-08-30 Delenex Therapeutics Ag Antigen-binding polypeptides against cartilage degeneration
WO2009097704A1 (en) * 2008-02-05 2009-08-13 Esbatech Ag Antigen-binding polypeptides against cartilage degeneration
JP2011510667A (en) * 2008-02-05 2011-04-07 デレネックス セラピューティクス アーゲー Antigen-binding polypeptide for cartilage degeneration
CN101939335A (en) * 2008-02-05 2011-01-05 德勒尼克斯治疗股份公司 Antigen-binding polypeptides against cartilage degeneration
EP3858854A1 (en) 2008-06-25 2021-08-04 Novartis AG Stable and soluble antibodies inhibiting tnf
WO2009155726A3 (en) * 2008-06-25 2010-07-15 Esbatech, An Alcon Biomedical Research Unit Llc Humanization of rabbit antibodies using a universal antibody framework
EP2752428A1 (en) * 2008-06-25 2014-07-09 ESBATech, an Alcon Biomedical Research Unit LLC Humanization of rabbit antibodies using a universal antibody framework
US11578123B2 (en) 2008-06-25 2023-02-14 Novartis Ag Stable and soluble antibodies inhibiting TNFα
CN107936117B (en) * 2008-06-25 2022-12-23 诺华股份有限公司 Stable and soluble antibodies inhibiting TNF alpha
WO2009155723A2 (en) * 2008-06-25 2009-12-30 Esbatech, An Alcon Biomedical Research Unit Llc STABLE AND SOLUBLE ANTIBODIES INHIBITING TNFα
US8937162B2 (en) 2008-06-25 2015-01-20 ESBATech, an Alcon Biomedical Research Unit, LLC Humanization of rabbit antibodies using a universal antibody framework
US11046757B2 (en) 2008-06-25 2021-06-29 Novartis Ag Solubility optimization of immunobinders
JP2015120746A (en) * 2008-06-25 2015-07-02 エスバテック − ア ノバルティスカンパニー エルエルシー Humanization of rabbit antibodies using universal antibody framework
AU2018285441B2 (en) * 2008-06-25 2020-12-24 Novartis Ag STABLE AND SOLUBLE ANTIBODIES INHIBITING TNFa
US8673310B2 (en) 2008-06-25 2014-03-18 ESBA Tech, an Alcon Biomedical Research Unit LLC Stable and soluble antibodies inhibiting TNFα
KR102050040B1 (en) 2008-06-25 2019-11-28 에스바테크 - 어 노바티스 컴파니 엘엘씨 STABLE AND SOLUBLE ANTIBODIES INHIBITING TNFα
RU2567100C2 (en) * 2008-06-25 2015-10-27 ИЭсБиЭйТЕК, ЭН АЛЬКОН БАЙОМЕДИКАЛ РИСЕРЧ ЮНИТ ЭлЭлСи STABLE AND SOLUBLE ANTIBODIES INHIBITING TNFα
JP2019195342A (en) * 2008-06-25 2019-11-14 エスバテック − ア ノバルティスカンパニー エルエルシー STABLE AND SOLUBLE ANTIBODY INHIBITING TNFα
RU2704838C1 (en) * 2008-06-25 2019-10-31 ИЭсБиЭйТЕК, ЭН АЛЬКОН БАЙОМЕДИКАЛ РИСЕРЧ ЮНИТ ЭлЭлСи Stable and soluble antibodies inhibiting tnfα
US11858981B2 (en) 2008-06-25 2024-01-02 Novartis Ag Humanization of rabbit antibodies using a universal antibody framework
US8293235B2 (en) 2008-06-25 2012-10-23 ESBATech, an Alcon Biomedical Research Unit, LLC Humanization of rabbit antibodies using a universal antibody framework
US10221237B2 (en) 2008-06-25 2019-03-05 Esbatech, An Alcon Biomedical Research Unit Llc Solubility optimization of immunobinders
EP3444274A1 (en) 2008-06-25 2019-02-20 ESBATech, an Alcon Biomedical Research Unit LLC Stable and soluble antibodies inhibiting tnf
US9422366B2 (en) 2008-06-25 2016-08-23 Esbatech, An Alcon Biomedical Research Unit Llc Stable and soluble antibodies inhibiting TNF alpha
