US20070237769A1 - Single domain antibodies directed against tumour necrosis factor-alpha and uses therefor - Google Patents

Single domain antibodies directed against tumour necrosis factor-alpha and uses therefor Download PDF

Info

Publication number
US20070237769A1
US20070237769A1 US11/788,832 US78883207A US2007237769A1 US 20070237769 A1 US20070237769 A1 US 20070237769A1 US 78883207 A US78883207 A US 78883207A US 2007237769 A1 US2007237769 A1 US 2007237769A1
Authority
US
United States
Prior art keywords
tnf
alpha
sequence
vhh
seq
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/788,832
Inventor
Karen Silence
Marc Lauwereys
Hans De Haard
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Ablynx NV
Original Assignee
Ablynx NV
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from PCT/BE2003/000192 external-priority patent/WO2004041862A2/en
Application filed by Ablynx NV filed Critical Ablynx NV
Priority to US11/788,832 priority Critical patent/US20070237769A1/en
Assigned to ABLYNX N.V. reassignment ABLYNX N.V. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: DE HAARD, HANS, LAUWEREYS, MARC, SILENCE, KAREN
Priority to US11/804,647 priority patent/US20090022721A1/en
Publication of US20070237769A1 publication Critical patent/US20070237769A1/en
Priority to US12/905,589 priority patent/US20110027281A1/en
Priority to US13/078,241 priority patent/US20110184150A1/en
Priority to US13/423,790 priority patent/US20120202977A1/en
Priority to US14/529,280 priority patent/US9371381B2/en
Priority to US15/157,830 priority patent/US20170096481A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/24Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against cytokines, lymphokines or interferons
    • C07K16/241Tumor Necrosis Factors
    • 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
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • 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
    • 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
    • C07K16/249Interferons
    • 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
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2863Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for growth factors, growth regulators
    • 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
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2875Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the NGF/TNF superfamily, e.g. CD70, CD95L, CD153, CD154
    • 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
    • C07K16/36Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against blood coagulation factors
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/40Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against enzymes
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/42Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against immunoglobulins
    • C07K16/4283Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against immunoglobulins against an allotypic or isotypic determinant on Ig
    • C07K16/4291Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against immunoglobulins against an allotypic or isotypic determinant on Ig against IgE
    • 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
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/22Immunoglobulins specific features characterized by taxonomic origin from camelids, e.g. camel, llama or dromedary
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/31Immunoglobulins specific features characterized by aspects of specificity or valency multispecific
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/34Identification of a linear epitope shorter than 20 amino acid residues or of a conformational epitope defined by amino acid residues
    • 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
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/569Single domain, e.g. dAb, sdAb, VHH, VNAR or nanobody®
    • 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
    • C07K2317/626Diabody or triabody
    • 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
    • C07K2319/00Fusion polypeptide

Definitions

  • the present invention provides polypeptides comprising one or more single domain antibodies directed towards tumor necrosis factor alpha (TNF-alpha).
  • TNF-alpha tumor necrosis factor alpha
  • the present invention further relates to their use in diagnosis and therapy.
  • Such antibodies may have a framework sequence with high homology to the human framework sequences.
  • Compositions comprising antibodies to tumor necrosis factor alpha (TNF-alpha) alone or in combination with other drugs are described.
  • Tumor necrosis factor alpha is believed to play an important role in various disorders, for example in inflammatory disorders such as rheumatoid arthritis, Crohn's disease, ulcerative colitis and multiple sclerosis. Both TNF-alpha and the receptors (CD120a, CD120b) have been studied in great detail. TNF-alpha in its bioactive form is a trimer and the groove formed by neighboring subunits is important for the cytokine-receptor interaction. Several strategies to antagonize the action of the cytokine have been developed and are currently used to treat various disease states.
  • a TNF-alpha inhibitor which has sufficient specificity and selectivity to TNF-alpha may be an efficient prophylactic or therapeutic pharmaceutical compound for preventing or treating disorders where TNF-alpha has been implicated as causative agent.
  • Methods of treating toxic shock EP 486526
  • tumor regression inhibition of cytotoxicity
  • U.S. Pat. No. 6,448,380 U.S. Pat. No. 6,451,983, U.S. Pat. No. 6,498,2307
  • autoimmune disease such as RA and Crohn's disease
  • EP 663836 U.S. Pat. No. 5,672,347, U.S. Pat. No. 5,656,272
  • graft versus host reaction U.S. Pat. No. 5,672,347
  • bacterial meningitis EP 585705
  • Heavy chain antibodies described in the invention which are derived from Camelidae are known to have cavity-binding propensity (WO97/49805; Lauwereys et al, EMBO J. 17, 5312, 1998)). Therefore, such heavy chain antibodies are inherently suited to bind to receptor binding domains of such ligands as TNF. In addition, such antibodies are known to be stable over long periods of time, therefore increasing their shelf-life (Perez et al, Biochemistry, 40, 74, 2001). Furthermore, such heavy chain antibody fragments can be produced ‘en-masse’ in fermentors using cheap expression systems compared to mammalian cell culture fermentation, such as yeast or other microorganisms (EP 0 698 097).
  • antibodies derived from sources such as mouse, sheep, goat, rabbit etc., and humanised derivatives thereof as a treatment for conditions which require a modulation of inflammation is problematic for several reasons.
  • Traditional antibodies are not stable at room temperature, and have to be refrigerated for preparation and storage, requiring necessary refrigerated laboratory equipment, storage and transport, which contribute towards time and expense. Refrigeration is sometimes not feasible in developing countries.
  • manufacture or small-scale production of said antibodies is expensive because the mammalian cellular systems necessary for the expression of intact and active antibodies require high levels of support in terms of time and equipment, and yields are very low.
  • the large size of conventional antibodies would restrict tissue penetration, for example, at the site of inflamed tissue.
  • traditional antibodies have a binding activity which depends upon pH, and hence are unsuitable for use in environments outside the usual physiological pH range such as, for example, in treating gastric bleeding, gastric surgery. Furthermore, traditional antibodies are unstable at low or high pH and hence are not suitable for oral administration. However, it has been demonstrated that camelidae antibodies resist harsh conditions, such as extreme pH, denaturing reagents and high temperatures (Dumoulin et al, Protein Science 11, 500, 2002), so making them suitable for delivery by oral administration. Furthermore, traditional antibodies have a binding activity, which depends upon temperature, and hence are unsuitable for use in assays or kits performed at temperatures outside biologically active-temperature ranges (e.g. 37 ⁇ 20° C.).
  • Single domain antibody-based therapeutics have significant potential as drugs because they have extraordinarily specificity to their target and a low inherent toxicity. However, improving further their intrinsic and functional affinity can lead to many benefits for a patient such as reduced dose of therapeutic, faster therapy, and reduced side effects.
  • polypeptides comprising one or more single domain antibodies which bind to TNF-alpha, homologues of said polypeptides, functional portions of homologues of said polypeptides. Said polypeptides modify the biological activity of TNF-alpha upon binding. Such polypeptides might bind into the receptor-binding groove of TNF-alpha, or might not bind in the receptor binding groove. Such polypeptides are single domain antibodies.
  • single domain antibodies which may be any of the art, or any future single domain antibodies.
  • examples include, but are not limited to, heavy chain antibodies, antibodies naturally devoid of light chains, single domain antibodies derived from conventional 4-chain antibodies, engineered antibodies and single domain scaffolds other than those derived from antibodies.
  • a single domain antibody as used herein is a naturally occurring single domain antibody known as heavy chain antibody devoid of light chains (WO 9404678).
  • this variable domain derived from a heavy chain antibody devoid of light chain will be called VHH or nanobody to distinguish it from the conventional VH of four chain immunoglobulins.
  • Such a VHH molecule can be derived from antibodies raised in Camelidae species, for example in camel, llama, dromedary, alpaca and guanaco.
  • One embodiment of the present invention is an anti-TNF-alpha polypeptide comprising at least one anti-TNF-alpha single domain antibody.
  • Another embodiment of the present invention is an anti-TNF-alpha polypeptide as described above wherein a single domain antibody corresponds to a sequence represented by any of SEQ ID NOs: 1 to 16 and 79 to 84.
  • Another embodiment of the present invention is an anti-TNF-alpha polypeptide as described above further comprising at least one single domain antibody directed against a serum protein.
  • Another embodiment of the present invention is an anti-TNF-alpha polypeptide as described above wherein said serum protein is any of serum albumin, serum immunoglobulins, thyroxine-binding protein, transferring, or fibrinogen.
  • Another embodiment of the present invention is an anti-TNF-alpha polypeptide as described above wherein a single domain anti-serum protein single domain antibody correspond to a sequence represented by any of SEQ ID NOs: 26 to 29 and 85 to 97.
  • Another embodiment of the present invention is an anti-TNF-alpha polypeptide as described above corresponding to a sequence represented by any of SEQ ID NOs: 30 to 43.
  • Another embodiment of the present invention is an anti-TNF-alpha polypeptide as described above further comprising at least one single domain antibody selected from the group consisting of anti-IFN-gamma single domain antibody, anti-TNF-alpha receptor single domain antibody and anti-IFN-gamma receptor single domain antibody.
  • Another embodiment of the present invention is an anti-TNF-alpha polypeptide as described above, wherein the number of single domain antibodies directed against TNF-alpha is at least two.
  • Another embodiment of the present invention is an anti-TNF-alpha polypeptide as described above corresponding to a sequence represented by any of SEQ ID NOs: 73 to 76.
  • Another embodiment of the present invention is an anti-TNF-alpha polypeptide as described above, wherein at least one single domain antibody is a humanized Camelidae VHHs.
  • Another embodiment of the present invention is an anti-TNF-alpha polypeptide as described above wherein a humanized Camelidae VHH corresponds to a sequence represented by any of SEQ ID NOs: 17 to 19 and 21 to 24.
  • compositions comprising an anti-TNF-alpha polypeptide as described above and at least one single domain antibody from the group consisting of anti-IFN-gamma single domain antibody, anti-TNF-alpha receptor single domain antibody and anti-IFN-gamma receptor single domain antibody, for simultaneous, separate or sequential administration to a subject.
  • Another embodiment of the present invention is an anti-TNF-alpha polypeptide as described above, or a composition as described above wherein at least one anti-IFN-gamma single domain antibody correspond to a sequence represented by any of SEQ ID NOs: 44 to 72.
  • Another embodiment of the present invention is an anti-TNF-alpha polypeptide as described above, or a composition as described above, wherein said single domain antibody is an homologous sequence, a functional portion, or a functional portion of an homologous sequence of the full length single domain antibody.
  • Another embodiment of the present invention is an anti-TNF-alpha polypeptide as described above, or a composition as described above, wherein the anti-TNF-alpha polypeptide is an homologous sequence, a functional portion, or a functional portion of an homologous sequence of the full length anti-TNF-alpha polypeptide.
  • Another embodiment of the present invention is an anti-TNF-alpha polypeptide as described above, or a composition as described above wherein at least one single domain antibody is a Camelidae VHH.
  • Another embodiment of the present invention is a nucleic acid encoding an anti-TNF-alpha polypeptide as described above.
  • Another embodiment of the present invention is a method of identifying an agent that modulates the binding of an anti-TNF-alpha polypeptide as described above, to Tumor Necrosis Factor-alpha comprising the steps of:
  • Another embodiment of the present invention is a kit for screening for agents that modulate Tumor Necrosis Factor-alpha-mediated disorders comprising an anti-TNF-alpha polypeptide as described above and Tumor Necrosis Factor-alpha.
  • Another embodiment of the present invention is an unknown agent that modulates the binding of an anti-TNF-alpha polypeptide as described above to Tumor Necrosis Factor-alpha, identified according to the method as described above.
  • Another embodiment of the present invention is an unknown agent that modulates Tumor Necrosis Factor-alpha-mediated disorders, identified according to the methods as described above.
  • Another embodiment of the present invention is an unknown agent as described above wherein said disorders are one or more of inflammation, rheumatoid arthritis, Crohn's disease, ulcerative colitis, inflammatory bowel syndrome and multiple sclerosis.
  • Another embodiment of the present invention is an anti-TNF-alpha polypeptide as described above, or a nucleic acid as described above, or a composition as described above, or an agent as described above for treating and/or preventing and/or alleviating disorders relating to inflammatory processes.
  • Another embodiment of the present invention is a use of an anti-TNF-alpha polypeptide as described above or a nucleic acid as described above, or a composition as described above, or an agent as described above for the preparation of a medicament for treating and/or preventing and/or alleviating disorders relating to inflammatory reactions.
  • Another embodiment of the present invention is an anti-TNF-alpha polypeptide as described above or a composition as described above, for treating and/or preventing and/or alleviating disorders susceptible to modulation by a TNF-alpha modulating substance which is able pass through the gastric environment without the substance being inactivated.
  • Another embodiment of the present invention is an use of an anti-TNF-alpha polypeptide as described above or a composition as described above, for the preparation of a medicament for treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by a TNF-alpha modulating substance which is able pass through the gastric environment without the substance being inactivated.
  • Another embodiment of the present invention is an anti-TNF-alpha polypeptide as described above or a composition as described above, for treating and/or preventing and/or alleviating disorders susceptible to modulation by a TNF-alpha modulating substance delivered to the vaginal and/or rectal tract.
  • Another embodiment of the present invention is a use of an anti-TNF-alpha polypeptide as described above or a composition as described above, for the preparation of a medicament for treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by a TNF-alpha modulating substance delivered to the vaginal and/or rectal tract.
  • Another embodiment of the present invention is an anti-TNF-alpha polypeptide as described above or a composition as described above, for treating and/or preventing and/or alleviating disorders susceptible to modulation by a TNF-alpha modulating substance delivered to the nose, upper respiratory tract and/or lung.
  • Another embodiment of the present invention is a use of an anti-TNF-alpha polypeptide as described above or a composition as described above, for the preparation of a medicament for treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by a TNF-alpha modulating substance delivered to the nose, upper respiratory tract and/or lung.
  • Another embodiment of the present invention is an anti-TNF-alpha polypeptide as described above or a composition as described above, for treating and/or preventing and/or alleviating disorders susceptible to modulation by a TNF-alpha modulating substance delivered to the intestinal mucosa, wherein said disorder increases the permeability of the intestinal mucosa.
  • Another embodiment of the present invention is a use of an anti-TNF-alpha polypeptide as described above or a composition as described above, for the preparation of a medicament for treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by a TNF-alpha modulating substance delivered to the intestinal mucosa, wherein said disorder increases the permeability of the intestinal mucosa.
  • Another embodiment of the present invention is an anti-TNF-alpha polypeptide as described above or a composition as described above, for treating and/or preventing and/or alleviating disorders susceptible to modulation by a TNF-alpha modulating substance which is able pass through the tissues beneath the tongue effectively.
  • Another embodiment of the present invention is a use of an anti-TNF-alpha polypeptide as described above or a composition as described above, for the preparation of a medicament for treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by a TNF-alpha modulating substance which is able pass through the tissues beneath the tongue effectively.
  • Another embodiment of the present invention is an anti-TNF-alpha polypeptide as described above or a composition as described above, for treating and/or preventing and/or alleviating disorders susceptible to modulation by a TNF-alpha modulating substance which is able pass through the skin effectively.
  • Another embodiment of the present invention is a use of an anti-TNF-alpha polypeptide as described above or a composition as described above, for the preparation of a medicament for treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by a TNF-alpha modulating substance which is able pass through the skin effectively.
  • Another embodiment of the present invention is a method as described above, a kit as described above, a nucleic acid or agent as described above, use of a nucleic acid or agent as described above, a composition as described above, use of a composition as described above, an anti-TNF-alpha polypeptide as described above, use of an anti-TNF-alpha polypeptide as described above wherein said disorders are any of inflammation, rheumatoid arthritis, Crohn's disease, ulcerative colitis, inflammatory bowel syndrome, multiple sclerosis, Addison's disease, Autoimmune hepatitis, Autoimmune parotitis, Diabetes Type I, Epididymitis, Glomerulonephritis, Graves' disease, Guillain-Barre syndrome, Hashimoto's disease, Hemolytic anemia, Systemic lupus erythematosus, Male infertility, Multiple sclerosis, Myasthenia Gravis, Pemphigus, Psoriasis
  • Another embodiment of the present invention is a composition
  • a composition comprising a nucleic acid or agent as described above, an anti-TNF-alpha polypeptide as described above, or a composition as described above, and a suitable pharmaceutical vehicle.
  • Another embodiment of the present invention is a kit for screening for a disorder as cited above, using a method as described above.
  • kits for screening for a disorder as cited above comprising an isolated anti-TNF-alpha polypeptide as described above.
  • Another embodiment of the present invention is a use of an anti-TNF-alpha polypeptide as described above for the purification of said Tumor Necrosis Factor-alpha.
  • Another embodiment of the present invention is a use of an anti-TNF-alpha polypeptide as described above for inhibiting the interaction between Tumor Necrosis Factor-alpha and one or more Tumor Necrosis Factor-alpha receptors.
  • Another embodiment of the present invention is a method as described above, wherein said host cells are bacterial or yeast.
  • Another embodiment of the present invention is a kit for screening for any of inflammation, rheumatoid arthritis, Crohn's disease, ulcerative colitis, inflammatory bowel syndrome or multiple sclerosis comprising an anti-TNF-alpha polypeptide as described above.
  • FIG. 1 Alignment of anti-human TNF VHH's as described in Example 1: VHH#3G (SEQ ID NO:121), VHH#3E (SEQ ID NO:122), VHH#1A (SEQ ID NO:123), VHH#2B (SEQ ID NO:124), VHH#12B (SEQ ID NO:125), VHH#7B (SEQ ID NO:126).
  • FIG. 2 Dilution series of anti-human TNF-alpha VHHs as tested in ELISA according to Example 1.
  • FIG. 3 Antagonistic effect of VHH as determined in cytotoxicity assay using human cell line KYM according to Example 1.
  • FIG. 4 In vitro receptor binding assay of wild type VHH#12B and mutant A74S+Y76N+K83R+P84A.
  • FIG. 5 In vitro receptor binding assay of wild type VHH#12B and mutant 1E+Q5LA74S+Y76N+K83R+P84A.
  • FIG. 6 Binding in ELISA of wild type VHH#3E and mutant VHH's.
  • FIG. 7 In vitro receptor binding assay of wild type VHH#3E and mutant VHH's.
  • FIG. 8 Alignment of antagonistic anti-mouse TNF's as described in Example 3: VHH#m3F (SEQ ID NO:127), VHH#m4B (SEQ ID NO:128), VHH#m9A (SEQ ID NO:129), VHH#m9E (SEQ ID NO:130).
  • FIG. 9 Antagonistic effect of anti-mouse TNF VHH as determined in cytotoxicity assay using murine cell line L929 according to Example 3.
  • FIG. 10 EcoRI—HindIII insert (SEQ ID NOs:131, 132) of vector pAX11 (pUC119 backbone) for production of bi-valent or bispecific VHH.
  • FIG. 11 Coomassie-stained PAGE (15%) of IMAC-purified mono-(lane 8), bi-(lane 1), tri-(lanes 2, 3 and 5) and tetravalent (lanes 4, 6 and 7) anti-TNFa VHH.
  • FIG. 12 Chromatogram of the analysis by gel filtration on Superdex 75HR of the mono-, bi-, tri and tetravalent VHH.
  • FIG. 13 Comparison of the antagonistic characteristics of the mono-, bi-, tri- and tetravalent form of the anti-human TNF VHH with the clinically used products Remicade and Enbrel.
  • FIG. 14 Antagonistic behaviour of the mono- and bivalent VHH's directed against mouse TNFalpha.
  • FIG. 15 Coomassie stained PAGE of VHH-Fc-fusion derived from human IgG1 described in Example 4.
  • FIG. 16 Antagonistic efficacy of VHH-Fc fusion derived from VHH#3E compared with bivalent format of VHH#3E as determined in bioassay.
  • FIG. 17 ELISA of reference and pepsin-treated TNF3E at pH2.2, pH3.2 and pH4.2 (100% is the signal measured at a 1/100 dilution).
  • FIG. 18 Experimental setting.
  • Table 1 Amino acid sequence listing of the peptides of aspects of present invention directed against TNF-alpha.
  • Table 2 List of mutagenesis reactions, mutagenic primers and templates used for mutagenesis of VHH#12B: mutation A74S+Y76N+K83R+P84A (SEQ ID NOs: 107, 108); mutation Q1E+Q5L+A74S+Y76N+K83R+P84A (SEQ ID NOs: 109, 110); mutation Q1E+Q5L+A74S+Y76N+K83R+P84A+T93A (SEQ ID NOs:111,112).
  • the present invention relates to an anti-tumour necrosis factor-alpha (TNF-alpha) polypeptide, comprising one or more single domain antibodies which are directed against TNF-alpha.
  • TNF-alpha anti-tumour necrosis factor-alpha
  • the invention also relates to nucleic acids capable of encoding said polypeptides.
  • Single domain antibodies are antibodies whose complementary determining regions are part of a single domain polypeptide. Examples include, but are not limited to, heavy chain antibodies, antibodies naturally devoid of light chains, single domain antibodies derived from conventional 4-chain antibodies, engineered antibodies and single domain scaffolds other than those derived from antibodies. Single domain antibodies may be any of the art, or any future single domain antibodies. Single domain antibodies may be derived from any species including, but not limited to mouse, human, camel, llama, goat, rabbit, bovine. According to one aspect of the invention, a single domain antibodies as used herein is a naturally occurring single domain antibody known as heavy chain antibody devoid of light chains. Such single domain antibodies are disclosed in WO 94/04678 for example.
  • variable domain derived from a heavy chain antibody naturally devoid of light chain is known herein as a VHH or nanobody to distinguish it from the conventional VH of four chain immunoglobulins.
  • a VHH molecule can be derived from antibodies raised in Camelidae species, for example in camel, dromedary, llama, alpaca and guanaco. bther species besides Camelidae may produce heavy chain antibodies naturally devoid of light chain; such VHHs are within the scope of the invention.
  • VHHs are heavy chain variable domains derived from immunoglobulins naturally devoid of light chains such as those derived from Camelidae as described in WO 94/04678 (and referred to hereinafter as VHH domains or nanobodies).
  • VHH molecules are about 10 ⁇ smaller than IgG molecules. They are single polypeptides and very stable, resisting extreme pH and temperature conditions. Moreover, they are resistant to the action of proteases which is not the case for conventional antibodies. Furthermore, in vitro expression of VHHs produces high yield, properly folded functional VHHs.
  • antibodies generated in Camelids will recognize epitopes other than those recognised by antibodies generated in vitro through the use of antibody libraries or via immunisation of mammals other than Camelids (WO 9749805).
  • anti-TNF-alpha VHH's may interact more efficiently with TNF-alpha than conventional antibodies, thereby blocking its interaction with the TNF-alpha receptor more efficiently.
  • TNF-alpha is derived from any species.
  • species relevant to the invention include as rabbits, goats, mice, rats, cows, calves, camels, llamas, monkeys, donkeys, guinea pigs, chickens, sheep, dogs, cats, horses, and preferably humans.
  • TNF-alpha is also a fragment of TNF-alpha, capable of eliciting an immune response.
  • TNF-alpha is also a fragment of TNF-alpha, capable of binding to a single domain antibody raised against the full length TNF-alpha.
  • a single domain antibody directed against TNF-alpha means single domain antibody that it is capable of binding to TNF-alpha with an affinity of better than 10 ⁇ 6 M.
  • One embodiment of the present invention is an anti-TNF polypeptide, wherein the single domain antibodies comprise Camelidae VHH directed against TNF-alpha.
  • the one or more single domain antibodies of the anti-TNF polypeptide which are directed against a TNF-alpha may be of the same sequence. Alternatively they may not all have the same sequence. It is within the scope of the invention that an anti-TNF polypeptide comprises anti-TNF-alpha single domain antibodies which do not all share the same sequence, but which are directed against the same target, one or more antigens thereof.
  • Another embodiment of the present invention is an anti-TNF-alpha polypeptide, wherein a single domain antibody corresponds to a sequence represented by any of SEQ ID NOs: 1 to 16 and 79 to 84 as shown in Table 1.
  • Said sequences are derived from Camelidae heavy chain antibodies (VHHs) which are directed against TNF-alpha.
  • the present invention further relates to an anti-TNF-alpha polypeptide, wherein said single domain antibody is a VHH directed against TNF-alpha, wherein the VHH belongs to a class having human-like sequences.
  • the class is characterised in that the VHHs carry an amino acid from the group consisting of glycine, alanine, valine, leucine, isoleucine, proline, phenylalanine, tyrosine, tryptophan, methionine, serine, threonine, asparagine, or glutamine at position 45, such as, for example, L45 and a tryptophan at position 103, according to the Kabat numbering.
  • VHH#2B SEQ ID NO: 3
  • VHH#12B SEQ ID No. 14
  • VHH#1A SEQ ID NO. 1
  • VHH#4B SEQ ID NO. 12
  • VHH#8-29 SEQ ID NO. 81
  • VHH#8-41 SEQ ID NO. 82
  • VHH#8-42 SEQ ID NO. 83
  • VHH#8-44 SEQ ID NO. 84
  • peptides belonging to these two classes show a high amino acid sequence homology to human VH framework regions and said peptides might be administered to a human directly without expectation of an unwanted immune response therefrom, and without the burden of further humanisation.
  • the invention also relates to nucleic acids capable of encoding said polypeptides.
  • one aspect of the present invention allows for the direct administration of an anti-TNF-alpha polypeptide, wherein the single domain antibodies belong to the humanized class of VHH, and comprise a sequence represented by any of SEQ ID NO:1, 3, 12, 14, 81, 82, 83, and 84 to a patient in need of the same.
  • VHHs as used by the invention may be of the traditional class or of the classes of human-like Camelidae antibodies. Said antibodies may be directed against whole TNF-alpha or a fragment thereof, or a fragment of a homologous sequence thereof.
  • These polypeptides include the full length Camelidae antibodies, namely Fc and VHH domains, chimeric versions of heavy chain Camelidae antibodies with a human Fc domain or VHH's by themselves or derived fragments.
  • Anti-serum albumin VHH's may interact in a more efficient way with serum albumin than conventional antibodies which is known to be a carrier protein.
  • a carrier protein some of the epitopes of serum albumin may be inaccessible by bound proteins, peptides and small chemical compounds. Since VHH's are known to bind into ‘unusual’ or non-conventional epitopes such as cavities (WO 97/49805), the affinity of such VHH's to circulating albumin may be increased.
  • the present invention also relates to the finding that an anti-TNF polypeptide as described herein further comprising one or more single domain antibodies directed against one or more serum proteins of a subject, surprisingly has significantly prolonged half-life in the circulation of said subject compared with the half-life of the anti-TNF-alpha single domain antibody when not part of said construct.
  • Examples of such polypeptides are represented in Table 5 by SEQ ID NOs: 30 to 43.
  • the said polypeptides were found to exhibit the same favourable properties of single domain antibodies such as high stability remaining intact in mice, extreme pH resistance, high temperature stability and high target affinity.
  • Another embodiment of the present invention is an anti-TNF-alpha polypeptide further comprising one or more single domain antibodies directed against one or more serum proteins, said anti-TNF alpha polypeptide comprising a sequence corresponding to any represented by SEQ ID NOs: 30 to 43 (Table 5).
  • Another embodiment of the present invention is an anti-TNF-alpha polypeptide, wherein an anti-serum protein single domain antibody corresponds to a sequence represented by any of SEQ ID NOs: 26 to 29 and 85 to 97 as shown in Table 5.
  • the serum protein may be any suitable protein found in the serum of subject.
  • the serum protein is serum albumin, serum immunoglobulins, thyroxine-binding protein, transferrin, or fibrinogen.
  • the VHH-partner can be directed to one of the above serum proteins.
  • Another aspect of the invention is an anti-TNF-alpha polypeptide as disclosed herein further comprising at least one polypeptide selected from the group consisting of an anti-IFN-gamma polypeptide, an anti-TNF-alpha receptor polypeptide and anti-IFN-gamma receptor polypeptide.
  • a single domain antibody directed against IFN-gamma corresponds to a sequence represented by any of SEQ ID NOs: 44 to 72 as shown in Table 6.
  • a single domain antibody is directed against TNF-alpha receptor.
  • Said single domain antibody may be a Camelidae VHH.
  • a single domain antibody is directed against IFN-gamma receptor.
  • Said single domain antibody may be a Camelidae VHH.
  • Another aspect of the invention is a method of treating an autoimmune disease or condition as cited herein, comprising administering to a patient an effective amount of an anti-TNF-alpha polypeptide further comprising a least one polypeptide selected from the group consisting of anti-IFN-gamma polypeptide, anti-TNF-alpha receptor polypeptide and anti-IFN-gamma receptor polypeptide, such polypeptides joined to each other as described below.
  • Such multi-specific constructs may have improved potency as inflammatory therapeutic compound over mono-specific constructs.
  • compositions comprising an anti-TNF-alpha polypeptide as disclosed herein and at least one polypeptide selected from the group consisting of anti-IFN-gamma polypeptide, anti-TNF-alpha receptor polypeptide and anti-IFN-gamma receptor polypeptide, for simultaneous, separate or sequential administration to a subject.
  • One aspect of the invention is a method for treating autoimmune disease comprising administering to an individual an effective amount of an anti-TNF-alpha polypeptide and a least one polypeptide selected from the group consisting of anti-IFN-gamma polypeptide, anti-TN F-alpha receptor polypeptide and anti-I FN-gamma receptor polypeptide, simultaneously, separately or sequentially.
  • kits containing an anti-TNF-alpha polypeptide and a least one polypeptide selected from the group consisting of anti-IFN-gamma polypeptide, anti-TNF-alpha receptor polypeptide and anti-IFN-gamma receptor polypeptide for simultaneous, separate or sequential administration to a subject. It is an aspect of the invention that the kit may be used according to the invention. It is an aspect of the invention that the kit may be used to treat the diseases as cited herein.
  • simultaneous administration means the polypeptides are administered to a subject at the same time.
  • a mixture of the polypeptides or a composition comprising said polypeptides examples include, but are not limited to a solution administered intraveneously, a tablet, liquid, topical cream, etc., wherein each preparation comprises the polypeptides of interest.
  • polypeptides are administered to a subject at the same time or substantially the same time.
  • the polypeptides are present in the kit as separate, unmixed preparations.
  • the different polypeptides may be present in the kit as individual tablets.
  • the tablets may be administered to the subject by swallowing both tablets at the same time, or one tablet directly following the other.
  • sequential administration means the polypeptides are administered to a subject sequentially.
  • the polypeptides are present in the kit as separate, unmixed preparations. There is a time interval between doses. For example, one polypeptide might be administered up to 336, 312, 288, 264, 240, 216, 192, 168, 144, 120, 96, 72, 48, 24, 20, 16, 12, 8, 4, 2, 1, or 0.5 hours after the other component.
  • one polypeptide may be administered once, or any number of times and in various doses before and/or after administration of another polypeptide.
  • Sequential administration may be combined with simultaneous or sequential administration.
  • composition comprising an anti-TNF-alpha polypeptide as disclosed herein and at least one polypeptide selected from the group consisting of anti-IFN-gamma polypeptide, anti-TNF-alpha receptor polypeptide and anti-IFN-gamma receptor polypeptide, for simultaneous, separate or sequential administration to a subject as disclosed here above.
  • an anti-IFN-gamma polypeptide anti-TNF-alpha a single domain antibody directed against IFN-gamma.
  • Said single domain antibody may be a Camelidae VHH.
  • a single domain antibody directed against IFN-gamma corresponds to a sequence represented by any of SEQ ID NOs: 44 to 72 as shown in Table 6.
  • anti-TNF-alpha a single domain antibody directed against TNF-alpha receptor.
  • Said single domain antibody may be a Camelidae VHH.
  • an anti-IFN-gamma receptor polypeptide anti-TNF-alpha a single domain antibody directed against IFN-gamma receptor.
  • Said single domain antibody may be a Camelidae VHH.
  • Another embodiment of the present invention is an anti-TNF-alpha polypeptide as disclosed herein, wherein the number of single domain antibodies directed against TNF-alpha is two or more.
  • Such multivalent anti-TNF-alpha polypeptides have the advantage of unusually high functional affinity for the target, displaying much higher than expected inhibitory properties compared to their monovalent counterparts.
  • the multivalent anti-TNF-alpha polypeptides have functional affinities that are several orders of magnitude higher than the monovalent parent anti-TNF-alpha polypeptides.
  • the inventors have found that the functional affinities of these multivalent polypeptides are much higher than those reported in the prior art for bivalent and multivalent antibodies.
  • anti-TNF-alpha polypeptides of the present invention linked to each other directly (SEQ ID No. 77 and 78) or via a short linker sequence show the high functional affinities expected theoretically with multivalent conventional four-chain antibodies.
  • the inventors have found that such large increased functional activities can be detected preferably with antigens composed of multidomain and multimeric proteins, either in straight binding assays or in functional assays, e.g. cytotoxicity assays.
  • Another embodiment of the present invention is an anti-TNF-alpha polypeptide as disclosed herein, wherein the number of single domain antibodies directed against TNF-alpha is two or more, said anti-TNF-alpha polypeptide comprising a sequence corresponding to any represented by SEQ ID NOs: 73 to 76.
  • the single domain antibodies may be joined to form any of the polypeptides disclosed herein comprising more than one single domain antibody using methods known in the art or any future method. For example, they may be fused by chemical cross-linking by reacting amino acid residues with an organic derivatising agent such as described by Blattler et al, Biochemistry 24,1517-1524; EP294703. Alternatively, the single domain antibody may be fused genetically at the DNA level i.e. a polynucleotide construct formed which encodes the complete polypeptide construct comprising one or more anti-target single domain antibodies and one or more anti-serum protein single domain antibodies.
  • a method for producing bivalent or multivalent VHH polypeptide constructs is disclosed in PCT patent application WO 96/34103.
  • One way of joining multiple single domain antibodies is via the genetic route by linking single domain antibody coding sequences either directly or via a peptide linker.
  • the C-terminal end of the first single domain antibody may be linked to the N-terminal end of the next single domain antibody.
  • This linking mode can be extended in order to link additional single domain antibodies for the construction and production of tri-, tetra-, etc. functional constructs.
  • the single domain antibodies are linked to each other directly, without use of a linker.
  • polypeptides of the invention can be linked directly (SEQ ID No. 77 and 78) thereby avoiding potential problems of the linker sequence, such as antigenicity when administered to a human subject, instability of the linker sequence leading to dissociation of the subunits.
  • the single domain antibodies are linked to each other via a peptide linker sequence.
  • linker sequence may be a naturally occurring sequence or a non-naturally occurring sequence.
  • the linker sequence is expected to be non-immunogenic in the subject to which the anti-TNF-alpha polypeptide is administered.
  • the linker sequence may provide sufficient flexibility to the multivalent anti-TNF-alpha polypeptide, at the same time being resistant to proteolytic degradation.
  • a non-limiting example of a linker sequences is one that can be derived from the hinge region of VHHs described in WO 96/34103.
  • multivalent single domain antibodies comprising more than two single domain antibodies can be linked to each other either directly or via a linker sequence.
  • Such constructs are difficult to produce with conventional antibodies and due to steric hindrance of the bulky subunits, functionality will be lost or greatly diminished rather than increased considerably as seen with VHH's of the invention compared to the monovalent construct (see FIG. 12 for gel filtration analyses of such multivalent VHH constructs).
  • VHHs may be obtained using methods known in the art such as by immunising a camel and obtaining hybridomas therefrom, or by cloning a library of single domain antibodies using molecular biology techniques known in the art and subsequent selection by using phage display.
  • an anti-TNF-alpha polypeptide may be a homologous sequence of a full-length anti-TNF-alpha polypeptide. According to another aspect of the invention, an anti-TNF-alpha polypeptide may be a functional portion of a full-length anti-TNF-alpha polypeptide. According to another aspect of the invention, an anti-TNF-alpha polypeptide may be a homologous sequence of a full-length anti-TNF-alpha polypeptide. According to another aspect of the invention, an anti-TNF-alpha polypeptide may be a functional portion of a homologous sequence of a full-length anti-TNF-alpha polypeptide. According to an aspect of the invention an anti-TNF-alpha polypeptide may comprise a sequence of an anti-TNF-alpha polypeptide.
  • a single domain antibody used to form an anti-TNF-alpha polypeptide may be a complete single domain antibody (e.g. a VHH) or a homologous sequence thereof.
  • a single domain antibody used to form the polypeptide construct may be a functional portion of a complete single domain antibody.
  • a single domain antibody used to form the polypeptide construct may be a homologous sequence of a complete single domain antibody.
  • a single domain antibody used to form the polypeptide construct may be a functional portion of a homologous sequence of a complete single domain antibody.
  • an homologous sequence of the present invention may comprise additions, deletions or substitutions of one or more amino acids, which do not substantially alter the functional characteristics of the polypeptides of the invention.
  • the number of amino acid deletions or substitutions is preferably up to 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69 or 70 amino acids.
  • a homologous sequence according to the present invention may a polypeptide modified by the addition, deletion or substitution of amino acids, said modification not substantially altering the functional characteristics compared with the unmodified polypeptide.
  • a homologous sequence according to the present invention may be a polypeptide modified by the addition, deletion or substitution of amino acids, said modification not substantially altering the functional characteristics compared with the unmodified polypeptide.
  • a homologous sequence according to the present invention may be a sequence which exists in other Camelidae species such as, for example, camel, dromedary, llama, alpaca, guanaco etc.
  • homologous sequence indicates sequence identity, it means a sequence which presents a high sequence identity (more than 70%, 75%, 80%, 85%, 90%, 95% or 98% sequence identity) with the parent sequence and is preferably characterised by similar properties of the parent sequence, namely affinity, said identity calculated using known methods.
  • an homologous sequence may also be any amino acid sequence resulting from allowed substitutions at any number of positions of the parent sequence according to the formula below:
  • Arg substituted by one of Arg, His, Gin, Lys, and Glu;
  • Pro substituted by one of Pro, Gly, Ala, and Thr;
  • Thr substituted by one of Thr, Pro, Ser, Ala, Gly, His, and Gln;
  • Ala substituted by one of Ala, Gly, Thr, and Pro;
  • Ile substituted by one of Ile, Met, Tyr, Phe, Val, and Leu;
  • Tyr substituted by one of Tyr, Trp, Met, Phe, Ile, Val, and Leu;
  • His substituted by one of His, Glu, Lys, Gln, Thr, and Arg;
  • Gln substituted by one of Gin, Glu, Lys, Asn, His, Thr, and Arg;
  • Lys substituted by one of Lys, Glu, Gln, His, and Arg;
  • Met substituted by one of Met, Phe, Ile, Val, Leu, and Tyr.
  • a homologous nucleotide sequence according to the present invention may refer to nucleotide sequences of more than 50, 100, 200, 300, 400, 500, 600, 800 or 1000 nucleotides able to hybridize to the reverse-complement of the nucleotide sequence capable of encoding the patent sequence, under stringent hybridisation conditions (such as the ones described by Sambrook et al., Molecular Cloning, Laboratory Manuel, Cold Spring, Harbor Laboratory press, New York).
  • a functional portion refers to a sequence of a single domain antibody that is of sufficient size such that the interaction of interest is maintained with affinity of 1 ⁇ 10 ⁇ 6 M or better.
  • a functional portion comprises a partial deletion of the complete amino acid sequence and still maintains the binding site(s) and protein domain(s) necessary for the binding of and interaction with the target.
  • a functional portion refers to less than 100% of the complete sequence (e.g., 99%, 90%, 80%, 70%, 60% 50%, 40%, 30%, 20%, 10%, 5%, 1% etc.), but comprises 5 or more amino acids or 15 or more nucleotides.
  • Targets as mentioned herein such as TNF-alpha, TNF-alpha receptor, serum proteins (e.g. serum albumin, serum immunoglobulins, thyroxine-binding protein, transferrin, fibrinogen) and IFN-gamma, IFN-gamma receptor may be fragments of said targets.
  • a target is also a fragment of said target, capable of eliciting an immune response.
  • a target is also a fragment of said target, capable of binding to a single domain antibody raised against the full length target.
  • a fragment as used herein refers to less than 100% of the sequence (e.g., 99%, 90%, 80%, 70%, 60%, 50%, 40%, 30%, 20%, 10% etc.), but comprising 5, 6, 7, 8, 9, 10, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25 or more amino acids.
  • a fragment is of sufficient length such that the interaction of interest is maintained with affinity of 1 ⁇ 10 ⁇ 6 M or better.
  • a fragment as used herein also refers to optional insertions, deletions and substitutions of one or more amino acids which do not substantially alter the ability of the target to bind to a single domain antibody raised against the wild-type target.
  • the number of amino acid insertions deletions or substitutions is preferably up to 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69 or 70 amino acids.
  • a homologous sequence of the present invention may include an anti-TNF-alpha polypeptide which has been humanised.
  • the humanisation of antibodies of the new class of VHHs would further reduce the possibility of unwanted immunological reaction in a human individual upon administration.
  • One embodiment of the present invention relates to a method for preparing modified polypeptides based upon llama antibodies by determining the amino acid residues of the antibody variable domain (VHH) which may be modified without diminishing the native affinity of the domain for antigen and while reducing its immunogenicity with respect to a heterologous species; the use of VHHs having modifications at the identified residues which are useful for administration to heterologous species; and to the VHH so modified.
  • VHH antibody variable domain
  • the invention relates to the preparation of modified VHHs, which are modified for administration to humans, the resulting VHH themselves, and the use of such “humanized” VHHs in the treatment of diseases in humans.
  • humanised is meant mutated so that immunogenicity upon administration in human patients is minor or nonexistent.
  • Humanising a polypeptide comprises a step of replacing one or more of the Camelidae amino acids by their human counterpart as found in the human consensus sequence, without that polypeptide losing its typical character, i.e. the humanisation does not significantly affect the antigen binding capacity of the resulting polypeptide.
  • Such methods are known by the skilled addressee.
  • Camelidae single domain antibodies requires the introduction and mutagenesis of a limited amount of amino acids in a single polypeptide chain. This is in contrast to humanization of scFv, Fab, (Fab)2 and IgG, which requires the introduction of amino acid changes in two chains, the light and the heavy chain and the preservation of the assembly of both chains.
  • the polypeptide of VHH#12B containing human-like residues in FR2 was humanized. Humanization required mutagenesis of residues in FR1 at position 1 and 5 which were introduced by the primer used for repertoire cloning and do not occur naturally in the llama sequence. Mutagenesis of those residues did not result in loss of binding and/or inhibition activity. Humanization also required mutagenesis of residues in FR3 at position 74, 76, 83, 84, 93. Mutagenesis of those residues did not result in a dramatic loss of binding and/or inhibition activity (see FIG. 4 ). Combining the mutations of FR1 and FR3 therefore did not affect the binding and/or inhibition activity ( FIG. 5 ).
  • the polypeptide represented in the VHH#3E containing camelid hallmark residues at position 37, 44, 45 and 47 with hydrophilic characteristics was humanized. Replacement of the hydrophilic residues by human hydrophobic residues at positions 44 and 45 (E44G and R45L), did not have an effect on binding and/or inhibition. However, loss of binding and/or inhibition activity was observed when F37V and F47W were introduced. Modeling data confirmed the critical residue 37 to preserve the integrity of the CDR3 loop conformation and hence on activity (see FIG. 6 )(all numbering according to the Kabat).
  • SEQ ID NO: 3 and 14 display more than 90% amino acid sequence homology to human VH framework regions and therefore said VHH might be administered to patients directly without expectation of an immune response therefrom, and without the additional burden of humanisation. Therefore, one aspect of the present invention allows for the direct administration of the polypeptide comprising SEQ ID NO: 3 and 14, homologous sequence thereof, or a functional portion of an homologous sequence thereof to a patient in need of the same.
  • One embodiment of the present invention is an anti-TNF-alpha polypeptide, or a nucleic acid capable of encoding said polypeptide for use in treating, preventing and/or alleviating the symptoms of disorders relating to inflammatory processes.
  • TNF-alpha is involved in inflammatory processes, and the blocking of TNF-alpha action can have an anti-inflammatory effect, which is highly desirable in certain disease states such as, for example, Crohn's disease.
  • Our Examples demonstrate VHHs according to the invention which bind TNF-alpha and moreover, block its binding to the TNF-alpha receptor.
  • the anti-TNF-alpha polypeptides of the present invention are applicable to autoimmune diseases, such as Addison's disease (adrenal), Autoimmune diseases of the ear (ear), Autoimmune diseases of the eye (eye), Autoimmune hepatitis (liver), Autoimmune parotitis (parotid glands), Crohn's disease (intestine), Diabetes Type I (pancreas), Epididymitis (epididymis), Glomerulonephritis (kidneys), Graves' disease (thyroid), Guillain-Barre syndrome (nerve cells), Hashimoto's disease (thyroid), Hemolytic anemia (red blood cells), Systemic lupus erythematosus (multiple tissues), Male infertility (sperm), Multiple sclerosis (nerve cells), Myasthenia Gravis (neuromuscular junction), Pemphigus (primarily skin), Psoriasis (skin), Rheumatic fever (
  • Autoimmune conditions for which the anti-TNF-alpha polypeptides of the present invention is applicable include, for example, AIDS, atopic allergy, bronchial asthma, eczema, leprosy, schizophrenia, inherited depression, transplantation of tissues and organs, chronic fatigue syndrome, Alzheimer's disease, Parkinson's disease, myocardial infarction, stroke, autism, epilepsy, Arthus's phenomenon, anaphylaxis, and alcohol and drug addiction.
  • the tissue affected is the primary target, in other cases it is the secondary target. These conditions are partly or mostly autoimmune syndromes.
  • Another embodiment of the present invention is a use of an anti-TNF-alpha polypeptide according to the invention, or a nucleic acid capable of encoding said polypeptide for the preparation of a medicament for treating a disorder relating to inflammatory processes.
  • disorders include rheumatoid arthritis, Crohn's disease, ulcerative colitis, inflammatory bowel syndrome and multiple sclerosis
  • Polypeptides and nucleic acids according to the present invention may be administered to a subject by conventional routes, such as intravenously.
  • a special property of the anti-TNF-alpha polypeptides of the invention is that they penetrate barriers such as tissue membranes and/or tumours and act locally and act locally thereon, and they are sufficiently stable to withstand extreme environments such as in the stomach. Therefore, another aspect of the present invention relates to the delivery of anti-TNF-alpha polypeptides.
  • a subject according to the invention can be any mammal susceptible to treatment by therapeutic polypeptides.
  • anti-TNF-alpha polypeptides of the invention results in the provision of such molecules in an active form in the colon at local sites that are affected by the disorder. These sites may be highly inflamed and contain TNF-alpha-producing cells.
  • the anti-TNF-alpha polypeptides of the invention which bind to TNF-alpha can neutralise the TNF-alpha locally, avoiding distribution throughout the whole body and thus limiting negative side-effects.
  • Genetically modified microorganisms such as Micrococcus lactis are able to secrete antibody or functional portions thereof. Such modified microorganisms can be used as vehicles for local production and delivery of antibodies or functional portions thereof in the intestine. By using a strain which produces an anti-TNF-alpha polypeptide, inflammatory bowel syndrome could be treated.
  • Another aspect of the invention involves delivering anti-TNF polypeptides by using surface expression on or secretion from non-invasive bacteria, such as Gram-positive host organisms like Lactococcus spec. using a vector such as described in WO00/23471.
  • One embodiment of the present invention is an anti-TNF-alpha polypeptide as disclosed herein for use in treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by a TNF-alpha modulating substance which is able pass through the gastric environment without the substance being inactivated.
  • disorders are any that cause inflammation, including, but not limited to rheumatoid arthritis, Crohn's disease, ulcerative colitis, inflammatory bowl syndrome, and multiple sclerosis.
  • formulation technology may be applied to release a maximum amount of polypeptide in the right location (in the stomach, in the colon, etc.). This method of delivery is important for treating, prevent and/or alleviate the symptoms of disorders whose targets are located in the gut system.
  • An aspect of the invention is a method for treating, preventing and/or alleviating the symptoms of a disorder susceptible to modulation by a TNF-alpha modulating substance which is able pass through the gastric environment without being inactivated, by orally administering to a subject an anti-TNF-alpha polypeptide as disclosed herein.
  • Another embodiment of the present invention is a use of an anti-TNF-alpha polypeptide as disclosed herein for the preparation of a medicament for treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by a TNF-alpha modulating substance which is able pass through the gastric environment without being inactivated.
  • An aspect of the invention is a method for delivering a TNF-alpha modulating substance to the gut system without said substance being inactivated, by orally administering to a subject an anti-TNF-alpha polypeptide as disclosed herein.
  • An aspect of the invention is a method for delivering a TNF-alpha modulating substance to the bloodstream of a subject without the substance being inactivated, by orally administering to a subject an anti-TNF-alpha polypeptide as disclosed herein.
  • Another embodiment of the present invention is an anti-TNF-alpha polypeptide as disclosed herein for use in treating, preventing and/or alleviating the symptoms or disorders susceptible to modulation by a TNF-alpha modulating substance delivered to the vaginal and/or rectal tract.
  • a formulation according to the invention comprises an anti-TNF-alpha polypeptide as disclosed herein, in the form of a gel, cream, suppository, film, or in the form of a sponge or as a vaginal ring that slowly releases the active ingredient over time (such formulations are described in EP 707473, EP 684814, U.S. Pat. No. 5,629,001).
  • An aspect of the invention is a method for treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by a TNF-alpha modulating substance delivered to the vaginal and/or rectal tract, by vaginally and/or rectally administering to a subject an anti-TNF-alpha polypeptide as disclosed herein.
  • Another embodiment of the present invention is a use of an anti-TNF-alpha polypeptide as disclosed herein for the preparation of a medicament for treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by a TNF-alpha modulating substance delivered to the vaginal and/or rectal tract.
  • An aspect of the invention is a method for delivering a TNF-alpha modulating substance to the vaginal and/or rectal tract without being said substance being inactivated, by administering to the vaginal and/or rectal tract of a subject an anti-TNF-alpha polypeptide as disclosed herein.
  • An aspect of the invention is a method for delivering a TNF-alpha modulating substance to the bloodstream of a subject without said substance being inactivated, by administering to the vaginal and/or rectal tract of a subject an anti-TNF-alpha polypeptide as disclosed herein.
  • Another embodiment of the present invention is an anti-TNF-alpha polypeptide as disclosed herein, for use in treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by a TNF-alpha modulating substance delivered to the nose, upper respiratory tract and/or lung.
  • a formulation according to the invention comprises an anti-TNF-alpha polypeptide as disclosed herein in the form of a nasal spray (e.g. an aerosol) or inhaler. Since the polypeptide construct is small, it can reach its target much more effectively than therapeutic IgG molecules.
  • An aspect of the invention is a method for treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by a TNF-alpha modulating substance delivered to the upper respiratory tract and lung, by administering to a subject an anti-TNF-alpha polypeptide as disclosed herein, by inhalation through the mouth or nose.