JP2011525358A (en) * 2008-06-25 2011-09-22 エスバテック、アン アルコン バイオメディカル リサーチ ユニット、エルエルシー Stable and soluble antibody that inhibits TNFα
KR20160128439A (en) * 2008-06-25 2016-11-07 에스바테크 - 어 노바티스 컴파니 엘엘씨 STABLE AND SOLUBLE ANTIBODIES INHIBITING TNFα
US9556265B2 (en) 2008-06-25 2017-01-31 Esbatech, An Alcon Biomedical Research Unit Llc Solubility optimization of immunobinders
JP2011525360A (en) * 2008-06-25 2011-09-22 エスバテック、アン アルコン バイオメディカル リサーチ ユニット、エルエルシー Humanization of rabbit antibodies using a versatile antibody framework
RU2678120C1 (en) * 2008-06-25 2019-01-23 ИЭсБиЭйТЕК, ЭН АЛЬКОН БАЙОМЕДИКАЛ РИСЕРЧ ЮНИТ ЭлЭлСи STABLE AND SOLUBLE ANTIBODIES INHIBITING TNFα
US10100111B2 (en) 2008-06-25 2018-10-16 Esbatech, An Alcon Biomedical Research Unit Llc Stable and soluble antibodies inhibiting TNF alpha
US9593161B2 (en) 2008-06-25 2017-03-14 Esbatech, An Alcon Biomedical Research Unit Llc Humanization of rabbit antibodies using a universal antibody framework
JP2017051201A (en) * 2008-06-25 2017-03-16 エスバテック − ア ノバルティス カンパニー エルエルシー STABLE AND SOLUBLE ANTIBODY INHIBITING TNFα
US10087244B2 (en) 2008-06-25 2018-10-02 Esbatech, An Alcon Biomedical Research Unit Llc Humanization of rabbit antibodies using a universal antibody framework
KR20180097792A (en) * 2008-06-25 2018-08-31 에스바테크 - 어 노바티스 컴파니 엘엘씨 STABLE AND SOLUBLE ANTIBODIES INHIBITING TNFα
KR101893010B1 (en) 2008-06-25 2018-08-29 에스바테크 - 어 노바티스 컴파니 엘엘씨 STABLE AND SOLUBLE ANTIBODIES INHIBITING TNFα
RU2653753C1 (en) * 2008-06-25 2018-05-14 ИЭсБиЭйТЕК, ЭН АЛЬКОН БАЙОМЕДИКАЛ РИСЕРЧ ЮНИТ ЭлЭлСи STABLE AND SOLUBLE ANTIBODIES INHIBITING TNFα
KR101773904B1 (en) 2008-06-25 2017-09-04 에스바테크 - 어 노바티스 컴파니 엘엘씨 STABLE AND SOLUBLE ANTIBODIES INHIBITING TNFα
CN107936117A (en) * 2008-06-25 2018-04-20 艾斯巴技术-诺华有限责任公司 Suppress the stabilization and soluble antibodies of TNF α
WO2009155726A2 (en) * 2008-06-25 2009-12-30 Esbatech, An Alcon Biomedical Research Unit Llc Humanization of rabbit antibodies using a universal antibody framework
WO2009155723A3 (en) * 2008-06-25 2010-04-22 Esbatech, An Alcon Biomedical Research Unit Llc STABLE AND SOLUBLE ANTIBODIES INHIBITING TNFα
EP3241843A1 (en) 2008-06-25 2017-11-08 ESBATech, an Alcon Biomedical Research Unit LLC Solubility optimization of immunobinders
JP2017190352A (en) * 2008-06-25 2017-10-19 エスバテック − ア ノバルティス カンパニー エルエルシー Humanization of rabbit antibodies using universal antibody framework
EP3130603A1 (en) 2008-06-30 2017-02-15 ESBATech, an Alcon Biomedical Research Unit LLC Functionalized polypeptides
US8637022B2 (en) 2008-06-30 2014-01-28 Esbatech, An Alcon Biomedical Research Unit Llc Functionalized polypeptides
US9371525B2 (en) 2008-06-30 2016-06-21 Esbatech, an Alcon Biomedical Reseach Unit LLC Functionalized polypeptides
CN104800147A (en) * 2008-07-10 2015-07-29 德勒尼克斯治疗股份公司 Methods and compositions for enhanced delivery of macromolecules
EP2769711A1 (en) 2008-07-10 2014-08-27 ESBATech, an Alcon Biomedical Research Unit LLC Methods and Compositions for Enhanced Delivery of Macromolecules
US8697074B2 (en) 2008-07-10 2014-04-15 Esbatech, An Alcon Biomedical Research Unit Llc Methods and compositions for enhanced delivery of macromolecules
US8841424B2 (en) 2009-05-11 2014-09-23 U3 Pharma Gmbh Humanized AXL antibodies
WO2010130751A1 (en) * 2009-05-11 2010-11-18 U3 Pharma Gmbh Humanized axl antibodies
EP2270053A1 (en) * 2009-05-11 2011-01-05 U3 Pharma GmbH Humanized AXL antibodies
US9226983B2 (en) 2010-04-07 2016-01-05 Abbvie Inc. TNF-α binding proteins
US10570198B2 (en) 2010-10-22 2020-02-25 Novartis Ag Stable and soluble antibodies
EP2649189A4 (en) * 2010-12-08 2014-05-14 Abbvie Inc Tnf- binding proteins
EP2649189A1 (en) * 2010-12-08 2013-10-16 AbbVie Inc. Tnf- binding proteins
EP3750917A1 (en) 2012-11-05 2020-12-16 Delenex Therapeutics AG Binding members to il-1 beta
US10077302B2 (en) 2012-11-05 2018-09-18 Cell Medica Switzerland Ag Method of inhibiting human IL-1 beta by administering an antibody to human IL-1 beta
US9404930B2 (en) 2012-11-05 2016-08-02 Delenex Therapeutics Ag Antibody to human IL-1 beta
WO2014068132A1 (en) 2012-11-05 2014-05-08 Delenex Therapeutics Ag Binding members to il-1 beta
US11267884B2 (en) 2012-11-05 2022-03-08 Cell Medica Inc. Antibody to human IL-1 beta
US11718667B2 (en) 2013-01-14 2023-08-08 Xencor, Inc. Optimized antibody variable regions
US11634506B2 (en) 2013-01-14 2023-04-25 Xencor, Inc. Heterodimeric proteins
US11299554B2 (en) 2013-03-15 2022-04-12 Xencor, Inc. Heterodimeric proteins
US11814423B2 (en) 2013-03-15 2023-11-14 Xencor, Inc. Heterodimeric proteins
US10370692B2 (en) 2013-12-20 2019-08-06 Hoffmann-La Roche Inc. Recombinant polypeptide production methods
RU2706551C2 (en) * 2014-03-26 2019-11-19 Селл Медика Свитзерлэнд Аг Binding members to tnf-alpha
WO2015144852A1 (en) * 2014-03-26 2015-10-01 Delenex Therapeutics Ag Binding members to tnf alpha
US10995138B2 (en) 2014-03-26 2021-05-04 Cell Medica Inc. Nucleic acids encoding binding members to TNF alpha
US10233238B2 (en) 2014-03-26 2019-03-19 Cell Medica Switzerland Ag TNF alpha antibody or fragment thereof and methods of use
CN106414498A (en) * 2014-03-26 2017-02-15 细胞药物瑞士股份公司 Binding members to TNF alpha
CN106414498B (en) * 2014-03-26 2020-09-08 细胞药物瑞士股份公司 Binding members for TNF alpha
US11840579B2 (en) 2014-03-28 2023-12-12 Xencor, Inc. Bispecific antibodies that bind to CD38 and CD3
US10426833B2 (en) 2014-05-23 2019-10-01 Fresenius Kabi Deutschland Gmbh Liquid pharmaceutical composition
US10729769B2 (en) 2014-05-23 2020-08-04 Fresenius Kabi Deutschland Gmbh Liquid pharmaceutical composition
US10426832B2 (en) 2014-05-23 2019-10-01 Fresenius Kabi Deutschland Gmbh Liquid pharmaceutical composition
US11752209B2 (en) 2014-05-23 2023-09-12 Fresenius Kabi Deutschland Gmbh Liquid pharmaceutical composition
US11752208B2 (en) 2014-05-23 2023-09-12 Fresenius Kabi Deutschland Gmbh Liquid pharmaceutical composition
EP3145487B1 (en) 2014-05-23 2018-08-22 Fresenius Kabi Deutschland GmbH Liquid pharmaceutical composition
US10493152B2 (en) 2014-05-23 2019-12-03 Fresenius Kabi Deutschland Gmbh Adalimumab formulations
US10772961B2 (en) 2014-05-23 2020-09-15 Fresenius Kabi Deutschland Gmbh Liquid pharmaceutical composition