  • Another embodiment of the present invention is a use of an anti-TNF-alpha polypeptide as disclosed herein for the preparation of a medicament for treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by a TNF-alpha modulating substance delivered to the nose, upper respiratory tract and/or lung, without said polypeptide being inactivated.
  • An aspect of the invention is a method for delivering a TNF-alpha modulating substance to the nose, upper respiratory tract and lung without inactivation, by administering to the nose, upper respiratory tract and/or lung of a subject an anti-TNF-alpha polypeptide as disclosed herein.
  • An aspect of the invention is a method for delivering a TNF-alpha modulating substance to the bloodstream of a subject without inactivation by administering to the nose, upper respiratory tract and/or lung of a subject an anti-TNF-alpha polypeptide as disclosed herein.
  • One embodiment of the present invention is an anti-TNF-alpha polypeptide as disclosed herein for use in treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by a TNF-alpha modulating substance delivered to the intestinal mucosa, wherein said disorder increases the permeability of the intestinal mucosa. Because of their small size, an anti-TNF-alpha polypeptide as disclosed herein can pass through the intestinal mucosa and reach the bloodstream more efficiently in subjects suffering from disorders which cause an increase in the permeability of the intestinal mucosa, for example Crohn's disease.
  • An aspect of the invention is a method for treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by a TNF-alpha modulating substance delivered to the intestinal mucosa, wherein said disorder increases the permeability of the intestinal mucosa, by orally administering to a subject an anti-TNF-alpha polypeptide as disclosed herein.
  • VHH is fused to a carrier that enhances the transfer through the intestinal wall into the bloodstream.
  • this “carrier” is a second VHH which is fused to the therapeutic VHH.
  • Such fusion constructs are made using methods known in the art.
  • the “carrier” VHH binds specifically to a receptor on the intestinal wall which induces an active transfer through the wall.
  • Another embodiment of the present invention is a use of an anti-TNF-alpha polypeptide as disclosed herein for the preparation of a medicament for treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by a TNF-alpha modulating substance delivered to the intestinal mucosa, wherein said disorder increases the permeability of the intestinal mucosa.
  • An aspect of the invention is a method for delivering a TNF-alpha modulating substance to the intestinal mucosa without being inactivated, by administering orally to a subject an anti-TNF-alpha polypeptide of the invention.
  • An aspect of the invention is a method for delivering a TNF-alpha modulating substance to the bloodstream of a subject without being inactivated, by administering orally to a subject an anti-TNF-alpha polypeptide of the invention.
  • an anti-TNF-alpha polypeptide as described herein is fused to a carrier that enhances the transfer through the intestinal wall into the bloodstream.
  • this “carrier” is a VHH which is fused to said polypeptide.
  • VHH binds specifically to a receptor on the intestinal wall which induces an active transfer through the wall.
  • One embodiment of the present invention is an anti-TNF-alpha polypeptide as disclosed herein for use in treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by a TNF-alpha modulating substance which is able pass through the tissues beneath the tongue effectively.
  • disorders are any that cause inflammation, including, but not limited to rheumatoid arthritis, Crohn's disease, ulcerative colitis, inflammatory bowl syndrome, and multiple sclerosis.
  • a formulation of said polypeptide construct as disclosed herein, for example, a tablet, spray, drop is placed under the tongue and adsorbed through the mucus membranes into the capillary network under the tongue.
  • An aspect of the invention is a method for treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by a TNF-alpha modulating substance which is able pass through the tissues beneath the tongue effectively, by sublingually administering to a subject an anti-TNF-alpha polypeptide as disclosed herein.
  • Another embodiment of the present invention is a use of an anti-TNF-alpha polypeptide as disclosed herein for the preparation of a medicament for treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by a TNF-alpha modulating substance which is able to pass through the tissues beneath the tongue.
  • An aspect of the invention is a method for delivering a TNF-alpha modulating substance to the tissues beneath the tongue without being inactivated, by administering sublingually to a subject an anti-TNF-alpha polypeptide as disclosed herein.
  • An aspect of the invention is a method for delivering a TNF-alpha modulating substance to the bloodstream of a subject without being inactivated, by administering orally to a subject an anti-TNF-alpha polypeptide as disclosed herein.
  • One embodiment of the present invention is an anti-TNF-alpha polypeptide as disclosed herein for use in treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by a TNF-alpha modulating substance which is able pass through the skin effectively.
  • disorders are any that cause inflammation, including, but not limited to rheumatoid arthritis, Crohn's disease, ulcerative colitis, inflammatory bowl syndrome, and multiple sclerosis.
  • a formulation of said polypeptide construct for example, a cream, film, spray, drop, patch, is placed on the skin and passes through.
  • An aspect of the invention is a method for treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by a TNF-alpha modulating substance which is able pass through the skin effectively, by topically administering to a subject an anti-TNF-alpha polypeptide as disclosed herein.
  • Another embodiment of the present invention is a use of an anti-TNF-alpha polypeptide as disclosed herein for the preparation of a medicament for treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by a TNF-alpha modulating substance which is able pass through the skin effectively.
  • An aspect of the invention is a method for delivering a TNF-alpha modulating substance to the skin without being inactivated, by administering topically to a subject an anti-TNF-alpha polypeptide as disclosed herein.
  • An aspect of the invention is a method for delivering a TNF-alpha modulating substance to the bloodstream of a subject, by administering topically to a subject an anti-TNF-alpha polypeptide as disclosed herein.
  • an anti-TNF-alpha polypeptide further comprises a carrier single domain antibody (e.g. VHH) which acts as an active transport carrier for transport said anti-TNF-alpha polypeptide, from the lung lumen to the blood.
  • a carrier single domain antibody e.g. VHH
  • An anti-TNF-alpha polypeptide further comprising a carrier binds specifically to a receptor present on the mucosal surface (bronchial epithelial cells) resulting in the active transport of the polypeptide from the lung lumen to the blood.
  • the carrier single domain antibody may be fused to the polypeptide construct. Such fusion constructs may be made using methods known in the art and are describe herein.
  • the “carrier” single domain antibody binds specifically to a receptor on the mucosal surface which induces an active transfer through the surface.
  • Another aspect of the present invention is a method to determine which single domain antibodies (e.g. VHHs) are actively transported into the bloodstream upon nasal administration.
  • a naive or immune VHH phage library can be administered nasally, and after different time points after administration, blood or organs can be isolated to rescue phages that have been actively transported to the bloodstream.
  • a non-limiting example of a receptor for active transport from the lung lumen to the bloodstream is the Fc receptor N (FcRn).
  • FcRn Fc receptor N
  • One aspect of the invention includes the VHH molecules identified by the method. Such VHH can then be used as a carrier VHH for the delivery of a therapeutic VHH to the corresponding target in the bloodstream upon nasal administration.
  • agents When identified in an assay that measures binding or said polypeptide displacement alone, agents will have to be subjected to functional testing to determine whether they would modulate the action of the antigen in vivo. Examples of screening assays are given below primarily in respect of SEQ ID NO: 3, though any anti-TNF-alpha polypeptide as disclosed herein as disclosed herein may be appropriate.
  • phage or cells expressing TNF-alpha or a fragment thereof are incubated in binding buffer with, for example, a polypeptide represented by SEQ ID NO: 3 which has been labeled, in the presence or absence of increasing concentrations of a candidate modulator.
  • control competition reactions using increasing concentrations of said polypeptide and which is unlabeled, can be performed.
  • cells are washed extensively, and bound, labeled polypeptide is measured as appropriate for the given label (e.g., scintillation counting, fluorescence, etc.).
  • a decrease of at least 10% in the amount of labeled polypeptide bound in the presence of candidate modulator indicates displacement of binding by the candidate modulator.
  • Candidate modulators are considered to bind specifically in this or other assays described herein if they displace 50% of labeled polypeptide (sub-saturating polypeptide dose) at a concentration of 1 ⁇ M or less.
  • binding or displacement of binding can be monitored by surface plasmon resonance (SPR).
  • SPR surface plasmon resonance
  • Surface plasmon resonance assays can be used as a quantitative method to measure binding between two molecules by the change in mass near an immobilized sensor caused by the binding or loss of binding of, for example, the polypeptide represented by SEQ ID NO: 3 from the aqueous phase to TNF-alpha immobilized in a membrane on the sensor. This change in mass is measured as resonance units versus time after injection or removal of the said polypeptide or candidate modulator and is measured using a Biacore Biosensor (Biacore AB).
  • Biacore Biosensor Biacore Biosensor
  • TNF-alpha can be for example immobilized on a sensor chip (for example, research grade CM5 chip; Biacore AB) in a thin film lipid membrane according to methods described by Salamon et al. (Salamon et al., 1996, Biophys J. 71: 283-294; Salamon et al., 2001, Biophys. J. 80: 1557-1567; Salamon et al., 1999, Trends Biochem. Sci. 24: 213-219, each of which is incorporated herein by reference.). Sarrio et al.
  • SPR can be used to detect ligand binding to the GPCR A(1) adenosine receptor immobilized in a lipid layer on the chip (Sarrio et al., 2000, Mol. Cell. Biol. 20: 5164-5174, incorporated herein by reference).
  • Conditions for the binding of SEQ ID NO:3 to TNF-alpha in an SPR assay can be fine-tuned by one of skill in the art using the conditions reported by Sarrio et al. as a starting point.
  • SPR can assay for modulators of binding in at least two ways.
  • a polypeptide represented by SEQ ID NO: 3 for example, can be pre-bound to immobilized TNF-alpha followed by injection of candidate modulator at a concentration ranging from 0.1 nM to 1 ⁇ M. Displacement of the bound polypeptide can be quantitated, permitting detection of modulator binding.
  • the membrane-bound TNF-alpha can be pre-incubated with a candidate modulator and challenged with, for example, a polypeptide represented by SEQ ID NO: 3.
  • a difference in binding affinity between said polypeptide and TNF-alpha pre-incubated with the modulator, compared with that between said polypeptide and TNF-alpha in absence of the modulator will demonstrate binding or displacement of said polypeptide in the presence of modulator.
  • a decrease of 10% or more in the amount of said polypeptide bound in the presence of candidate modulator, relative to the amount of said polypeptide bound in the absence of candidate modulator indicates that the candidate modulator inhibits the interaction of TNF-alpha and said polypeptide.
  • FRET fluorescence resonance energy transfer
  • the fluorescence emitted upon excitation of the donor fluorophore will have a different wavelength from that emitted in response to that excitation wavelength when the said polypeptide and TNF-alpha are not bound, providing for quantitation of bound versus unbound molecules by measurement of emission intensity at each wavelength.
  • Donor fluorophores with which to label the TNF-alpha are well known in the art. Of particular interest are variants of the A. Victoria GFP known as Cyan FP (CFP, Donor (D)) and Yellow FP (YFP, Acceptor (A)).
  • the YFP variant can be made as a fusion protein with TNF-alpha.
  • a sample as used herein may be any biological sample containing TNF-alpha such as clinical (e.g. cell fractions, whole blood, plasma, serum, tissue, cells, etc.), derived from clinical, agricultural, forensic, research, or other possible samples.
  • the clinical samples may be from human or animal origin.
  • the sample analysed can be both solid or liquid in nature. It is evident when solid materials are used, these are first dissolved in a suitable solution.
  • FRET fluorescence quenching to monitor molecular interactions.
  • One molecule in the interacting pair can be labelled with a fluorophore, and the other with a molecule that quenches the fluorescence of the fluorophore when brought into close apposition with it.
  • a change in fluorescence upon excitation is indicative of a change in the association of the molecules tagged with the fluorophore:quencher pair.
  • an increase in fluorescence of the labelled TNF-alpha is indicative that anti-TNF-alpha polypeptide bearing the quencher has been displaced.
  • fluorescence polarization measurement is useful to quantitate binding.
  • the fluorescence polarization value for a fluorescently-tagged molecule depends on the rotational correlation time or tumbling rate.
  • Complexes such as those formed by TNF-alpha associating with a fluorescently labelled anti-TNF-alpha polypeptide, have higher polarization values than uncomplexed, labelled polypeptide.
  • the inclusion of a candidate inhibitor of the TNF-alpha:anti-TNF-alpha polypeptide interaction results in a decrease in fluorescence polarization, relative to a mixture without the candidate inhibitor, if the candidate inhibitor disrupts or inhibits the interaction of TNF-alpha with said polypeptide.
  • Fluorescence polarization is well suited for the identification of small molecules that disrupt the formation of TNF-alpha:anti-TNF-alpha polypeptide complexes.
  • TNF-alpha:anti-TNF-alpha polypeptide interactions uses a biosensor assay.
  • ICS biosensors have been described in the art (Australian Membrane Biotechnology Research Institute; Cornell B, Braach-Maksvytis V, King L, Osman P, Raguse B, Wieczorek L, and Pace R. “A biosensor that uses ion-channel switches” Nature 1997, 387, 580).
  • the association of TNF-alpha and a anti-TNF-alpha polypeptide is coupled to the closing of gramacidin-facilitated ion channels in suspended membrane bilayers and thus to a measurable change in the admittance (similar to impedence) of the biosensor.
  • a modulator will interact at a location removed from the site of interaction and cause, for example, a conformational change in the TNF-alpha.
  • Modulators that act in this manner are nonetheless of interest as agents to modulate the binding of TNF-alpha to its receptor.
  • any of the binding assays described can be used to determine the presence of an agent in a sample, e.g., a tissue sample, that binds to TNF-alpha, or that affects the binding of, for example, a polypeptide represented by SEQ ID NO: 3 to the TNF-alpha.
  • a TNF-alpha is reacted with said polypeptide in the presence or absence of the sample, and polypeptide binding is measured as appropriate for the binding assay being used.
  • a decrease of 10% or more in the binding of said polypeptide indicates that the sample contains an agent that modulates the binding of said polypeptide to the TNF-alpha.
  • the above-generalized method might easily be applied to screening for candidate modulators which alter the binding between any anti-TNF-alpha polypeptide of the invention, an homologous sequence thereof, a functional portion thereof or a functional portion of an homologous sequence thereof, and TNF-alpha or a fragment thereof.
  • One embodiment of the present invention is an unknown agent identified by the method disclosed herein.
  • One embodiment of the present invention is an unknown agent identified by the method disclosed herein for use in treating, preventing and/or alleviating the symptoms of disorders relating to inflammatory processes.
  • Another embodiment of the present invention is a use of an unknown agent identified by the method disclosed herein for use in treating, preventing and/or alleviating the symptoms of disorders relating to inflammatory processes.
  • disorders include rheumatoid arthritis, Crohn's disease, ulcerative colitis, inflammatory bowel syndrome and multiple sclerosis
  • a cell that is useful according to the invention is preferably selected from the group consisting of bacterial cells such as, for example, E. coli, yeast cells such as, for example, S. cerevisiae, P. pastoris, insect cells or mammal cells.
  • a cell that is useful according to the invention can be any cell into which a nucleic acid sequence encoding a polypeptide comprising an anti-TNF-alpha of the invention, an homologous sequence thereof, a functional portion thereof or a functional portion of an homologous sequence thereof according to the invention can be introduced such that the polypeptide is expressed at natural levels or above natural levels, as defined herein.
  • a polypeptide of the invention that is expressed in a cell exhibits normal or near normal pharmacology, as defined herein.
  • a polypeptide of the invention that is expressed in a cell comprises the nucleotide sequence capable of encoding any one of the amino acid sequences presented in Table 1 or capable of encoding an amino acid sequence that is at least 70% identical to the amino acid sequence presented in Table 1.
  • a cell is selected from the group consisting of COS7-cells, a CHO cell, a LM (TK-) cell, a NIH-3T3 cell, HEK-293 cell, K-562 cell or a 1321N1 astrocytoma cell but also other transfectable cell lines.
  • terapéuticaally effective amount means the amount needed to achieve the desired result or results (modulating TNF-alpha binding; treating or preventing inflammation).
  • an “effective amount” can vary for the various compounds that modulate TNF-alpha binding used in the invention.
  • One skilled in the art can readily assess the potency of the compound.
  • the term “compound” refers to an anti-TNF-alpha polypeptide of the present invention, a composition, or a nucleic acid capable of encoding said polypeptide or an agent identified according to the screening method described herein or said polypeptide comprising one or more derivatised amino acids.
  • pharmaceutically acceptable is meant a material that is not biologically or otherwise undesirable, i.e., the material may be administered to an individual along with the compound without causing any undesirable biological effects or interacting in a deleterious manner with any of the other components of the pharmaceutical composition in which it is contained.
  • Anti-TNF-alpha polypeptides as disclosed herein is useful for treating or preventing conditions in a subject and comprises administering a pharmaceutically effective amount of a compound or composition.
  • Anti-TNF polypeptides of the present invention are useful for treating or preventing conditions relating to rheumatoid arthritis, Crohn's disease, ulcerative colitis, inflammatory bowel syndrome and multiple sclerosis in a subject and comprises administering a pharmaceutically effective amount of a compound or composition that binds TNF-alpha.
  • Anti-TNF-alpha polypeptides as disclosed here in are useful for treating or preventing conditions in a subject and comprises administering a pharmaceutically effective amount of a compound combination with another, such as, for example, aspirin.
  • the anti-TNF-alpha polypeptides as disclosed here in are useful for treating or preventing conditions relating to rheumatoid arthritis, Crohn's disease, ulcerative colitis, inflammatory bowel syndrome and multiple sclerosis in a subject and comprises administering a pharmaceutically effective amount of a compound combination with another, such as, for example, aspirin.
  • the present invention is not limited to the administration of formulations comprising a single compound of the invention. It is within the scope of the invention to provide combination treatments wherein a formulation is administered to a patient in need thereof that comprises more than one compound of the invention.
  • Conditions mediated by TNF-alpha include, but are not limited rheumatoid arthritis, Crohn's disease, ulcerative colitis, inflammatory bowel syndrome and multiple sclerosis.
  • a compound useful in the present invention can be formulated as pharmaceutical compositions and administered to a mammalian host, such as a human patient or a domestic animal in a variety of forms adapted to the chosen route of administration, i.e., orally or parenterally, by intranassally by inhalation, intravenous, intramuscular, topical or subcutaneous routes.
  • a compound of the present invention can also be administered using gene therapy methods of delivery. See, e.g., U.S. Pat. No. 5,399,346, which is incorporated by reference in its entirety.
  • gene therapy methods of delivery See, e.g., U.S. Pat. No. 5,399,346, which is incorporated by reference in its entirety.
  • primary cells transfected with the gene for the compound of the present invention can additionally be transfected with tissue specific promoters to target specific organs, tissue, grafts, tumors, or cells.
  • the present compound may be systemically administered, e.g., orally, in combination with a pharmaceutically acceptable vehicle such as an inert diluent or an assimilable edible carrier. They may be enclosed in hard or soft shell gelatin capsules, may be compressed into tablets, or may be incorporated directly with the food of the patient's diet.
  • a pharmaceutically acceptable vehicle such as an inert diluent or an assimilable edible carrier.
  • the active compound may be combined with one or more excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, and the like.
  • Such compositions and preparations should contain at least 0.1% of active compound.
  • the percentage of the compositions and preparations may, of course, be varied and may conveniently be between about 2 to about 60% of the weight of a given unit dosage form.
  • the amount of active compound in such therapeutically useful compositions is such that an effective dosage level will be obtained.
  • the tablets, troches, pills, capsules, and the like may also contain the following: binders such as gum tragacanth, acacia, corn starch or gelatin; excipients such as dicalcium phosphate; a disintegrating agent such as corn starch, potato starch, alginic acid and the like; a lubricant such as magnesium stearate; and a sweetening agent such as sucrose, fructose, lactose or aspartame or a flavoring agent such as peppermint, oil of wintergreen, or cherry flavoring may be added.
  • a liquid carrier such as a vegetable oil or a polyethylene glycol.
  • any material used in preparing any unit dosage form should be pharmaceutically acceptable and substantially non-toxic in the amounts employed.
  • the active compound may be incorporated into sustained-release preparations and devices.
  • the active compound may also be administered intravenously or intraperitoneally by infusion or injection.
  • Solutions of the active compound or its salts can be prepared in water, optionally mixed with a nontoxic surfactant.
  • Dispersions can also be prepared in glycerol, liquid polyethylene glycols, triacetin, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms.
  • the pharmaceutical dosage forms suitable for injection or infusion can include sterile aqueous solutions or dispersions or sterile powders comprising the active ingredient which are adapted for the extemporaneous preparation of sterile injectable or infusible solutions or dispersions, optionally encapsulated in liposomes.
  • the liquid carrier or vehicle can be a solvent or liquid dispersion medium comprising, for example, water, ethanol, a polyol (for example, glycerol, propylene glycol, liquid polyethylene glycols, and the like), vegetable oils, nontoxic glyceryl esters, and suitable mixtures thereof.
  • the proper fluidity can be maintained, for example, by the formation of liposomes, by the maintenance of the required particle size in the case of dispersions or by the use of surfactants.
  • the prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars, buffers or sodium chloride. Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin.
  • Sterile injectable solutions are prepared by incorporating the active compound in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by filter sterilization.
  • the preferred methods of preparation are vacuum drying and the freeze drying techniques, which yield a powder of the active ingredient plus any additional desired ingredient present in the previously sterile-filtered solutions.
  • the present compound may be applied in pure form, i.e., when they are liquids. However, it will generally be desirable to administer them to the skin as compositions or formulations, in combination with a dermatologically acceptable carrier, which may be a solid or a liquid.
  • a dermatologically acceptable carrier which may be a solid or a liquid.
  • Useful solid carriers include finely divided solids such as talc, clay, microcrystalline cellulose, silica, alumina and the like.
  • Useful liquid carriers include water, hydroxyalkyls or glycols or water-alcohol/glycol blends, in which the present compound can be dissolved or dispersed at effective levels, optionally with the aid of non-toxic surfactants.
  • Adjuvants such as fragrances and additional antimicrobial agents can be added to optimize the properties for a given use.
  • the resultant liquid compositions can be applied from absorbent pads, used to impregnate bandages and other dressings, or sprayed onto the affected area using pump-type or aerosol sprayers.
  • Thickeners such as synthetic polymers, fatty acids, fatty acid salts and esters, fatty alcohols, modified celluloses or modified mineral materials can also be employed with liquid carriers to form spreadable pastes, gels, ointments, soaps, and the like, for application directly to the skin of the user.
  • Examples of useful dermatological compositions which can be used to deliver the compound to the skin are known to the art; for example, see Jacquet et al. (U.S. Pat. No. 4,608,392), Geria (U.S. Pat. No. 4,992,478), Smith et al. (U.S. Pat. No. 4,559,157) and Wortzman (U.S. Pat. No. 4,820,508).
  • Useful dosages of the compound can be determined by comparing their in vitro activity, and in vivo activity in animal models. Methods for the extrapolation of effective dosages in mice, and other animals, to humans are known to the art; for example, see U.S. Pat. No. 4,938,949.
  • the concentration of the compound(s) in a liquid composition will be from about 0.1-25 wt- %, preferably from about 0.5-10 wt- %.
  • concentration in a semi-solid or solid composition such as a gel or a powder will be about 0.1-5 wt- %, preferably about 0.5-2.5 wt- %.
  • the amount of the compound, or an active salt or derivative thereof, required for use in treatment will vary not only with the particular salt selected but also with the route of administration, the nature of the condition being treated and the age and condition of the patient and will be ultimately at the discretion of the attendant physician or clinician. Also the dosage of the compound varies depending on the target cell, tumor, tissue, graft, or organ.
  • the desired dose may conveniently be presented in a single dose or as divided doses administered at appropriate intervals, for example, as two, three, four or more sub-doses per day.
  • the sub-dose itself may be further divided, e.g., into a number of discrete loosely spaced administrations; such as multiple inhalations from an insufflator or by application of a plurality of drops into the eye.
  • An administration regimen could include long-term, daily treatment.
  • long-term is meant at least two weeks and preferably, several weeks, months, or years of duration. Necessary modifications in this dosage range may be determined by one of ordinary skill in the art using only routine experimentation given the teachings herein. See Remington's Pharmaceutical Sciences (Martin, E. W., ed. 4), Mack Publishing Co., Easton, Pa. The dosage can also be adjusted by the individual physician in the event of any complication.
  • the invention provides for an agent that is a modulator of TNF-alpha/TNF-alpha-receptor interactions.
  • the candidate agent may be a synthetic agent, or a mixture of agents, or may be a natural product (e.g. a plant extract or culture supernatant).
  • a candidate agent according to the invention includes a small molecule that can be synthesized, a natural extract, peptides, proteins, carbohydrates, lipids etc.
  • Candidate modulator agents from large libraries of synthetic or natural agents can be screened. Numerous means are currently used for random and directed synthesis of saccharide, peptide, and nucleic acid based agents. Synthetic agent libraries are commercially available from a number of companies including Maybridge Chemical Co. (Trevillet, Cornwall, UK), Comgenex (Princeton, N.J.), Brandon Associates (Merrimack, N.H.), and Microsource (New Milford, Conn.). A rare chemical library is available from Aldrich (Milwaukee, Wis.). Combinatorial libraries are available and can be prepared.
  • libraries of natural agents in the form of bacterial, fungal, plant and animal extracts are available from e.g., Pan Laboratories (Bothell, Wash.) or MycoSearch (N.C.), or are readily producible by methods well known in the art. Additionally, natural and synthetically produced libraries and agents are readily modified through conventional chemical, physical, and biochemical means.
  • Useful agents may be found within numerous chemical classes. Useful agents may be organic agents, or small organic agents. Small organic agents have a molecular weight of more than 50 yet less than about 2,500 daltons, preferably less than about 750, more preferably less than about 350 daltons. Exemplary classes include heterocycles, peptides, saccharides, steroids, and the like. The agents may be modified to enhance efficacy, stability, pharmaceutical compatibility, and the like. Structural identification of an agent may be used to identify, generate, or screen additional agents.
  • peptide agents may be modified in a variety of ways to enhance their stability, such as using an unnatural amino acid, such as a D-amino acid, particularly D-alanine, by functionalizing the amino or carboxylic terminus, e.g. for the amino group, acylation or alkylation, and for the carboxyl group, esterification or amidification, or the like.
  • an unnatural amino acid such as a D-amino acid, particularly D-alanine
  • a useful concentration of a candidate agent according to the invention is from about 10 mM to about 100 ⁇ M or more (i.e. 1 mM, 10 mM, 100 mM, 1 M etc.).
  • the primary screening concentration will be used as an upper limit, along with nine additional concentrations, wherein the additional concentrations are determined by reducing the primary screening concentration at half-log intervals (e.g. for 9 more concentrations) for secondary screens or for generating concentration curves.
  • a high throughput screening kit comprises all the necessary means and media for performing the detection of an agent that modulates TNF-alpha/TNF-alpha receptor interactions by interacting with TNF-alpha in the presence of a polypeptide, preferably at a concentration in the range of 1 ⁇ M to 1 mM.
  • the kit comprises the following.
  • Recombinant cells of the invention comprising and expressing the nucleotide sequence encoding TNF-alpha, which are grown according to the kit on a solid support, such as a microtiter plate, more preferably a 96 well microtiter plate, according to methods well known to the person skilled in the art especially as described in WO 00/02045.
  • TNF-alpha is supplied in a purified form to be immobilized on, for example, a 96 well microtiter plate by the person skilled in the art.
  • TNF-alpha is supplied in the kit pre-immobilized on, for example, a 96 well microtiter plate.
  • the TNF-alpha may be whole TNF-alpha or a fragment thereof.
  • Modulator agents according to the invention at concentrations from about 1 ⁇ M to 1 mM or more, are added to defined wells in the presence of an appropriate concentration of anti-TNF-alpha polypeptide, an homologous sequence thereof, a functional portion thereof or a functional portion of an homologous sequence thereof, said concentration of said polypeptide preferably in the range of 1 ⁇ M to 1 mM.
  • Kits may contain one or more anti-TNF-alpha polypeptide (e.g. one or more of a polypeptide represented by any of the SEQ ID NOs: 1 to 15 or other anti-TNF-alpha polypeptides, an homologous sequence thereof, a functional portion thereof or a functional portion of an homologous sequence thereof).
  • Binding assays are performed as according to the methods already disclosed herein and the results are compared to the baseline level of, for example TNF-alpha binding to an anti-TNF-alpha polypeptide, an homologous sequence thereof, a functional portion thereof or a functional portion of an homologous sequence thereof, but in the absence of added modulator agent.
  • Wells showing at least 2 fold, preferably 5 fold, more preferably 10 fold and most preferably a 100 fold or more increase or decrease in TNF-alpha-polypeptide binding (for example) as compared to the level of activity in the absence of modulator, are selected for further analysis.
  • kits useful for screening for modulators of TNF-alpha/TNF-alpha receptor binding as well as kits useful for diagnosis of disorders characterised by dysfunction of TNF-alpha.
  • the invention also provides for kits useful for screening for modulators of disorders as well as kits for their diagnosis, said disorders characterised by one or more process involving TNF-alpha.
  • Kits useful according to the invention can include an isolated TNF-alpha.
  • a kit can comprise cells transformed to express TNF-alpha.
  • a kit according to the invention can comprise a polynucleotide encoding TNF-alpha.
  • a kit according to the invention may comprise the specific primers useful for amplification of TNF-alpha.
  • Kits useful according to the invention can comprise an isolated TNF-alpha polypeptide, a homologue thereof, or a functional portion thereof.
  • a kit according to the invention can comprise cells transformed to express said polypeptide. Kits may contain more than one polypeptide.
  • a kit according to the invention can comprise a polynucleotide encoding TNF-alpha.
  • a kit according to the invention may comprise the specific primers useful for amplification of a macromolecule such as, for example, TNF-alpha. All kits according to the invention will comprise the stated items or combinations of items and packaging materials therefore. Kits will also include instructions for use.
  • a llama ( Llama glama ) was immunized with human TNF-alpha.
  • the cytokine was formulated as an emulsion with an appropriate, animal-friendly adjuvant (Specoll, CEDI Diagnostics B.V.).
  • the antigen cocktail was administered by double-spot injections intramuscularly in the neck.
  • the animal received 6 injections of the emulsion, containing 100 ⁇ g of TNF-alpha at weekly intervals.
  • 10-ml blood samples were collected from the animal and sera were prepared. The induction of an antigen specific humoral immune response was verified using the serum samples in an ELISA experiment with TNF (data not shown).
  • HcAbs conventional and heavy-chain antibodies
  • the maximal diversity of antibodies is expected to be equal to the number of sampled B-lymphocytes, which is about 10% of the number of PBLs (5 ⁇ 10 7 ).
  • the fraction of heavy-chain antibodies in llama is up to 20% of the number of B-lymphocytes. Therefore, the maximal diversity of HcAbs in the 150 ml blood sample is calculated as 10 7 different molecules.
  • Total RNA (around 400 ⁇ g) was isolated from these cells using an acid guanidinium thiocyanate extraction (Chomczynski and Sacchi, 1987).
  • cDNA was prepared on 100 ⁇ g total RNA with M-MLV Reverse Transcriptase (Gibco BRL) and oligo-dT-primer or hexanucleotide random primers (Amersham Biosciences) as described before (de Haard et al., 1999).
  • the cDNA was purified with a phenol/chloroform extraction combined with an ethanol precipitation and subsequently used as template to specifically amplify the VHH repertoire.
  • the VHH repertoire was amplified using oligo-dT primed cDNA as template with a single degenerated framework1 (FR1) primer ABL013 (5′-GAGGTBCARCTGCAGGASTCYGG-3′) (SEQ ID NO:98), introducing a PstI restriction site (in bold), in combination with the oligo-dT primer as is described in EP01205100.9.
  • This amplification yields two fragments of 1650 bp and 1300 bp, the latter being the product derived from the CH1-deleted HcAb genes.
  • the smaller PCR-product was gel purified and subsequently digested with PstI and BstEII. The BstEII-site frequently occurs within the FR4 of heavy-chain derived VHH encoding DNA-fragments.
  • VHH-repertoire was amplified in a hinge-dependent approach using two IgG specific oligonucleotide primers.
  • PstI and NotI (bold underlined) restriction site was introduced within the FR1 and hinge primers respectively, to allow cloning. Subsequently, the DNA fragments were ligated into PstI-BstEII or PstI-NotI digested phagemid vector pAX004, which is identical to pHEN1 (Hoogenboom et al., 1991), but encodes a carboxyterminal (His) 6 - and c-myc-tag for purification and detection, respectively. The ligation mixture was desalted on a Microcon filter (YM-50, Millipore) and electroporated into E. coli TG1 cells to obtain a library containing 1.8 ⁇ 10 7 clones.
  • the transformed cells were grown overnight at 37° C. on a single 20 ⁇ 20 cm plate with LB containing 100 ⁇ g/ml ampicillin and 2% glucose. The colonies were scraped from plates using 2 ⁇ TY medium and stored at ⁇ 80° C. in 20% glycerol.
  • the library was selected using in vitro biotinylated TNF-alpha.
  • the biotinylation was carried out as described by Magni et al (Anal Biochem 2001, 298, 181-188).
  • the incorporation of biotin in TNF was evaluated by SDS-PAGE analysis and detection with Extravidin-alkaline phosphatase conjugate (Sigma).
  • the functionality of the modified protein was evaluated for its ability to bind to the solid phase coated recombinant a p75 receptor.
  • VHH were selected by capturing biotinylated TNF-alpha (10 to 400 ng per well during 2 hours at room temperature) on streptavidin coated microtiter plates (coated with 100 ⁇ l of 10 ⁇ g/ml streptavidin during 16 hours at +4° C.). Antagonistic VHH were obtained by elution with an excess of receptor, either the extracellular ligand binding domain or with cells expressing the receptor.
  • VHH#1A, #2B, #3E, #3G, #7B and #12B Five clones, coded VHH#1A, #2B, #3E, #3G, #7B and #12B, with different sequences ( FIG. 1 ) were characterized in more detail.
  • VHH#3E, #3G and #7B are single-domain antibody fragments carrying the typical hydrophilic residue at position 45 (arginine) and the phenylalanine to tryptophan substitution in position 47 in FR2 thereby conferring the advantageous characteristics in terms of solubility.
  • VHH#1A contains the hydrophobic FR2 residues typically found in double-chain antibodies of human origin or from other species, but compensating this loss in hydrophilicity by the charged arginine residue on position 103 that substitutes the conserved tryptophan residue present in VH from double-chain antibodies (PCT/EP02/07804).
  • a new class of humanised Camelidae single-domain antibodies described in this invention is represented by VHH#2B and VHH#12B, which contains the hydrophobic residues in FR2 in combination with the hydrophobic residue tryptophan at position 103. Larger amounts of antibody fragments were expressed by cultivation on 50 ml scale and purified by IMAC using TALON resin (Clontech). After dialysis against PBS to remove the eluent imidazol the amount of VHH was determined by OD280; approximately 300 ⁇ g of VHH was obtained from each clone.
  • This material was used for determining the sensitivity of detection of (biotinylated) TNF in ELISA.
  • a streptavidin (10 ⁇ g/ml) coated microtiterplate was employed for capture of biotinylated TNF (1 ⁇ g/ml), VHH was diluted in 0.2% casein/PBS and incubated for 2 hours at room temperature. Bound VHH was detected with anti-MYC mAB 9E10 (0.5 ⁇ g/ml) and anti-mouse AP conjugate (1000-fold diluted, Sigma). The results are shown in FIG. 2 .
  • TNF-alpha-induced cytostasis/cytotoxicity was determined by the colorimetric MTT assay as described by Vandenabeele and colleagues (Vandenabeele, P., Declercq, W., Vercammen, D., Van de Craen, M., Grooten, J., Loetscher, H., Brockhaus, M., Lesslauer, W., Fiers, W. (1992) Functional characterization of the human tumor necrosis factor receptor p75 in a transfected rat/mouse T cell hybridoma. J. Exp. Med. 176, 1015-1024.).
  • MTT (3-(4,5-cimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide) is a pale yellow substrate that is cleaved by living cells to yield a dark blue formazan product. This process requires active mitochondria, and even freshly dead cells do not cleave significant amounts of MTT.
  • KYM cells (Sekiguchi M, Shiroko Y, Suzuki T, Imada M, Miyahara M, Fujii G. (1985) Characterization of a human rhabdomyosarcoma cell strain in tissue culture. Biomed. Pharmacother.
  • TNF-alpha (0.216 ng/ml or approx. 5 pM of trimer).
  • TNF variable amounts of antibody VHH or Remicade
  • MTT was added to the culture medium at a final concentration of 500 ⁇ g/ml and the plates were incubated at 37° C. to achieve cleavage of MTT by mitochondrial enzymes.
  • the formed formazon product which appear as black, fuzzy crystals on the bottom of the well were dissolved by addition of acid isopropanol (40 nM HCl in isopropanol) or DMSO.
  • the absorbance is measured at 570 nm.
  • the MTT assay shows that VHH#1A, which has arginine on position 103 in combination with the human-like hydrophobic residues in FR2, has a moderate antagonistic effect (IC50 ⁇ 100 nM).
  • VHH#7B with the characteristic hydrophilic residues in FR2 does not prevent binding of TNF- ⁇ to its ligand in spite of its sensitive detection of the cytokine in ELISA (curve not shown).
  • VHH#3E and #3G with hydrophilic FR2 hallmark residues are very potent antagonistic VHH's (IC50 of 20 nM).
  • VHH#3E and #3G have a high degree of homology and are clonally related (Harmsen et al., Mol.
  • VHH#3E is more potent, probably due to the fact that it has a higher affinity than VHH#3G ( FIG. 2 ).
  • the (chimeric) monoclonal antibody Remicade is very potent (IC50 of 80 pM), but its derived Fab fragment lost most of this efficacy (IC50 is 3 nM, 30 fold less than the intact mAB).
  • IC50 is 3 nM, 30 fold less than the intact mAB.
  • DP-47 is SEQ ID NO:101 and VHH#12B is SEQ ID NO:102: DP-47 E VQL L ESGGGLVQPGGSLRLSCAASGFTFS SYAMS WVRQAPGKGLEWVS AISGSGGSTYY VHH#12B Q VQL Q ESGGGLVQPGGSLRLSCAASGF E F E NHWMY WVRQAPGKGLEWVS TVNTNGLITRY
  • a specific inhibitor for the TNF-alpha cytokine, with high homology to the human germline gene DP-47 was therefore an ideal candidate to further humanize and evaluate the influence of mutagenesis on inhibition capacity in ELISA.
  • VHH#3E and a human VH3 germline revealed the presence of hydrophilic amino acid residues in FR2 of VHH#3E compared to hydrophobic residues in DP-47.
  • DP-47 E VQL L ESGGGLVQPGGSLRLSCAAS G F TFS SYAMS----- WVRQAPGK GL E W V S AISGSGGSTYY VHH#3E Q VQL Q ESGGGLVQPGGSLRLSCAASG R TFS DHSGYTYTIG W F RQAPGK ER E F V A RIYWSSGNTYY
  • VHH#12B was mutated by using a non-PCR based site-directed mutagenesis method as described by Chen and Ruffner and commercialized by Stratagene (Quickchange site-directed mutagenesis). Plasmid DNA was used as template in combination with 2 mutagenic primers introducing the desired mutation(s). The 2 primers are each complementary to opposite strands of the template plasmid DNA. In a polymerase reaction using the Pfu DNA polymerase each strand is extended from the primer sequence during a cycling program using a limited number of cycles. This results in a mixture of wild type and mutated strands. Digestion with Dpnl results in selection of the mutated in vitro synthesized DNA strand, since only the template strand is sensitive for digestion. The DNA was precipitated and transformed to E. coli and analyzed for the required mutation by sequence analysis. The generated mutant VHH's and the mutagenic primers are listed in Table 2.
  • a microtiter plate was coated overnight at 4° C. with Enbrel (Wyeth) at 2 ⁇ g/ml in PBS. The plate was washed five times with PBS-Tween and blocked for 1 hour at room temperature with PBS containing 1% casein. The plate was washed five times with PBS-Tween. Biotinylated human TNF-alpha (80 ⁇ g/ml) was pre-incubated with a dilution series of mutant or wild type VHH#12B for 1 hr at RT and the mixture was incubated for 1 hr at room temperature in the wells of the microtiterplate. The plate was washed five times with PBS-Tween.
  • Bound human TNF- ⁇ was detected using Extravidin-AP (1/1,000 dilution) and paranitrophenylphosphate (pNPP). Signals were measured after 30 minutes at 405 nm. The results are presented in FIG. 4 and 5 .
  • the IC50 increased 3-fold from 66 nM (wild type) to 200 nM (mutant Q1E+Q5L+A74S+Y76N+K83R+P84A). Mutation of position T93A resulted in loss of inhibition (data not shown).
  • the positions that still need to be humanized are: E28, E30 and Q108. However, E28 and E30 are part of the H1 canonical structure and thus part of the CDR1 according to Chothia numbering system.
  • amino acid sequences of mutant VHHs are presented in Table 4 SEQ ID NOs: 17 to 19.
  • VHH#3E was mutated by using a non-PCR based site-directed mutagenesis method as described above.
  • the obtained mutant VHH's and the mutagenic primers are listed in Table 3.
  • amino acid sequences of mutant VHHs are presented in Table 4 SEQ ID NOs 21 to 24.
  • a library containing 8.5 ⁇ 10 7 clones was generated from the first PBL's, and a library with 7 ⁇ 10 6 clones for the second PBL sample and 5.8 ⁇ 10 8 clones for the lymph node.
  • Using the combination of the oligo-dT primer and the Ig primer libraries from the first PBL sample were made containing 1.2 ⁇ 10 8 clones, from the second sample of PBL's a library of 5.7 ⁇ 10 7 clones and the lymph node derived library contained 2 ⁇ 10 8 clones.
  • the libraries were pooled dependent on the used combination of primers and the resulting two libraries were grown for propagation of phage as was described before.
  • VHH#m3F and VHH#m9E Two distinct mouse TNF-alpha specific VHH (VHH#m3F and VHH#m9E) were selected from the library obtained by amplification with 1 g derived primers, while two closely related VHH's were retrieved from the library constructed by PCR with oligo-dT primer and Ig-primer ( FIG. 8 ).
  • VHH#m3F and VHH#m4B ( FIG. 9 ) turned out to be 10-fold more potent then the other two VHH's.
  • the E. coli production vector pAX11 ( FIG. 10 ) was designed, which allows the two-step cloning of bivalent or bispecific VHH.
  • the carboxy terminal VHH is cloned first with PstI and BstEII, while in the second step the other VHH is inserted by SfiI and NotI, which do not cut within the first gene fragment.
  • the procedure avoids the enforcement of new sites by amplification and thus the risk of introducing PCR errors.
  • the plasmid vector encoding the bivalent VHH was used to generate a tri- and tetrameric derivative, which was accomplished by partial digestion of the plasmid with BstEII, which occurs in both VHH gene segments.
  • the linearized vector was purified from gel, subsequently de-phosphorylated and used as acceptor for cloning of the BstEII fragment of approx. 350 bp that was obtained by complete digestion of the same plasmid. Ligation of the BstEII fragment alone prior to addition to the vector enhances the insertion of multimeric VHH encoding gene segments. After transformation in E.
  • the clones were grown and induced on 50 ml scale, periplasmic fractions prepared and used for IMAC purification with TALON resin. Analysis of the purified products on Coomassie stained PAGE revealed good production levels (between 2 and 10 mg per liter cell culture) of intact multivalent VHH (see FIG. 11 ). The molecular appearance of the IMAC purified VHH was determined by gel filtration on a Superdex 75HR column and as expected the molecules with higher avidities came earlier from the column (see FIG. 12 ).
  • the antagonistic efficacy was analyzed with the cell based assay using KYM cells.
  • the cells were seeded in microtiterplates and cultured in the presence or absence of TNF-alpha (1.29 ng/ml or approx. 25 pM of trimer).
  • the assays revealed that the monovalent molecules used in this study had the poorest antagonistic characteristics, what is reflected by their IC50 values: the Fab derived from the chimeric antibody Remicade has an IC50 of 2 nM and for VHH#3E it is 12 nM (see also FIG. 3 ).
  • TNF-alpha is a trimeric molecule, which interacts to a dimeric receptor and therefore it can be expected that the avidity of the IgG permits the mutual binding to two epitopes on the cytokine and supports the formation of large complexes as has been described before (Santora et al, Anal. Biochem. 299, 119-129).
  • the trimeric VHH has an IC50 of 20 pM and the tetravalent format 6 pM. All higher avidity formats of the VHH are more efficient than Remicade, while the tetravalent format is even better than Enbrel, which consists of the extracellular domain of the receptor p75 fused to the Fc of an IgG and therefore has a bivalent binding mode.
  • VHH generated against mouse TNF FIG. 14
  • the same type of cytotoxicity assay was performed using MTT as substrate and mouse TNF-alpha (65 pg/ml or 1.3 pM), but with the murine cell line L929, which expresses the mouse specific receptor.
  • Three different antagonistic (monovalent) VHH were identified coded 9E and 3F, of which the first two have IC50's of 25 nM and the latter 2 nM (see also Example 3).
  • VHH#3E directed against human TNF, was cloned via PstI and BstElI in an adapted vector derived from pCDNA3, thereby generating a genetic fusion to the CH1 deleted Fc portion of human IgG1.
  • the plasmid construct was transfected to the myeloma cell line NS0. The obtained cell line was grown and the VHH-Fc fusion was secreted into the culture supernatant.
  • the product was purified with an anti-human Fc VHH resin and analyzed on a Coomassie stained gel ( FIG. 15 ).
  • VHH-fusion was tested in the bioassay with the human cell line KYM and turned out to be 5-fold less effective than the bivalent VHH in spite of the fact that both molecules have the same avidity and that they both originate from VHH#3E ( FIG. 16 ). Probably steric hindrance by the bulky Fc tail might cause this discrepancy.
  • the degree of amino acid sequence homology between anti-target single domain antibodies of the invention was calculated using the Bioedit Sequence Alignment Editor. The calculations indicate the proportion of identical residues between all of the sequences as they are aligned by ClustalW. (Thompson, J. D., Higgins, D. G. and Gibson, T. J. (1994) CLUSTAL W: improving the sensitivity of progressive multiple sequence alignment through sequence weighting, position specific gap penalties and weight matrix choice. Nucleic Acids Research, submitted, June 1994).
  • Table 8 indicates the fraction homology between anti-serum albumin VHHs of the invention.
  • Table 9 indicates the fraction homology between anti-TNF-alpha VHHs of the invention.
  • Table 10 indicates the percentage homology between anti-IFN-gamma VHHs of the invention.
  • the CDR3 region of VHH#3E was amplified by using a sense primer located in the framework 4 region (Forward: CCCCTGGCCCCAGTAGTTATACG) (SEQ ID NO:103) and an anti-sense primer located in the framework 3 region (Reverse: TGTGCAGCAAGAGACGG) (SEQ ID NO:104).
  • Reverse primer Sfi1 (SEQ ID NO:105) GTCCTCGCAACTGCGGCCCAGCCGGCCTGTGCAGCAAGAGACGG
  • Forward primer Not1 (SEQ ID NO:106) GTCCTCGCAACTGCGCGGCCGCCCCCTGGCCCCAGTAGTTATACG
  • the PCR reactions were performed in 50 ml reaction volume using 50 pmol of each primer.
  • the reaction conditions for the primary PCR were 11 min at 94° C., followed by 30/60/120 sec at 94/55/72° C. for 30 cycles, and 5 min at 72° C. All reaction were performed with 2.5 mM MgCl2, 200 mM dNTP and 1.25 U AmpliTaq God DNA Polymerase (Roche Diagnostics, Brussels, Belgium).
  • VHH#3E a VHH specific to human TNF ⁇ was produced as recombinant protein in E. coli and purified to homogeneity by IMAC and gel filtration chromatography. The protein concentration after purification was determined spectrophotometrically by using the calculated molar extinction coefficient at 280 nm. Diluted solutions at 100 microgram/ml were prepared in Mcllvaine buffer (J. Biol. Chem. 49, 1921, 183) at pH 2, pH3 and 4 respectively.
  • An antibody solution containing the anti-human TNF ⁇ specific VHH#3E (100 microgram per milliliter in 100-fold diluted PBS) was prepared. Three mice which were first deprived from drinking water for 12 hours and subsequently allowed to freely access the antibody solution during the next two hours. Afterwards the mice were sacrificed and their stomachs were dissected. Immediately the content of the stomachs was collected by flushing the stomach with 500 microliter PBS containing 1% BSA. This flushed material was subsequently used to prepare serial three-fold dilutions, starting at a 1/5 dilution from the undiluted material. One hundred microliter of these samples was transferred to individual wells of a microtiter plate coated with human TNF ⁇ .
  • Each cycle consisted of a DSS treatment interval (7 days) where DSS was provided with the drinking water at a concentration of 5% (w/v) and a recovery interval (12 days) with no DSS present in the drinking water.
  • the last recovery period was prolonged from 12 to 21 days to provide for an inflammation status rather representing a chronic than an acute inflammation at the time of the treatment.
  • mice were randomly assigned to groups of 8 mice and treatment with the VHH-constructs was started.
  • the treatment interval was 2 weeks.
  • One week after the end of the treatment interval the animals were sacrificed, the intestine was dissected and histologically examined.
  • the experimental setting is shown schematically in FIG. 18 .
  • mice 8 animals per group were treated daily for 14 consecutive days with bivalent VHH#3F (VHH#m3F-VHH#m3F; SEQ ID No. 76) by intra-gastric or intra-venous application of 100 ⁇ g bivalent VHH 3F.
  • An additional group of animals was treated rectally with the bivalent VHH#3F every other day for a period of 14 days.
  • a dose of 100 ⁇ g of the bivalent VHH#3F was applied at a concentration of 1 mg/ml in a buffered solution.
  • the negative control groups received 100 ⁇ l of PBS under otherwise identical conditions.
  • the treatment schedule is shown in Table 11.

Abstract

The present invention relates to polypeptides derived from single domain heavy chain antibodies directed to Tumor Necrosis Factor-alpha. It further relates to single domain antibodies that are Camelidae VHHs. It further relates to methods of administering said polypeptides. It further relates to protocols for screening for agents that modulate the TNF-alpha receptor, and the agents resulting from said screening.

Description

    RELATED APPLICATIONS
  • This application is a continuation of U.S. patent application Ser. No. 11/636,300 filed Dec. 8, 2006, currently pending, which is a continuation of U.S. patent application Ser. No. 10/534,348 filed May 9, 2005, currently pending, which is a National Stage of PCT/BE03/00192, filed Nov. 7, 2003, which claims priority to PCT/EP03/06581, filed Jun. 23, 2003 and PCT/EP03/07313, filed Jul. 8, 2003; this application also claims the benefit of U.S. provisional application Ser. No. 60/425,073, filed Nov. 8, 2002 and U.S. provisional application Ser. No. 60/425,063, filed Nov. 8, 2002; all of the applications are incorporated herein by reference.
  • FIELD OF THE INVENTION
  • The present invention provides polypeptides comprising one or more single domain antibodies directed towards tumor necrosis factor alpha (TNF-alpha). The present invention further relates to their use in diagnosis and therapy. Such antibodies may have a framework sequence with high homology to the human framework sequences. Compositions comprising antibodies to tumor necrosis factor alpha (TNF-alpha) alone or in combination with other drugs are described.
  • BACKGROUND TO THE INVENTION
  • Tumor necrosis factor alpha (TNF-alpha) is believed to play an important role in various disorders, for example in inflammatory disorders such as rheumatoid arthritis, Crohn's disease, ulcerative colitis and multiple sclerosis. Both TNF-alpha and the receptors (CD120a, CD120b) have been studied in great detail. TNF-alpha in its bioactive form is a trimer and the groove formed by neighboring subunits is important for the cytokine-receptor interaction. Several strategies to antagonize the action of the cytokine have been developed and are currently used to treat various disease states.
  • A TNF-alpha inhibitor which has sufficient specificity and selectivity to TNF-alpha may be an efficient prophylactic or therapeutic pharmaceutical compound for preventing or treating disorders where TNF-alpha has been implicated as causative agent. Methods of treating toxic shock (EP 486526), tumor regression, inhibition of cytotoxicity (U.S. Pat. No. 6,448,380, U.S. Pat. No. 6,451,983, U.S. Pat. No. 6,498,237), autoimmune disease such as RA and Crohn's disease (EP 663836, U.S. Pat. No. 5,672,347, U.S. Pat. No. 5,656,272), graft versus host reaction (U.S. Pat. No. 5,672,347), bacterial meningitis (EP 585705) by means of an antibody to TNF-alpha have been described.
  • Yet none of the presently available drugs are completely effective for the treatment of autoimmune disease, and most are limited by severe toxicity. In addition, it is extremely difficult and a lengthy process to develop a new chemical entity (NCE) with sufficient potency and selectivity to such target sequence. Antibody-based therapeutics on the other hand have significant potential as drugs because they have exquisite specificity to their target and a low inherent toxicity. In addition, the development time can be reduced considerably when compared to the development of new chemical entities (NCE's). However, conventional antibodies are difficult to raise against multimeric proteins where the receptor-binding domain of the ligand is embedded in a groove, as is the case with TNF-alpha. Heavy chain antibodies described in the invention which are derived from Camelidae, are known to have cavity-binding propensity (WO97/49805; Lauwereys et al, EMBO J. 17, 5312, 1998)). Therefore, such heavy chain antibodies are inherently suited to bind to receptor binding domains of such ligands as TNF. In addition, such antibodies are known to be stable over long periods of time, therefore increasing their shelf-life (Perez et al, Biochemistry, 40, 74, 2001). Furthermore, such heavy chain antibody fragments can be produced ‘en-masse’ in fermentors using cheap expression systems compared to mammalian cell culture fermentation, such as yeast or other microorganisms (EP 0 698 097).
  • The use of antibodies derived from sources such as mouse, sheep, goat, rabbit etc., and humanised derivatives thereof as a treatment for conditions which require a modulation of inflammation is problematic for several reasons. Traditional antibodies are not stable at room temperature, and have to be refrigerated for preparation and storage, requiring necessary refrigerated laboratory equipment, storage and transport, which contribute towards time and expense. Refrigeration is sometimes not feasible in developing countries. Furthermore, the manufacture or small-scale production of said antibodies is expensive because the mammalian cellular systems necessary for the expression of intact and active antibodies require high levels of support in terms of time and equipment, and yields are very low. Furthermore the large size of conventional antibodies, would restrict tissue penetration, for example, at the site of inflamed tissue. Furthermore, traditional antibodies have a binding activity which depends upon pH, and hence are unsuitable for use in environments outside the usual physiological pH range such as, for example, in treating gastric bleeding, gastric surgery. Furthermore, traditional antibodies are unstable at low or high pH and hence are not suitable for oral administration. However, it has been demonstrated that camelidae antibodies resist harsh conditions, such as extreme pH, denaturing reagents and high temperatures (Dumoulin et al, Protein Science 11, 500, 2002), so making them suitable for delivery by oral administration. Furthermore, traditional antibodies have a binding activity, which depends upon temperature, and hence are unsuitable for use in assays or kits performed at temperatures outside biologically active-temperature ranges (e.g. 37±20° C.).
  • Polypeptide therapeutics and in particular antibody-based therapeutics have significant potential as drugs because they have exquisite specificity to their target and a low inherent toxicity. However, it is known by the skilled addressee that an antibody which has been obtained for a therapeutically useful target requires additional modification in order to prepare it for human therapy, so as to avoid an unwanted immunological reaction in a human individual upon administration thereto. The modification process is commonly termed “humanisation”. It is known by the skilled artisan that antibodies raised in species, other than in humans, require humanisation to render the antibody therapeutically useful in humans ((1) CDR grafting: Protein Design Labs: U.S. Pat. No. 6,180,370, U.S. Pat. No. 5,693,761; Genentech U.S. Pat. No. 6,054,297; Celltech: 460167, EP 626390, U.S. Pat. No. 5,859,205; (2) Veneering: Xoma: U.S. Pat. No. 5,869,619, U.S. Pat. No. 5,766,886, U.S. Pat. No. 5,821,123). There is a need for a method for producing antibodies which avoids the requirement for substantial humanisation, or which completely obviates the need for humanisation. There is a need for a new class of antibodies which have defined framework regions or amino acid residues and which can be administered to a human subject without the requirement for substantial humanisation, or the need for humanisation at all.
  • Another important drawback of conventional antibodies is that they are complex, large molecules and therefore relatively unstable, and they are sensitive to breakdown by proteases. This means that conventional antibody drugs cannot be administered orally, sublingually, topically, nasally, vaginally, rectally or by inhalation because they are not resistant to the low pH at these sites, the action of proteases at these sites and in the blood and/or because of their large size. They have to be administered by injection (intravenously, subcutaneously, etc.) to overcome some of these problems. Administration by injection requires specialist training in order to use a hypodermic syringe or needle correctly and safely. It further requires sterile equipment, a liquid formulation of the therapeutic polypeptide, vial packing of said polypeptide in a sterile and stable form and, of the subject, a suitable site for entry of the needle. Furthermore, subjects commonly experience physical and psychological stress prior to and upon receiving an injection. Therefore, there is need for a method for the delivery of therapeutic polypeptides which avoids the need for injection which is not only cost/time saving, but which would also be more convenient and more comfortable for the subject.
  • Single domain antibody-based therapeutics have significant potential as drugs because they have exquisite specificity to their target and a low inherent toxicity. However, improving further their intrinsic and functional affinity can lead to many benefits for a patient such as reduced dose of therapeutic, faster therapy, and reduced side effects.
  • THE AIMS OF THE PRESENT INVENTION
  • It is an aim of the present invention is to provide polypeptides comprising one or more single domain antibodies which bind to TNF-alpha, homologues of said polypeptides, functional portions of homologues of said polypeptides. Said polypeptides modify the biological activity of TNF-alpha upon binding. Such polypeptides might bind into the receptor-binding groove of TNF-alpha, or might not bind in the receptor binding groove. Such polypeptides are single domain antibodies.
  • It is a further aim of the present invention to provide single domain antibodies which may be any of the art, or any future single domain antibodies. Examples include, but are not limited to, heavy chain antibodies, antibodies naturally devoid of light chains, single domain antibodies derived from conventional 4-chain antibodies, engineered antibodies and single domain scaffolds other than those derived from antibodies. According to one aspect of the invention, a single domain antibody as used herein is a naturally occurring single domain antibody known as heavy chain antibody devoid of light chains (WO 9404678). For clarity reasons, this variable domain derived from a heavy chain antibody devoid of light chain will be called VHH or nanobody to distinguish it from the conventional VH of four chain immunoglobulins. Such a VHH molecule can be derived from antibodies raised in Camelidae species, for example in camel, llama, dromedary, alpaca and guanaco.
  • It is a further aim of the invention to provide a method of administering anti-TNF-alpha polypeptides intravenously, subcutaneously, orally, sublingually, topically, nasally, vaginally, rectally or by inhalation.
  • It is a further aim of the invention to enhance the binding affinity of monovalent single domain antibodies.
  • SUMMARY OF THE INVENTION
  • One embodiment of the present invention is an anti-TNF-alpha polypeptide comprising at least one anti-TNF-alpha single domain antibody.
  • Another embodiment of the present invention is an anti-TNF-alpha polypeptide as described above wherein a single domain antibody corresponds to a sequence represented by any of SEQ ID NOs: 1 to 16 and 79 to 84.
  • Another embodiment of the present invention is an anti-TNF-alpha polypeptide as described above further comprising at least one single domain antibody directed against a serum protein.
  • Another embodiment of the present invention is an anti-TNF-alpha polypeptide as described above wherein said serum protein is any of serum albumin, serum immunoglobulins, thyroxine-binding protein, transferring, or fibrinogen.
  • Another embodiment of the present invention is an anti-TNF-alpha polypeptide as described above wherein a single domain anti-serum protein single domain antibody correspond to a sequence represented by any of SEQ ID NOs: 26 to 29 and 85 to 97.
  • Another embodiment of the present invention is an anti-TNF-alpha polypeptide as described above corresponding to a sequence represented by any of SEQ ID NOs: 30 to 43.
  • Another embodiment of the present invention is an anti-TNF-alpha polypeptide as described above further comprising at least one single domain antibody selected from the group consisting of anti-IFN-gamma single domain antibody, anti-TNF-alpha receptor single domain antibody and anti-IFN-gamma receptor single domain antibody.
  • Another embodiment of the present invention is an anti-TNF-alpha polypeptide as described above, wherein the number of single domain antibodies directed against TNF-alpha is at least two.
  • Another embodiment of the present invention is an anti-TNF-alpha polypeptide as described above corresponding to a sequence represented by any of SEQ ID NOs: 73 to 76.
  • Another embodiment of the present invention is an anti-TNF-alpha polypeptide as described above, wherein at least one single domain antibody is a humanized Camelidae VHHs.
  • Another embodiment of the present invention is an anti-TNF-alpha polypeptide as described above wherein a humanized Camelidae VHH corresponds to a sequence represented by any of SEQ ID NOs: 17 to 19 and 21 to 24.
  • Another embodiment of the present invention is a composition comprising an anti-TNF-alpha polypeptide as described above and at least one single domain antibody from the group consisting of anti-IFN-gamma single domain antibody, anti-TNF-alpha receptor single domain antibody and anti-IFN-gamma receptor single domain antibody, for simultaneous, separate or sequential administration to a subject.
  • Another embodiment of the present invention is an anti-TNF-alpha polypeptide as described above, or a composition as described above wherein at least one anti-IFN-gamma single domain antibody correspond to a sequence represented by any of SEQ ID NOs: 44 to 72.
  • Another embodiment of the present invention is an anti-TNF-alpha polypeptide as described above, or a composition as described above, wherein said single domain antibody is an homologous sequence, a functional portion, or a functional portion of an homologous sequence of the full length single domain antibody.
  • Another embodiment of the present invention is an anti-TNF-alpha polypeptide as described above, or a composition as described above, wherein the anti-TNF-alpha polypeptide is an homologous sequence, a functional portion, or a functional portion of an homologous sequence of the full length anti-TNF-alpha polypeptide.
  • Another embodiment of the present invention is an anti-TNF-alpha polypeptide as described above, or a composition as described above wherein at least one single domain antibody is a Camelidae VHH.
  • Another embodiment of the present invention is a nucleic acid encoding an anti-TNF-alpha polypeptide as described above.
  • Another embodiment of the present invention is a method of identifying an agent that modulates the binding of an anti-TNF-alpha polypeptide as described above, to Tumor Necrosis Factor-alpha comprising the steps of:
    • (a) contacting an anti-TNF-alpha polypeptide as described above with a target that is Tumor Necrosis Factor alpha, in the presence and absence of a candidate modulator under conditions permitting binding between said polypeptide and target, and
    • (b) measuring the binding between the polypeptide and target of step (a), wherein a decrease in binding in the presence of said candidate modulator, relative to the binding in the absence of said candidate modulator identified said candidate modulator as an agent that modulates the binding of an anti-TNF-alpha polypeptide as described above and Tumor Necrosis Factor-alpha.
  • Another embodiment of the present invention is a method of identifying an agent that modulates Tumor Necrosis Factor-alpha-mediated disorders through the binding of an anti-TNF-alpha polypeptide as described above to Tumor Necrosis Factor-alpha comprising:
    • (a) contacting an anti-TNF-alpha polypeptide as described above with a target that is Tumor Necrosis Factor alpha, in the presence and absence of a candidate modulator under conditions permitting binding between said polypeptide and target, and
    • (b) measuring the binding between the polypeptide and target of step (a), wherein a decrease in binding in the presence of said candidate modulator, relative to the binding in the absence of said candidate modulator identified, said candidate modulator as an agent that modulates Tumor Necrosis Factor alpha-mediated disorders.
  • Another embodiment of the present invention is a method of identifying an agent that modulates the binding of Tumor Necrosis Factor alpha to its receptor through the binding of an anti-TNF-alpha polypeptide as described above to Tumor Necrosis Factor-alpha comprising:
    • (a) contacting an anti-TNF-alpha polypeptide as described above with a target that is Tumor Necrosis Factor-alpha, in the presence and absence of a candidate modulator under conditions permitting binding between said polypeptide and target, and
    • (b) measuring the binding between the polypeptide and target of step (a), wherein a decrease in binding in the presence of said candidate modulator, relative to the binding in the absence of said candidate modulator identified said candidate modulator as an agent that modulates the binding of Tumor Necrosis Factor-alpha to its receptor.
  • Another embodiment of the present invention is a kit for screening for agents that modulate Tumor Necrosis Factor-alpha-mediated disorders comprising an anti-TNF-alpha polypeptide as described above and Tumor Necrosis Factor-alpha.
  • Another embodiment of the present invention is an unknown agent that modulates the binding of an anti-TNF-alpha polypeptide as described above to Tumor Necrosis Factor-alpha, identified according to the method as described above.
  • Another embodiment of the present invention is an unknown agent that modulates Tumor Necrosis Factor-alpha-mediated disorders, identified according to the methods as described above.
  • Another embodiment of the present invention is an unknown agent as described above wherein said disorders are one or more of inflammation, rheumatoid arthritis, Crohn's disease, ulcerative colitis, inflammatory bowel syndrome and multiple sclerosis.
  • Another embodiment of the present invention is an anti-TNF-alpha polypeptide as described above, or a nucleic acid as described above, or a composition as described above, or an agent as described above for treating and/or preventing and/or alleviating disorders relating to inflammatory processes.
  • Another embodiment of the present invention is a use of an anti-TNF-alpha polypeptide as described above or a nucleic acid as described above, or a composition as described above, or an agent as described above for the preparation of a medicament for treating and/or preventing and/or alleviating disorders relating to inflammatory reactions.
  • Another embodiment of the present invention is an anti-TNF-alpha polypeptide as described above or a composition as described above, for treating and/or preventing and/or alleviating disorders susceptible to modulation by a TNF-alpha modulating substance which is able pass through the gastric environment without the substance being inactivated.
  • Another embodiment of the present invention is an use of an anti-TNF-alpha polypeptide as described above or a composition as described above, for the preparation of a medicament for treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by a TNF-alpha modulating substance which is able pass through the gastric environment without the substance being inactivated.
  • Another embodiment of the present invention is an anti-TNF-alpha polypeptide as described above or a composition as described above, for treating and/or preventing and/or alleviating disorders susceptible to modulation by a TNF-alpha modulating substance delivered to the vaginal and/or rectal tract.
  • Another embodiment of the present invention is a use of an anti-TNF-alpha polypeptide as described above or a composition as described above, for the preparation of a medicament for treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by a TNF-alpha modulating substance delivered to the vaginal and/or rectal tract.
  • Another embodiment of the present invention is an anti-TNF-alpha polypeptide as described above or a composition as described above, for treating and/or preventing and/or alleviating disorders susceptible to modulation by a TNF-alpha modulating substance delivered to the nose, upper respiratory tract and/or lung.
  • Another embodiment of the present invention is a use of an anti-TNF-alpha polypeptide as described above or a composition as described above, for the preparation of a medicament for treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by a TNF-alpha modulating substance delivered to the nose, upper respiratory tract and/or lung.
  • Another embodiment of the present invention is an anti-TNF-alpha polypeptide as described above or a composition as described above, for treating and/or preventing and/or alleviating disorders susceptible to modulation by a TNF-alpha modulating substance delivered to the intestinal mucosa, wherein said disorder increases the permeability of the intestinal mucosa.
  • Another embodiment of the present invention is a use of an anti-TNF-alpha polypeptide as described above or a composition as described above, for the preparation of a medicament for treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by a TNF-alpha modulating substance delivered to the intestinal mucosa, wherein said disorder increases the permeability of the intestinal mucosa.
  • Another embodiment of the present invention is an anti-TNF-alpha polypeptide as described above or a composition as described above, for treating and/or preventing and/or alleviating disorders susceptible to modulation by a TNF-alpha modulating substance which is able pass through the tissues beneath the tongue effectively.
  • Another embodiment of the present invention is a use of an anti-TNF-alpha polypeptide as described above or a composition as described above, for the preparation of a medicament for treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by a TNF-alpha modulating substance which is able pass through the tissues beneath the tongue effectively.
  • Another embodiment of the present invention is an anti-TNF-alpha polypeptide as described above or a composition as described above, for treating and/or preventing and/or alleviating disorders susceptible to modulation by a TNF-alpha modulating substance which is able pass through the skin effectively.
  • Another embodiment of the present invention is a use of an anti-TNF-alpha polypeptide as described above or a composition as described above, for the preparation of a medicament for treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by a TNF-alpha modulating substance which is able pass through the skin effectively.
  • Another embodiment of the present invention is a method as described above, a kit as described above, a nucleic acid or agent as described above, use of a nucleic acid or agent as described above, a composition as described above, use of a composition as described above, an anti-TNF-alpha polypeptide as described above, use of an anti-TNF-alpha polypeptide as described above wherein said disorders are any of inflammation, rheumatoid arthritis, Crohn's disease, ulcerative colitis, inflammatory bowel syndrome, multiple sclerosis, Addison's disease, Autoimmune hepatitis, Autoimmune parotitis, Diabetes Type I, Epididymitis, Glomerulonephritis, Graves' disease, Guillain-Barre syndrome, Hashimoto's disease, Hemolytic anemia, Systemic lupus erythematosus, Male infertility, Multiple sclerosis, Myasthenia Gravis, Pemphigus, Psoriasis, Rheumatic fever, Rheumatoid arthritis, Sarcoidosis, Scleroderma, Sjogren's syndrome, Spondyloarthropathies, Thyroiditis, and Vasculitis.
  • Another embodiment of the present invention is a composition comprising a nucleic acid or agent as described above, an anti-TNF-alpha polypeptide as described above, or a composition as described above, and a suitable pharmaceutical vehicle.
  • Another embodiment of the present invention is a method of diagnosing a disorder characterised by the dysfunction of Tumor Necrosis Factor-alpha comprising:
    • (a) contacting a sample with an anti-TNF-alpha polypeptide as described above,
    • (b) detecting binding of said polypeptide to said sample, and
    • (c) comparing the binding detected in step (b) with a standard, wherein a difference in binding relative to said sample is diagnostic of a disorder characterised by dysfunction of Tumor Necrosis Factor-alpha.
  • Another embodiment of the present invention is a kit for screening for a disorder as cited above, using a method as described above.
  • Another embodiment of the present invention is a kit for screening for a disorder as cited above comprising an isolated anti-TNF-alpha polypeptide as described above.
  • Another embodiment of the present invention is a use of an anti-TNF-alpha polypeptide as described above for the purification of said Tumor Necrosis Factor-alpha.
  • Another embodiment of the present invention is a use of an anti-TNF-alpha polypeptide as described above for inhibiting the interaction between Tumor Necrosis Factor-alpha and one or more Tumor Necrosis Factor-alpha receptors.
  • Another embodiment of the present invention is a method for producing an anti-TNF-alpha polypeptide as described above comprising the steps of:
    • (a) obtaining double stranded DNA encoding a Camelidae VHH directed to Tumor Necrosis Factor alpha,
    • (b) cloning and expressing the DNA selected in step (b).
  • Another embodiment of the present invention is a method of producing an anti-TNF-alpha polypeptide as described above comprising:
    • (a) culturing host cells comprising nucleic acid capable of encoding an anti-TNF-alpha polypeptide as described above, under conditions allowing the expression of the polypeptide, and,
    • (b) recovering the produced polypeptide from the culture.
  • Another embodiment of the present invention is a method as described above, wherein said host cells are bacterial or yeast.
  • Another embodiment of the present invention is a kit for screening for any of inflammation, rheumatoid arthritis, Crohn's disease, ulcerative colitis, inflammatory bowel syndrome or multiple sclerosis comprising an anti-TNF-alpha polypeptide as described above.
  • BRIEF DESCRIPTION OF FIGURES AND TABLES
  • FIG. 1 Alignment of anti-human TNF VHH's as described in Example 1: VHH#3G (SEQ ID NO:121), VHH#3E (SEQ ID NO:122), VHH#1A (SEQ ID NO:123), VHH#2B (SEQ ID NO:124), VHH#12B (SEQ ID NO:125), VHH#7B (SEQ ID NO:126).
  • FIG. 2 Dilution series of anti-human TNF-alpha VHHs as tested in ELISA according to Example 1.
  • FIG. 3 Antagonistic effect of VHH as determined in cytotoxicity assay using human cell line KYM according to Example 1.
  • FIG. 4 In vitro receptor binding assay of wild type VHH#12B and mutant A74S+Y76N+K83R+P84A.
  • FIG. 5 In vitro receptor binding assay of wild type VHH#12B and mutant 1E+Q5LA74S+Y76N+K83R+P84A.
  • FIG. 6 Binding in ELISA of wild type VHH#3E and mutant VHH's.
  • FIG. 7 In vitro receptor binding assay of wild type VHH#3E and mutant VHH's.
  • FIG. 8 Alignment of antagonistic anti-mouse TNF's as described in Example 3: VHH#m3F (SEQ ID NO:127), VHH#m4B (SEQ ID NO:128), VHH#m9A (SEQ ID NO:129), VHH#m9E (SEQ ID NO:130).
  • FIG. 9 Antagonistic effect of anti-mouse TNF VHH as determined in cytotoxicity assay using murine cell line L929 according to Example 3.
  • FIG. 10 EcoRI—HindIII insert (SEQ ID NOs:131, 132) of vector pAX11 (pUC119 backbone) for production of bi-valent or bispecific VHH.
  • FIG. 11 Coomassie-stained PAGE (15%) of IMAC-purified mono-(lane 8), bi-(lane 1), tri-( lanes 2, 3 and 5) and tetravalent ( lanes 4, 6 and 7) anti-TNFa VHH.
  • FIG. 12 Chromatogram of the analysis by gel filtration on Superdex 75HR of the mono-, bi-, tri and tetravalent VHH.
  • FIG. 13 Comparison of the antagonistic characteristics of the mono-, bi-, tri- and tetravalent form of the anti-human TNF VHH with the clinically used products Remicade and Enbrel.
  • FIG. 14 Antagonistic behaviour of the mono- and bivalent VHH's directed against mouse TNFalpha.
  • FIG. 15 Coomassie stained PAGE of VHH-Fc-fusion derived from human IgG1 described in Example 4.
  • FIG. 16 Antagonistic efficacy of VHH-Fc fusion derived from VHH#3E compared with bivalent format of VHH#3E as determined in bioassay.
  • FIG. 17 ELISA of reference and pepsin-treated TNF3E at pH2.2, pH3.2 and pH4.2 (100% is the signal measured at a 1/100 dilution).
  • FIG. 18 Experimental setting.
  • Table 1 Amino acid sequence listing of the peptides of aspects of present invention directed against TNF-alpha.
  • Table 2 List of mutagenesis reactions, mutagenic primers and templates used for mutagenesis of VHH#12B: mutation A74S+Y76N+K83R+P84A (SEQ ID NOs: 107, 108); mutation Q1E+Q5L+A74S+Y76N+K83R+P84A (SEQ ID NOs: 109, 110); mutation Q1E+Q5L+A74S+Y76N+K83R+P84A+T93A (SEQ ID NOs:111,112).
  • Table 3 List of mutagenesis reactions, mutagenic primers and templates used for mutagenesis of VHH#3E: mutation F37V (SEQ ID NOs: 113, 114); mutation E44G (SEQ ID NOs: 115, 116); mutation R45L (SEQ ID NOs: 117, 118); mutation F47W (SEQ ID NOs: 119, 120).
  • Table 4 Overview of humanised and wild type VHH.
  • Table 5 Anti-mouse serum albumin/anti TNF-alpha
  • Table 6 Amino acid sequence listing of VHH's directed against human IFN-gamma.
  • Table 7 Sequences of bivalent (BIV 3E, BIV#m3F), trivalent (TRI3E) or tetravalent (TETRA 3E) VHH directed against TNF-alpha.
  • Table 8 Fractional homologies between the amino acid sequences of anti-mouse serum albumin VHHs of the invention.
  • Table 9 Fractional homologies between anti-TNF-alpha VHHs of the invention.
  • Table 10 Percentage homologies between anti-IFN-gamma VHHs of the invention.
  • Table 11 Treatment schedule.
  • DETAILED DESCRIPTION
  • The present invention relates to an anti-tumour necrosis factor-alpha (TNF-alpha) polypeptide, comprising one or more single domain antibodies which are directed against TNF-alpha. The invention also relates to nucleic acids capable of encoding said polypeptides.
  • Single domain antibodies are antibodies whose complementary determining regions are part of a single domain polypeptide. Examples include, but are not limited to, heavy chain antibodies, antibodies naturally devoid of light chains, single domain antibodies derived from conventional 4-chain antibodies, engineered antibodies and single domain scaffolds other than those derived from antibodies. Single domain antibodies may be any of the art, or any future single domain antibodies. Single domain antibodies may be derived from any species including, but not limited to mouse, human, camel, llama, goat, rabbit, bovine. According to one aspect of the invention, a single domain antibodies as used herein is a naturally occurring single domain antibody known as heavy chain antibody devoid of light chains. Such single domain antibodies are disclosed in WO 94/04678 for example. For clarity reasons, this variable domain derived from a heavy chain antibody naturally devoid of light chain is known herein as a VHH or nanobody to distinguish it from the conventional VH of four chain immunoglobulins. Such a VHH molecule can be derived from antibodies raised in Camelidae species, for example in camel, dromedary, llama, alpaca and guanaco. bther species besides Camelidae may produce heavy chain antibodies naturally devoid of light chain; such VHHs are within the scope of the invention.
  • VHHs, according to the present invention, and as known to the skilled addressee are heavy chain variable domains derived from immunoglobulins naturally devoid of light chains such as those derived from Camelidae as described in WO 94/04678 (and referred to hereinafter as VHH domains or nanobodies). VHH molecules are about 10× smaller than IgG molecules. They are single polypeptides and very stable, resisting extreme pH and temperature conditions. Moreover, they are resistant to the action of proteases which is not the case for conventional antibodies. Furthermore, in vitro expression of VHHs produces high yield, properly folded functional VHHs. In addition, antibodies generated in Camelids will recognize epitopes other than those recognised by antibodies generated in vitro through the use of antibody libraries or via immunisation of mammals other than Camelids (WO 9749805). As such, anti-TNF-alpha VHH's may interact more efficiently with TNF-alpha than conventional antibodies, thereby blocking its interaction with the TNF-alpha receptor more efficiently.
  • According to the invention, TNF-alpha is derived from any species. Examples of species relevant to the invention include as rabbits, goats, mice, rats, cows, calves, camels, llamas, monkeys, donkeys, guinea pigs, chickens, sheep, dogs, cats, horses, and preferably humans.
  • TNF-alpha is also a fragment of TNF-alpha, capable of eliciting an immune response. TNF-alpha is also a fragment of TNF-alpha, capable of binding to a single domain antibody raised against the full length TNF-alpha.
  • A single domain antibody directed against TNF-alpha means single domain antibody that it is capable of binding to TNF-alpha with an affinity of better than 10−6 M.
  • One embodiment of the present invention is an anti-TNF polypeptide, wherein the single domain antibodies comprise Camelidae VHH directed against TNF-alpha.
  • The one or more single domain antibodies of the anti-TNF polypeptide which are directed against a TNF-alpha may be of the same sequence. Alternatively they may not all have the same sequence. It is within the scope of the invention that an anti-TNF polypeptide comprises anti-TNF-alpha single domain antibodies which do not all share the same sequence, but which are directed against the same target, one or more antigens thereof.
  • Another embodiment of the present invention is an anti-TNF-alpha polypeptide, wherein a single domain antibody corresponds to a sequence represented by any of SEQ ID NOs: 1 to 16 and 79 to 84 as shown in Table 1. Said sequences are derived from Camelidae heavy chain antibodies (VHHs) which are directed against TNF-alpha.
  • The present invention further relates to an anti-TNF-alpha polypeptide, wherein said single domain antibody is a VHH directed against TNF-alpha, wherein the VHH belongs to a class having human-like sequences. The class is characterised in that the VHHs carry an amino acid from the group consisting of glycine, alanine, valine, leucine, isoleucine, proline, phenylalanine, tyrosine, tryptophan, methionine, serine, threonine, asparagine, or glutamine at position 45, such as, for example, L45 and a tryptophan at position 103, according to the Kabat numbering. The new class of Camelidae single-domain antibodies described in this invention (Table 1, Example 1) is represented by VHH#2B (SEQ ID NO: 3) and VHH#12B (SEQ ID No. 14) containing the hydrophobic residues in FR2 in combination with the hydrophobic residue tryptophan at position 103.
  • Another human-like class of Camelidae single domain antibodies represented by sequences VHH#1A (SEQ ID NO. 1), VHH#4B (SEQ ID NO. 12), VHH#8-29 (SEQ ID NO. 81), VHH#8-41 (SEQ ID NO. 82), VHH#8-42 (SEQ ID NO. 83) and VHH#8-44 (SEQ ID NO. 84) (Table 1, Example 1) have been described in WO03035694 and contain the hydrophobic FR2 residues typically found in conventional antibodies of human origin or from other species, but compensating this loss in hydrophilicity by the charged arginine residue on position 103 that substitutes the conserved tryptophan residue present in VH from double-chain antibodies. As such, peptides belonging to these two classes show a high amino acid sequence homology to human VH framework regions and said peptides might be administered to a human directly without expectation of an unwanted immune response therefrom, and without the burden of further humanisation. The invention also relates to nucleic acids capable of encoding said polypeptides.
  • Therefore, one aspect of the present invention allows for the direct administration of an anti-TNF-alpha polypeptide, wherein the single domain antibodies belong to the humanized class of VHH, and comprise a sequence represented by any of SEQ ID NO:1, 3, 12, 14, 81, 82, 83, and 84 to a patient in need of the same.
  • Any of the VHHs as used by the invention may be of the traditional class or of the classes of human-like Camelidae antibodies. Said antibodies may be directed against whole TNF-alpha or a fragment thereof, or a fragment of a homologous sequence thereof. These polypeptides include the full length Camelidae antibodies, namely Fc and VHH domains, chimeric versions of heavy chain Camelidae antibodies with a human Fc domain or VHH's by themselves or derived fragments.
  • Anti-serum albumin VHH's may interact in a more efficient way with serum albumin than conventional antibodies which is known to be a carrier protein. As a carrier protein some of the epitopes of serum albumin may be inaccessible by bound proteins, peptides and small chemical compounds. Since VHH's are known to bind into ‘unusual’ or non-conventional epitopes such as cavities (WO 97/49805), the affinity of such VHH's to circulating albumin may be increased.
  • The present invention also relates to the finding that an anti-TNF polypeptide as described herein further comprising one or more single domain antibodies directed against one or more serum proteins of a subject, surprisingly has significantly prolonged half-life in the circulation of said subject compared with the half-life of the anti-TNF-alpha single domain antibody when not part of said construct. Examples of such polypeptides are represented in Table 5 by SEQ ID NOs: 30 to 43. Furthermore, the said polypeptides were found to exhibit the same favourable properties of single domain antibodies such as high stability remaining intact in mice, extreme pH resistance, high temperature stability and high target affinity.
  • Another embodiment of the present invention is an anti-TNF-alpha polypeptide further comprising one or more single domain antibodies directed against one or more serum proteins, said anti-TNF alpha polypeptide comprising a sequence corresponding to any represented by SEQ ID NOs: 30 to 43 (Table 5).
  • Another embodiment of the present invention is an anti-TNF-alpha polypeptide, wherein an anti-serum protein single domain antibody corresponds to a sequence represented by any of SEQ ID NOs: 26 to 29 and 85 to 97 as shown in Table 5.
  • The serum protein may be any suitable protein found in the serum of subject. In one aspect of the invention, the serum protein is serum albumin, serum immunoglobulins, thyroxine-binding protein, transferrin, or fibrinogen. Depending on the intended use such as the required half-life for effective treatment and/or compartimentalisation of the target antigen, the VHH-partner can be directed to one of the above serum proteins.
  • Another aspect of the invention is an anti-TNF-alpha polypeptide as disclosed herein further comprising at least one polypeptide selected from the group consisting of an anti-IFN-gamma polypeptide, an anti-TNF-alpha receptor polypeptide and anti-IFN-gamma receptor polypeptide.
  • It is an embodiment of the invention that a single domain antibody directed against IFN-gamma corresponds to a sequence represented by any of SEQ ID NOs: 44 to 72 as shown in Table 6.
  • According to one aspect of the invention, a single domain antibody is directed against TNF-alpha receptor. Said single domain antibody may be a Camelidae VHH.
  • According to one aspect of the invention, a single domain antibody is directed against IFN-gamma receptor. Said single domain antibody may be a Camelidae VHH.
  • Another aspect of the invention is a method of treating an autoimmune disease or condition as cited herein, comprising administering to a patient an effective amount of an anti-TNF-alpha polypeptide further comprising a least one polypeptide selected from the group consisting of anti-IFN-gamma polypeptide, anti-TNF-alpha receptor polypeptide and anti-IFN-gamma receptor polypeptide, such polypeptides joined to each other as described below.
  • Such multi-specific constructs may have improved potency as inflammatory therapeutic compound over mono-specific constructs.
  • One aspect of the invention is a composition comprising an anti-TNF-alpha polypeptide as disclosed herein and at least one polypeptide selected from the group consisting of anti-IFN-gamma polypeptide, anti-TNF-alpha receptor polypeptide and anti-IFN-gamma receptor polypeptide, for simultaneous, separate or sequential administration to a subject.
  • One aspect of the invention is a method for treating autoimmune disease comprising administering to an individual an effective amount of an anti-TNF-alpha polypeptide and a least one polypeptide selected from the group consisting of anti-IFN-gamma polypeptide, anti-TN F-alpha receptor polypeptide and anti-I FN-gamma receptor polypeptide, simultaneously, separately or sequentially.
  • Another aspect of the invention is a kit containing an anti-TNF-alpha polypeptide and a least one polypeptide selected from the group consisting of anti-IFN-gamma polypeptide, anti-TNF-alpha receptor polypeptide and anti-IFN-gamma receptor polypeptide for simultaneous, separate or sequential administration to a subject. It is an aspect of the invention that the kit may be used according to the invention. It is an aspect of the invention that the kit may be used to treat the diseases as cited herein.
  • By simultaneous administration means the polypeptides are administered to a subject at the same time. For example, as a mixture of the polypeptides or a composition comprising said polypeptides. Examples include, but are not limited to a solution administered intraveneously, a tablet, liquid, topical cream, etc., wherein each preparation comprises the polypeptides of interest.
  • By separate administration means the polypeptides are administered to a subject at the same time or substantially the same time. The polypeptides are present in the kit as separate, unmixed preparations. For example, the different polypeptides may be present in the kit as individual tablets. The tablets may be administered to the subject by swallowing both tablets at the same time, or one tablet directly following the other.
  • By sequential administration means the polypeptides are administered to a subject sequentially. The polypeptides are present in the kit as separate, unmixed preparations. There is a time interval between doses. For example, one polypeptide might be administered up to 336, 312, 288, 264, 240, 216, 192, 168, 144, 120, 96, 72, 48, 24, 20, 16, 12, 8, 4, 2, 1, or 0.5 hours after the other component.
  • In sequential administration, one polypeptide may be administered once, or any number of times and in various doses before and/or after administration of another polypeptide. Sequential administration may be combined with simultaneous or sequential administration.
  • The medical uses of the anti-TNF-alpha polypeptide described below, also apply to the composition comprising an anti-TNF-alpha polypeptide as disclosed herein and at least one polypeptide selected from the group consisting of anti-IFN-gamma polypeptide, anti-TNF-alpha receptor polypeptide and anti-IFN-gamma receptor polypeptide, for simultaneous, separate or sequential administration to a subject as disclosed here above.
  • According to one aspect of the invention, an anti-IFN-gamma polypeptide anti-TNF-alpha a single domain antibody directed against IFN-gamma. Said single domain antibody may be a Camelidae VHH.
  • It is an embodiment of the invention that a single domain antibody directed against IFN-gamma corresponds to a sequence represented by any of SEQ ID NOs: 44 to 72 as shown in Table 6.
  • According to one aspect of the invention, anti-TNF-alpha a single domain antibody directed against TNF-alpha receptor. Said single domain antibody may be a Camelidae VHH.
  • According to one aspect of the invention, an anti-IFN-gamma receptor polypeptide anti-TNF-alpha a single domain antibody directed against IFN-gamma receptor. Said single domain antibody may be a Camelidae VHH.
  • Another embodiment of the present invention is an anti-TNF-alpha polypeptide as disclosed herein, wherein the number of single domain antibodies directed against TNF-alpha is two or more. Such multivalent anti-TNF-alpha polypeptides have the advantage of unusually high functional affinity for the target, displaying much higher than expected inhibitory properties compared to their monovalent counterparts.
  • The multivalent anti-TNF-alpha polypeptides have functional affinities that are several orders of magnitude higher than the monovalent parent anti-TNF-alpha polypeptides. The inventors have found that the functional affinities of these multivalent polypeptides are much higher than those reported in the prior art for bivalent and multivalent antibodies. Surprisingly, anti-TNF-alpha polypeptides of the present invention linked to each other directly (SEQ ID No. 77 and 78) or via a short linker sequence show the high functional affinities expected theoretically with multivalent conventional four-chain antibodies.
  • The inventors have found that such large increased functional activities can be detected preferably with antigens composed of multidomain and multimeric proteins, either in straight binding assays or in functional assays, e.g. cytotoxicity assays.
  • Another embodiment of the present invention is an anti-TNF-alpha polypeptide as disclosed herein, wherein the number of single domain antibodies directed against TNF-alpha is two or more, said anti-TNF-alpha polypeptide comprising a sequence corresponding to any represented by SEQ ID NOs: 73 to 76.
  • The single domain antibodies may be joined to form any of the polypeptides disclosed herein comprising more than one single domain antibody using methods known in the art or any future method. For example, they may be fused by chemical cross-linking by reacting amino acid residues with an organic derivatising agent such as described by Blattler et al, Biochemistry 24,1517-1524; EP294703. Alternatively, the single domain antibody may be fused genetically at the DNA level i.e. a polynucleotide construct formed which encodes the complete polypeptide construct comprising one or more anti-target single domain antibodies and one or more anti-serum protein single domain antibodies. A method for producing bivalent or multivalent VHH polypeptide constructs is disclosed in PCT patent application WO 96/34103. One way of joining multiple single domain antibodies is via the genetic route by linking single domain antibody coding sequences either directly or via a peptide linker. For example, the C-terminal end of the first single domain antibody may be linked to the N-terminal end of the next single domain antibody. This linking mode can be extended in order to link additional single domain antibodies for the construction and production of tri-, tetra-, etc. functional constructs.
  • According to one aspect of the present invention, the single domain antibodies are linked to each other directly, without use of a linker. Contrary to joining bulky conventional antibodies where a linker sequence is needed to retain binding activity in the two subunits, polypeptides of the invention can be linked directly (SEQ ID No. 77 and 78) thereby avoiding potential problems of the linker sequence, such as antigenicity when administered to a human subject, instability of the linker sequence leading to dissociation of the subunits.
  • According to another aspect of the present invention, the single domain antibodies are linked to each other via a peptide linker sequence. Such linker sequence may be a naturally occurring sequence or a non-naturally occurring sequence. The linker sequence is expected to be non-immunogenic in the subject to which the anti-TNF-alpha polypeptide is administered. The linker sequence may provide sufficient flexibility to the multivalent anti-TNF-alpha polypeptide, at the same time being resistant to proteolytic degradation. A non-limiting example of a linker sequences is one that can be derived from the hinge region of VHHs described in WO 96/34103.
  • According to another aspect of the invention, multivalent single domain antibodies comprising more than two single domain antibodies can be linked to each other either directly or via a linker sequence. Such constructs are difficult to produce with conventional antibodies and due to steric hindrance of the bulky subunits, functionality will be lost or greatly diminished rather than increased considerably as seen with VHH's of the invention compared to the monovalent construct (see FIG. 12 for gel filtration analyses of such multivalent VHH constructs).
  • The polypeptide constructs disclosed herein may be made by the skilled artisan according to methods known in the art or any future method. For example, VHHs may be obtained using methods known in the art such as by immunising a camel and obtaining hybridomas therefrom, or by cloning a library of single domain antibodies using molecular biology techniques known in the art and subsequent selection by using phage display.
  • According to an aspect of the invention an anti-TNF-alpha polypeptide may be a homologous sequence of a full-length anti-TNF-alpha polypeptide. According to another aspect of the invention, an anti-TNF-alpha polypeptide may be a functional portion of a full-length anti-TNF-alpha polypeptide. According to another aspect of the invention, an anti-TNF-alpha polypeptide may be a homologous sequence of a full-length anti-TNF-alpha polypeptide. According to another aspect of the invention, an anti-TNF-alpha polypeptide may be a functional portion of a homologous sequence of a full-length anti-TNF-alpha polypeptide. According to an aspect of the invention an anti-TNF-alpha polypeptide may comprise a sequence of an anti-TNF-alpha polypeptide.
  • According to an aspect of the invention a single domain antibody used to form an anti-TNF-alpha polypeptide may be a complete single domain antibody (e.g. a VHH) or a homologous sequence thereof. According to another aspect of the invention, a single domain antibody used to form the polypeptide construct may be a functional portion of a complete single domain antibody. According to another aspect of the invention, a single domain antibody used to form the polypeptide construct may be a homologous sequence of a complete single domain antibody. According to another aspect of the invention, a single domain antibody used to form the polypeptide construct may be a functional portion of a homologous sequence of a complete single domain antibody.
  • As used herein, an homologous sequence of the present invention may comprise additions, deletions or substitutions of one or more amino acids, which do not substantially alter the functional characteristics of the polypeptides of the invention. The number of amino acid deletions or substitutions is preferably up to 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69 or 70 amino acids.
  • A homologous sequence according to the present invention may a polypeptide modified by the addition, deletion or substitution of amino acids, said modification not substantially altering the functional characteristics compared with the unmodified polypeptide.
  • A homologous sequence according to the present invention may be a polypeptide modified by the addition, deletion or substitution of amino acids, said modification not substantially altering the functional characteristics compared with the unmodified polypeptide.
  • A homologous sequence according to the present invention may be a sequence which exists in other Camelidae species such as, for example, camel, dromedary, llama, alpaca, guanaco etc.
  • Where homologous sequence indicates sequence identity, it means a sequence which presents a high sequence identity (more than 70%, 75%, 80%, 85%, 90%, 95% or 98% sequence identity) with the parent sequence and is preferably characterised by similar properties of the parent sequence, namely affinity, said identity calculated using known methods.
  • Alternatively, an homologous sequence may also be any amino acid sequence resulting from allowed substitutions at any number of positions of the parent sequence according to the formula below:
  • Ser substituted by Ser, Thr, Gly, and Asn;
  • Arg substituted by one of Arg, His, Gin, Lys, and Glu;
  • Leu substituted by one of Leu, Ile, Phe, Tyr, Met, and Val;
  • Pro substituted by one of Pro, Gly, Ala, and Thr;
  • Thr substituted by one of Thr, Pro, Ser, Ala, Gly, His, and Gln;
  • Ala substituted by one of Ala, Gly, Thr, and Pro;
  • Val substituted by one of Val, Met, Tyr, Phe, Ile, and Leu;
  • Gly substituted by one of Gly, Ala, Thr, Pro, and Ser;
  • Ile substituted by one of Ile, Met, Tyr, Phe, Val, and Leu;
  • Phe substituted by one of Phe, Trp, Met, Tyr, Ile, Val, and Leu;
  • Tyr substituted by one of Tyr, Trp, Met, Phe, Ile, Val, and Leu;
  • His substituted by one of His, Glu, Lys, Gln, Thr, and Arg;
  • Gln substituted by one of Gin, Glu, Lys, Asn, His, Thr, and Arg;
  • Asn substituted by one of Asn, Glu, Asp, Gln, and Ser;
  • Lys substituted by one of Lys, Glu, Gln, His, and Arg;
  • Asp substituted by one of Asp, Glu, and Asn;
  • Glu substituted by one of Glu, Asp, Lys, Asn, Gln, His, and Arg;
  • Met substituted by one of Met, Phe, Ile, Val, Leu, and Tyr.
  • A homologous nucleotide sequence according to the present invention may refer to nucleotide sequences of more than 50, 100, 200, 300, 400, 500, 600, 800 or 1000 nucleotides able to hybridize to the reverse-complement of the nucleotide sequence capable of encoding the patent sequence, under stringent hybridisation conditions (such as the ones described by Sambrook et al., Molecular Cloning, Laboratory Manuel, Cold Spring, Harbor Laboratory press, New York).
  • As used herein, a functional portion refers to a sequence of a single domain antibody that is of sufficient size such that the interaction of interest is maintained with affinity of 1×10−6 M or better.
  • Alternatively, a functional portion comprises a partial deletion of the complete amino acid sequence and still maintains the binding site(s) and protein domain(s) necessary for the binding of and interaction with the target.
  • As used herein, a functional portion refers to less than 100% of the complete sequence (e.g., 99%, 90%, 80%, 70%, 60% 50%, 40%, 30%, 20%, 10%, 5%, 1% etc.), but comprises 5 or more amino acids or 15 or more nucleotides.
  • Targets as mentioned herein such as TNF-alpha, TNF-alpha receptor, serum proteins (e.g. serum albumin, serum immunoglobulins, thyroxine-binding protein, transferrin, fibrinogen) and IFN-gamma, IFN-gamma receptor may be fragments of said targets. Thus a target is also a fragment of said target, capable of eliciting an immune response. A target is also a fragment of said target, capable of binding to a single domain antibody raised against the full length target.
  • A fragment as used herein refers to less than 100% of the sequence (e.g., 99%, 90%, 80%, 70%, 60%, 50%, 40%, 30%, 20%, 10% etc.), but comprising 5, 6, 7, 8, 9, 10, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25 or more amino acids. A fragment is of sufficient length such that the interaction of interest is maintained with affinity of 1×10−6 M or better.
  • A fragment as used herein also refers to optional insertions, deletions and substitutions of one or more amino acids which do not substantially alter the ability of the target to bind to a single domain antibody raised against the wild-type target. The number of amino acid insertions deletions or substitutions is preferably up to 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69 or 70 amino acids.
  • A homologous sequence of the present invention may include an anti-TNF-alpha polypeptide which has been humanised. The humanisation of antibodies of the new class of VHHs would further reduce the possibility of unwanted immunological reaction in a human individual upon administration.
  • One embodiment of the present invention relates to a method for preparing modified polypeptides based upon llama antibodies by determining the amino acid residues of the antibody variable domain (VHH) which may be modified without diminishing the native affinity of the domain for antigen and while reducing its immunogenicity with respect to a heterologous species; the use of VHHs having modifications at the identified residues which are useful for administration to heterologous species; and to the VHH so modified.
  • More specifically, the invention relates to the preparation of modified VHHs, which are modified for administration to humans, the resulting VHH themselves, and the use of such “humanized” VHHs in the treatment of diseases in humans. By humanised is meant mutated so that immunogenicity upon administration in human patients is minor or nonexistent. Humanising a polypeptide, according to the present invention, comprises a step of replacing one or more of the Camelidae amino acids by their human counterpart as found in the human consensus sequence, without that polypeptide losing its typical character, i.e. the humanisation does not significantly affect the antigen binding capacity of the resulting polypeptide. Such methods are known by the skilled addressee.