US11712471B2 (en) 2014-05-23 2023-08-01 Fresenius Kabi Deustschland GmbH Liquid pharmaceutical composition
US11707524B2 (en) 2014-05-23 2023-07-25 Fresenius Kabi Deutschland Gmbh Liquid pharmaceutical composition
US11352442B2 (en) 2014-11-26 2022-06-07 Xencor, Inc. Heterodimeric antibodies that bind CD3 and CD38
US11859011B2 (en) 2014-11-26 2024-01-02 Xencor, Inc. Heterodimeric antibodies that bind CD3 and tumor antigens
US11945880B2 (en) 2014-11-26 2024-04-02 Xencor, Inc. Heterodimeric antibodies that bind CD3 and tumor antigens
US11673972B2 (en) 2014-11-26 2023-06-13 Xencor, Inc. Heterodimeric antibodies that bind CD3 and tumor antigens
US11584793B2 (en) 2015-06-24 2023-02-21 Hoffmann-La Roche Inc. Anti-transferrin receptor antibodies with tailored affinity
US11787868B2 (en) 2015-10-02 2023-10-17 Hoffmann-La Roche Inc. Bispecific anti-human A-beta/human transferrin receptor antibodies and methods of use
US11603411B2 (en) 2015-10-02 2023-03-14 Hoffmann-La Roche Inc. Bispecific anti-human CD20/human transferrin receptor antibodies and methods of use
US11229702B1 (en) 2015-10-28 2022-01-25 Coherus Biosciences, Inc. High concentration formulations of adalimumab
US11630111B2 (en) 2015-11-06 2023-04-18 Promise Proteomics Method for quantifying anti-TNF antibodies
EP3165928A1 (en) * 2015-11-06 2017-05-10 Promise Advanced Proteomics A method for quantifying anti-tnf antibodies
US11543416B2 (en) 2015-11-06 2023-01-03 Promise Proteomics Method for quantifying therapeutic antibodies
WO2017077080A1 (en) * 2015-11-06 2017-05-11 Promise Advanced Proteomics A method for quantifying anti-tnf antibodies
US11623957B2 (en) 2015-12-07 2023-04-11 Xencor, Inc. Heterodimeric antibodies that bind CD3 and PSMA
WO2017158097A1 (en) 2016-03-17 2017-09-21 Tillotts Pharma Ag Anti-tnfalpha-antibodies and functional fragments thereof
WO2017158101A1 (en) 2016-03-17 2017-09-21 Numab Innovation Ag ANTI-TNFα-ANTIBODIES AND FUNCTIONAL FRAGMENTS THEREOF
WO2017158084A1 (en) 2016-03-17 2017-09-21 Numab Innovation Ag ANTI-TNFα-ANTIBODIES AND FUNCTIONAL FRAGMENTS THEREOF
WO2017158092A1 (en) 2016-03-17 2017-09-21 Tillotts Pharma Ag Anti-tnfalpha-antibodies and functional fragments thereof
EP3219726A1 (en) 2016-03-17 2017-09-20 Tillotts Pharma AG Anti-tnf alpha-antibodies and functional fragments thereof
WO2017158079A1 (en) 2016-03-17 2017-09-21 Numab Innovation Ag Anti-tnfalpha-antibodies and functional fragments thereof
EP4275745A2 (en) 2016-03-17 2023-11-15 Tillotts Pharma Ag Anti-tnfalpha-antibodies and functional fragments thereof
EP3219727A1 (en) 2016-03-17 2017-09-20 Tillotts Pharma AG Anti-tnf alpha-antibodies and functional fragments thereof
US11492407B2 (en) 2016-06-14 2022-11-08 Xencor, Inc. Bispecific checkpoint inhibitor antibodies
US11426566B2 (en) 2016-12-14 2022-08-30 Biora Therapeutics, Inc. Treatment of a disease of the gastrointestinal tract with a TLR modulator
US11597762B2 (en) 2016-12-14 2023-03-07 Biora Therapeutics, Inc. Treatment of a disease of the gastrointestinal tract with an IL-12/IL-23 inhibitor released using an ingestible device