  • Humanization of Camelidae single domain antibodies requires the introduction and mutagenesis of a limited amount of amino acids in a single polypeptide chain. This is in contrast to humanization of scFv, Fab, (Fab)2 and IgG, which requires the introduction of amino acid changes in two chains, the light and the heavy chain and the preservation of the assembly of both chains.
  • As a non-limited example, the polypeptide of VHH#12B containing human-like residues in FR2 was humanized. Humanization required mutagenesis of residues in FR1 at position 1 and 5 which were introduced by the primer used for repertoire cloning and do not occur naturally in the llama sequence. Mutagenesis of those residues did not result in loss of binding and/or inhibition activity. Humanization also required mutagenesis of residues in FR3 at position 74, 76, 83, 84, 93. Mutagenesis of those residues did not result in a dramatic loss of binding and/or inhibition activity (see FIG. 4). Combining the mutations of FR1 and FR3 therefore did not affect the binding and/or inhibition activity (FIG. 5).
  • Humanization also required mutagenesis of residues in FR4 at position 108. Mutagenesis of Q108L resulted in lower production level in Escherichia coli. Position 108 is solvent exposed in camelid VHH, while in human antibodies this position is buried at the VH-VL interface (Spinelli, 1996; Nieba, 1997). In isolated VHs position 108 is solvent exposed. The introduction of a non-polar hydrophobic Leu instead of polar uncharged Gln can have a drastic effect on the intrinsic folding/stability of the molecule.
  • As a non-limited example, the polypeptide represented in the VHH#3E containing camelid hallmark residues at position 37, 44, 45 and 47 with hydrophilic characteristics, was humanized. Replacement of the hydrophilic residues by human hydrophobic residues at positions 44 and 45 (E44G and R45L), did not have an effect on binding and/or inhibition. However, loss of binding and/or inhibition activity was observed when F37V and F47W were introduced. Modeling data confirmed the critical residue 37 to preserve the integrity of the CDR3 loop conformation and hence on activity (see FIG. 6)(all numbering according to the Kabat).
  • SEQ ID NO: 3 and 14 display more than 90% amino acid sequence homology to human VH framework regions and therefore said VHH might be administered to patients directly without expectation of an immune response therefrom, and without the additional burden of humanisation. Therefore, one aspect of the present invention allows for the direct administration of the polypeptide comprising SEQ ID NO: 3 and 14, homologous sequence thereof, or a functional portion of an homologous sequence thereof to a patient in need of the same.
  • One embodiment of the present invention is a method for humanizing a VHH comprising the steps of replacing of any of the following residues either alone or in combination:
      • FR1 position 1, 5, 28 and 30,
      • the hallmark amino acid at position 44 and 45 in FR2,
      • FR3 residues 74, 75, 76, 83, 84, 93 and 94,
      • and positions 103, 104, 108 and 111 in FR4;
        numbering according to the Kabat numbering.
  • One embodiment of the present invention is an anti-TNF-alpha polypeptide, or a nucleic acid capable of encoding said polypeptide for use in treating, preventing and/or alleviating the symptoms of disorders relating to inflammatory processes. TNF-alpha is involved in inflammatory processes, and the blocking of TNF-alpha action can have an anti-inflammatory effect, which is highly desirable in certain disease states such as, for example, Crohn's disease. Our Examples demonstrate VHHs according to the invention which bind TNF-alpha and moreover, block its binding to the TNF-alpha receptor.
  • The anti-TNF-alpha polypeptides of the present invention are applicable to autoimmune diseases, such as Addison's disease (adrenal), Autoimmune diseases of the ear (ear), Autoimmune diseases of the eye (eye), Autoimmune hepatitis (liver), Autoimmune parotitis (parotid glands), Crohn's disease (intestine), Diabetes Type I (pancreas), Epididymitis (epididymis), Glomerulonephritis (kidneys), Graves' disease (thyroid), Guillain-Barre syndrome (nerve cells), Hashimoto's disease (thyroid), Hemolytic anemia (red blood cells), Systemic lupus erythematosus (multiple tissues), Male infertility (sperm), Multiple sclerosis (nerve cells), Myasthenia Gravis (neuromuscular junction), Pemphigus (primarily skin), Psoriasis (skin), Rheumatic fever (heart and joints), Rheumatoid arthritis (joint lining), Sarcoidosis (multiple tissues and organs), Scleroderma (skin and connective tissues), Sjogren's syndrome (exocrine glands, and other tissues), Spondyloarthropathies (axial skeleton, and other tissues), Thyroiditis (thyroid), Vasculitis (blood vessels). Within parenthesis is the tissue affected by the disease. This listing of autoimmune diseases is intended to be exemplary rather than inclusive.
  • Autoimmune conditions for which the anti-TNF-alpha polypeptides of the present invention is applicable include, for example, AIDS, atopic allergy, bronchial asthma, eczema, leprosy, schizophrenia, inherited depression, transplantation of tissues and organs, chronic fatigue syndrome, Alzheimer's disease, Parkinson's disease, myocardial infarction, stroke, autism, epilepsy, Arthus's phenomenon, anaphylaxis, and alcohol and drug addiction. In the above-identified autoimmune conditions, the tissue affected is the primary target, in other cases it is the secondary target. These conditions are partly or mostly autoimmune syndromes.
  • Therefore, in treating them, it is possible to use the same methods, or aspects of the same methods that are herein disclosed, sometimes in combination with other methods.
  • Another embodiment of the present invention is a use of an anti-TNF-alpha polypeptide according to the invention, or a nucleic acid capable of encoding said polypeptide for the preparation of a medicament for treating a disorder relating to inflammatory processes. Examples of disorders include rheumatoid arthritis, Crohn's disease, ulcerative colitis, inflammatory bowel syndrome and multiple sclerosis
  • Polypeptides and nucleic acids according to the present invention may be administered to a subject by conventional routes, such as intravenously. However, a special property of the anti-TNF-alpha polypeptides of the invention is that they penetrate barriers such as tissue membranes and/or tumours and act locally and act locally thereon, and they are sufficiently stable to withstand extreme environments such as in the stomach. Therefore, another aspect of the present invention relates to the delivery of anti-TNF-alpha polypeptides.
  • A subject according to the invention can be any mammal susceptible to treatment by therapeutic polypeptides.
  • Oral delivery of anti-TNF-alpha polypeptides of the invention results in the provision of such molecules in an active form in the colon at local sites that are affected by the disorder. These sites may be highly inflamed and contain TNF-alpha-producing cells. The anti-TNF-alpha polypeptides of the invention which bind to TNF-alpha can neutralise the TNF-alpha locally, avoiding distribution throughout the whole body and thus limiting negative side-effects. Genetically modified microorganisms such as Micrococcus lactis are able to secrete antibody or functional portions thereof. Such modified microorganisms can be used as vehicles for local production and delivery of antibodies or functional portions thereof in the intestine. By using a strain which produces an anti-TNF-alpha polypeptide, inflammatory bowel syndrome could be treated.
  • Another aspect of the invention involves delivering anti-TNF polypeptides by using surface expression on or secretion from non-invasive bacteria, such as Gram-positive host organisms like Lactococcus spec. using a vector such as described in WO00/23471.
  • One embodiment of the present invention is an anti-TNF-alpha polypeptide as disclosed herein for use in treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by a TNF-alpha modulating substance which is able pass through the gastric environment without the substance being inactivated.
  • Examples of disorders are any that cause inflammation, including, but not limited to rheumatoid arthritis, Crohn's disease, ulcerative colitis, inflammatory bowl syndrome, and multiple sclerosis. As known by persons skilled in the art, once in possession of said polypeptide construct, formulation technology may be applied to release a maximum amount of polypeptide in the right location (in the stomach, in the colon, etc.). This method of delivery is important for treating, prevent and/or alleviate the symptoms of disorders whose targets are located in the gut system.
  • An aspect of the invention is a method for treating, preventing and/or alleviating the symptoms of a disorder susceptible to modulation by a TNF-alpha modulating substance which is able pass through the gastric environment without being inactivated, by orally administering to a subject an anti-TNF-alpha polypeptide as disclosed herein.
  • Another embodiment of the present invention is a use of an anti-TNF-alpha polypeptide as disclosed herein for the preparation of a medicament for treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by a TNF-alpha modulating substance which is able pass through the gastric environment without being inactivated.
  • An aspect of the invention is a method for delivering a TNF-alpha modulating substance to the gut system without said substance being inactivated, by orally administering to a subject an anti-TNF-alpha polypeptide as disclosed herein.
  • An aspect of the invention is a method for delivering a TNF-alpha modulating substance to the bloodstream of a subject without the substance being inactivated, by orally administering to a subject an anti-TNF-alpha polypeptide as disclosed herein.
  • Another embodiment of the present invention is an anti-TNF-alpha polypeptide as disclosed herein for use in treating, preventing and/or alleviating the symptoms or disorders susceptible to modulation by a TNF-alpha modulating substance delivered to the vaginal and/or rectal tract.
  • Examples of disorders are any that cause inflammation, including, but not limited to rheumatoid arthritis, Crohn's disease, ulcerative colitis, inflammatory bowl syndrome, and multiple sclerosis. In a non-limiting example, a formulation according to the invention comprises an anti-TNF-alpha polypeptide as disclosed herein, in the form of a gel, cream, suppository, film, or in the form of a sponge or as a vaginal ring that slowly releases the active ingredient over time (such formulations are described in EP 707473, EP 684814, U.S. Pat. No. 5,629,001).
  • An aspect of the invention is a method for treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by a TNF-alpha modulating substance delivered to the vaginal and/or rectal tract, by vaginally and/or rectally administering to a subject an anti-TNF-alpha polypeptide as disclosed herein.
  • Another embodiment of the present invention is a use of an anti-TNF-alpha polypeptide as disclosed herein for the preparation of a medicament for treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by a TNF-alpha modulating substance delivered to the vaginal and/or rectal tract.
  • An aspect of the invention is a method for delivering a TNF-alpha modulating substance to the vaginal and/or rectal tract without being said substance being inactivated, by administering to the vaginal and/or rectal tract of a subject an anti-TNF-alpha polypeptide as disclosed herein.
  • An aspect of the invention is a method for delivering a TNF-alpha modulating substance to the bloodstream of a subject without said substance being inactivated, by administering to the vaginal and/or rectal tract of a subject an anti-TNF-alpha polypeptide as disclosed herein.
  • Another embodiment of the present invention is an anti-TNF-alpha polypeptide as disclosed herein, for use in treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by a TNF-alpha modulating substance delivered to the nose, upper respiratory tract and/or lung.
  • Examples of disorders are any that cause inflammation, including, but not limited to rheumatoid arthritis, Crohn's disease, ulcerative colitis, inflammatory bowl syndrome, and multiple sclerosis. In a non-limiting example, a formulation according to the invention, comprises an anti-TNF-alpha polypeptide as disclosed herein in the form of a nasal spray (e.g. an aerosol) or inhaler. Since the polypeptide construct is small, it can reach its target much more effectively than therapeutic IgG molecules.
  • An aspect of the invention is a method for treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by a TNF-alpha modulating substance delivered to the upper respiratory tract and lung, by administering to a subject an anti-TNF-alpha polypeptide as disclosed herein, by inhalation through the mouth or nose.
  • Another embodiment of the present invention is a use of an anti-TNF-alpha polypeptide as disclosed herein for the preparation of a medicament for treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by a TNF-alpha modulating substance delivered to the nose, upper respiratory tract and/or lung, without said polypeptide being inactivated.
  • An aspect of the invention is a method for delivering a TNF-alpha modulating substance to the nose, upper respiratory tract and lung without inactivation, by administering to the nose, upper respiratory tract and/or lung of a subject an anti-TNF-alpha polypeptide as disclosed herein.
  • An aspect of the invention is a method for delivering a TNF-alpha modulating substance to the bloodstream of a subject without inactivation by administering to the nose, upper respiratory tract and/or lung of a subject an anti-TNF-alpha polypeptide as disclosed herein.
  • One embodiment of the present invention is an anti-TNF-alpha polypeptide as disclosed herein for use in treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by a TNF-alpha modulating substance delivered to the intestinal mucosa, wherein said disorder increases the permeability of the intestinal mucosa. Because of their small size, an anti-TNF-alpha polypeptide as disclosed herein can pass through the intestinal mucosa and reach the bloodstream more efficiently in subjects suffering from disorders which cause an increase in the permeability of the intestinal mucosa, for example Crohn's disease.
  • An aspect of the invention is a method for treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by a TNF-alpha modulating substance delivered to the intestinal mucosa, wherein said disorder increases the permeability of the intestinal mucosa, by orally administering to a subject an anti-TNF-alpha polypeptide as disclosed herein.
  • This process can be even further enhanced by an additional aspect of the present invention—the use of active transport carriers. In this aspect of the invention, VHH is fused to a carrier that enhances the transfer through the intestinal wall into the bloodstream. In a non-limiting example, this “carrier” is a second VHH which is fused to the therapeutic VHH. Such fusion constructs are made using methods known in the art. The “carrier” VHH binds specifically to a receptor on the intestinal wall which induces an active transfer through the wall.
  • Another embodiment of the present invention is a use of an anti-TNF-alpha polypeptide as disclosed herein for the preparation of a medicament for treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by a TNF-alpha modulating substance delivered to the intestinal mucosa, wherein said disorder increases the permeability of the intestinal mucosa.
  • An aspect of the invention is a method for delivering a TNF-alpha modulating substance to the intestinal mucosa without being inactivated, by administering orally to a subject an anti-TNF-alpha polypeptide of the invention.
  • An aspect of the invention is a method for delivering a TNF-alpha modulating substance to the bloodstream of a subject without being inactivated, by administering orally to a subject an anti-TNF-alpha polypeptide of the invention.
  • This process can be even further enhanced by an additional aspect of the present invention—the use of active transport carriers. In this aspect of the invention, an anti-TNF-alpha polypeptide as described herein is fused to a carrier that enhances the transfer through the intestinal wall into the bloodstream. In a non-limiting example, this “carrier” is a VHH which is fused to said polypeptide. Such fusion constructs made using methods known in the art. The “carrier” VHH binds specifically to a receptor on the intestinal wall which induces an active transfer through the wall.
  • One embodiment of the present invention is an anti-TNF-alpha polypeptide as disclosed herein for use in treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by a TNF-alpha modulating substance which is able pass through the tissues beneath the tongue effectively.
  • Examples of disorders are any that cause inflammation, including, but not limited to rheumatoid arthritis, Crohn's disease, ulcerative colitis, inflammatory bowl syndrome, and multiple sclerosis. A formulation of said polypeptide construct as disclosed herein, for example, a tablet, spray, drop is placed under the tongue and adsorbed through the mucus membranes into the capillary network under the tongue.
  • An aspect of the invention is a method for treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by a TNF-alpha modulating substance which is able pass through the tissues beneath the tongue effectively, by sublingually administering to a subject an anti-TNF-alpha polypeptide as disclosed herein.
  • Another embodiment of the present invention is a use of an anti-TNF-alpha polypeptide as disclosed herein for the preparation of a medicament for treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by a TNF-alpha modulating substance which is able to pass through the tissues beneath the tongue.
  • An aspect of the invention is a method for delivering a TNF-alpha modulating substance to the tissues beneath the tongue without being inactivated, by administering sublingually to a subject an anti-TNF-alpha polypeptide as disclosed herein.
  • An aspect of the invention is a method for delivering a TNF-alpha modulating substance to the bloodstream of a subject without being inactivated, by administering orally to a subject an anti-TNF-alpha polypeptide as disclosed herein.
  • One embodiment of the present invention is an anti-TNF-alpha polypeptide as disclosed herein for use in treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by a TNF-alpha modulating substance which is able pass through the skin effectively.
  • Examples of disorders are any that cause inflammation, including, but not limited to rheumatoid arthritis, Crohn's disease, ulcerative colitis, inflammatory bowl syndrome, and multiple sclerosis. A formulation of said polypeptide construct, for example, a cream, film, spray, drop, patch, is placed on the skin and passes through.
  • An aspect of the invention is a method for treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by a TNF-alpha modulating substance which is able pass through the skin effectively, by topically administering to a subject an anti-TNF-alpha polypeptide as disclosed herein.
  • Another embodiment of the present invention is a use of an anti-TNF-alpha polypeptide as disclosed herein for the preparation of a medicament for treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by a TNF-alpha modulating substance which is able pass through the skin effectively.
  • An aspect of the invention is a method for delivering a TNF-alpha modulating substance to the skin without being inactivated, by administering topically to a subject an anti-TNF-alpha polypeptide as disclosed herein.
  • An aspect of the invention is a method for delivering a TNF-alpha modulating substance to the bloodstream of a subject, by administering topically to a subject an anti-TNF-alpha polypeptide as disclosed herein.
  • In another embodiment of the present invention, an anti-TNF-alpha polypeptide further comprises a carrier single domain antibody (e.g. VHH) which acts as an active transport carrier for transport said anti-TNF-alpha polypeptide, from the lung lumen to the blood.
  • An anti-TNF-alpha polypeptide further comprising a carrier binds specifically to a receptor present on the mucosal surface (bronchial epithelial cells) resulting in the active transport of the polypeptide from the lung lumen to the blood. The carrier single domain antibody may be fused to the polypeptide construct. Such fusion constructs may be made using methods known in the art and are describe herein. The “carrier” single domain antibody binds specifically to a receptor on the mucosal surface which induces an active transfer through the surface.
  • Another aspect of the present invention is a method to determine which single domain antibodies (e.g. VHHs) are actively transported into the bloodstream upon nasal administration. Similarly, a naive or immune VHH phage library can be administered nasally, and after different time points after administration, blood or organs can be isolated to rescue phages that have been actively transported to the bloodstream. A non-limiting example of a receptor for active transport from the lung lumen to the bloodstream is the Fc receptor N (FcRn). One aspect of the invention includes the VHH molecules identified by the method. Such VHH can then be used as a carrier VHH for the delivery of a therapeutic VHH to the corresponding target in the bloodstream upon nasal administration.
  • In one aspect of the invention, one can use an anti-TNF-alpha polypeptide as disclosed herein, in order to screen for agents that modulate the binding of the polypeptide to TNF-alpha. When identified in an assay that measures binding or said polypeptide displacement alone, agents will have to be subjected to functional testing to determine whether they would modulate the action of the antigen in vivo. Examples of screening assays are given below primarily in respect of SEQ ID NO: 3, though any anti-TNF-alpha polypeptide as disclosed herein as disclosed herein may be appropriate.
  • In an example of a displacement experiment, phage or cells expressing TNF-alpha or a fragment thereof are incubated in binding buffer with, for example, a polypeptide represented by SEQ ID NO: 3 which has been labeled, in the presence or absence of increasing concentrations of a candidate modulator. To validate and calibrate the assay, control competition reactions using increasing concentrations of said polypeptide and which is unlabeled, can be performed. After incubation, cells are washed extensively, and bound, labeled polypeptide is measured as appropriate for the given label (e.g., scintillation counting, fluorescence, etc.). A decrease of at least 10% in the amount of labeled polypeptide bound in the presence of candidate modulator indicates displacement of binding by the candidate modulator. Candidate modulators are considered to bind specifically in this or other assays described herein if they displace 50% of labeled polypeptide (sub-saturating polypeptide dose) at a concentration of 1 μM or less.
  • Alternatively, binding or displacement of binding can be monitored by surface plasmon resonance (SPR). Surface plasmon resonance assays can be used as a quantitative method to measure binding between two molecules by the change in mass near an immobilized sensor caused by the binding or loss of binding of, for example, the polypeptide represented by SEQ ID NO: 3 from the aqueous phase to TNF-alpha immobilized in a membrane on the sensor. This change in mass is measured as resonance units versus time after injection or removal of the said polypeptide or candidate modulator and is measured using a Biacore Biosensor (Biacore AB). TNF-alpha can be for example immobilized on a sensor chip (for example, research grade CM5 chip; Biacore AB) in a thin film lipid membrane according to methods described by Salamon et al. (Salamon et al., 1996, Biophys J. 71: 283-294; Salamon et al., 2001, Biophys. J. 80: 1557-1567; Salamon et al., 1999, Trends Biochem. Sci. 24: 213-219, each of which is incorporated herein by reference.). Sarrio et al. demonstrated that SPR can be used to detect ligand binding to the GPCR A(1) adenosine receptor immobilized in a lipid layer on the chip (Sarrio et al., 2000, Mol. Cell. Biol. 20: 5164-5174, incorporated herein by reference). Conditions for the binding of SEQ ID NO:3 to TNF-alpha in an SPR assay can be fine-tuned by one of skill in the art using the conditions reported by Sarrio et al. as a starting point.
  • SPR can assay for modulators of binding in at least two ways. First, a polypeptide represented by SEQ ID NO: 3, for example, can be pre-bound to immobilized TNF-alpha followed by injection of candidate modulator at a concentration ranging from 0.1 nM to 1 μM. Displacement of the bound polypeptide can be quantitated, permitting detection of modulator binding. Alternatively, the membrane-bound TNF-alpha can be pre-incubated with a candidate modulator and challenged with, for example, a polypeptide represented by SEQ ID NO: 3. A difference in binding affinity between said polypeptide and TNF-alpha pre-incubated with the modulator, compared with that between said polypeptide and TNF-alpha in absence of the modulator will demonstrate binding or displacement of said polypeptide in the presence of modulator. In either assay, a decrease of 10% or more in the amount of said polypeptide bound in the presence of candidate modulator, relative to the amount of said polypeptide bound in the absence of candidate modulator indicates that the candidate modulator inhibits the interaction of TNF-alpha and said polypeptide.
  • Another method of detecting inhibition of binding of, for example, a polypeptide represented by SEQ ID NO: 3, to TNF-alpha uses fluorescence resonance energy transfer (FRET). FRET is a quantum mechanical phenomenon that occurs between a fluorescence donor (D) and a fluorescence acceptor (A) in close proximity to each other (usually <100 Å of separation) if the emission spectrum of D overlaps with the excitation spectrum of A. The molecules to be tested, e.g. a polypeptide represented by SEQ ID NO: 3 and a TNF-alpha are labelled with a complementary pair of donor and acceptor fluorophores. While bound closely together by the TNF-alpha: polypeptide interaction, the fluorescence emitted upon excitation of the donor fluorophore will have a different wavelength from that emitted in response to that excitation wavelength when the said polypeptide and TNF-alpha are not bound, providing for quantitation of bound versus unbound molecules by measurement of emission intensity at each wavelength. Donor fluorophores with which to label the TNF-alpha are well known in the art. Of particular interest are variants of the A. Victoria GFP known as Cyan FP (CFP, Donor (D)) and Yellow FP (YFP, Acceptor (A)). As an example, the YFP variant can be made as a fusion protein with TNF-alpha. Vectors for the expression of GFP variants as fusions (Clontech) as well as fluorophore-labeled reagents (Molecular Probes) are known in the art. The addition of a candidate modulator to the mixture of fluorescently-labelled polypeptide and YFP-TNF-alpha will result in an inhibition of energy transfer evidenced by, for example, a decrease in YFP fluorescence relative to a sample without the candidate modulator. In an assay using FRET for the detection of TNF-alpha: polypeptide interaction, a 10% or greater decrease in the intensity of fluorescent emission at the acceptor wavelength in samples containing a candidate modulator, relative to samples without the candidate modulator, indicates that the candidate modulator inhibits the TNF-alpha:polypeptide interaction.
  • A sample as used herein may be any biological sample containing TNF-alpha such as clinical (e.g. cell fractions, whole blood, plasma, serum, tissue, cells, etc.), derived from clinical, agricultural, forensic, research, or other possible samples. The clinical samples may be from human or animal origin. The sample analysed can be both solid or liquid in nature. It is evident when solid materials are used, these are first dissolved in a suitable solution.
  • A variation on FRET uses fluorescence quenching to monitor molecular interactions. One molecule in the interacting pair can be labelled with a fluorophore, and the other with a molecule that quenches the fluorescence of the fluorophore when brought into close apposition with it. A change in fluorescence upon excitation is indicative of a change in the association of the molecules tagged with the fluorophore:quencher pair. Generally, an increase in fluorescence of the labelled TNF-alpha is indicative that anti-TNF-alpha polypeptide bearing the quencher has been displaced. For quenching assays, a 10% or greater increase in the intensity of fluorescent emission in samples containing a candidate modulator, relative to samples without the candidate modulator, indicates that the candidate modulator inhibits TNF-alpha:anti-TNF-alpha polypeptide interaction.
  • In addition to the surface plasmon resonance and FRET methods, fluorescence polarization measurement is useful to quantitate binding. The fluorescence polarization value for a fluorescently-tagged molecule depends on the rotational correlation time or tumbling rate. Complexes, such as those formed by TNF-alpha associating with a fluorescently labelled anti-TNF-alpha polypeptide, have higher polarization values than uncomplexed, labelled polypeptide. The inclusion of a candidate inhibitor of the TNF-alpha:anti-TNF-alpha polypeptide interaction results in a decrease in fluorescence polarization, relative to a mixture without the candidate inhibitor, if the candidate inhibitor disrupts or inhibits the interaction of TNF-alpha with said polypeptide. Fluorescence polarization is well suited for the identification of small molecules that disrupt the formation of TNF-alpha:anti-TNF-alpha polypeptide complexes. A decrease of 10% or more in fluorescence polarization in samples containing a candidate modulator, relative to fluorescence polarization in a sample lacking the candidate modulator, indicates that the candidate modulator inhibits the TNF-alpha:anti-TNF-alpha polypeptide interaction.
  • Another alternative for monitoring TNF-alpha:anti-TNF-alpha polypeptide interactions uses a biosensor assay. ICS biosensors have been described in the art (Australian Membrane Biotechnology Research Institute; Cornell B, Braach-Maksvytis V, King L, Osman P, Raguse B, Wieczorek L, and Pace R. “A biosensor that uses ion-channel switches” Nature 1997, 387, 580). In this technology, the association of TNF-alpha and a anti-TNF-alpha polypeptide is coupled to the closing of gramacidin-facilitated ion channels in suspended membrane bilayers and thus to a measurable change in the admittance (similar to impedence) of the biosensor. This approach is linear over six orders of magnitude of admittance change and is ideally suited for large scale, high throughput screening of small molecule combinatorial libraries. A 10% or greater change (increase or decrease) in admittance in a sample containing a candidate modulator, relative to the admittance of a sample lacking the candidate modulator, indicates that the candidate modulator inhibits the interaction of TNF-alpha and said polypeptide. It is important to note that in assays testing the interaction of TNF-alpha with an anti-TNF-alpha polypeptide, it is possible that a modulator of the interaction need not necessarily interact directly with the domain(s) of the proteins that physically interact with said polypeptide. It is also possible that a modulator will interact at a location removed from the site of interaction and cause, for example, a conformational change in the TNF-alpha. Modulators (inhibitors or agonists) that act in this manner are nonetheless of interest as agents to modulate the binding of TNF-alpha to its receptor.
  • Any of the binding assays described can be used to determine the presence of an agent in a sample, e.g., a tissue sample, that binds to TNF-alpha, or that affects the binding of, for example, a polypeptide represented by SEQ ID NO: 3 to the TNF-alpha. To do so a TNF-alpha is reacted with said polypeptide in the presence or absence of the sample, and polypeptide binding is measured as appropriate for the binding assay being used. A decrease of 10% or more in the binding of said polypeptide indicates that the sample contains an agent that modulates the binding of said polypeptide to the TNF-alpha. Of course, the above-generalized method might easily be applied to screening for candidate modulators which alter the binding between any anti-TNF-alpha polypeptide of the invention, an homologous sequence thereof, a functional portion thereof or a functional portion of an homologous sequence thereof, and TNF-alpha or a fragment thereof.
  • One embodiment of the present invention is an unknown agent identified by the method disclosed herein.
  • One embodiment of the present invention is an unknown agent identified by the method disclosed herein for use in treating, preventing and/or alleviating the symptoms of disorders relating to inflammatory processes.
  • Another embodiment of the present invention is a use of an unknown agent identified by the method disclosed herein for use in treating, preventing and/or alleviating the symptoms of disorders relating to inflammatory processes.
  • Examples of disorders include rheumatoid arthritis, Crohn's disease, ulcerative colitis, inflammatory bowel syndrome and multiple sclerosis
  • A cell that is useful according to the invention is preferably selected from the group consisting of bacterial cells such as, for example, E. coli, yeast cells such as, for example, S. cerevisiae, P. pastoris, insect cells or mammal cells.
  • A cell that is useful according to the invention can be any cell into which a nucleic acid sequence encoding a polypeptide comprising an anti-TNF-alpha of the invention, an homologous sequence thereof, a functional portion thereof or a functional portion of an homologous sequence thereof according to the invention can be introduced such that the polypeptide is expressed at natural levels or above natural levels, as defined herein. Preferably a polypeptide of the invention that is expressed in a cell exhibits normal or near normal pharmacology, as defined herein. Most preferably a polypeptide of the invention that is expressed in a cell comprises the nucleotide sequence capable of encoding any one of the amino acid sequences presented in Table 1 or capable of encoding an amino acid sequence that is at least 70% identical to the amino acid sequence presented in Table 1.
  • According to a preferred embodiment of the present invention, a cell is selected from the group consisting of COS7-cells, a CHO cell, a LM (TK-) cell, a NIH-3T3 cell, HEK-293 cell, K-562 cell or a 1321N1 astrocytoma cell but also other transfectable cell lines.
  • In general, “therapeutically effective amount”, “therapeutically effective dose” and “effective amount” means the amount needed to achieve the desired result or results (modulating TNF-alpha binding; treating or preventing inflammation). One of ordinary skill in the art will recognize that the potency and, therefore, an “effective amount” can vary for the various compounds that modulate TNF-alpha binding used in the invention. One skilled in the art can readily assess the potency of the compound.
  • As used herein, the term “compound” refers to an anti-TNF-alpha polypeptide of the present invention, a composition, or a nucleic acid capable of encoding said polypeptide or an agent identified according to the screening method described herein or said polypeptide comprising one or more derivatised amino acids.
  • By “pharmaceutically acceptable” is meant a material that is not biologically or otherwise undesirable, i.e., the material may be administered to an individual along with the compound without causing any undesirable biological effects or interacting in a deleterious manner with any of the other components of the pharmaceutical composition in which it is contained.
  • Anti-TNF-alpha polypeptides as disclosed herein is useful for treating or preventing conditions in a subject and comprises administering a pharmaceutically effective amount of a compound or composition.
  • Anti-TNF polypeptides of the present invention are useful for treating or preventing conditions relating to rheumatoid arthritis, Crohn's disease, ulcerative colitis, inflammatory bowel syndrome and multiple sclerosis in a subject and comprises administering a pharmaceutically effective amount of a compound or composition that binds TNF-alpha.
  • Anti-TNF-alpha polypeptides as disclosed here in are useful for treating or preventing conditions in a subject and comprises administering a pharmaceutically effective amount of a compound combination with another, such as, for example, aspirin.
  • The anti-TNF-alpha polypeptides as disclosed here in are useful for treating or preventing conditions relating to rheumatoid arthritis, Crohn's disease, ulcerative colitis, inflammatory bowel syndrome and multiple sclerosis in a subject and comprises administering a pharmaceutically effective amount of a compound combination with another, such as, for example, aspirin.
  • The present invention is not limited to the administration of formulations comprising a single compound of the invention. It is within the scope of the invention to provide combination treatments wherein a formulation is administered to a patient in need thereof that comprises more than one compound of the invention.
  • Conditions mediated by TNF-alpha include, but are not limited rheumatoid arthritis, Crohn's disease, ulcerative colitis, inflammatory bowel syndrome and multiple sclerosis.
  • A compound useful in the present invention can be formulated as pharmaceutical compositions and administered to a mammalian host, such as a human patient or a domestic animal in a variety of forms adapted to the chosen route of administration, i.e., orally or parenterally, by intranassally by inhalation, intravenous, intramuscular, topical or subcutaneous routes.
  • A compound of the present invention can also be administered using gene therapy methods of delivery. See, e.g., U.S. Pat. No. 5,399,346, which is incorporated by reference in its entirety. Using a gene therapy method of delivery, primary cells transfected with the gene for the compound of the present invention can additionally be transfected with tissue specific promoters to target specific organs, tissue, grafts, tumors, or cells.
  • Thus, the present compound may be systemically administered, e.g., orally, in combination with a pharmaceutically acceptable vehicle such as an inert diluent or an assimilable edible carrier. They may be enclosed in hard or soft shell gelatin capsules, may be compressed into tablets, or may be incorporated directly with the food of the patient's diet. For oral therapeutic administration, the active compound may be combined with one or more excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, and the like. Such compositions and preparations should contain at least 0.1% of active compound. The percentage of the compositions and preparations may, of course, be varied and may conveniently be between about 2 to about 60% of the weight of a given unit dosage form. The amount of active compound in such therapeutically useful compositions is such that an effective dosage level will be obtained.
  • The tablets, troches, pills, capsules, and the like may also contain the following: binders such as gum tragacanth, acacia, corn starch or gelatin; excipients such as dicalcium phosphate; a disintegrating agent such as corn starch, potato starch, alginic acid and the like; a lubricant such as magnesium stearate; and a sweetening agent such as sucrose, fructose, lactose or aspartame or a flavoring agent such as peppermint, oil of wintergreen, or cherry flavoring may be added. When the unit dosage form is a capsule, it may contain, in addition to materials of the above type, a liquid carrier, such as a vegetable oil or a polyethylene glycol. Various other materials may be present as coatings or to otherwise modify the physical form of the solid unit dosage form. For instance, tablets, pills, or capsules may be coated with gelatin, wax, shellac or sugar and the like. A syrup or elixir may contain the active compound, sucrose or fructose as a sweetening agent, methyl and propylparabens as preservatives, a dye and flavoring such as cherry or orange flavor. Of course, any material used in preparing any unit dosage form should be pharmaceutically acceptable and substantially non-toxic in the amounts employed. In addition, the active compound may be incorporated into sustained-release preparations and devices.
  • The active compound may also be administered intravenously or intraperitoneally by infusion or injection. Solutions of the active compound or its salts can be prepared in water, optionally mixed with a nontoxic surfactant. Dispersions can also be prepared in glycerol, liquid polyethylene glycols, triacetin, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms.
  • The pharmaceutical dosage forms suitable for injection or infusion can include sterile aqueous solutions or dispersions or sterile powders comprising the active ingredient which are adapted for the extemporaneous preparation of sterile injectable or infusible solutions or dispersions, optionally encapsulated in liposomes. In all cases, the ultimate dosage form must be sterile, fluid and stable under the conditions of manufacture and storage. The liquid carrier or vehicle can be a solvent or liquid dispersion medium comprising, for example, water, ethanol, a polyol (for example, glycerol, propylene glycol, liquid polyethylene glycols, and the like), vegetable oils, nontoxic glyceryl esters, and suitable mixtures thereof. The proper fluidity can be maintained, for example, by the formation of liposomes, by the maintenance of the required particle size in the case of dispersions or by the use of surfactants. The prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars, buffers or sodium chloride. Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin.
  • Sterile injectable solutions are prepared by incorporating the active compound in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by filter sterilization. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum drying and the freeze drying techniques, which yield a powder of the active ingredient plus any additional desired ingredient present in the previously sterile-filtered solutions.
  • For topical administration, the present compound may be applied in pure form, i.e., when they are liquids. However, it will generally be desirable to administer them to the skin as compositions or formulations, in combination with a dermatologically acceptable carrier, which may be a solid or a liquid.
  • Useful solid carriers include finely divided solids such as talc, clay, microcrystalline cellulose, silica, alumina and the like. Useful liquid carriers include water, hydroxyalkyls or glycols or water-alcohol/glycol blends, in which the present compound can be dissolved or dispersed at effective levels, optionally with the aid of non-toxic surfactants. Adjuvants such as fragrances and additional antimicrobial agents can be added to optimize the properties for a given use. The resultant liquid compositions can be applied from absorbent pads, used to impregnate bandages and other dressings, or sprayed onto the affected area using pump-type or aerosol sprayers.
  • Thickeners such as synthetic polymers, fatty acids, fatty acid salts and esters, fatty alcohols, modified celluloses or modified mineral materials can also be employed with liquid carriers to form spreadable pastes, gels, ointments, soaps, and the like, for application directly to the skin of the user.
  • Examples of useful dermatological compositions which can be used to deliver the compound to the skin are known to the art; for example, see Jacquet et al. (U.S. Pat. No. 4,608,392), Geria (U.S. Pat. No. 4,992,478), Smith et al. (U.S. Pat. No. 4,559,157) and Wortzman (U.S. Pat. No. 4,820,508).
  • Useful dosages of the compound can be determined by comparing their in vitro activity, and in vivo activity in animal models. Methods for the extrapolation of effective dosages in mice, and other animals, to humans are known to the art; for example, see U.S. Pat. No. 4,938,949.
  • Generally, the concentration of the compound(s) in a liquid composition, such as a lotion, will be from about 0.1-25 wt- %, preferably from about 0.5-10 wt- %. The concentration in a semi-solid or solid composition such as a gel or a powder will be about 0.1-5 wt- %, preferably about 0.5-2.5 wt- %.
  • The amount of the compound, or an active salt or derivative thereof, required for use in treatment will vary not only with the particular salt selected but also with the route of administration, the nature of the condition being treated and the age and condition of the patient and will be ultimately at the discretion of the attendant physician or clinician. Also the dosage of the compound varies depending on the target cell, tumor, tissue, graft, or organ.
  • The desired dose may conveniently be presented in a single dose or as divided doses administered at appropriate intervals, for example, as two, three, four or more sub-doses per day. The sub-dose itself may be further divided, e.g., into a number of discrete loosely spaced administrations; such as multiple inhalations from an insufflator or by application of a plurality of drops into the eye.
  • An administration regimen could include long-term, daily treatment. By “long-term” is meant at least two weeks and preferably, several weeks, months, or years of duration. Necessary modifications in this dosage range may be determined by one of ordinary skill in the art using only routine experimentation given the teachings herein. See Remington's Pharmaceutical Sciences (Martin, E. W., ed. 4), Mack Publishing Co., Easton, Pa. The dosage can also be adjusted by the individual physician in the event of any complication.
  • The invention provides for an agent that is a modulator of TNF-alpha/TNF-alpha-receptor interactions.
  • The candidate agent may be a synthetic agent, or a mixture of agents, or may be a natural product (e.g. a plant extract or culture supernatant). A candidate agent according to the invention includes a small molecule that can be synthesized, a natural extract, peptides, proteins, carbohydrates, lipids etc.
  • Candidate modulator agents from large libraries of synthetic or natural agents can be screened. Numerous means are currently used for random and directed synthesis of saccharide, peptide, and nucleic acid based agents. Synthetic agent libraries are commercially available from a number of companies including Maybridge Chemical Co. (Trevillet, Cornwall, UK), Comgenex (Princeton, N.J.), Brandon Associates (Merrimack, N.H.), and Microsource (New Milford, Conn.). A rare chemical library is available from Aldrich (Milwaukee, Wis.). Combinatorial libraries are available and can be prepared. Alternatively, libraries of natural agents in the form of bacterial, fungal, plant and animal extracts are available from e.g., Pan Laboratories (Bothell, Wash.) or MycoSearch (N.C.), or are readily producible by methods well known in the art. Additionally, natural and synthetically produced libraries and agents are readily modified through conventional chemical, physical, and biochemical means.
  • Useful agents may be found within numerous chemical classes. Useful agents may be organic agents, or small organic agents. Small organic agents have a molecular weight of more than 50 yet less than about 2,500 daltons, preferably less than about 750, more preferably less than about 350 daltons. Exemplary classes include heterocycles, peptides, saccharides, steroids, and the like. The agents may be modified to enhance efficacy, stability, pharmaceutical compatibility, and the like. Structural identification of an agent may be used to identify, generate, or screen additional agents. For example, where peptide agents are identified, they may be modified in a variety of ways to enhance their stability, such as using an unnatural amino acid, such as a D-amino acid, particularly D-alanine, by functionalizing the amino or carboxylic terminus, e.g. for the amino group, acylation or alkylation, and for the carboxyl group, esterification or amidification, or the like.
  • For primary screening, a useful concentration of a candidate agent according to the invention is from about 10 mM to about 100 μM or more (i.e. 1 mM, 10 mM, 100 mM, 1 M etc.). The primary screening concentration will be used as an upper limit, along with nine additional concentrations, wherein the additional concentrations are determined by reducing the primary screening concentration at half-log intervals (e.g. for 9 more concentrations) for secondary screens or for generating concentration curves.
  • High throughput Screening Kit
  • A high throughput screening kit according to the invention comprises all the necessary means and media for performing the detection of an agent that modulates TNF-alpha/TNF-alpha receptor interactions by interacting with TNF-alpha in the presence of a polypeptide, preferably at a concentration in the range of 1 μM to 1 mM.
  • The kit comprises the following. Recombinant cells of the invention, comprising and expressing the nucleotide sequence encoding TNF-alpha, which are grown according to the kit on a solid support, such as a microtiter plate, more preferably a 96 well microtiter plate, according to methods well known to the person skilled in the art especially as described in WO 00/02045. Alternatively TNF-alpha is supplied in a purified form to be immobilized on, for example, a 96 well microtiter plate by the person skilled in the art. Alternatively TNF-alpha is supplied in the kit pre-immobilized on, for example, a 96 well microtiter plate. The TNF-alpha may be whole TNF-alpha or a fragment thereof.
  • Modulator agents according to the invention, at concentrations from about 1 μM to 1 mM or more, are added to defined wells in the presence of an appropriate concentration of anti-TNF-alpha polypeptide, an homologous sequence thereof, a functional portion thereof or a functional portion of an homologous sequence thereof, said concentration of said polypeptide preferably in the range of 1 μM to 1 mM. Kits may contain one or more anti-TNF-alpha polypeptide (e.g. one or more of a polypeptide represented by any of the SEQ ID NOs: 1 to 15 or other anti-TNF-alpha polypeptides, an homologous sequence thereof, a functional portion thereof or a functional portion of an homologous sequence thereof).
  • Binding assays are performed as according to the methods already disclosed herein and the results are compared to the baseline level of, for example TNF-alpha binding to an anti-TNF-alpha polypeptide, an homologous sequence thereof, a functional portion thereof or a functional portion of an homologous sequence thereof, but in the absence of added modulator agent. Wells showing at least 2 fold, preferably 5 fold, more preferably 10 fold and most preferably a 100 fold or more increase or decrease in TNF-alpha-polypeptide binding (for example) as compared to the level of activity in the absence of modulator, are selected for further analysis.
  • Other Kits Useful According to the Invention
  • The invention provides for kits useful for screening for modulators of TNF-alpha/TNF-alpha receptor binding, as well as kits useful for diagnosis of disorders characterised by dysfunction of TNF-alpha. The invention also provides for kits useful for screening for modulators of disorders as well as kits for their diagnosis, said disorders characterised by one or more process involving TNF-alpha. Kits useful according to the invention can include an isolated TNF-alpha. Alternatively, or in addition, a kit can comprise cells transformed to express TNF-alpha. In a further embodiment, a kit according to the invention can comprise a polynucleotide encoding TNF-alpha. In a still further embodiment, a kit according to the invention may comprise the specific primers useful for amplification of TNF-alpha. Kits useful according to the invention can comprise an isolated TNF-alpha polypeptide, a homologue thereof, or a functional portion thereof. A kit according to the invention can comprise cells transformed to express said polypeptide. Kits may contain more than one polypeptide. In a further embodiment, a kit according to the invention can comprise a polynucleotide encoding TNF-alpha. In a still further embodiment, a kit according to the invention may comprise the specific primers useful for amplification of a macromolecule such as, for example, TNF-alpha. All kits according to the invention will comprise the stated items or combinations of items and packaging materials therefore. Kits will also include instructions for use.
  • EXAMPLES
  • The invention is illustrated by the following non-limiting examples.
  • Example 1 Example of Camelidae Antibodies against Human Tumor Necrosis Factor Alpha
  • 1) Immunization and Library Constructions
  • A llama (Llama glama) was immunized with human TNF-alpha. For immunization, the cytokine was formulated as an emulsion with an appropriate, animal-friendly adjuvant (Specoll, CEDI Diagnostics B.V.). The antigen cocktail was administered by double-spot injections intramuscularly in the neck. The animal received 6 injections of the emulsion, containing 100 μg of TNF-alpha at weekly intervals. At different time points during immunization, 10-ml blood samples were collected from the animal and sera were prepared. The induction of an antigen specific humoral immune response was verified using the serum samples in an ELISA experiment with TNF (data not shown). Five days after the last immunization, a blood sample of 150 ml was collected. From this sample, conventional and heavy-chain antibodies (HcAbs) were fractionated (Lauwereys et al. 1998) and used in an ELISA, which revealed that the HcAbs were responsible for the antigen specific humoral immune response (data not shown). Peripheral blood lymphocytes (PBLs), as the genetic source of the llama heavy chain immunoglobulins (HcAbs), were isolated from the 150-ml blood sample using a Ficoll-Paque gradient (Amersham Biosciences) yielding 5×108 PBLs. The maximal diversity of antibodies is expected to be equal to the number of sampled B-lymphocytes, which is about 10% of the number of PBLs (5×107). The fraction of heavy-chain antibodies in llama is up to 20% of the number of B-lymphocytes. Therefore, the maximal diversity of HcAbs in the 150 ml blood sample is calculated as 107 different molecules. Total RNA (around 400 μg) was isolated from these cells using an acid guanidinium thiocyanate extraction (Chomczynski and Sacchi, 1987).
  • cDNA was prepared on 100 μg total RNA with M-MLV Reverse Transcriptase (Gibco BRL) and oligo-dT-primer or hexanucleotide random primers (Amersham Biosciences) as described before (de Haard et al., 1999). The cDNA was purified with a phenol/chloroform extraction combined with an ethanol precipitation and subsequently used as template to specifically amplify the VHH repertoire.
  • The VHH repertoire was amplified using oligo-dT primed cDNA as template with a single degenerated framework1 (FR1) primer ABL013 (5′-GAGGTBCARCTGCAGGASTCYGG-3′) (SEQ ID NO:98), introducing a PstI restriction site (in bold), in combination with the oligo-dT primer as is described in EP01205100.9. This amplification yields two fragments of 1650 bp and 1300 bp, the latter being the product derived from the CH1-deleted HcAb genes. The smaller PCR-product was gel purified and subsequently digested with PstI and BstEII. The BstEII-site frequently occurs within the FR4 of heavy-chain derived VHH encoding DNA-fragments.
  • Alternatively, the VHH-repertoire was amplified in a hinge-dependent approach using two IgG specific oligonucleotide primers. In a single PCR reaction a short (5′-AACAGTTAAGCTTCCGCTTGCGGCCGCGGAGCTGGGGTCTTCGCTGTGGTGCG-3′) (SEQ ID NO:99) or long (5′-AACAGTTAAGCTTCCGCTTGCGGCCGCTGGTTGTGG TTTTGGTGTCTTGGGTT-3′) (SEQ ID NO:100) hinge primer known to be specific for HcAbs was combined with the FR1-primer ABL013 (see above). A PstI and NotI (bold underlined) restriction site was introduced within the FR1 and hinge primers respectively, to allow cloning. Subsequently, the DNA fragments were ligated into PstI-BstEII or PstI-NotI digested phagemid vector pAX004, which is identical to pHEN1 (Hoogenboom et al., 1991), but encodes a carboxyterminal (His)6- and c-myc-tag for purification and detection, respectively. The ligation mixture was desalted on a Microcon filter (YM-50, Millipore) and electroporated into E. coli TG1 cells to obtain a library containing 1.8×107 clones. The transformed cells were grown overnight at 37° C. on a single 20×20 cm plate with LB containing 100 μg/ml ampicillin and 2% glucose. The colonies were scraped from plates using 2×TY medium and stored at −80° C. in 20% glycerol.