US11523772B2 (en) 2016-12-14 2022-12-13 Biora Therapeutics, Inc. Treatment of a disease of the gastrointestinal tract with an immunosuppressant
US11857669B2 (en) 2016-12-14 2024-01-02 Biora Therapeutics, Inc. Treatment of a disease of the gastrointestinal tract with a JAK inhibitor and devices
US11596670B2 (en) 2017-03-30 2023-03-07 Biora Therapeutics, Inc. Treatment of a disease of the gastrointestinal tract with IL-10 or an IL-10 agonist
EP3409688A1 (en) 2017-05-31 2018-12-05 Tillotts Pharma Ag Topical treatment of inflammatory bowel disease using anti-tnf-alpha antibodies and fragments thereof
WO2018219559A1 (en) 2017-05-31 2018-12-06 Tillotts Pharma Ag Topical treatment of inflammatory bowel disease using antibodies and fragments thereof
WO2019057560A1 (en) 2017-09-19 2019-03-28 Tillotts Pharma Ag USE OF ANTI-TNFα ANTIBODIES FOR TREATING WOUNDS
EP3456739A1 (en) 2017-09-19 2019-03-20 Tillotts Pharma Ag Use of anti-tnfalpha antibodies for treating wounds
EP3459529A1 (en) 2017-09-20 2019-03-27 Tillotts Pharma Ag Preparation of sustained release solid dosage forms comprising antibodies by spray drying
EP3459527A1 (en) 2017-09-20 2019-03-27 Tillotts Pharma Ag Method for preparing a solid dosage form comprising antibodies by wet granulation, extrusion and spheronization
WO2019057563A1 (en) 2017-09-20 2019-03-28 Tillotts Pharma Ag Method for preparing a solid dosage form comprising antibodies by wet granulation, extrusion and spheronization
WO2019057566A1 (en) 2017-09-20 2019-03-28 Tillotts Pharma Ag Preparation of sustained release solid dosage forms comprising antibodies by spray drying
WO2019057562A1 (en) 2017-09-20 2019-03-28 Tillotts Pharma Ag Preparation of solid dosage forms comprising antibodies by solution/suspension layering
EP3459528A1 (en) 2017-09-20 2019-03-27 Tillotts Pharma Ag Preparation of solid dosage forms comprising antibodies by solution/suspension layering
WO2020114616A1 (en) 2018-12-07 2020-06-11 Tillotts Pharma Ag Topical treatment of immune checkpoint inhibitor induced diarrhoea, colitis or enterocolitis using antibodies and fragments thereof
WO2020115277A1 (en) 2018-12-07 2020-06-11 Tillotts Pharma Ag Topical treatment of immune checkpoint inhibitor induced diarrhoea, colitis or enterocolitis using antibodies and fragments thereof
WO2020157305A1 (en) 2019-01-31 2020-08-06 Numab Therapeutics AG Multispecific antibodies having specificity for tnfa and il-17a, antibodies targeting il-17a, and methods of use thereof
US11472890B2 (en) 2019-03-01 2022-10-18 Xencor, Inc. Heterodimeric antibodies that bind ENPP3 and CD3
US11919956B2 (en) 2020-05-14 2024-03-05 Xencor, Inc. Heterodimeric antibodies that bind prostate specific membrane antigen (PSMA) and CD3
US11919958B2 (en) 2020-08-19 2024-03-05 Xencor, Inc. Anti-CD28 compositions
US11591401B2 (en) 2020-08-19 2023-02-28 Xencor, Inc. Anti-CD28 compositions
US11739144B2 (en) 2021-03-09 2023-08-29 Xencor, Inc. Heterodimeric antibodies that bind CD3 and CLDN6
US11859012B2 (en) 2021-03-10 2024-01-02 Xencor, Inc. Heterodimeric antibodies that bind CD3 and GPC3