  • As quality control the percentage of insert containing clones was verified on 24 clones for each library by PCR using a combination of vector based primers. This analysis revealed that 95% of the clones contained a VHH encoding insert. The variability was examined by HinfI fingerprint analysis of the amplified VHH fragment of these 24 clones, thereby showing that all clones were indeed different (data not shown).
  • 2) Selection of Antagonistic Anti-TNF VHH's
  • From both libraries phage was prepared. To rescue the polyclonal phage repertoire, libraries were grown to logarithmic phase (OD600=0.5) at 37° C. in 2×TY containing 100 μg/ml ampicillin and 2% glucose and subsequently superinfected with M13K07 helper phage for 30 minutes at 37° C. Infected cells were pelleted for 5 minutes at 4000 rpm and resuspended in 2×TY containing 100 μg/ml ampicillin and 25 μg/ml kanamycin. Bacteriophage was propagated by overnight growth at 37° C. and 250 rpm. Overnight cultures were centrifuged for 15 minutes at 4500 rpm and phage was precipitated with one fifth volume of a [20% polyethyleneglycol 6000, 1.5 M NaCl]-solution by incubation for 30 minutes on ice. Phage was pelleted by centrifugation for 15 minutes at 4000×g and 4° C. After resuspension of the phages in PBS, cell debris was pelleted by centrifugation for 1 minute at maximal speed (15000×g) in microcentrifuge tubes. The supernatant containing the phage particles was transferred to a new tube and again phage was precipitated as described above. Phage was dissolved in PBS and separated from remaining cell debris as mentioned above. The titer of phage was determined by infection of logarithmic TG1 cells followed by plating on selective medium.
  • The library was selected using in vitro biotinylated TNF-alpha. The biotinylation was carried out as described by Magni et al (Anal Biochem 2001, 298, 181-188). The incorporation of biotin in TNF was evaluated by SDS-PAGE analysis and detection with Extravidin-alkaline phosphatase conjugate (Sigma). The functionality of the modified protein was evaluated for its ability to bind to the solid phase coated recombinant a p75 receptor.
  • VHH were selected by capturing biotinylated TNF-alpha (10 to 400 ng per well during 2 hours at room temperature) on streptavidin coated microtiter plates (coated with 100 μl of 10 μg/ml streptavidin during 16 hours at +4° C.). Antagonistic VHH were obtained by elution with an excess of receptor, either the extracellular ligand binding domain or with cells expressing the receptor. After 2 hours incubation of phage with captured cytokine, the non-specific phage was washed away, while specific phage displaying antagonistic VHH was eluted for 30 minutes with receptor (extracellular domain of CD120b or p75; 10 μM) or with receptor displaying cells (>105 KYM cells per well). High enrichments, i.e. the ratio of the number of phage eluted with receptor and those eluted by serum albumin (50 μg per well), of more than a factor of 20 suggested the successful selection of TNF-alpha specific clones. Alternatively, instead of elution with receptor a standard procedure was applied, in which a low pH causes the denaturation of VHH and/or antigen (0.1 M glycine buffer pH 2.5). Log phase growing E. coli cells were infected with the eluted and neutralized phage and plated on selective medium.
  • Individual clones were picked and grown in microtiter plate for the production of VHH in culture supernatant. ELISA screening with TNF-alpha captured on Extravidin coated plates revealed about 50% positive clones. HinfI-fingerprint analysis showed that 13 different clones were selected, which were grown and induced on 50 ml scale. The sequences of said clones are shown in Table 1.
  • Five clones, coded VHH#1A, #2B, #3E, #3G, #7B and #12B, with different sequences (FIG. 1) were characterized in more detail. VHH#3E, #3G and #7B are single-domain antibody fragments carrying the typical hydrophilic residue at position 45 (arginine) and the phenylalanine to tryptophan substitution in position 47 in FR2 thereby conferring the advantageous characteristics in terms of solubility. VHH#1A contains the hydrophobic FR2 residues typically found in double-chain antibodies of human origin or from other species, but compensating this loss in hydrophilicity by the charged arginine residue on position 103 that substitutes the conserved tryptophan residue present in VH from double-chain antibodies (PCT/EP02/07804). A new class of humanised Camelidae single-domain antibodies described in this invention is represented by VHH#2B and VHH#12B, which contains the hydrophobic residues in FR2 in combination with the hydrophobic residue tryptophan at position 103. Larger amounts of antibody fragments were expressed by cultivation on 50 ml scale and purified by IMAC using TALON resin (Clontech). After dialysis against PBS to remove the eluent imidazol the amount of VHH was determined by OD280; approximately 300 μg of VHH was obtained from each clone.
  • This material was used for determining the sensitivity of detection of (biotinylated) TNF in ELISA. For this purpose a streptavidin (10 μg/ml) coated microtiterplate was employed for capture of biotinylated TNF (1 μg/ml), VHH was diluted in 0.2% casein/PBS and incubated for 2 hours at room temperature. Bound VHH was detected with anti-MYC mAB 9E10 (0.5 μg/ml) and anti-mouse AP conjugate (1000-fold diluted, Sigma). The results are shown in FIG. 2.
  • 3) Determination of Antagonistic Effect in Cytotoxicity Assay with KYM Cell Line
  • TNF-alpha-induced cytostasis/cytotoxicity was determined by the colorimetric MTT assay as described by Vandenabeele and colleagues (Vandenabeele, P., Declercq, W., Vercammen, D., Van de Craen, M., Grooten, J., Loetscher, H., Brockhaus, M., Lesslauer, W., Fiers, W. (1992) Functional characterization of the human tumor necrosis factor receptor p75 in a transfected rat/mouse T cell hybridoma. J. Exp. Med. 176, 1015-1024.). MTT (3-(4,5-cimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide) is a pale yellow substrate that is cleaved by living cells to yield a dark blue formazan product. This process requires active mitochondria, and even freshly dead cells do not cleave significant amounts of MTT. KYM cells (Sekiguchi M, Shiroko Y, Suzuki T, Imada M, Miyahara M, Fujii G. (1985) Characterization of a human rhabdomyosarcoma cell strain in tissue culture. Biomed. Pharmacother. 39, 372-380.) were seeded in 96 well microtiterplates and cultured in the presence or absence of TNF-alpha (0.216 ng/ml or approx. 5 pM of trimer). In addition to TNF variable amounts of antibody (VHH or Remicade) were included during cultivation. For the assay MTT was added to the culture medium at a final concentration of 500 μg/ml and the plates were incubated at 37° C. to achieve cleavage of MTT by mitochondrial enzymes.
  • The formed formazon product, which appear as black, fuzzy crystals on the bottom of the well were dissolved by addition of acid isopropanol (40 nM HCl in isopropanol) or DMSO. The absorbance is measured at 570 nm.
  • The MTT assay (FIG. 3) shows that VHH#1A, which has arginine on position 103 in combination with the human-like hydrophobic residues in FR2, has a moderate antagonistic effect (IC50˜100 nM). VHH#7B with the characteristic hydrophilic residues in FR2 does not prevent binding of TNF-α to its ligand in spite of its sensitive detection of the cytokine in ELISA (curve not shown). In contrast, VHH#3E and #3G with hydrophilic FR2 hallmark residues are very potent antagonistic VHH's (IC50 of 20 nM). VHH#3E and #3G have a high degree of homology and are clonally related (Harmsen et al., Mol. Immunol. 37, 579-590), but VHH#3E is more potent, probably due to the fact that it has a higher affinity than VHH#3G (FIG. 2). The (chimeric) monoclonal antibody Remicade is very potent (IC50 of 80 pM), but its derived Fab fragment lost most of this efficacy (IC50 is 3 nM, 30 fold less than the intact mAB). This clearly shows the avidity effect of the interaction between the antibody and the cytokine: the mAB with two binding sites interacts more efficiently with the trimeric TNF molecule via cooperative binding. VHH fragments are strictly monovalent and therefore it was speculated that increasing the avidity by genetically fusing VHH genes might increase their antagonistic efficacy (see Example 4).
  • These experiments show that a new class of human-like VHH has bona fide binding and functional characteristics, thereby enabling their application for therapeutic purposes.
  • Example 2 Humanization of VHH#12B and VHH#3E by Site Directed Mutagenesis
  • 1) Homoloqy between VHH#3E/VHH#12B and Human Germlne Heavy Chain V-Region DP-47
  • Alignment of VHH#12B and a human VH3 germline sequence (DP-47) revealed a high degree of homology:
      • 4 AA changes in FR1 on position 1, 5, 28 and 30
      • 5 AA changes in FR3 on position 74, 76, 83, 84 and 93
      • 1 AA change in FR4 on position 108
  • as represented in the following sequence alignment in which DP-47 is SEQ ID NO:101 and VHH#12B is SEQ ID NO:102:
    DP-47 EVQLLESGGGLVQPGGSLRLSCAASGFTFS SYAMS WVRQAPGKGLEWVS AISGSGGSTYY
    VHH#
    12B QVQLQESGGGLVQPGGSLRLSCAASGFEFE NHWMY WVRQAPGKGLEWVS TVNTNGLITRY
  • DP-47 ADSVKG RFTISRDNSKNTLYLQMNSLRAEDTAVYYCAK --------------------------
    VHH#12B ADSVKG RFTISRDNAKYTLYLQMNSLKSEDTAVYYCTK VLPPYSDDSRTNAD WGQGTQVTVSS
  • A specific inhibitor for the TNF-alpha cytokine, with high homology to the human germline gene DP-47 was therefore an ideal candidate to further humanize and evaluate the influence of mutagenesis on inhibition capacity in ELISA.
  • Alignment of VHH#3E and a human VH3 germline (DP-47) revealed the presence of hydrophilic amino acid residues in FR2 of VHH#3E compared to hydrophobic residues in DP-47.
    DP-47 EVQLLESGGGLVQPGGSLRLSCAASGFTFS SYAMS----- WVRQAPGKGLEWVS AISGSGGSTYY
    VHH#
    3E QVQLQESGGGLVQPGGSLRLSCAASGRTFS DHSGYTYTIG WFRQAPGKEREFVA RIYWSSGNTYY
  • DP-47 ADSVKG RFTISRDNSKNTLYLQMNSLRAEDTAVYYCAK ------------- ------------
    VHH#3E ADSVKG RFAISRDIAKNTVDLTMNNLEPEDTAVYYCAA RDGIPTSRSVESYNY WGQGTQVTVSS
  • Evaluation of the effect of substituting the hydrophilic by hydrophobic residues as present in human VH is important, since the majority of camelid VHH sequences contain hydrophilic residues.
  • 2) Mutagenesis of VHH#12B
  • VHH#12B was mutated by using a non-PCR based site-directed mutagenesis method as described by Chen and Ruffner and commercialized by Stratagene (Quickchange site-directed mutagenesis). Plasmid DNA was used as template in combination with 2 mutagenic primers introducing the desired mutation(s). The 2 primers are each complementary to opposite strands of the template plasmid DNA. In a polymerase reaction using the Pfu DNA polymerase each strand is extended from the primer sequence during a cycling program using a limited number of cycles. This results in a mixture of wild type and mutated strands. Digestion with Dpnl results in selection of the mutated in vitro synthesized DNA strand, since only the template strand is sensitive for digestion. The DNA was precipitated and transformed to E. coli and analyzed for the required mutation by sequence analysis. The generated mutant VHH's and the mutagenic primers are listed in Table 2.
  • Plasmid was prepared from mutant clones and was transformed into WK6 electrocompetent cells. A single colony was used to start an overnight culture in LB containing 2% glucose and 100 μg/ml ampicillin. This overnight culture was diluted 100-fold in 300 ml TB medium containing 100 μg/ml ampicillin, and incubated at 37° C. until OD600 nm=2, when 1 mM IPTG and 5 mM MgSO4 (final concentrations) was added and the culture was incubated for 3 more hours at 37° C.
  • Cultures were centrifuged for 20 minutes at 4,500 rpm at 4° C. The pellet was frozen overnight or for 1 hour at −20° C. Next, the pellet was thawed at room temperature for 40 minutes, re-suspended in 20 ml PBS/1 mM EDTA/1M NaCl and shaken on ice for 1 hour. Periplasmic fraction was isolated by centrifugation for 20 minutes at 4° C. at 4,500 rpm. The supernatant containing the VHH was loaded on TALON (ClonTech) and purified to homogeneity. The yield of VHH was determined using the calculated extinction coefficient.
  • All mutant VHH's expressed comparable to the wild type. The mutants were analyzed for their inhibition capacity in an in vitro receptor binding assay.
  • A microtiter plate was coated overnight at 4° C. with Enbrel (Wyeth) at 2 μg/ml in PBS. The plate was washed five times with PBS-Tween and blocked for 1 hour at room temperature with PBS containing 1% casein. The plate was washed five times with PBS-Tween. Biotinylated human TNF-alpha (80 μg/ml) was pre-incubated with a dilution series of mutant or wild type VHH#12B for 1 hr at RT and the mixture was incubated for 1 hr at room temperature in the wells of the microtiterplate. The plate was washed five times with PBS-Tween. Bound human TNF-α was detected using Extravidin-AP (1/1,000 dilution) and paranitrophenylphosphate (pNPP). Signals were measured after 30 minutes at 405 nm. The results are presented in FIG. 4 and 5. The IC50 increased 3-fold from 66 nM (wild type) to 200 nM (mutant Q1E+Q5L+A74S+Y76N+K83R+P84A). Mutation of position T93A resulted in loss of inhibition (data not shown). The positions that still need to be humanized are: E28, E30 and Q108. However, E28 and E30 are part of the H1 canonical structure and thus part of the CDR1 according to Chothia numbering system.
  • The amino acid sequences of mutant VHHs are presented in Table 4 SEQ ID NOs: 17 to 19.
  • 3) Mutagenesis of VHH#3E
  • VHH#3E was mutated by using a non-PCR based site-directed mutagenesis method as described above. The obtained mutant VHH's and the mutagenic primers are listed in Table 3.
  • All mutant VHH's expressed comparable to the wild type. The purified mutant VHH's were analyzed for binding in ELISA and inhibition capacity in receptor binding assay identical to the method described above.
  • The results of the ELISA are shown in FIG. 6, those from the receptor binding assay in FIG. 7.
  • The amino acid sequences of mutant VHHs are presented in Table 4 SEQ ID NOs 21 to 24.
  • Example 3 Isolation of Antagonistic VHH against Mouse TNF-Alpha
  • 1) Selection of anti-mouse TNF-alpha VHH
  • In order to perform efficacy studies in mouse models for IBD or Crohn's disease mouse TNF specific VHH were selected. Therefore a llama was immunized with mouse TNF-alpha as described in Example 1. RNA was extracted from PBL's sampled 4 and 10 days after the last immunization, as well as from a biopsy taken from a lymph node after day 4. Total RNA was converted in either random primed or oligo-dT primed cDNA and used as template for the amplification of the VHH encoding gene segments using Ig derived primers or a combination of oligo-dT primer and a single Ig primer (see example 1). With the Ig primers a library containing 8.5×107 clones was generated from the first PBL's, and a library with 7×106 clones for the second PBL sample and 5.8×108 clones for the lymph node. Using the combination of the oligo-dT primer and the Ig primer libraries from the first PBL sample were made containing 1.2×108 clones, from the second sample of PBL's a library of 5.7×107 clones and the lymph node derived library contained 2×108 clones. The libraries were pooled dependent on the used combination of primers and the resulting two libraries were grown for propagation of phage as was described before. Selections were performed on biotinylated mouse TNF-alpha captured on coated streptavidin, bound phage was eluted by competition with the human receptor p75, which is known to cross-react with mouse TNF-alpha. Two distinct mouse TNF-alpha specific VHH (VHH#m3F and VHH#m9E) were selected from the library obtained by amplification with 1 g derived primers, while two closely related VHH's were retrieved from the library constructed by PCR with oligo-dT primer and Ig-primer (FIG. 8).
  • 2) Determination Antagonistic Efficacy in Cytotoxicity Assay with L929 Cell Line (FIG. 9)
  • The same type of assay was applied as described in Example 1, but with the murine cell line L929. VHH#m3F and VHH#m4B (FIG. 9) turned out to be 10-fold more potent then the other two VHH's.
  • Example 4 Enhancing the Antagonistic Efficacy by Increasing the Avidity Using Multivalent Camelidae Antibodies
  • 1) Antagonistic Efficacy of Bi-, Tri- and Tetravalent VHH against Human and Mouse TNF-Alpha
  • The E. coli production vector pAX11 (FIG. 10) was designed, which allows the two-step cloning of bivalent or bispecific VHH. The carboxy terminal VHH is cloned first with PstI and BstEII, while in the second step the other VHH is inserted by SfiI and NotI, which do not cut within the first gene fragment. The procedure avoids the enforcement of new sites by amplification and thus the risk of introducing PCR errors.
  • With this vector the bivalent derivative of the antagonistic anti- human TNF-alpha VHH#3E was generated. The plasmid vector encoding the bivalent VHH was used to generate a tri- and tetrameric derivative, which was accomplished by partial digestion of the plasmid with BstEII, which occurs in both VHH gene segments. The linearized vector was purified from gel, subsequently de-phosphorylated and used as acceptor for cloning of the BstEII fragment of approx. 350 bp that was obtained by complete digestion of the same plasmid. Ligation of the BstEII fragment alone prior to addition to the vector enhances the insertion of multimeric VHH encoding gene segments. After transformation in E. coli TG1 the resulting clones were screened by PCR with M13Rev and M13Fwd primers; since BstEII is an a-symmetric cutter (5 nt overhang) only correctly oriented inserts were obtained as was confirmed by digesting the plasmids with PstI alone (350 bp) or double digesting with EcoRI and HindlIl (1000 bp for bivalent (BIV 3E, SEQ ID NO: 73), 1350 bp for trivalent (TRI 3E, SEQ ID NO: 74) and 1700 bp for tetravalent (TETRA 3E, SEQ ID NO: 75), data not shown). The sequences are listed in Table 7.
  • The clones were grown and induced on 50 ml scale, periplasmic fractions prepared and used for IMAC purification with TALON resin. Analysis of the purified products on Coomassie stained PAGE revealed good production levels (between 2 and 10 mg per liter cell culture) of intact multivalent VHH (see FIG. 11). The molecular appearance of the IMAC purified VHH was determined by gel filtration on a Superdex 75HR column and as expected the molecules with higher avidities came earlier from the column (see FIG. 12).
  • The antagonistic efficacy was analyzed with the cell based assay using KYM cells. The cells were seeded in microtiterplates and cultured in the presence or absence of TNF-alpha (1.29 ng/ml or approx. 25 pM of trimer). The assays (FIG. 13) revealed that the monovalent molecules used in this study had the poorest antagonistic characteristics, what is reflected by their IC50 values: the Fab derived from the chimeric antibody Remicade has an IC50 of 2 nM and for VHH#3E it is 12 nM (see also FIG. 3). The avidity of the used molecules turned out to have a dramatic influence on the antagonistic efficacy as was observed with the bivalent IgG molecule Remicade, which is 40-fold more effective (IC50 50 pM) than the Fab. TNF-alpha is a trimeric molecule, which interacts to a dimeric receptor and therefore it can be expected that the avidity of the IgG permits the mutual binding to two epitopes on the cytokine and supports the formation of large complexes as has been described before (Santora et al, Anal. Biochem. 299, 119-129). Surprisingly, increasing the avidity of the VHH from monomer to dimer has a far more spectacular effect than observed with Remicade, since the IC50 of the dimer (30 pM) is 400 fold lower than of the monomer. Increasing the avidity even more leads to a still better antagonistic behaviour: the trimeric VHH has an IC50 of 20 pM and the tetravalent format 6 pM. All higher avidity formats of the VHH are more efficient than Remicade, while the tetravalent format is even better than Enbrel, which consists of the extracellular domain of the receptor p75 fused to the Fc of an IgG and therefore has a bivalent binding mode.
  • The same unexpected effect of avidity on antagonistic behaviour was observed with VHH generated against mouse TNF (FIG. 14). The same type of cytotoxicity assay was performed using MTT as substrate and mouse TNF-alpha (65 pg/ml or 1.3 pM), but with the murine cell line L929, which expresses the mouse specific receptor. Three different antagonistic (monovalent) VHH were identified coded 9E and 3F, of which the first two have IC50's of 25 nM and the latter 2 nM (see also Example 3). Conversion of 3F into the bivalent format (BIV#m3F, SEQ ID NO: 76) yielded a 1000 fold increase in IC50 (2 μM), thereby demonstrating once more that the increased avidity of the antibody leads to an unexpected improvement of the antagonistic characteristics.
  • 2) Comparison with VHH-Fc Fusion
  • VHH#3E, directed against human TNF, was cloned via PstI and BstElI in an adapted vector derived from pCDNA3, thereby generating a genetic fusion to the CH1 deleted Fc portion of human IgG1. After confirmation by sequencing, the plasmid construct was transfected to the myeloma cell line NS0. The obtained cell line was grown and the VHH-Fc fusion was secreted into the culture supernatant. The product was purified with an anti-human Fc VHH resin and analyzed on a Coomassie stained gel (FIG. 15). In the presence of DTT the fusion was visible as a 45 kDa protein, in the absence of DTT the dimeric molecule with a molecular weight of 90 kDa could be observed. This dimeric product results from the linkage of two chains by two disulfide bridges, which originate from cysteine residues located in the hinge region.
  • The VHH-fusion was tested in the bioassay with the human cell line KYM and turned out to be 5-fold less effective than the bivalent VHH in spite of the fact that both molecules have the same avidity and that they both originate from VHH#3E (FIG. 16). Probably steric hindrance by the bulky Fc tail might cause this discrepancy.
  • Example 5 Calculation of Homologies between Anti-Target-Single Domain Antibodies of the Invention
  • The degree of amino acid sequence homology between anti-target single domain antibodies of the invention was calculated using the Bioedit Sequence Alignment Editor. The calculations indicate the proportion of identical residues between all of the sequences as they are aligned by ClustalW. (Thompson, J. D., Higgins, D. G. and Gibson, T. J. (1994) CLUSTAL W: improving the sensitivity of progressive multiple sequence alignment through sequence weighting, position specific gap penalties and weight matrix choice. Nucleic Acids Research, submitted, June 1994). Table 8 indicates the fraction homology between anti-serum albumin VHHs of the invention. Table 9 indicates the fraction homology between anti-TNF-alpha VHHs of the invention. Table 10 indicates the percentage homology between anti-IFN-gamma VHHs of the invention.
  • Example 6 Expression of a VHH-CDR3 Fragment of VHH#3E
  • The CDR3 region of VHH#3E was amplified by using a sense primer located in the framework 4 region (Forward: CCCCTGGCCCCAGTAGTTATACG) (SEQ ID NO:103) and an anti-sense primer located in the framework 3 region (Reverse: TGTGCAGCAAGAGACGG) (SEQ ID NO:104).
  • In order to clone the CDR-3 fragment in pAX10, a second round PCR amplification was performed with following primers introducing the required restriction sites:
    Reverse primer Sfi1:
    (SEQ ID NO:105)
    GTCCTCGCAACTGCGGCCCAGCCGGCCTGTGCAGCAAGAGACGG
    Forward primer Not1:
    (SEQ ID NO:106)
    GTCCTCGCAACTGCGCGGCCGCCCCCTGGCCCCAGTAGTTATACG
  • The PCR reactions were performed in 50 ml reaction volume using 50 pmol of each primer. The reaction conditions for the primary PCR were 11 min at 94° C., followed by 30/60/120 sec at 94/55/72° C. for 30 cycles, and 5 min at 72° C. All reaction were performed with 2.5 mM MgCl2, 200 mM dNTP and 1.25 U AmpliTaq God DNA Polymerase (Roche Diagnostics, Brussels, Belgium).
  • After cleavage with Sfi1 and Not1 the PCR product was cloned in pAX10.
  • Example 7 Stability Testing of Antibody Fragments Specific for Human TNFα
  • Orally administered proteins are subject to denaturation at the acidic pH of the stomach and as well to degradation by pepsin. We have selected conditions to study the resistance of the VHH#3E to pepsin which are supposed to mimick the gastric environment. VHH#3E a VHH specific to human TNFα was produced as recombinant protein in E. coli and purified to homogeneity by IMAC and gel filtration chromatography. The protein concentration after purification was determined spectrophotometrically by using the calculated molar extinction coefficient at 280 nm. Diluted solutions at 100 microgram/ml were prepared in Mcllvaine buffer (J. Biol. Chem. 49, 1921, 183) at pH 2, pH3 and 4 respectively. These solutions were subsequently incubated for 15 minutes at 37° C., prior the addition of porcine gastric mucosa pepsin at a 1/30 w/w ratio. Sixty minutes after adding the protease a sample was collected and immediately diluted 100-fold in PBS pH7.4 containing 0.1% casein to inactivate the pepsin. Seven additional 3-fold dilutions were prepared from this sample for assessing the presence of functional antibody fragment by ELISA. Identical dilutions prepared from an aliquot collected prior the addition of the protease served as a reference. In the ELISA assay biotinylated TNFα was captured in wells of a microtiter plate coated with neutravidin. For both the pepsin-treated and reference samples similar serial dilutions of the samples were prepared and 100 microliter of those dilutions were added to the wells. After incubation for 1 hour the plates were washed. For the detection of VHH binding to of the captured TNFα a polyclonal rabbit anti-VHH antiserum (R42) and an anti-rabbit IgG alkaline phosphatase conjugate was used. After washing, the plates were developed with paranitrophenyl phosphate. The data plotted in FIG. 17 shows similar curves for all of the samples exposed to digestive conditions as well as for the reference samples. This indicates that the VHH#3E essentially retains its functional activity under all of the chosen conditions.
  • Example 8 Oral Administration of an Anti-Human TNFα Specific VHH in Mice
  • An antibody solution containing the anti-human TNFα specific VHH#3E (100 microgram per milliliter in 100-fold diluted PBS) was prepared. Three mice which were first deprived from drinking water for 12 hours and subsequently allowed to freely access the antibody solution during the next two hours. Afterwards the mice were sacrificed and their stomachs were dissected. Immediately the content of the stomachs was collected by flushing the stomach with 500 microliter PBS containing 1% BSA. This flushed material was subsequently used to prepare serial three-fold dilutions, starting at a 1/5 dilution from the undiluted material. One hundred microliter of these samples was transferred to individual wells of a microtiter plate coated with human TNFα. After incubation for 1 hour and following extensive washing the presence of immuno-reactive material was assessed with a polyclonal rabbit anti-VHH antiserum (R42) followed by incubation with an anti-rabbit alkaline-phosphatase conjugate. The ELISA was developed with paranitrophenyl acetate. The ELISA signals obtained after 10 minutes clearly demonstrated the presence of functional VHH#3E in the gastric flushings of these mice. By comparing to the standard curve we determined the concentration of the functional antibody fragment in the gastric flushing fluid to be 1.5, 12.6 and 8.6 microgram/ml for the three mice tested.
  • Example 9 Efficacy in an Animal Model for IBD
  • 1) Animal Model of Chronic Colitis
  • The efficacy of bivalent VHH constructs applied via various routes of administration was assessed in a DSS (dextran sodium sulfate) induced model of chronic colitis in BALB/c mice. This model was originally described by Okayasu et al. [Okayasu et al. Gastroenterology 1990; 98: 694-702] and modified by Kojouharoff et. al. [G. Kojouharoff et al. Clin. Exp. Immunol. 1997; 107: 353-8]. The animals were obtained from Charles River Laboratories, Germany, at an age of 11 weeks and kept in the animal facility until they reached a body weight between 21 and 22 g. Chronic colitis was induced in the animals by four DSS treatment cycles. Each cycle consisted of a DSS treatment interval (7 days) where DSS was provided with the drinking water at a concentration of 5% (w/v) and a recovery interval (12 days) with no DSS present in the drinking water. The last recovery period was prolonged from 12 to 21 days to provide for an inflammation status rather representing a chronic than an acute inflammation at the time of the treatment. Subsequent to the last recovery interval the mice were randomly assigned to groups of 8 mice and treatment with the VHH-constructs was started. The treatment interval was 2 weeks. One week after the end of the treatment interval the animals were sacrificed, the intestine was dissected and histologically examined. The experimental setting is shown schematically in FIG. 18.
  • 2) VHH Treatment Schedule
  • During the VHH treatment period the mice (8 animals per group) were treated daily for 14 consecutive days with bivalent VHH#3F (VHH#m3F-VHH#m3F; SEQ ID No. 76) by intra-gastric or intra-venous application of 100 μg bivalent VHH 3F. An additional group of animals was treated rectally with the bivalent VHH#3F every other day for a period of 14 days. In all treatment groups a dose of 100 μg of the bivalent VHH#3F was applied at a concentration of 1 mg/ml in a buffered solution. The negative control groups received 100 μl of PBS under otherwise identical conditions. The treatment schedule is shown in Table 11.
  • 3) Results
  • After the mice were sacrificed the body weight was determined and the colon was dissected. The length of the dissected colon was determined and the histology of the colon was assessed by Haematoxilin-Eosin (HE) stain (standard conditions). As compared to the negative controls (PBS treatment) the groups treated with bivalent nanobody 3F showed a prorogued colon length as well as an improved histological score [G. Kojouharoff et al. Clin. Exp. Immunol. 1997; 107: 353-8] thereby demonstrating efficacy of the treatment.
    TABLE 1
    Amino acid sequence listing of the peptides of aspects of present invention directed
    against TNF-alpha.
    SEQ
    ID
    NAME NO SEQUENCE
    VHH#1A
    1 QVQLQESGGGLVQPGGSLRLSCATSGFDFSVSWMYWVRQAPGKGLEWVS
    EINTNGLITKYVDSVKGRFTISRDNAKNTLYLQMDSLIPEDTALYYCAR
    SPSGSFRGQGTQVTVSS
    VHH#7B
    2 QVQLQESGGGLVQPGGSLRLSCAASGSIFRVNAMGWYRQVPGNQREFVA
    IITSGDNLNYADAVKGRFTISTDNVKKTVYLQMNVLKPEDTAVYYCNAI
    LQTSRWSIPSNYWGQGTQVTVSS
    VHH#2B
    3 QVQLQESGGGLVQPGGSLRLSCATSGFTFSDYWMYWVRQAPGKGLEWVS
    TVNTNGLITRYADSVKGRFTISRDNAKYTLYLQMNSLKSEDTAVYYCTK
    VVPPYSDDSRTNADWGQGTQVTVSS
    VHH#3E
    4 QVQLQESGGGLVQPGGSLRLSCAASGRTFSDHSGYTYTIGWFRQAPGKE
    REFVARIYWSSGNTYYADSVKGRFAISRDIAKNTVDLTMNNLEPEDTAV
    YYCAARDGIPTSRSVESYNYWGQGTQVTVSS
    VHH#3G
    5 QVQLQDSGGGLVQAGGSLRLSCAVSGRTFSAHSVYTMGWFRQAPGKERE
    FVARIYWSSANTYYADSVKGRFTISRDNAKNTVDLLMNSLKPEDTAVYY
    CAARDGIPTSRTVGSYNYWGQGTQVTVSS
    VHH#10A 6 QVQLQESGGGLVQPGGSLRLSCAASGSIFRVNAMGWYRQVPGNQREFVA
    IITSSDTNDTTNYADAVKGRFTISTDNVKKTVYLQMNVLKPEDTAVYYC
    NAVLQTSRWSIPSNYWGQGTQVTVSS
    VHH#2G 7 QVQLQDSGGGLVQAGGSLRLSCTTSGRTISVYAMGWFRQAPGKEREFVA
    SISGSGAITPYADSVKGRFTISRDNAKNTVYLQMNSLNPEDTAVYYCAA
    SRYARYRDVHAYDYWGQGTQVTVSS
    VHH#1F 8 QVQLQDSGGGLVQAGGSLRLSCAASTRTFSRYVVGWFRQAPGKEREFVA
    TISWNGEHTYYADSVKGRYTISRDNAKNTVYLQMGSLKPEDTAVYYCAA
    RSFWGYNVEQRDFGSWGQGTPVTVSS
    VHH#9C 9 QVQLQESGGGLVQPGGSLRLSCAASGSIFRVNAMGWYRQVPGNQREFVA
    IITNDTTNYADAVKGRFTISTDNVKKTVYLQMNVLKPEDTAVYYCNTVL
    QTSRWNIPTNYWGQGTQVTVSS
    VHH#11E 10 QVQLQESGGGLVQPGGSLRLSCAASGSIFRVNAMGWYRQVPGNQREFVA
    IISGDTTNYADAVKGRFTISTDNVKKTVYLQMNVLESEDTAVYYCNAVL
    QTSRWSIPSNYWGQGTQVTVSS
    VHH#10C 11 QVQLQDSGGGLVQPGGSLRLACVASGSIFSIDVMGWYRQAPGQQRELVA
    TITNSWTTNYADSVKGRFTISRDNAKNVVYLQMNSLKLEDTAVYYCNAR
    RWYQPEAWGQGTQVTVSS
    VHH#4B 12 QVQLQDSGGGLVQPGGSLRLSCAASGFTFSTHWMYWVRQAPGKGLEWVS
    TINTNGLITDYIHSVKGRFTISRDNAKNTLYLQMNSLKSEDTAVYYCAL
    NQAGLSRGQGTQVTVSS
    VHH#10D 13 QVQLQESGGGLVQAGGSLRLSCAASRRTFSGYAMGWFRQAPGKEREFVA
    VVSGTGTIAYYADSVKGRFTISRDNAENTVYLQMNSLKPEDTGLYYCAV
    GPSSSRWYYRGASLVDYWGKGTLVTVSS
    VHH#12B
    14 QVQLQESGGGLVQPGGSLRLSCAASGFEFENHWMYWVRQAPGKGLEWVS
    TVNTNGLITRYADSVKGRFTISRDNAKYTLYLQMNSLKSEDTAVYYCTK
    VLPPYSDDSRTNADWGQGTQVTVSS
    VHH#m9A 79 EVQLVESGGGLVQAGGSLRLSCAASGGTLSSYITGWFRQAPGKEREFVG
    AVSWSSSTIVYADSVEGRFTISRDNHQNTVYLQMDSLKPEDTAVYYCAA
    RPYQKYNWASASYNVWGQGTQVTVSS
    VHH#m9E 15 EVQLVESGGGLVQAGGSLRLSCAASGGTLSSYITGWFRQAPGKEREFVG
    AVSWSSSTIVYADSVEGRFTISRDNHQNTVYLQMDSLKPEDTAVYYCAA
    RPYQKYNWASASYNVWGQGTQVTVSS
    VHH#m3F 16 QVQLQDSGGGLVQAGGSLRLSCAASGGTFSSIIMAWFRQAPGKEREFVG
    AVSWSGGTTVYADSVLGRFEISRDSARKSVYLQMNSLKPEDTAVYYCAA
    RPYQKYNWASASYNVWGQGTQVTVSS
    VHH#m4B 80 QVQLQDSGGGLVQAGGSLRLSCGVSGLSFSGYTMGWFRQAPGKEREFAA
    AIGWNSGTTEYRNSVKGRFTISRDNAKNTVYLQMNSLKPEDTAVYYCAA
    SPKYMTAYERSYDFWGQGTQVTVSS
    VHH#8-29 81 QVQLVESGGGLVQPGGSLRLSCAASGFAFGDSWMYWVRQAPGKGLEWVS
    EINTNGLITKYKDSVTGRFTISRDNAKNTLHLEMNRLKPEDTALYYCAR
    DPSGKLRGPGTQVTVSS
    VHH#8-41 82 QVQLVESGGGLVQPGGPLRLSCAASGFAFGDSWMYWVRQAPGKGLEWVS
    EINTNGLITKYKDSVTGRFTISRDNAKNTLHLEMNRLKPEDTALYYCAR
    DPSGKLRGPGTQVTVSS
    VHH#8-42 83 QVQLVESGGGLVQPGGSLRLSCAASGFAFGDSWMYWVRQAPGKGLEWVS
    EINTNGLITKYKDSVTGRFTISRDNAKNTLHLEMNRLKPEDTALYYCAR
    DPSGKLRGPGTQVTVSS
    VHH#8-44 84 QVQLVESGGGLVQPGGSLRLSCAASGFTFSDHWMYWVRQAPGKGLEWVS
    TINTNGLITNYIHSVKGRFTISRDNAKNTLYLQMNSLKSEDTAVYYCAL
    NQAGLSRGQGTQVTVSS
  • TABLE 2
    List of mutagenesis reactions, mutagenic primers and templates used for
    mutagenesis of VHH#12B
    Mutation Template Primer sequence
    A74S + Y76N + Wild type 5′-AGA GAC AAC TCC AAG AAC ACG CTG TAT
    K83R + P84A CTG CAA ATG AAC AGC CTG AGA GCT GAG GAC
    ACG-3′
    Arg Asp Asn Ser Lys Asn Thr Leu Tyr
    Leu Gln Met Asn Ser Leu Arg Ala Glu Asp
    Thr
    Q1E + Q5L + A74S + A74S + Y76N + 5′-C ATG GCT GAG GTG CAG CTG CTC GAG TCT
    Y76N + K83R + P84A K83R + P84A GG-3′
    Met Ala Glu Val Gln Leu Leu Glu Ser
    Q1E + Q5L + A74S + Y76N + Q1E + Q5L + A74S + 5′-G GAC ACG GCC GTC TAT TAC TGT GCA AAA
    K83R + P84A + T93A Y76N + K83R + P84A GTA CTT C-3′
    Asp Thr Ala Val Tyr Tyr Cys Ala Lys
    Val Leu
  • TABLE 3
    List of mutagenesis reactions, mutagenic primers and templates used for
    mutagenesis of VHH#3E
    Mutation Template Primer sequence
    F37V Wild type 5′-ACC TAT ACC ATT GGC TGG GTC CGC CAG GCT-3′
    Thr Tyr Thr Ile Gly Trp Val Arg Gln Ala
    E44G Wild type 5′-CGC CAG GCT CCA GGG AAG GGG CGT GAG TTT-3′
    Arg Gln Ala Pro Gly Lys Gly Arg Glu Phe
    R45L Wild type 5′-A GGG AAG GAG CTT GAG TTT GTA GCG CGT AT-3′
    Gly Lys Glu Leu Glu Phe Val Ala Arg
    F47W Wild type 5′-A GGG AAG GAG CGT GAG TGG GTA GCG CGT AT-3′
    Gly Lys Glu Arg Glu Trp Val Ala Arg
  • TABLE 4
    Overview of humanized and wild type anti-TNF-alpha VHH
    SEQ ID Name Sequence
    17 VHH#12B QVQLQESGGGLVQPGGSLRLSCAASGFEFENHWMYWVRQAPGKGLEW
    A74S + Y76N + K83R + P84A VSTVNTNGLITRYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVY
    YCTKVLPPYSDDSRTNADWGQGTQVTVSS
    18 VHH#12B EVQLLESGGGLVQPGGSLRLSCAASGFEFENHWMYWVRQAPGKGLEW
    Q1E + Q5L + A74S + Y76N + K83R + P84A VSTVNTNGLITRYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVY
    YCTKVLPPYSDDSRTNADWGQGTQVTVSS
    19 VHH#12B EVQLLESGGGLVQPGGSLRLSCAASGFEFENHWMYWVRQAPGKGLEW
    Q1E + Q5L + A74S + Y76N + VSTVNTNGLITRYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVY
    K83R + P84A + T93A YCAKVLPPYSDDSRTNADWGQGTQVTVSS
    20 VHH#12B QVQLQESGGGLVQPGGSLRLSCAASGFEFENHWMYWVRQAPGKGLEW
    Wild type VSTVNTNGLITRYADSVKGRFTISRDNAKYTLYLQMNSLKSEDTAVY
    YCTKVLPPYSDDSRTNADWGQGTQVTVSS
    21 VHH#3E QVQLQESGGGLVQPGGSLRLSCAASGRTFSDHSGYTYTIGWVRQAPG
    F37V KEREFVARIYWSSGNTYYADSVKGRFAISRDIAKNTVDLTMNNLEPE
    DTAVYYCAARDGIPTSRSVESYNYWGQGTQVTVSS
    22 VHH#3E QVQLQESGGGLVQPGGSLRLSCAASGRTFSDHSGYTYTIGWFRQAPG
    E44G KGREFVARIYWSSGNTYYADSVKGRFAISRDIAKNTVDLTMNNLEPE
    DTAVYYCAARDGIPTSRSVESYNYWG
    QGTQVTVSS
    23 VHH#3E QVQLQESGGGLVQPGGSLRLSCAASGRTFSDHSGYTYTIGWFRQAPG
    R45L KELEFVARIYWSSGNTYYADSVKGRFAISRDIAKNTVDLTMNNLEPE
    DTAVYYCAARDGIPTSRSVESYNYWGQGTQVTVSS
    24 VHH#3E QVQLQESGGGLVQPGGSLRLSCAASGRTFSDHSGYTYTIGWFRQAPG
    F47W KEREWVARIYWSSGNTYYADSVKGRFAISRDIAKNTVDLTMNNLEPE
    DTAVYYCAARDGIPTSRSVESYNYWGQGTQVTVSS
    25 VHH#3E QVQLQESGGGLVQPGGSLRLSCAASGRTFSDHSGYTYTIGWFRQAPG
    Wild type KEREFVARIYWSSGNTYYADSVKGRFAISRDIAKNTVDLTMNNLEPE
    DTAVYYCAARDGIPTSRSVESYNYWGQGTQVTVSS
  • TABLE 5
    Anti-mouse serum albumin, and anti-mouse serum albumin + anti TNF-alpha VHH
    Name SEQ ID Sequence
    Anti-mouse serum albumin
    MSA21 26 QVQLQESGGGLVQPGGSLRLSCEASGFTFSRFGMTWVRQAPGKGVEWV
    SGISSLGDSTLYADSVKGRFTISRDNAKNTLYLQMNSLKPEDTAVYYC
    TIGGSLNPGGQGTQVTVSS
    MSA24 27 QVQLQESGGGLVQPGNSLRLSCAASGFTFRNFGMSWVRQAPGKEPEWV
    SSISGSGSNTIYADSVKDRFTISRDNAKSTLYLQMNSLKPEDTAVYYC
    TIGGSLSRSSQGTQVTVSS
    MSA210 28 QVQLQESGGGLVQPGGSLRLTCTASGFTFSSFGMSWVRQAPGKGLEWV
    SAISSDSGTKNYADSVKGRFTISRDNAKKMLFLQMNSLRPEDTAVYYC
    VIGRGSPSSQGTQVTVSS
    MSA212 29 QVQLQESGGGLVQPGGSLRLTCTASGFTFRSFGMSWVRQAPGKGLEWV
    SAISADGSDKRYADSVKGRFTISRDNGKKMLTLDMNSLKPEDTAVYYC
    VIGRGSPASQGTQVTVSS
    MSAcl6 85 AVQLVESGGGLVQAGDSLRLSCVVSGTTFSSAAMGWFRQAPGKEREFV
    GAIKWSGTSTYYTDSVKGRFTISRDNVKNTVYLQMNNLKPEDTGVYTC
    AADRDRYRDRMGPMTTTDFRFWGQGTQVTVSS
    MSAcl12 86 QVKLEESGGGLVQTGGSLRLSCAASGRTFSSFAMGWFRQAPGREREFV
    ASIGSSGITTNYADSVKGRFTISRDNAKNTVYLQMNSLKPEDTGLCYC
    AVNRYGIPYRSGTQYQNWGQGTQVTVSS
    MSAcl10 87 EVQLEESGGGLVQPGGSLRLSCAASGLTFNDYAMGWYRQAPGKERDMV
    ATISIGGRTYYADSVKGRFTISRDNAKNTVYLQMNSLKPEDTAIYYCV
    AHRQTVVRGPYLLWGQGTQVTVSS
    MSAcl14 88 QVQLVESGGKLVQAGGSLRLSCAASGRTFSNYAMGWFRQAPGKEREFV
    AGSGRSNSYNYYSDSVKGRFTISRDNAKNTVYLQMNSLKPEDTAVYYC
    AASTNLWPRDRNLYAYWGQGTQVTVSS
    MSAcl16 89 EVQLVESGGGLVQAGDSLRLSCAASGRSLGIYRMGWFRQVPGKEREFV
    AAISWSGGTTRYLDSVKGRFTISRDSTKNAVYLQMNSLKPEDTAVYYC
    AVDSSGRLYWTLSTSYDYWGQGTQVTVSS
    MSAcl19 90 QVQLVEFGGGLVQAGDSLRLSCAASGRSLGIYKMAWFRQVPGKEREFV
    AAISWSGGTTRYIDSVKGRFTLSRDNTKNMVYLQMNSLKPDDTAVYYC
    AVDSSGRLYWTLSTSYDYWGQGTQVTVSS
    MSAcl5 91 EVQLVESGGGLVQAGGSLSLSCAASGRTFSPYTMGWFRQAPGKEREFL
    AGVTWSGSSTFYGDSVKGRFTASRDSAKNTVTLEMNSLNPEDTAVYYC
    AAAYGGGLYRDPRSYDYWGRGTQVTVSS
    MScl11 92 AVQLVESGGGLVQAGGSLRLSCAASGFTLDAWPIAWFRQAPGKEREGV
    SCIRDGTTYYADSVKGRFTISSDNANNTVYLQTNSLKPEDTAVYYCAA
    PSGPATGSSHTFGIYWNLRDDYDNWGQGTQVTVSS
    MSAc115 93 EVQLVESGGGLVQAGGSLRLSCAASGFTFDHYTIGWFRQVPGKEREGV
    SCISSSDGSTYYADSVKGRFTISSDNAKNTVYLQMNTLEPDDTAVYYC
    AAGGLLLRVEELQASDYDYWGQGIQVTVSS
    MSAc18 94 AVQLVDSGGGLVQPGGSLRLSCTASGFTLDYYAIGWFRQAPGKEREGV
    ACISNSDGSTYYGDSVKGRFTISRDNAKTTVYLQMNSLKPEDTAVYYC
    ATADRHYSASHHPFADFAFNSWGQGTQVTVSS
    MSAc17 95 EVQLVESGGGLVQAGGSLRLSCAAYGLTFWRAAMAWFRRAPGKERELV
    VARNWGDGSTRYADSVKGRFTISRDNAKNTVYLQMNSLKPEDTAVYYC
    AAVRTYGSATYDIWGQGTQVTVSS
    MSAc120 96 EVQLVESGGGLVQDGGSLRLSCIFSGRTFANYAMGWFRQAPGKEREFV
    AAINRNGGTTNYADALKGRFTISRDNTKNTAFLQMNSLKPDDTAVYYC
    AAREWPFSTIPSGWRYWGQGTQVTVSS
    MSAc14 97 DVQLVESGGGWVQPGGSLRLSCAASGPTASSHAIGWFRQAPGKEREFV
    VGINRGGVTRDYADSVKGRFAVSRDNVKNTVYLQMNRLKPEDSAIYIC
    AARPEYSFTAMSKGDMDYWGKGTLVTVSS
    Anti-mouse serum albumin/anti TNF-alpha
    MSA21/ 30 QVQLQESGGGLVQPGGSLRLSCEASGFTFSRFGMTWVRQAPGKGVEWV
    VHH#3E SGISSLGDSTLYADSVKGRFTISRDNAKNTLYLQMNSLKPEDTAVYYC
    TIGGSLNPGGQGTQVTVSSEPKTPKPQPAAAQVQLQESGGGLVQPGGSLRL
    SCAASGRTFSDHSGYTYTIGWFRQAPGKEREFVARIYWSSGNTYYADSVKGRF
    AISRDIAKNTVDLTMNNLEPEDTAVYYCAARDGIPTSRSVESYNYWGQGTQVT
    VSS
    MSA24/ 31 QVQLQESGGGLVQPGNSLRLSCAASGFTFRNFGMSWVRQAPGKEPEWV
    VHH#3E SSISGSGSNTIYADSVKDRFTISRDNAKSTLYLQMNSLKPEDTAVYYC
    TIGGSLSRSSQGTQVTVSSEPKTPKPQPAAAQVQLQESGGGLVQPGGSLRL
    SCAASGRTFSDHSGYTYTIGWFRQAPGKEREFVARIYWSSGNTYYADSVKGRF
    AISRDIAKNTVDLTMNNLEPEDTAVYYCAARDGIPTSRSVESYNYWGQGTQVT
    VSS
    MSA210/ 32 QVQLQESGGGLVQPGGSLRLTCTASGFTFSSFGMSWVRQAPGKGLEWV
    VHH#3E SAISSDSGTKNYADSVKGRFTISRDNAKKMLFLQMNSLRPEDTAVYYC
    VIGRGSPSSQGTQVTVSSEPKTPKPQPAAAQVQLQESGGGLVQPGGSLRLS
    CAASGRTFSDHSGYTYTIGWFRQAPGKEREFVARIYWSSGNTYYADSVKGRFA
    ISRDIAKNTVDLTMNNLEPEDTAVYYCAARDGIPTSRSVESYNYWGQGTQVTV
    SS
    MSA212/ 33 QVQLQESGGGLVQPGGSLRLTCTASGFTFRSFGMSWVRQAPGKGLEWV
    VHH#3E SAISADGSDKRYADSVKGRFTISRDNGKKMLTLDMNSLKPEDTAVYYC
    VIGRGSPASQGTQVTVSSEPKTPKPQPAAAQVQLQESGGGLVQPGGSLRLS
    CAASGRTFSDHSGYTYTIGWFRQAPGKEREFVARIYWSSGNTYYADSVKGRFA
    ISRDIAKNTVDLTMNNLEPEDTAVYYCAARDGIPTSRSVESYNYWGQGTQVTV
    SS
    MSA21/ 34 QVQLQESGGGLVQPGGSLRLSCEASGFTFSRFGMTWVRQAPGKGVEWV
    MSA21/ SGISSLGDSTLYADSVKGRFTISRDNAKNTLYLQMNSLKPEDTAVYYC
    VHH#3E TIGGSLNPGGQGTQVTVSSEPKTPKPQPAAAQVQLQESGGGLVQPGGS
    LRLSCEASGFTFSRFGMTWVRQAPGKGVEWVSGISSLGDSTLYADSVK
    GRFTISRDNAKNTLYLQMNSLKPEDTAVYYCTIGGSLNPGGQGTQVTV
    SSEPKTPKPQPAAAQVQLQESGGGLVQPGGSLRLSCAASGRTFSDHSG
    YTYTIGWFRQAPGKEREFVARIYWSSGNTYYADSVKGRFAISRDIAKN
    TVDLTMNNLEPEDTAVYYCAARDGIPTSRSVESYNYWGQGTQVTVSS
    MSA210/ 35 QVQLQESGGGLVQPGGSLRLTCTASGFTFSSFGMSWVRQAPGKGLEWV
    VHH#1A SAISSDSGTKNYADSVKGRFTISRDNAKKMLFLQMNSLRPEDTAVYYC
    VIGRGSPSSQGTQVTVSSEPKTPKPQPAAAQVQLQESGGGLVQPGGSL
    RLSCATSGFDFSVSWMYWVRQAPGKGLEWVSEINTNGLITKYVDSVKG
    RFTISRDNAKNTLYLQMDSLIPEDTALYYCARSPSGSFRGQGTQVTVSS
    MSA210/ 36 QVQLQESGGGLVQPGGSLRLTCTASGFTFSSFGMSWVRQAPGKGLEWV
    VHH#7B SAISSDSGTKNYADSVKGRFTISRDNAKKMLFLQMNSLRPEDTAVYYC
    VIGRGSPSSQGTQVTVSSEPKTPKPQPAAAQVQLQESGGGLVQPGGSL
    RLSCAASGSIFRVNAMGWYRQVPGNQREFVAIITSGDNLNYADAVKGR
    FTISTDNVKKTVYLQMNVLKPEDTAVYYCNAILQTSRWSIPSNYWGQG
    TQVTVSS
    MSA210/ 37 QVQLQESGGGLVQPGGSLRLTCTASGFTFSSFGMSWVRQAPGKGLEWV
    VHH#2B SAISSDSGTKNYADSVKGRFTISRDNAKKMLFLQMNSLRPEDTAVYYC
    VIGRGSPSSQGTQVTVSSEPKTPKPQPAAAQVQLQESGGGLVQPGGSL
    RLSCATSGFTFSDYWMYWVRQAPGKGLEWVSTVNTNGLITRYADSVKG
    RFTISRDNAKYTLYLQMNSLKSEDTAVYYCTKVVPPYSDDSRTNADWG
    QGTQVTVSS
    MSA210/ 38 QVQLQESGGGLVQPGGSLRLTCTASGFTFSSFGMSWVRQAPGKGLEWV
    VHH#3E SAISSDSGTKNYADSVKGRFTISRDNAKKMLFLQMNSLRPEDTAVYYC
    VIGRGSPSSQGTQVTVSSEPKTPKPQPAAAQVQLQESGGGLVQPGGSL
    RLSCAASGRTFSDHSGYTYTIGWFRQAPGKEREFVARIYWSSGNTYYA
    DSVKGRFAISRDIAKNTVDLTMNNLEPEDTAVYYCAARDGIPTSRSVE
    SYNYWGQGTQVTVSS
    MSA210/ 39 QVQLQESGGGLVQPGGSLRLTCTASGFTFSSFGMSWVRQAPGKGLEWV
    VHH#3G SAISSDSGTKNYADSVKGRFTISRDNAKKMLFLQMNSLRPEDTAVYYC
    VIGRGSPSSQGTQVTVSSEPKTPKPQPAAAQVQLQDSGGGLVQAGGSL
    RLSCAVSGRTFSAHSVYTMGWFRQAPGKEREFVARIYWSSANTYYADS
    VKGRFTISRDNAKNTVDLLMNSLKPEDTAVYYCAARDGIPTSRTVGSY
    NYWGQGTQVTVSS
    MSA21/ 40 QVQLQESGGGLVQPGGSLRLSCEASGFTFSRFGMTWVRQAPGKGVEWV
    VHH#12B SGISSLGDSTLYADSVKGRFTISRDNAKNTLYLQMNSLKPEDTAVYYC
    TIGGSLNPGGQGTQVTVSSEPKTPKPQPAAAQVQLQESGGGLVQPGGS
    LRLSCAASGFEFENHWMYWVRQAPGKGLEWVSTVNTNGLITRYADSVK
    GRFTISRDNAKYTLYLQMNSLKSEDTAVYYCTKVLPPYSDDSRTNADW
    GQGTQVTVSS
    MSA24/ 41 QVQLQESGGGLVQPGNSLRLSCAASGFTFRNFGMSWVRQAPGKEPEWV
    VHH#12B SSISGSGSNTIYADSVKDRFTISRDNAKSTLYLQMNSLKPEDTAVYYC
    TIGGSLSRSSQGTQVTVSSEPKTPKPQPAAAQVQLQESGGGLVQPGGS
    LRLSCAASGFEFENHWMYWVRQAPGKGLEWVSTVNTNGLITRYADSVK
    GRFTISRDNAKYTLYLQMNSLKSEDTAVYYCTKVLPPYSDDSRTNADW
    GQGTQVTVSS
    MSA210/ 42 QVQLQESGGGLVQPGGSLRLTCTASGFTFSSFGMSWVRQAPGKGLEWV
    VHH#12B SAISSDSGTKNYADSVKGRFTISRDNAKKMLFLQMNSLRPEDTAVYYC
    VIGRGSPSSQGTQVTVSSEPKTPKPQPAAAQVQLQESGGGLVQPGGSL
    RLSCAASGFEFENHWMYWVRQAPGKGLEWVSTVNTNGLITRYADSVKG
    RFTISRDNAKYTLYLQMNSLKSEDTAVYYCTKVLPPYSDDSRTNADWG
    QGTQVTVSS
    MSA212/ 43 QVQLQESGGGLVQPGGSLRLTCTASGFTFRSFGMSWVRQAPGKGLEWV
    VHH#12B SAISADGSDKRYADSVKGRFTISRDNGKKMLTLDMNSLKPEDTAVYYC
    VIGRGSPASQGTQVTVSSEPKTPKPQPAAAQVQLQESGGGLVQPGGSL
    RLSCAASGFEFENHWMYWVRQAPGKGLEWVSTVNTNGLITRYADSVKG
    RFTISRDNAKYTLYLQMNSLKSEDTAVYYCTKVLPPYSDDSRTNADWG
    QGTQVTVSS
  • TABLE 6
    Amino acid sequence listing of VHH's directed against human IFN-gamma.
    Seq. Seq.
    Family Name Id Sequence
    1 MP3D2SRA 44 QVQLQDSGGGTVQAGGSLRLSCAASGRTFSDYAVGWFRQA
    PGKEREFVARILWTGASRSYANSVDGRFTVSTDNAKNTVY
    LQMNSLKPEDTAIYYCAALPSNIITTDYLRVYYWGQGTQV
    TVSS
    1 MP3A3SR 45 QVQLQDSGGGTVQAGGSLRLSCAASGRTFSNYAVGWFRQA
    PGKEREFVARIKWSGGSRSYANSVDGRFTVSTDNAKNTVY
    LQMNSLKPEDTAIYYCA?LPSNIITTDYLRVYYWGQGTQV
    TVSS
    2 MP3C5SR 46 QVQLQESGGGLVQAGGSLRLSCAAAGISGSVFSRTPMGWY
    RQAPGKQRELVAGILTSGATSYAESVKGRFTISRDNAKNT
    VYLQMNSLSPEDTAEYYCNTYPTWVLSWGQGTQVTVSS
    2 MP3C1SR 47 QVQLQDSGGGLVQAGGSLRLSCAAAGISGSVFSRTPMGWY
    RQAPGKQRELVAGILSSGATVYAESVKGRFTISRDNAKNT
    VYLQMNSLSPEDTAEYYCNTYPTWVLSWGQGTQVTVSS
    2 MP3G8SR 48 QVQLQESGGGLVQAGGSLRLSCAAAGISGSVFSRTPMGWY
    RQAPGKQRELVAGILSSGATAYAESVKGRFTISRDNAKNT
    VYLQMNSLSPEDTAEYYCNTYPTWVLSWGQGTQVTVSS
    3 MP3D2BR 49 QVQLQESGGGLVQPGESLRLSCAASRGIFRFNAGGWYRQA
    PGKQRELVAFIGVDNTTRYIDSVKGRFTISRDNAKTTVYL
    QMNSLQPEDTAVYYCNKVPYIDWGQGTQVTVSS
    4 MP3H6SRA 50 QVQLQESGGGLVQAGGSLRLSCAASGRTFSTYNMGWFRQA
    PGKEREFVAGISWNGGSIYYTSSVEGRFTISRDNAENTVY
    LQMNSLKPEDTGVYYCASKGRPYGVPSPRQGDYDYWGQGT
    QVTVSS
    4 MP3B4SRA 51 QVQLQESGGGLVQAGGSLRLSCAASGRTFSTYNMGWFRQA
    PGKEREFVAGISWNGGSIYYTSSVEGRFTISRDNAENTVY
    LQMNSLKPEDTGVYYCASKGRPYGVPSPRQGDYDYWGQGT
    QVTVSS
    4 MP4E4BR 52 QVQLQESGGGLVQAGGSLRLSCAASGRTFSIYNMGWFRQA
    PGKEREFVAAISWNGGSIYYTSSVEGRFTISRDNAINTVY
    LQMNSLKPEDTGVYYCASKGRPYGVPSPRQGEYDYWGQGT
    QVTVSS
    4 MP4H8SR 53 QVQLQESGGGLVQAGGSLRLSCAASGRTFNIYNMGWFRQA
    PGKERDFVAAISWNGGSIYYTSSVEGRFTISRDNAENTVY
    LQMNSLKPEDTGVYYCASKGRPYGVPSPRQGDYDYWGQGT
    QVTVSS
    5 MP2F6SR 54 QVKLEESGGGLVQAGGSLRLSCAASGRTFNNYNMGWFRQA
    PGKEREFVAAISWNGGSTYYDDSVKGRFTISRDNANNLVY
    LQMNSLNFEDTAVYYCACAANPYGIPQYRENRYDFWGQGT
    QVTVSS
    5 MP3D1BR 55 QVQLQESGGGLVQAGGSLRLSCAASGRTFDNYNMGWFRQA
    PGKEREFVAAISWNGGSTYYDDSVKGRFTISRDNFQKLVY
    LQMNSLKLEDTAVYYCACAANPYGIPQYRENRYDFWGQGT
    QVTVSS
    6 MP2B5BR 56 QVQLVESGGRLVQAGGSLRLSCIASGRTISDYAAGWFRQA
    PGKEREFLASVTWGFGSTSYADSVKGRFTISRDKAKDTVY
    LQMNTLEPDDTSVYYCASSPRYCAGYRCYVTASEFDSWGQ
    GTQVTVSS
    6 MP2C1BR 57 QVKLEESGGRLVQAGGSLRLSCIASGRTISDYAAGWFRQA
    PGKEREFLASVSWGFGSTYYADSVKGRFTISRDTAKDTVY
    LQMNTLEPDDTSVYYCASSPRYCAGYRCYATASEFDSWGQ
    GTQVTVSS
    6 MP4A12SR 58 QVQLQESGGRLVQAGGSLRLSCIASGRTISDYAAGWFRQA
    PGKEREFLASVTWGFGSTYYADSVKGRFTISRDKAKDTVY
    LQMNTLEPDDTSAYYCASSPRYCAGYRCYVTASEFDSWGP
    GTQVTVSS
    7 MP3F4SRA 59 QVQLQDSGGGLVQAGDSLRLSCAASGRSFSSYGMGWFRQA
    PGKEHEFVAGIWRSGVSLYYTDSVKGRFTISRDDAKMTVS
    LQMNSLKPEDTAVYYCAAEATFPTWSRGRFADYDYRGQGT
    QVTVSS
    7 MP3D3BR 60 QVQLQESGGGLVQAGDSLRLSCTASGRSFSSYGMGWFRQA
    PGKDHEFVAGIWRSGVSLYYADSVKGRFTISRDDAKMTVS
    LQMNGLKPEDTAVYYCAAEATFPTWNRGTFADYDYRGQGT
    QVTVSS
    7 MP3E5BR 61 QVQLQESGGGLVQAGDSLRLSCAASGRSFSSYGMGWFRQA
    PGKEHEFVAGIWRSGVSLYYADSVKGRFTISRDDAKMTVS
    LQMNGLKPEDTAVYYCAAEATFPTWNRGSFADYDYRGQGT
    QVTVSS
    7 MP3C7SRA 62 QVQLQESGGGLVQAGDSLRLSCAASGRSFSSYGMGWFRQA
    PGKEHEFVAGIWRSGVSLYYADSVKGRFTISRDDAKMTVS
    LQMNSLKPEDTAVYYCAAEATFPTWNRGRFADYDYSGQGT
    QVTVSS
    8 MP2F1BR 63 AVQLVESGGGLVQTGDSLRLSCVASGGTFSRYAMGWFRQA
    PGKEREFVARIGYSGRSISYATSVEGRFAISRDNAKNTVY
    LQMNSLKPEDTAVYYCASLVSGTLYQADYWGQGTQVTVSS
    8 MP2C5BR 64 QVQLVESGGGLVQTGDSLRLSCVASGGTFSRYAMGWFRQP
    PGKERDFVARIGYSGQSISYATSVEGRFAISRDNAKNTVY
    LQMNSLKPEDTAVYYCASLVSGTLYKPNYWGQGTQVTVSS
    9 MP2C10BR 65 QVKLEESGGGLVQAGGSLRLSCAASGLTYTVGWFRQAPGK
    EREFVAAISWSGGSALYADSVKGRFTISRDNAKNTVYLQM
    GSLEPEDTAYYSCAAPGTRYYGSNQVNYNYWGQGTQVTVSS
    9 MP2G5SR 66 QVKLEESGGGLVQAGDSLRLSCAASGLTYTVGWFRQAPGK
    EREFVAAIDWSGGSALYADSVKGRFTISRDNTKNTVYLQM
    GSLEPEDTAVYWCAAPGTRYHGRNQVNYNYWGQGTQVTVSS
    10 MP3B1SRA 67 QVQLQESGGGLVQPGGSLRLSCAASGFTSSNYAMSWVRQA
    PGKGLEWVSSINSRTGSITYADSVKGRFTITLDNAKNTLY
    LQMNSLKPEDTAVYYCASRVDDRVSRGQGTQVTVSS
    11 MP2F10SR 68 QVQLVESGGGLVQAGGSLRLSCAASGRTISSFRMGWFRRA
    PGEEREFVAFVRSNGTSTYYADSVEGRFTITRDNAKNTVY
    LRMDSLKPEDTAVYYCAAATRDYGGSFDYWGQGTQVTVSS
    11 MP3A7SRA 69 QVQLQDSGGGLVQAGGSLRLSCAASGRTFSSFRMGWFRRA
    PGEEREFVAFVRSNGTSTYYADSVEGRFTITRDNAKNTVY
    LRMDSLKPEDTAVYYCAAATRDYGGSFDYWGQGTQVIVSS
    12 MP4C10SR 70 QVQLQESGGGLVQPGGSLRLSCAASGFTVSNYAMSWVRQP
    PGKGIEWVSSINNRNDHITYADSVKGRFTIARDNANNILY
    LQMNSLKPEDTAVYYCASRVDDRVSRGQGTQVTVSS
    13 MP4D5BR 71 QVQLQDSGGGLVQPGGSLRLSCAASGRTFSSYGMAWFRQA
    PGKERELVVAINRSGGATSYATSVRGRFTISRDNAKNTMY
    LQMNSLNPEDTAVYYCAARDPTRTYSSYFEYTYWGQGTQV
    TVSS
    14 MP3F1SRA 72 QVQLQESGGGLVQAGGSLTLSCVASGRTISDYAVGWFRQA
    PGKEREFVASISWGGGFTAFADSMKGRFTISRDNAKNTVY
    LQTHTLEPDDTSVYYCASSRRYCTGYRCYATASEFDSWGQ
    GTQVTVSS
  • TABLE 7
    Sequences of bivalent (BIV 3E, BIV#m3F), trivalent (TRI3E) or tetravalent (TETRA
    3E) VHH directed against TNF-alpha.
    SEQ
    Name ID Sequence
    With linker sequence (underlined)
    BIV 3E 73 QVQLQDSGGGLVQAGGSLRLSCAASGGTFSSIIMAWFRQAPGKEREFVGAVSWSG
    GTTVYADSVLGRFEISRDSARKSVYLQMNSLKPEDTAVYYCAARPYQKYNWASAS
    YNVWGQGTQVTVSSEPKTPKPQPAAAQVQLQDSGGGLVQAGGSLRLSCAASGGTF
    SSIIMAWFRQAPGKEREFVGAVSWSGGTTVYADSVLGRFEISRDSARKSVYLQMN
    SLKPEDTAVYYCAARPYQKYNWASASYNVWGQGTQVTVSS
    TRI
    3E 74 QVQLQESGGGLVQPGGSLRLSCAASGRTFSDHSGYTYTIGWFRQAPGKEREFVAR
    IYWSSGNTYYADSVKGRFAISRDIAKNTVDLTMNNLEPEDTAVYYCAARDGIPTS
    RSVESYNYWGQGTQVTVSSEPKTPKPQPAAAQVQLQESGGGLVQPGGSLRLSCAA
    SGRTFSDHSGYTYTIGWFRQAPGKEREFVARIYWSSGNTYYADSVKGRFAISRDI
    AKNTVDLTMNNLEPEDTAVYYCAARDGIPTSRSVESYNYWGQGTQVTVSSEPKTP
    KPQPAAAQVQLQESGGGLVQPGGSLRLSCAASGRTFSDHSGYTYTIGWFRQAPGK
    EREFVARIYWSSGNTYYADSVKGRFAISRDIAKNTVDLTMNNLEPEDTAVYYCAA
    RDGIPTSRSVESYNYWGQGTQVTVSS
    TETRA 75 QVQLQESGGGLVQPGGSLRLSCAASGRTFSDHSGYTYTIGWFRQAPGKEREFVAR
    3E IYWSSGNTYYADSVKGRFAISRDIAKNTVDLTMNNLEPEDTAVYYCAARDGIPTS
    RSVESYNYWGQGTQVTVSSEPKTPKPQPAAAQVQLQESGGGLVQPGGSLRLSCAA
    SGRTFSDHSGYTYTIGWFRQAPGKEREFVARIYWSSGNTYYADSVKGRFAISRDI
    AKNTVDLTMNNLEPEDTAVYYCAARDGIPTSRSVESYNYWGQGTQVTVSSEPKTP
    KPQPAAAQVQLQESGGGLVQPGGSLRLSCAASGRTFSDHSGYTYTIGWFRQAPGK
    EREFVARIYWSSGNTYYADSVKGRFAISRDIAKNTVDLTMNNLEPEDTAVYYCAA
    RDGIPTSRSVESYNYWGQGTQVTVSSEPKTPKPQPAAAQVQLQESGGGLVQPGGS
    LRLSCAASGRTFSDHSGYTYTIGWFRQAPGKEREFVARIYWSSGNTYYADSVKGR
    FAISRDIAKNTVDLTMNNLEPEDTAVYYCAARDGIPTSRSVESYNYWGQGTQVTV
    SS
    BIV#m 76 QVQLQDSGGGLVQAGGSLRLSCAASGGTFSSIIMAWFRQAPGKEREFVGAVSWSG
    3F GTTVYADSVLGRFEISRDSARKSVYLQMNSLKPEDTAVYYCAARPYQKYNWASAS
    YNVWGQGTQVTVSSEPKTPKPQPAAAQVQLQDSGGGLVQAGGSLRLSCAASGGTF
    SSIIMAWFRQAPGKEREFVGAVSWSGGTTVYADSVLGRFEISRDSARKSVYLQMN
    SLKPEDTAVYYCAARPYQKYNWASASYNVWGQGTQVTVSS
    Without linker sequence
    BIV#
    3 77 QVQLQDSGGGLVQAGGSLRLSCAASGGTFSSIIMAWFRQAPGKEREFVGAVSWSG
    Edir GTTVYADSVLGRFEISRDSARKSVYLQMNSLKPEDTAVYYCAARPYQKYNWASAS
    YNVWGQGTQVTVSSQVQLQDSGGGLVQAGGSLRLSCAASGGTFSSIIMAWFRQAP
    GKEREFVGAVSWSGGTTVYADSVLGRFEISRDSARKSVYLQMNSLKPEDTAVYYC
    AARPYQKYNWASASYNVWGQGTQVTVSS
    BIV#
    12 78 QVQLQESGGGLVQPGGSLRLSCAASGFEFENHWMYWVRQAPGKGLEWVSTVNTNG
    Bdir LITRYADSVKGRFTISRDNAKYTLYLQMNSLKSEDTAVYYCTKVLPPYSDDSRTN
    ADWGQGTQVTVSSQVQLQESGGGLVQPGGSLRLSCAASGFEFENHWMYWVRQAPG
    KGLEWVSTVNTNGLITRYADSVKGRFTISRDNAKYTLYLQMNSLKSEDTAVYYCT
    KVLPPYSDDSRTNADWGQGTQVTVSS
  • TABLE 8
    Fractional homologies between the amino acid sequences of
    anti-mouse serum albumin VHHs of the invention.
    SEQ MSA21 MSA24 MSA210 MSA212
    MSA21 1.000 0.834 0.800 0.782
    MSA24 1.000 0.782 0.791
    MSA210 1.000 0.903
    MSA212 1.000
  • TABLE 9
    Fractional homologies between anti-TNF-alpha VHHs of the invention
    SEQ VHH#1A VHH#7B VHH#2B VHH#3E VHH#3G VHH#10A VHH#2G VHH#1F VHH#9C VHH#11E
    VHH#1A 1.000 0.601 0.764 0.596 0.622 0.600 0.682 0.629 0.609 0.601
    VHH#7B 1.000 0.604 0.635 0.645 0.943 0.653 0.616 0.933 0.933
    VHH#2B 1.000 0.620 0.645 0.611 0.682 0.661 0.629 0.620
    VHH#3E 1.000 0.875 0.641 0.713 0.689 0.620 0.643
    VHH#3G 1.000 0.651 0.779 0.740 0.637 0.637
    VHH#10A 1.000 0.658 0.614 0.935 0.935
    VHH#2G 1.000 0.741 0.653 0.669
    VHH#1F 1.000 0.616 0.616
    VHH#9C 1.000 0.941
    VHH#11E 1.000
    VHH#10C
    VHH#4B
    VHH#10D
    VHH#12B
    VHH#9E
    VHH#3F
    SEQ VHH#10C VHH#4B VHH#10D VHH#12B VHH#9E VHH#3F
    VHH#1A 0.614 0.818 0.642 0.747 0.596 0.604
    VHH#7B 0.719 0.593 0.614 0.620 0.616 0.624
    VHH#2B 0.637 0.796 0.634 0.951 0.620 0.645
    VHH#3E 0.612 0.604 0.648 0.596 0.674 0.682
    VHH#3G 0.653 0.645 0.689 0.622 0.708 0.716
    VHH#10A 0.725 0.592 0.612 0.626 0.622 0.637
    VHH#2G 0.685 0.666 0.746 0.650 0.701 0.717
    VHH#1F 0.664 0.661 0.714 0.645 0.709 0.717
    VHH#9C 0.743 0.601 0.622 0.645 0.600 0.616
    VHH#11E 0.719 0.601 0.622 0.637 0.608 0.624
    VHH#10C 1.000 0.650 0.606 0.637 0.600 0.632
    VHH#4B 1.000 0.611 0.796 0.588 0.629
    VHH#10D 1.000 0.619 0.674 0.674
    VHH#12B 1.000 0.604 0.637
    VHH#9E 1.000 0.854
    VHH#3F 1.000
  • TABLE 10
    Percentage homologies between anti-IFN-gamma VHHs of the invention
    % Homology
    MP3D2SRA MP3A3SR MP3C5SR MP3C1SR MP3G8SR P3D2BR MP3H6SRA MP3B4SRA MP4E4BR
    MP3D2SRA X 96 66 66 66 62 71 71 71
    MP3A3SR X 66 66 66 62 72 72 72
    MP3C5SR X 97 98 73 65 65 64
    MP3C1SR X 98 72 64 64 64
    MP3G8SR X 73 65 65 64
    MP3D2BR X 63 63 63
    MP3H6SRA X 100  97
    MP3B4SRA X 97
    MP4E4BR X
    MP4H8SR
    MP2F6SR
    MP3D1BR
    MP2B5BR
    MP2C1BR
    MP4A12SR
    MP3F4SRA
    MP3D3BR
    MP3E5BR
    MP3C7SRA
    MP2F1BR
    MP2C5BR
    MP2C10BR
    MP2G5SR
    MP3B1SRA
    MP2F10SR
    MP3A7SRA
    MP4C10SR
    MP4D5BR
    MP3F1SRA
    MP6D6BR
    MP6B1BR
    MP6A8BR
    MP6B12BR
    MP6C11BR
    MP6B10BR
    % Homology
    MP4H8SR MP2F6SR MP3D1BR MP2B5BR MP2C1BR MP4A12SR MP3F4SRA MP3D3BR
    MP3D2SRA 70 68 69 65 63 64 68 66
    MP3A3SR 71 70 71 65 63 64 68 66
    MP3C5SR 63 63 63 60 58 59 64 64
    MP3C1SR 62 62 62 58 57 58 65 64
    MP3G8SR 63 63 63 59 58 59 64 64
    MP3D2BR 62 63 64 59 58 58 62 61
    MP3H6SRA 97 80 81 67 68 67 75 71
    MP3B4SRA 97 80 81 67 68 67 75 71
    MP4E4BR 97 81 82 68 69 68 73 70
    MP4H8SR X 81 81 66 66 66 72 69
    MP2F6SR X 94 65 68 64 70 67
    MP3D1BR X 65 66 65 71 69
    MP2B5BR X 95 97 63 64
    MP2C1BR X 95 63 64
    MP4A12SR X 63 64
    MP3F4SRA X 94
    MP3D3BR X
    MP3E5BR
    MP3C7SRA
    MP2F1BR
    MP2C5BR
    MP2C10BR
    MP2G5SR
    MP3B1SRA
    MP2F10SR
    MP3A7SRA
    MP4C10SR
    MP4D5BR
    MP3F1SRA
    MP6D6BR
    MP6B1BR
    MP6A8BR
    MP6B12BR
    MP6C11BR
    MP6B10BR
    % Homology
    MP3E5BR MP3C7SRA MP2F1BR MP2C5BR MP2C10BR MP2G5SR MP3B1SRA MP2F10SR
    MP3D2SRA 67 68 71 70 68 67 63 67
    MP3A3SR 67 68 72 72 69 67 64 66
    MP3C5SR 65 66 65 65 65 63 63 64
    MP3C1SR 64 65 64 63 64 62 63 64
    MP3G8SR 65 66 65 64 65 63 63 65
    MP3D2BR 62 63 64 63 63 63 64 63
    MP3H6SRA 73 75 73 71 73 71 66 75
    MP3B4SRA 73 75 73 71 73 71 66 75
    MP4E4BR 71 73 73 71 73 71 66 75
    MP4H8SR 71 72 71 71 72 71 64 73
    MP2F6SR 69 71 67 65 73 71 63 71
    MP3D1BR 71 72 67 65 70 69 63 71
    MP2B5BR 64 64 65 63 64 63 60 66
    MP2C1BR 64 64 63 61 66 65 59 66
    MP4A12SR 64 64 62 60 63 62 59 65
    MP3F4SRA 96 97 69 67 68 68 62 67
    MP3D3BR 98 96 70 68 67 67 62 67
    MP3E5BR X 98 70 68 68 69 63 68
    MP3C7SRA X 71 69 69 70 63 69
    MP2F1BR X 94 66 67 63 68
    MP2C5BR X 66 67 63 67
    MP2C10BR X 94 62 68
    MP2G5SR X 62 67
    MP3B1SRA X 66
    MP2F10SR X
    MP3A7SRA
    MP4C10SR
    MP4D5BR
    MP3F1SRA
    MP6D6BR
    MP6B1BR
    MP6A8BR
    MP6B12BR
    MP6C11BR
    MP6B10BR
    % Homology
    MP3A7SRA MP4C10SR MP4D5BR MP3F1SRA MP6D6BR
    MP3D2SRA 68 60 72 65 68
    MP3A3SR 67 60 73 65 67
    MP3C5SR 64 61 67 60 74
    MP3C1SR 65 60 67 59 73
    MP3G8SR 65 61 66 60 73
    MP3D2BR 63 63 65 58 73
    MP3H6SRA 75 63 71 69 71
    MP3B4SRA 75 63 71 69 71
    MP4E4BR 75 63 72 70 71
    MP4H8SR 73 62 70 67 69
    MP2F6SR 70 62 69 66 67
    MP3D1BR 71 62 68 66 67
    MP2B5BR 63 57 63 84 65
    MP2C1BR 63 56 61 85 65
    MP4A12SR 63 56 61 84 64
    MP3F4SRA 69 60 72 63 67
    MP3D3BR 67 60 70 64 66
    MP3E5BR 68 60 72 64 67
    MP3C7SRA 69 61 72 64 68
    MP2F1BR 67 61 70 64 68
    MP2C5BR 65 62 69 63 67
    MP2C10BR 66 59 67 66 69
    MP2G5SR 65 59 67 65 67
    MP3B1SRA 65 91 67 60 67
    MP2F10SR 97 61 67 65 71
    MP3A7SRA X 61 68 63 71
    MP4C10SR X 64 58 65
    MP4D5BR X 64 69
    MP3F1SRA X 65
    MP6D6BR X
    MP6B1BR
    MP6A8BR
    MP6B12BR
    MP6C11BR
    MP6B10BR
    % Homology
    MP6B1BR MP6A8BR MP6B12BR MP6C11BR MP6B10BR
    MP3D2SRA 67 66 67 76 70
    MP3A3SR 67 65 66 77 71
    MP3C5SR 63 60 63 70 64
    MP3C1SR 63 60 62 70 65
    MP3G8SR 63 61 63 71 64
    MP3D2BR 64 60 63 68 67
    MP3H6SRA 71 68 70 82 70
    MP3B4SRA 71 68 70 82 70
    MP4E4BR 71 68 70 80 71
    MP4H8SR 70 67 70 79 71
    MP2F6SR 69 68 67 78 69
    MP3D1BR 71 69 69 79 70
    MP2B5BR 63 63 62 70 65
    MP2C1BR 64 63 62 70 65
    MP4A12SR 63 63 62 70 65
    MP3F4SRA 68 65 65 76 71
    MP3D3BR 66 64 64 75 69
    MP3E5BR 68 65 66 77 71
    MP3C7SRA 68 66 66 78 71
    MP2F1BR 65 64 64 74 67
    MP2C5BR 64 62 63 73 67
    MP2C10BR 68 64 68 74 73
    MP2G5SR 66 64 66 73 73
    MP3B1SRA 69 68 69 69 65
    MP2F10SR 66 65 67 77 68
    MP3A7SRA 65 65 67 77 69
    MP4C10SR 64 63 66 66 63
    MP4D5BR 68 65 67 76 73
    MP3F1SRA 64 64 63 71 68
    MP6D6BR 70 65 70 77 73
    MP6B1BR X 78 81 76 71
    MP6A8BR X 75 74 66
    MP6B12BR X 73 68
    MP6C11BR X 77
    MP6B10BR X
  • TABLE 11
    Treatment schedule
    Group Animals Description Schedule
    1 8 negative control 1 daily 100 μl PBS i.p. +
    ip
    2 8 negative control 2 every other day 100 μl PBS rectal for 2
    rectal weeks
    3 8 negative control 3 daily 100 μl PBS intragastric for 14
    intragastric consecutive days
    4 8 positive control 1 5 μg i.p. for 7 consecutive days
    dexamethasone
    5 8 positive control 2 applied orally once per day for 14
    IL10 expressing I.lactis consecutive days
    6 8 bivalent VHH 3F daily 100 μg bivalent VHH 3F2
    intra-gastric intragastric on 14 consecutive days
    7 8 bivalent VHH 3F daily 100 μg bivalent VHH 3F i.p. for 14
    i.p. consecutive days
    8 8 bivalent VHH 3F 100 μg bivalent VHH 3F rectally in 100 μl
    rectally PBS every other day for two weeks