Also Published As

Publication number Publication date
PH12012500758B1 (en) 2016-01-11
HK1218653A1 (en) 2017-03-03
JP5089582B2 (en) 2012-12-05
EP2390267A1 (en) 2011-11-30
BRPI0611765B1 (en) 2022-09-27
CN102924597A (en) 2013-02-13
US20130171143A1 (en) 2013-07-04
JP5645890B2 (en) 2014-12-24
JP2013027390A (en) 2013-02-07
US8067547B2 (en) 2011-11-29
WO2006131013A3 (en) 2007-01-25
NZ563580A (en) 2010-09-30
BRPI0611765A2 (en) 2011-12-20
KR20140071450A (en) 2014-06-11
US9315572B2 (en) 2016-04-19
PH12012500758A1 (en) 2016-01-11
MX2007015280A (en) 2008-04-08
US8691228B2 (en) 2014-04-08
US20090263382A1 (en) 2009-10-22
PL2390267T3 (en) 2013-09-30
IL243270B (en) 2020-09-30
DK2390267T3 (en) 2013-08-26
CA2609999A1 (en) 2006-12-14
EP2390267B1 (en) 2013-06-05
KR20080031003A (en) 2008-04-07
US20120064077A1 (en) 2012-03-15
US20160185850A1 (en) 2016-06-30
EP1891110A2 (en) 2008-02-27
JP2008545433A (en) 2008-12-18
RU2415151C2 (en) 2011-03-27
EP3260465A1 (en) 2017-12-27
RU2007149255A (en) 2009-07-20
CA2609999C (en) 2017-05-30
IL187503A0 (en) 2008-03-20
HK1164891A1 (en) 2012-09-28
ES2424042T3 (en) 2013-09-26
US8389693B2 (en) 2013-03-05
AU2006255415A1 (en) 2006-12-14
KR101457223B1 (en) 2014-11-04
US20140171634A1 (en) 2014-06-19
HRP20130632T1 (en) 2013-11-22
IL243270A0 (en) 2016-02-29
PT2390267E (en) 2013-07-16
CN105153306B (en) 2020-12-11
US9683034B2 (en) 2017-06-20
RS52861B (en) 2013-12-31
KR101539545B1 (en) 2015-07-24
AU2006255415B2 (en) 2011-10-06
CN105153306A (en) 2015-12-16

Similar Documents

Publication Publication Date Title
US9683034B2 (en) Stable and soluble antibodies inhibiting TNFα
JP6100463B2 (en) Stable and soluble antibody that inhibits TNFα
AU2016204739C1 (en) Stable and soluble antibodies inhibiting TNFalpha
AU2011265593B2 (en) Stable and soluble antibodies inhibiting TNFalpha
AU2013207650B2 (en) Stable and soluble antibodies inhibiting TNFalpha

Legal Events

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

Ref document number: 187503

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 563580

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 12007502664

Country of ref document: PH

WWE Wipo information: entry into national phase

Ref document number: 2609999

Country of ref document: CA

REEP Request for entry into the european phase

Ref document number: 2006741625

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2006741625

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: MX/a/2007/015280

Country of ref document: MX

Ref document number: 2006255415

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2008515021

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

WWW Wipo information: withdrawn in national office

Ref document number: DE

WWE Wipo information: entry into national phase

Ref document number: 9603/DELNP/2007

Country of ref document: IN

ENP Entry into the national phase

Ref document number: 2006255415

Country of ref document: AU

Date of ref document: 20060606

Kind code of ref document: A

WWP Wipo information: published in national office

Ref document number: 2006255415

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 1020087000199

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: 2007149255

Country of ref document: RU

WWE Wipo information: entry into national phase

Ref document number: 200680028415.3

Country of ref document: CN

WWP Wipo information: published in national office

Ref document number: 2006741625

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 11916793

Country of ref document: US

ENP Entry into the national phase

Ref document number: PI0611765

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20071207

WWE Wipo information: entry into national phase

Ref document number: 242304

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 243270

Country of ref document: IL