Claims (89)

1. Anti-TNF-alpha polypeptide comprising one or more single domain antibodies directed against tumor necrosis factor alpha (TNF-alpha) and an Fc domain.
2. Anti-TNF-alpha polypeptide according to claim 1, in which the Fc domain is a human Fc domain.
3. Anti-TNF-alpha polypeptide according to claim 1, in which the one or more single domain antibodies directed against tumor necrosis factor alpha (TNF-alpha) are fused to the Fc domain.
4. Anti-TNF-alpha polypeptide according to claim 1, which inhibits the interaction between TNF alpha and one or more TNF alpha receptors.
5. Anti-TNF-alpha polypeptide according to claim 1, which modifies the biological activity of TNF-alpha after binding to TNF alpha.
6. Anti-TNF-alpha polypeptide according to claim 1, which binds into the receptor-binding groove of TNF-alpha.
7. Anti-TNF-alpha polypeptide according to claim 1, in which said one or more single domain antibodies directed against TNF-alpha bind to TNF-alpha with an affinity of better than 10−6 M.
8. Anti-TNF-alpha polypeptide according to claim 1, in which said one or more single domain antibodies directed against TNF-alpha have an amino acid selected from glycine, alanine, valine, leucine, isoleucine, proline, phenylalanine, tyrosine, tryptophan, methionine, serine, threonine, asparagine and glutamine at position 45 and a tryptophan at position 103 according to the Kabat numbering.
9. Anti-TNF-alpha polypeptide according to claim 1, in which said one or more single domain antibodies directed against TNF-alpha have an arginine residue at position 103 according to the Kabat numbering.
10. Anti-TN F-alpha polypeptide according to claim 1, in which said one or more single domain antibodies directed against TNF-alpha are one or more Camelidae VHHs.
11. Anti-TNF-alpha polypeptide according to claim 10, in which said one or more single domain antibodies directed against TNF-alpha are one or more humanized Camelidae VHHS.
12. Anti-TN F polypeptide according to claim 1, wherein the one or more one single domain antibodies directed against TNF-alpha comprises a sequence represented by any of SEQ ID NOs: 1 to 16 or 79 to 84, or comprises:
a) a sequence that is more than 70% identical to the sequence of any of SEQ ID NOs: 1 to 16 or 79 to 84;
b) a functional portion of any of SEQ ID NOs: 1 to 16 or 79 to 84 that maintains the interaction with the target with affinity of 1×10−6 M or better;
c) a functional portion of any of SEQ ID NOs: 1 to 7, 23 to 31, and 62 to 65 that comprises a partial deletion of the complete amino acid sequence and still maintains the binding site(s) and protein domain(s) necessary for the binding of and interaction with the target.
13. Anti-TNF polypeptide according to claim 12, wherein the one or more one single domain antibodies directed against TNF-alpha comprises a sequence that is more than 80% identical to the sequence of any of SEQ ID NOs: 1 to 16 or 79 to 84.
14. Anti-TNF polypeptide according to claim 13, wherein the one or more one single domain antibodies directed against TNF-alpha comprises a sequence that is more than 90% identical to the sequence of any of SEQ ID NOs: 1 to 16 or 79 to 84.
15. Anti-TNF polypeptide according to claim 12, wherein the one or more one single domain antibodies directed against TNF-alpha comprises a sequence represented by any of SEQ ID NOs: 1 to 16 or 79 to 84.
16. Anti-TNF polypeptide according to claim 1, wherein the one or more one single domain antibodies directed against TNF-alpha comprises the sequence of SEQ ID NO: 4, or comprises:
a) a sequence that is more than 70% identical to the sequence of SEQ ID NO: 4;
b) a functional portion of SEQ ID NO: 4 that maintains the interaction with the target with affinity of 1×10−6 M or better;
c) a functional portion of SEQ ID NO: 4 that comprises a partial deletion of the complete amino acid sequence and still maintains the binding site(s) and protein domain(s) necessary for the binding of and interaction with the target.
17. Anti-TNF polypeptide according to claim 16, wherein the one or more one single domain antibodies directed against TNF-alpha comprises a sequence that is more than 80% identical to the sequence of SEQ ID NO:4.
18. Anti-TNF polypeptide according to claim 17, wherein the one or more one single domain antibodies directed against TNF-alpha comprises a sequence that is more than 90% identical to the sequence of SEQ ID NO:4.
19. Anti-TNF polypeptide according to claim 16, wherein the one or more one single domain antibodies directed against TNF-alpha comprises the sequence of SEQ ID NO:4.
20. Method for treating and/or preventing and/or alleviating disorders relating to inflammatory processes, comprising administering to a subject a therapeutically effective amount of an anti-TNF polypeptide according to claim 1.
21. Method for treating and/or preventing and/or alleviating disorders relating to inflammation, rheumatoid arthritis, Crohn's disease, ulcerative colitis, inflammatory bowel syndrome, multiple sclerosis, Addison's disease, Autoimmune hepatitis, Autoimmune parotitis, Diabetes Type I, Epididymitis, Glomerulonephritis, Graves' disease, Guillain-Barre syndrome, Hashimoto's disease, Hemolytic anemia, Systemic lupus erythematosus, Male infertility, Multiple sclerosis, Myasthenia Gravis, Pemphigus, Psoriasis, Rheumatic fever, Rheumatoid arthritis, Sarcoidosis, Scleroderma, Sjogren's syndrome, Spondyloarthropathies, Thyroiditis, or Vasculitis, comprising administering to a subject a therapeutically effective amount of an anti-TNF polypeptide according to claim 1.
22. Composition comprising a polypeptide according to claim 1 and a suitable pharmaceutical vehicle.
23. Method for treating and/or preventing and/or alleviating disorders relating to inflammatory processes, comprising administering to a subject a therapeutically effective amount of a composition according to claim 22.
24. Method for treating and/or preventing and/or alleviating disorders relating to inflammation, rheumatoid arthritis, Crohn's disease, ulcerative colitis, inflammatory bowel syndrome, multiple sclerosis, Addison's disease, Autoimmune hepatitis, Autoimmune parotitis, Diabetes Type I, Epididymitis, Glomerulonephritis, Graves' disease, Guillain-Barre syndrome, Hashimoto's disease, Hemolytic anemia, Systemic lupus erythematosus, Male infertility, Multiple sclerosis, Myasthenia Gravis, Pemphigus, Psoriasis, Rheumatic fever, Rheumatoid arthritis, Sarcoidosis, Scleroderma, Sjogren's syndrome, Spondyloarthropathies, Thyroiditis, or Vasculitis, comprising administering to a subject a therapeutically effective amount of a composition according to claim 22.
25. Anti-TNF-alpha polypeptide comprising one or more single domain antibodies directed against tumor necrosis factor alpha (TNF-alpha) and an effector group.
26. Anti-TNF-alpha polypeptide according to claim 25, in which the effector group is a human Fc domain.
27. Anti-TNF-alpha polypeptide according to claim 26, in which the one or more single domain antibodies directed against tumor necrosis factor alpha (TNF-alpha) are fused to the Fc domain.
28. Anti-TNF-alpha polypeptide according to claim 25, which inhibits the interaction between TNF alpha and one or more TNF alpha receptors.
29. Anti-TNF-alpha polypeptide according to claim 25, which modifies the biological activity of TNF-alpha after binding to TNF alpha.
30. Anti-TNF-alpha polypeptide according to claim 25, which binds into the receptor-binding groove of TNF-alpha.
31. Anti-TNF-alpha polypeptide according to claim 25, in which said one or more single domain antibodies directed against TNF-alpha bind to TNF-alpha with an affinity of better than 10−6 M.
32. Anti-TNF-alpha polypeptide according to claim 25, in which said one or more single domain antibodies directed against TNF-alpha have an amino acid selected from glycine, alanine, valine, leucine, isoleucine, proline, phenylalanine, tyrosine, tryptophan, methionine, serine, threonine, asparagine and glutamine at position 45 and a tryptophan at position 103 according to the Kabat numbering.
33. Anti-TNF-alpha polypeptide according to claim 25, in which said one or more single domain antibodies directed against TNF-alpha have an arginine residue at position 103 according to the Kabat numbering.
34. Anti-TNF-alpha polypeptide according to claim 25, in which said one or more single domain antibodies directed against TNF-alpha are one or more Camelidae VHHs.
35. Anti-TNF-alpha polypeptide according to claim 34, in which said one or more single domain antibodies directed against TNF-alpha are one or more humanized Camelidae VHHs.
36. Anti-TNF polypeptide according to claim 35, wherein the one or more one single domain antibodies directed against TNF-alpha comprises a sequence represented by any of SEQ ID NOs: 1 to 16 or 79 to 84, or comprises:
a) a sequence that is more than 70% identical to the sequence of any of SEQ ID NOs: 1 to 16 or 79 to 84;
b) a functional portion of any of SEQ ID NOs: 1 to 16 or 79 to 84 that maintains the interaction with the target with affinity of 1×10−6 M or better;
c) a functional portion of any of SEQ ID NOs: 1 to 7, 23 to 31, and 62 to 65 that comprises a partial deletion of the complete amino acid sequence and still maintains the binding site(s) and protein domain(s) necessary for the binding of and interaction with the target.
37. Anti-TNF polypeptide according to claim 36, wherein the one or more one single domain antibodies directed against TNF-alpha comprises a sequence that is more than 80% identical to the sequence of any of SEQ ID NOs: 1 to 16 or 79 to 84.
38. Anti-TNF polypeptide according to claim 37, wherein the one or more one single domain antibodies directed against TNF-alpha comprises a sequence that is more than 90% identical to the sequence of any of SEQ ID NOs: 1 to 16 or 79 to 84.
39. Anti-TNF polypeptide according to claim 36, wherein the one or more one single domain antibodies directed against TNF-alpha comprises a sequence represented by any of SEQ ID NOs: 1 to 16 or79 to 84.
40. Anti-TNF polypeptide according to claim 25, wherein the one or more one single domain antibodies directed against TNF-alpha comprises the sequence of SEQ ID NO: 4, or comprises:
a) a sequence that is more than 70% identical to the sequence of SEQ ID NO: 4;
b) a functional portion of SEQ ID NO: 4 that maintains the interaction with the target with affinity of 1×10−6 M or better;
c) a functional portion of SEQ ID NO: 4 that comprises a partial deletion of the complete amino acid sequence and still maintains the binding site(s) and protein domain(s) necessary for the binding of and interaction with the target.
41. Anti-TNF polypeptide according to claim 40, wherein the one or more one single domain antibodies directed against TNF-alpha comprises a sequence that is more than 80% identical to the sequence of SEQ ID NO:4.
42. Anti-TNF polypeptide according to claim 41, wherein the one or more one single domain antibodies directed against TNF-alpha comprises a sequence that is more than 90% identical to the sequence of SEQ ID NO:4.
43. Anti-TNF polypeptide according to claim 40, wherein the one or more one single domain antibodies directed against TNF-alpha comprises the sequence of SEQ ID NO:4.
44. Method for treating and/or preventing and/or alleviating disorders relating to inflammatory processes, comprising administering to a subject a therapeutically effective amount of an anti-TNF polypeptide according to claim 25.
45. Method for treating and/or preventing and/or alleviating disorders relating to inflammation, rheumatoid arthritis, Crohn's disease, ulcerative colitis, inflammatory bowel syndrome, multiple sclerosis, Addison's disease, Autoimmune hepatitis, Autoimmune parotitis, Diabetes Type I, Epididymitis, Glomerulonephritis, Graves' disease, Guillain-Barre syndrome, Hashimoto's disease, Hemolytic anemia, Systemic lupus erythematosus, Male infertility, Multiple sclerosis, Myasthenia Gravis, Pemphigus, Psoriasis, Rheumatic fever, Rheumatoid arthritis, Sarcoidosis, Scleroderma, Sjogren's syndrome, Spondyloarthropathies, Thyroiditis, or Vasculitis, comprising administering to a subject a therapeutically effective amount of an anti-TNF polypeptide according to claim 25.
46. Composition comprising a polypeptide according to claim 25 and a suitable pharmaceutical vehicle.
47. Method for treating and/or preventing and/or alleviating disorders relating to inflammatory processes, comprising administering to a subject a therapeutically effective amount of a composition according to claim 46.
48. Method for treating and/or preventing and/or alleviating disorders relating to inflammation, rheumatoid arthritis, Crohn's disease, ulcerative colitis, inflammatory bowel syndrome, multiple sclerosis, Addison's disease, Autoimmune hepatitis, Autoimmune parotitis, Diabetes Type I, Epididymitis, Glomerulonephritis, Graves' disease, Guillain-Barre syndrome, Hashimoto's disease, Hemolytic anemia, Systemic lupus erythematosus, Male infertility, Multiple sclerosis, Myasthenia Gravis, Pemphigus, Psoriasis, Rheumatic fever, Rheumatoid arthritis, Sarcoidosis, Scleroderma, Sjogren's syndrome, Spondyloarthropathies, Thyroiditis, or Vasculitis, comprising administering to a subject a therapeutically effective amount of a composition according to claim 46.
49. A method for synthesizing a single-domain-effector group (dAb-effector group) suitable for in vivo use comprising the steps of: (a) selecting an antibody single variable domain having an epitope binding specificity; and (b) attaching the single domain of step (a) to an effector group.
50. A method according to claim 49 wherein the antibody single variable domain is a heavy chain variable domain.
51. A method according to claim 49 wherein the effector group comprises any one or more of those groups selected from the group consisting of: an antibody CH1 heavy chain domain, an antibody CH2 heavy chain domain, an antibody CH3 heavy chain domain, an Fc region of an antibody and a hinge region of an antibody molecule.
52. A method according to claim 49, wherein the effector group constitutes an Fc region of an antibody.
53. A method according to claim 49, wherein the effector group consists of a CH2 and CH3 domain.
54. A method according to claim 51, wherein the effector group consists of a CH2 domain, a CH3 domain and the hinge region of an antibody molecule.
55. A method according to claim 49, wherein the antibody single variable domain is a non-Camelid variable domain.
56. A method according to claim 55, wherein the antibody single variable domain is a human variable domain.
57. A method according to claim 49, wherein the antibody single variable domain comprises one or more human framework regions.
58. A method according to claim 49, wherein the antibody single variable domain comprises four framework regions as defined by Kabat, which are derived from a human.
59. A method according to claim 58, wherein one or more of the human framework regions as defined by Kabat are identical on the amino acid level to those encoded by human germline antibody genes.
60. A method according to claim 49, wherein the antibody single variable domain is isolated, in part, by human immunization.
61. A method according to claim 49, wherein the antibody single variable domain is not isolated by animal immunization.
62. A method according to claim 49, wherein the effector group is of Camelid or human origin.
63. A method according to claim 49, wherein the single variable domain comprises one or more human framework regions and the immunoglobulin effector group is of human origin.
64. A method according to claim 63, wherein the single variable domain comprises four human framework regions and the immunoglobulin effector group is of human origin.
65. A method according to claim 49, wherein attaching of the single variable domain to the effector group in step (b) is effected by expressing the single-domain-effector group as a fusion polypeptide.
66. A dAb-effector group comprising: (a) an antibody single variable domain having an epitope binding specificity; and (b) an effector group attached to said antibody single variable domain.
67. A medicament comprising the dAb-effector group of claim 66.
68. A dAb-effector group according to claim 66, wherein the antibody single variable domain is a heavy chain variable domain.
69. A dAb-effector group according to claim 66, wherein the effector group comprises any one or more of those groups selected from the group consisting of: an antibody CH1 heavy chain domain, an antibody CH2 heavy chain domain, an antibody CH3 heavy chain domain, an Fc region of an antibody and a hinge region of an antibody molecule.
70. A dAb-effector group according to claim 69 wherein the effector group consists of a CH2 and CH3 domain.
71. A dAb-effector group according to claim 69 wherein the effector group consists of a CH2 domain, a CH3 domain and the hinge region of an antibody molecule.
72. A dAb-effector group according to claim 69 wherein the effector group constitutes an Fc region of an antibody.
73. A dAb-effector group according to claim 66, wherein the antibody single variable domain is of human origin.
74. A dAb-effector group according to claim 66, wherein the antibody single variable domain comprises human framework regions.
75. A dAb-effector group according to claim 66, wherein the effector group is of Camelid or human origin.
76. A dAb-effector group according to claim 66, wherein the single variable domain comprises one or more human framework regions and the immunoglobulin effector group is of human origin.
77. Two or more dAb-effector groups according to claim 66 provided as a higher order structure selected from the group consisting of the following: dimers, trimers and multimers.
78. Two dAb-effector groups according to claim 77 provided as a heterodimer or a homodimer.
79. Two dAb-effector groups according to claim 78 provided as a homodimer.
80. A nucleic acid molecule encoding a dAb-effector group according to claim 66.
81. A nucleic acid molecule according to claim 80 further encoding a signal sequence for export of the dAb and effector group from the cytoplasm of a host cell upon expression.
82. A vector comprising nucleic acid according to claim 80.
83. A host cell transfected with a vector according to claim 82.
84. A composition comprising a dAb-effector group(s) according to claim 66 and a pharmaceutically acceptable carrier, diluent or excipient.
85. A method of treating and/or preventing disease in a patient, wherein the method comprises administering to the patient a dAb-effector group(s) according to claim 66 or a composition according to claims 84.
86. A medicament for the treatment and/or prevention of disease, comprising the dAb-effector group of claim 66 or the composition of claim 84.
87. A method for the treatment and/or prophylaxis of an inflammatory disease in a patient in need of such treatment and/or prophylaxis which comprises the step of administering to that patient a therapeutically effective amount of a dAb-effector group according to claim 66.
88. A method according to claim 87 wherein the inflammatory disease is mediated by TNF alpha and is selected from the group consisting of the following: rheumatoid arthritis, psoriasis, Crohn's disease, inflammatory bowel disease (IBD), multiple sclerosis, Alzheimer's, and glomerular nephritis.
89. A method according to claim 88, wherein the TNF alpha is human TNF alpha and the patient is a human.
US11/788,832 2002-11-08 2007-04-20 Single domain antibodies directed against tumour necrosis factor-alpha and uses therefor Abandoned US20070237769A1 (en)

Priority Applications (7)

Application Number Priority Date Filing Date Title
US11/788,832 US20070237769A1 (en) 2002-11-08 2007-04-20 Single domain antibodies directed against tumour necrosis factor-alpha and uses therefor
US11/804,647 US20090022721A1 (en) 2002-11-08 2007-05-18 Single domain antibodies directed against tumour necrosis factor-alpha and uses therefor
US12/905,589 US20110027281A1 (en) 2002-11-08 2010-10-15 Single domain antibodies directed against tumor necrosis factor-alpha and uses therefor
US13/078,241 US20110184150A1 (en) 2002-11-08 2011-04-01 Single domain antibodies directed against tumor necrosis factor-alpha and uses therefor
US13/423,790 US20120202977A1 (en) 2002-11-08 2012-03-19 Single domain antibodies directed against tumor necrosis factor-alpha and uses therefor
US14/529,280 US9371381B2 (en) 2002-11-08 2014-10-31 Single domain antibodies directed against tumor necrosis factor-alpha and uses therefor
US15/157,830 US20170096481A1 (en) 2002-11-08 2016-05-18 Single domain antibodies directed against tumor necrosis factor-alpha and uses therefor

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
US42507302P 2002-11-08 2002-11-08
US42506302P 2002-11-08 2002-11-08
EP03447005 2003-01-10
EP03447005.4 2003-01-10
PCT/BE2003/000192 WO2004041862A2 (en) 2002-11-08 2003-11-07 Single domain antibodies directed against tumour necrosis factor-alpha and uses therefor
US10/534,348 US20060034845A1 (en) 2002-11-08 2003-11-07 Single domain antibodies directed against tumor necrosis factor alpha and uses therefor
US11/636,300 US20070077249A1 (en) 2002-11-08 2006-12-08 Single domain antibodies directed against tumour necrosis factor-alpha and uses therefor
US11/788,832 US20070237769A1 (en) 2002-11-08 2007-04-20 Single domain antibodies directed against tumour necrosis factor-alpha and uses therefor

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US11/636,300 Continuation US20070077249A1 (en) 2002-11-08 2006-12-08 Single domain antibodies directed against tumour necrosis factor-alpha and uses therefor

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US11/804,647 Continuation US20090022721A1 (en) 2002-11-08 2007-05-18 Single domain antibodies directed against tumour necrosis factor-alpha and uses therefor

Publications (1)

Publication Number Publication Date
US20070237769A1 true US20070237769A1 (en) 2007-10-11

Family

ID=35800205

Family Applications (9)

Application Number Title Priority Date Filing Date
US10/534,348 Abandoned US20060034845A1 (en) 2002-11-08 2003-11-07 Single domain antibodies directed against tumor necrosis factor alpha and uses therefor
US11/636,300 Abandoned US20070077249A1 (en) 2002-11-08 2006-12-08 Single domain antibodies directed against tumour necrosis factor-alpha and uses therefor
US11/788,832 Abandoned US20070237769A1 (en) 2002-11-08 2007-04-20 Single domain antibodies directed against tumour necrosis factor-alpha and uses therefor
US11/804,647 Abandoned US20090022721A1 (en) 2002-11-08 2007-05-18 Single domain antibodies directed against tumour necrosis factor-alpha and uses therefor
US12/905,589 Abandoned US20110027281A1 (en) 2002-11-08 2010-10-15 Single domain antibodies directed against tumor necrosis factor-alpha and uses therefor
US13/078,241 Abandoned US20110184150A1 (en) 2002-11-08 2011-04-01 Single domain antibodies directed against tumor necrosis factor-alpha and uses therefor
US13/423,790 Abandoned US20120202977A1 (en) 2002-11-08 2012-03-19 Single domain antibodies directed against tumor necrosis factor-alpha and uses therefor
US14/529,280 Active US9371381B2 (en) 2002-11-08 2014-10-31 Single domain antibodies directed against tumor necrosis factor-alpha and uses therefor
US15/157,830 Abandoned US20170096481A1 (en) 2002-11-08 2016-05-18 Single domain antibodies directed against tumor necrosis factor-alpha and uses therefor

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US10/534,348 Abandoned US20060034845A1 (en) 2002-11-08 2003-11-07 Single domain antibodies directed against tumor necrosis factor alpha and uses therefor
US11/636,300 Abandoned US20070077249A1 (en) 2002-11-08 2006-12-08 Single domain antibodies directed against tumour necrosis factor-alpha and uses therefor

Family Applications After (6)

Application Number Title Priority Date Filing Date
US11/804,647 Abandoned US20090022721A1 (en) 2002-11-08 2007-05-18 Single domain antibodies directed against tumour necrosis factor-alpha and uses therefor
US12/905,589 Abandoned US20110027281A1 (en) 2002-11-08 2010-10-15 Single domain antibodies directed against tumor necrosis factor-alpha and uses therefor
US13/078,241 Abandoned US20110184150A1 (en) 2002-11-08 2011-04-01 Single domain antibodies directed against tumor necrosis factor-alpha and uses therefor
US13/423,790 Abandoned US20120202977A1 (en) 2002-11-08 2012-03-19 Single domain antibodies directed against tumor necrosis factor-alpha and uses therefor
US14/529,280 Active US9371381B2 (en) 2002-11-08 2014-10-31 Single domain antibodies directed against tumor necrosis factor-alpha and uses therefor
US15/157,830 Abandoned US20170096481A1 (en) 2002-11-08 2016-05-18 Single domain antibodies directed against tumor necrosis factor-alpha and uses therefor

Country Status (1)

Country Link
US (9) US20060034845A1 (en)

Cited By (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040253638A1 (en) * 1992-08-21 2004-12-16 Vrije Universiteit Brussel Immunoglobulins devoid of light chains
US20050054001A1 (en) * 2001-10-24 2005-03-10 Serge Muyldermans Functional heavy chain antibodies, fragments thereof, library thereof and methods of production thereof
US20060034845A1 (en) * 2002-11-08 2006-02-16 Karen Silence Single domain antibodies directed against tumor necrosis factor alpha and uses therefor
US20070178082A1 (en) * 2002-11-08 2007-08-02 Ablynx N.V. Stabilized single domain antibodies
US20070269422A1 (en) * 2006-05-17 2007-11-22 Ablynx N.V. Serum albumin binding proteins with long half-lives
US20090238829A1 (en) * 2002-11-08 2009-09-24 Ablynx N.V. Stabilized single domain antibodies
US20090324512A1 (en) * 2002-11-08 2009-12-31 Ablynx N.V. Polypeptide constructs for nasal administration
US20100003253A1 (en) * 2002-11-08 2010-01-07 Ablynx N.V. Single domain antibodies directed against epidermal growth factor receptor and uses therefor
US20100172894A1 (en) * 2008-10-29 2010-07-08 Wyeth Methods for purification of single domain antigen binding molecules
US20100297111A1 (en) * 2005-05-21 2010-11-25 Els Anna Alice Beirnaert Nanobodies against tumor necrosis factor-alpha
US8188223B2 (en) 2005-05-18 2012-05-29 Ablynx N.V. Serum albumin binding proteins
US8603824B2 (en) 2004-07-26 2013-12-10 Pfenex, Inc. Process for improved protein expression by strain engineering
WO2014033313A1 (en) 2012-09-03 2014-03-06 Vib Vzw Protective anti-etec antibody
US9320792B2 (en) 2002-11-08 2016-04-26 Ablynx N.V. Pulmonary administration of immunoglobulin single variable domains and constructs thereof
US9393304B2 (en) 2008-10-29 2016-07-19 Ablynx N.V. Formulations of single domain antigen binding molecules
US9394571B2 (en) 2007-04-27 2016-07-19 Pfenex Inc. Method for rapidly screening microbial hosts to identify certain strains with improved yield and/or quality in the expression of heterologous proteins
WO2016156474A1 (en) * 2015-03-31 2016-10-06 Vhsquared Limited Polypeptide comprising an immunoglobulin chain variable domain which binds to clostridium difficile toxin a
US9580719B2 (en) 2007-04-27 2017-02-28 Pfenex, Inc. Method for rapidly screening microbial hosts to identify certain strains with improved yield and/or quality in the expression of heterologous proteins
US10041102B2 (en) 2002-10-08 2018-08-07 Pfenex Inc. Expression of mammalian proteins in Pseudomonas fluorescens
US10633438B2 (en) 2015-03-31 2020-04-28 Vhsquared Limited Polypeptides
US11623952B2 (en) 2019-06-21 2023-04-11 Sorriso Pharmaceuticals, Inc. IL-23 and TNF-alpha binding bi-specific heavy chain polypeptides
US11667719B2 (en) 2019-06-21 2023-06-06 Sorriso Pharmaceuticals, Inc. VHH immunoglobulin chain variable domain that binds to IL-7R and methods of use thereof for treating autoimmune and/or inflammatory diseases
US11684677B2 (en) 2016-09-30 2023-06-27 Sorriso Pharmaceuticals, Inc. Compositions

Families Citing this family (31)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060034833A1 (en) * 2002-11-08 2006-02-16 Els Beirnaert Single domain antibodies directed against interferron-gamma and uses therefor
US9017662B2 (en) * 2005-08-30 2015-04-28 Actogenix N.V. Anti-TNF alpha producing lactic acid bacteria for the treatment of chronic enterocolitis
JP2007172129A (en) * 2005-12-20 2007-07-05 Sony Corp Nonvolatile memory access control device and nonvolatile memory control system
AU2006326867A1 (en) * 2005-12-20 2007-06-28 Cephalon Australia Pty Ltd Chimeric antibodies with part New World primate binding regions
MX2008009792A (en) 2006-02-01 2008-09-01 Arana Therapeutics Ltd Domain antibody construct.
US20100226920A1 (en) * 2006-03-27 2010-09-09 Ablynx N.V. Medical delivery device for therapeutic proteins based on single domain antibodies
EP2081960B1 (en) * 2006-10-27 2018-06-27 Ablynx N.V. Intranasal delivery of polypeptides and proteins
GB0809069D0 (en) 2008-05-19 2008-06-25 Univ Leuven Kath Gene signatures
WO2010047419A1 (en) * 2008-10-24 2010-04-29 Fujifilm Corporation Immobilization substrate and method for producing the same
WO2010063113A1 (en) * 2008-12-05 2010-06-10 University Of Guelph Anti-cobra toxin antibody fragments and method of producing a vhh library
KR101881714B1 (en) 2009-10-26 2018-07-24 네스텍 소시에테아노님 Assays for the detection of anti-tnf drugs and autoantibodies
WO2011098518A2 (en) 2010-02-11 2011-08-18 Ablynx Nv Delivery of immunoglobulin variable domains and constructs thereof
WO2011161263A1 (en) 2010-06-25 2011-12-29 Ablynx Nv Pharmaceutical compositions for cutaneous administration
AU2011317149B2 (en) 2010-10-18 2015-05-07 Société des Produits Nestlé S.A. Methods for determining anti-drug antibody isotypes
US9580480B2 (en) 2011-05-31 2017-02-28 Massachusetts Institute Of Technology Cell-directed synthesis of multifunctional nanopatterns and nanomaterials
US20130266963A1 (en) * 2011-07-06 2013-10-10 Nestec S.A. Assay for detecting neutralizing autoantibodies to biologic therapy
EP2729807B1 (en) 2011-07-06 2016-08-17 Nestec S.A. Assays for detecting neutralizing autoantibodies to biologic therapy with tnf alpha
WO2013059159A1 (en) 2011-10-21 2013-04-25 Crystal Bioscience, Inc. In vivo method for generating diversity in a protein scaffold
US9230932B2 (en) * 2012-02-09 2016-01-05 Taiwan Semiconductor Manufacturing Company, Ltd. Interconnect crack arrestor structure and methods
AU2013372026B2 (en) * 2013-01-03 2018-11-08 Merck Patent Gmbh Method for producing secretable antibodies by expression in saccharomyces cerevisiae
CA2962275C (en) 2014-10-23 2019-11-05 Singh Biotechnology, Llc Single domain antibodies directed against intracellular antigens
US20170267784A1 (en) 2014-10-23 2017-09-21 Singh Molecular Medicine, Llc Single domain antibodies directed against intracellular antigens
MX2017007232A (en) 2014-12-05 2017-10-16 Nestec Sa Indirect homogeneous mobility shift assays for the detection of biologics in patient samples.
WO2016110595A1 (en) 2015-01-09 2016-07-14 Atonomics A/S A UNIVERSAL ASSAY FOR DETERMINING THE QUANTITY OF TNFα INHIBITORY DRUGS AND THEIR CORRESPONDING ANTI-DRUG-ANTIBODIES
GB201500463D0 (en) 2015-01-12 2015-02-25 Cresendo Biolog Ltd Therapeutic molecules
WO2017059397A1 (en) 2015-10-01 2017-04-06 Whitehead Institute For Biomedical Research Labeling of antibodies
TWI746473B (en) 2015-11-02 2021-11-21 美商辛分子醫藥有限公司 Single domain antibodies directed against intracellular antigens
US11119096B2 (en) 2016-07-08 2021-09-14 W. Health L.P. Universal assay for determining the quantity of therapeutic monoclonal antibodies and their corresponding anti-drug-antibodies in samples
RU2652876C1 (en) * 2016-12-21 2018-05-03 Федеральное государственное автономное образовательное учреждение высшего образования "Национальный исследовательский Нижегородский государственный университет им. Н.И. Лобачевского" Recombinant antibody specific to tumor nucleus factor and myeloid immune cell marker
CN109970860A (en) * 2017-12-27 2019-07-05 信达生物制药(苏州)有限公司 Three chain antibodies, Its Preparation Method And Use
WO2024003873A1 (en) 2022-06-30 2024-01-04 Intrexon Actobiotics Nv D/B/A Precigen Actobio Single variable domain antibodies against tumor necrosis factor-alpha

Citations (42)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4405306A (en) * 1981-12-08 1983-09-20 Beecham Inc. Medicated disposable douche product
US5091513A (en) * 1987-05-21 1992-02-25 Creative Biomolecules, Inc. Biosynthetic antibody binding sites
US5196193A (en) * 1989-10-31 1993-03-23 Ophidian Pharmaceuticals, Inc. Antivenoms and methods for making antivenoms
US5487890A (en) * 1990-10-04 1996-01-30 University Of Virginia Patent Foundation Mammalian primate erythrocyte bound heteropolymerized monoclonal antibodies and methods of use thereof
US5656273A (en) * 1994-01-18 1997-08-12 Genentech, Inc. Method of treatment of parasitic infection using IgE antagonists
US5672347A (en) * 1984-07-05 1997-09-30 Genentech, Inc. Tumor necrosis factor antagonists and their use
US5837243A (en) * 1995-06-07 1998-11-17 Medarex, Inc. Therapeutic compounds comprised of anti-Fc receptor antibodies
US5843440A (en) * 1990-10-03 1998-12-01 Redcell Canada, Inc. Cellular and serum protein anchors for modulating pharmacokinetics
US5891996A (en) * 1972-09-17 1999-04-06 Centro De Inmunologia Molecular Humanized and chimeric monoclonal antibodies that recognize epidermal growth factor receptor (EGF-R); diagnostic and therapeutic use
US5976532A (en) * 1988-05-18 1999-11-02 Centocor, Inc. Method of antithrombotic therapy using anti-GPIIb/IIIa antibodies or fragments thereof, including c7E3
US5994511A (en) * 1997-07-02 1999-11-30 Genentech, Inc. Anti-IgE antibodies and methods of improving polypeptides
US6066718A (en) * 1992-09-25 2000-05-23 Novartis Corporation Reshaped monoclonal antibodies against an immunoglobulin isotype
US6251393B1 (en) * 1998-10-23 2001-06-26 The Brigham And Women's Hospital, Inc. Conformation-specific anti-von Willebrand Factor antibodies
US20020009453A1 (en) * 2000-06-16 2002-01-24 Symphogen A/S Polyclonal antibody composition for treating allergy
US20020028204A1 (en) * 1994-11-30 2002-03-07 Ajinomoto Co., Inc. Antithrombotic agent and anti-von willebrand factor monoclonal antibody
US20020058033A1 (en) * 2000-10-13 2002-05-16 Raisch Kevin Paul Human anti-epidermal growth factor receptor single-chain antibodies
US20020076404A1 (en) * 1998-01-29 2002-06-20 Chang Tse Wen Treating atopic dermatitis with IgE antagonists
US6419934B1 (en) * 1999-02-24 2002-07-16 Edward L. Tobinick TNF modulators for treating neurological disorders associated with viral infection
US20020132275A1 (en) * 2000-11-03 2002-09-19 Board Of Regents, The University Of Texas System Methods for detecting the efficacy of anticancer treatments
US6504013B1 (en) * 2000-02-01 2003-01-07 Idexx Laboratories, Inc. Canine allergy therapeutic recombinant chimeric anti-IgE monoclonal antibody
US20030092892A1 (en) * 1997-10-27 2003-05-15 Frenken Leon Gerardus Multivalent antigen-binding proteins
US20030118592A1 (en) * 2001-01-17 2003-06-26 Genecraft, Inc. Binding domain-immunoglobulin fusion proteins
US6759518B1 (en) * 1998-04-09 2004-07-06 Vectron Therapeutics Ag Single-chain multiple antigen-binding molecule, its preparation and use
US20040180046A1 (en) * 2000-04-26 2004-09-16 Jeff Himawan Bispecific molecules and uses thereof
US20040197326A1 (en) * 1995-07-27 2004-10-07 Genentech, Inc. Method for treatment of allergic asthma
US20040219643A1 (en) * 2001-06-28 2004-11-04 Greg Winter Dual-specific ligand
US6902734B2 (en) * 2000-08-07 2005-06-07 Centocor, Inc. Anti-IL-12 antibodies and compositions thereof
US20060034833A1 (en) * 2002-11-08 2006-02-16 Els Beirnaert Single domain antibodies directed against interferron-gamma and uses therefor
US20060034845A1 (en) * 2002-11-08 2006-02-16 Karen Silence Single domain antibodies directed against tumor necrosis factor alpha and uses therefor
US7054257B2 (en) * 2003-04-29 2006-05-30 International Business Machines Corporation AFM-based data storage and microscopy
US20060115470A1 (en) * 2002-11-08 2006-06-01 Ablynx N. V. Camelidae antibodies against imminoglobulin e and use thereof for the treatment of allergic disorders
US20060228355A1 (en) * 2003-11-07 2006-10-12 Toon Laeremans Camelidae single domain antibodies vhh directed against epidermal growth factor receptor and uses therefor
US20070031424A1 (en) * 2001-10-24 2007-02-08 Vlaams Interuniversitair Instituut Voor Biotechnogie Vzw Functional heavy chain antibodies, fragments thereof, library thereof and methods of production thereof
US20070178082A1 (en) * 2002-11-08 2007-08-02 Ablynx N.V. Stabilized single domain antibodies
US20070264253A1 (en) * 2004-03-19 2007-11-15 Meilin Liu Human Anti-Epidermal Growth Factor Receptor Antibody
US7300655B2 (en) * 2002-08-01 2007-11-27 Immunomedics, Inc. Alpha-fetoprotein Immu31 antibodies and fusion proteins and methods of use thereof
US7368111B2 (en) * 1995-10-06 2008-05-06 Cambridge Antibody Technology Limited Human antibodies specific for TGFβ2
US7589180B2 (en) * 2001-05-11 2009-09-15 Abbott Laboratories Inc. Specific binding proteins and uses thereof
US20090238829A1 (en) * 2002-11-08 2009-09-24 Ablynx N.V. Stabilized single domain antibodies
US20100003258A1 (en) * 2008-07-01 2010-01-07 Zhigang Weng Fibroblast growth factor receptor 3 (fgfr3) binding proteins
US7897151B2 (en) * 2000-08-30 2011-03-01 Pharmacia & Upjohn Company, Llc Anti-IgE vaccines
US20110123529A1 (en) * 2002-11-08 2011-05-26 Ablynx N.V. Single domain antibodies directed against epidermal growth factor receptor and uses therefor

Family Cites Families (79)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2007A (en) * 1841-03-16 Improvement in the mode of harvesting grain
SE307996B (en) 1965-11-30 1969-01-27 Asea Ab
JPS62175426A (en) 1986-01-27 1987-08-01 Kazufumi Shimizu Antibody and spraying agent containing said substance as active component
EP0288088B1 (en) 1987-04-24 1994-03-09 Teijin Limited Detection of tumor necrosis factor; monoclonal antibody and kit
ATE121299T1 (en) 1987-12-31 1995-05-15 Tanox Biosystems Inc ANTIGEN EPITOPES LOCATED EXCLUSIVELY ON IGE-BEARING B-LYMPHOCYTES.
JPH01268645A (en) 1988-04-18 1989-10-26 Teijin Ltd Agent for suppressing schwartzman reaction
DE68914244T2 (en) 1988-10-24 1994-10-27 Otsuka Pharma Co Ltd Monoclonal antibody.
JP2919890B2 (en) 1988-11-11 1999-07-19 メディカル リサーチ カウンスル Single domain ligand, receptor consisting of the ligand, method for producing the same, and use of the ligand and the receptor
GB8905400D0 (en) 1989-03-09 1989-04-19 Jonker Margreet Medicaments
SE509359C2 (en) 1989-08-01 1999-01-18 Cemu Bioteknik Ab Use of stabilized protein or peptide conjugates for the preparation of a drug
AU640400B2 (en) 1989-08-07 1993-08-26 Peptide Technology Ltd. Tumour necrosis factor binding ligands
US5644034A (en) * 1989-08-07 1997-07-01 Peptide Technology Ltd. Tumour necrosis factor binding ligands
GB8921123D0 (en) 1989-09-19 1989-11-08 Millar Ann B Treatment of ards
GB9016299D0 (en) 1990-07-25 1990-09-12 Brien Caroline J O Binding substances
JP2988635B2 (en) 1990-09-18 1999-12-13 塩野義製薬株式会社 Monoclonal antibody against human IgE
US5612034A (en) 1990-10-03 1997-03-18 Redcell, Inc. Super-globuling for in vivo extended lifetimes
DE69230998T2 (en) 1991-03-21 2000-12-21 Masimo Corp LOW-NOISE OPTICAL CONVERTER
WO1993004173A1 (en) 1991-08-14 1993-03-04 Genentech, Inc. Immunoglobulin variants for specific fc epsilon receptors
EP0584421A1 (en) 1992-08-21 1994-03-02 Cécile Casterman Immunoglobulins devoid of light chains
PT1621554E (en) 1992-08-21 2009-07-13 Univ Bruxelles Immunoglobulins devoid of light chains
US6765087B1 (en) 1992-08-21 2004-07-20 Vrije Universiteit Brussel Immunoglobulins devoid of light chains
EP1452542A3 (en) 1992-09-24 2007-05-02 Novartis AG Reshaped human monoclonal antibodies specific for IgE
AU5669694A (en) 1992-11-20 1994-06-22 Schering Corporation Antagonists of human gamma interferon
ATE204299T1 (en) 1993-03-05 2001-09-15 Bayer Ag HUMAN MONOCLONAL ANTI-TNF ALPHA ANTIBODIES
DE69427974T2 (en) 1993-04-29 2001-12-06 Unilever Nv PRODUCTION OF ANTIBODIES OR FUNCTIONAL PARTS THEREOF, DERIVED FROM HEAVY CHAINS OF IMMUNOGLOBULINES FROM CAMELIDAE
AU1441395A (en) 1993-12-21 1995-07-10 St. Louis University Ocular diagnostics and therapies
US5747148A (en) 1994-09-12 1998-05-05 Minnesota Mining And Manufacturing Company Ink jet printing sheet
US6096871A (en) 1995-04-14 2000-08-01 Genentech, Inc. Polypeptides altered to contain an epitope from the Fc region of an IgG molecule for increased half-life
EP0739981A1 (en) 1995-04-25 1996-10-30 Vrije Universiteit Brussel Variable fragments of immunoglobulins - use for therapeutic or veterinary purposes
EP0823941A4 (en) 1995-04-28 2001-09-19 Abgenix Inc Human antibodies derived from immunized xenomice
JPH11507535A (en) 1995-06-07 1999-07-06 イムクローン システムズ インコーポレイテッド Antibodies and antibody fragments that suppress tumor growth
GB2302024B (en) 1995-06-09 1998-10-07 Malix Polymer Engineering Limi Obstetric vacuum extractor
US5637038A (en) 1995-07-10 1997-06-10 Davis; James F. Apparatus and method for skinning poultry
DE69609188T2 (en) 1995-09-18 2000-12-21 Intracel Corp NEUTRALIZING MONOCLONAL ANTIBODIES AGAINST RESPIRATORY SYNCYTIAL VIRUS
TR199801532T2 (en) 1996-02-09 1998-11-23 Basf Aktiengesellschaft Human antibodies that bind human TNFalpha.
US6417337B1 (en) * 1996-10-31 2002-07-09 The Dow Chemical Company High affinity humanized anti-CEA monoclonal antibodies
IT1289608B1 (en) 1997-02-05 1998-10-15 Angelini Ricerche Spa COMPOSITION FOR THERAPEUTIC OR DIAGNOSTIC USE ADMINISTRABLE VIA INTRANASAL, SUBLINGUAL OR VAGINAL
US5942602A (en) 1997-02-13 1999-08-24 Schering Aktiengessellschaft Growth factor receptor antibodies
AUPO561697A0 (en) 1997-03-13 1997-04-10 Crc For Cardiac Technology Haemocompatible surfaces
DE29712318U1 (en) 1997-07-07 1997-10-02 Arnold Guenter Paring knife
US6350860B1 (en) 1997-08-18 2002-02-26 Innogenetics N.V. Interferon-gamma-binding molecules for treating septic shock, cachexia, immune diseases and skin disorders
EP0954978B1 (en) 1998-03-12 2011-11-30 VHsquared Limited New products comprising inactivated yeasts or moulds provided with active antibodies
WO1999064069A1 (en) 1998-06-10 1999-12-16 Ophidian Pharmaceuticals, Inc. Antibodies to cytokines in the prevention and treatment of inflammatory bowel disease
EP1002861A1 (en) 1998-10-26 2000-05-24 Unilever Plc Antigen-binding proteins comprising a linker which confers restricted conformational flexibility
IL127127A0 (en) 1998-11-18 1999-09-22 Peptor Ltd Small functional units of antibody heavy chain variable regions
DE19852800C1 (en) 1998-11-16 2000-04-13 Univ Albert Ludwigs Freiburg Production of antibodies to a polypeptide encoded by a known DNA sequence comprises binding of antibodies produced by DNA vaccination to immobilized recombinantly expressed polypeptide
NZ511219A (en) 1999-01-05 2003-05-30 Univ Southern Australia Novel agents and methods for treatment and diagnosis of ocular disorders
CA2669512A1 (en) 1999-03-25 2000-09-28 Abbott Gmbh & Co. Kg Human antibodies that bind human il-12 and methods for producing
AU776824B2 (en) 1999-04-22 2004-09-23 Unilever Plc Inhibition of viral infection using monovalent antigen-binding proteins
AU7648900A (en) 1999-09-16 2001-04-17 Unilever Plc Delivery system for antidandruff agent
EP1118669A3 (en) 1999-12-17 2001-08-29 Unilever Plc Production of camelid antibodies in plants
DE60030323T2 (en) 1999-12-24 2007-10-11 Genentech, Inc., South San Francisco METHOD AND CONNECTIONS FOR EXTENDING THE HALF-TIME PERIODS FOR THE DISPOSAL OF BIOWIRESAMIC COMPOUNDS
KR20120011898A (en) 2000-02-10 2012-02-08 아보트 러보러터리즈 Antibodies that bind human interleukin-18 and methods of making and using
ATE428733T1 (en) 2000-03-14 2009-05-15 Unilever Nv VARIABLE DOMAIN OF THE HEAVY CHAIN OF AN ANTIBODY TO HUMAN NUTRITIONAL LIPASES AND THEIR USES
US7097840B2 (en) * 2000-03-16 2006-08-29 Genentech, Inc. Methods of treatment using anti-ErbB antibody-maytansinoid conjugates
AU2001264747A1 (en) 2000-05-22 2001-12-03 Idec Pharmaceuticals Corporation Identification of unique binding interactions between certain antibodies and thehuman b7.1 and b7.2 co-stimulatory antigens
EP1328626B1 (en) 2000-05-26 2013-04-17 National Research Council Of Canada Single-domain brain-targeting antibody fragments derived from llama antibodies
UA81743C2 (en) 2000-08-07 2008-02-11 Центокор, Инк. HUMAN MONOCLONAL ANTIBODY WHICH SPECIFICALLY BINDS TUMOR NECROSIS FACTOR ALFA (TNFα), PHARMACEUTICAL MIXTURE CONTAINING THEREOF, AND METHOD FOR TREATING ARTHRITIS
DK1360207T3 (en) 2000-12-13 2011-09-05 Bac Ip B V Protein array of variable domains of heavy immunoglobulin chains from camels
GB0031448D0 (en) 2000-12-22 2001-02-07 Leuven K U Res & Dev Inhibition of the vWF-collagen interaction by anti-human vWF monoclonal antibody (82D6A3) results in abolition of in vivo arterial platelet thrombus formation
CA2442074C (en) 2001-03-28 2014-07-22 Heska Corporation Methods of detecting early renal disease in animals
IL158126A0 (en) 2001-03-29 2004-03-28 Univ Ramot Peptides and antibodies to muc 1 proteins
US8178098B2 (en) 2001-04-03 2012-05-15 National Jewish Health Method to inhibit airway hyperresponsiveness using aerosolized T cell receptor antibodies
JP4398644B2 (en) 2001-04-06 2010-01-13 ザ トラスティーズ オブ ザ ユニバーシティ オブ ペンシルベニア ErbB interfacial peptide mimics and methods of use thereof
US7084257B2 (en) * 2001-10-05 2006-08-01 Amgen Inc. Fully human antibody Fab fragments with human interferon-gamma neutralizing activity
CN101143221A (en) 2002-03-15 2008-03-19 布赖汉姆妇女医院 Central airway administration for systemic delivery of therapeutics
US20040063912A1 (en) 2002-03-15 2004-04-01 The Brigham And Women's Hospital, Inc. Central airway administration for systemic delivery of therapeutics
CN1678634A (en) * 2002-06-28 2005-10-05 多曼蒂斯有限公司 Immunoglobulin single variable antigen combination area and its opposite constituent
US9320792B2 (en) 2002-11-08 2016-04-26 Ablynx N.V. Pulmonary administration of immunoglobulin single variable domains and constructs thereof
AU2004220325B2 (en) 2003-06-30 2011-05-12 Domantis Limited Polypeptides
AP2007004019A0 (en) 2004-11-25 2007-06-30 Unilever Plc Heavy chain and single domain antibodies
BRPI0518622A2 (en) 2004-12-02 2008-12-02 Domantis Ltd uses of interleukin-1 (il-1r1) type 1 receptor antagonists for the manufacture of a medicament for the treatment of a respiratory disease; A pharmaceutical composition comprising an IL-1R1 antagonist and a physiologically acceptable carrier and drug delivery device.
DK2949668T3 (en) 2005-05-18 2019-11-11 Ablynx Nv IMPROVED NANOBODIES TM AGAINST TUMOR CANCER FACTOR-ALFA
DE102005023617A1 (en) 2005-05-21 2006-11-23 Aspre Ag Method for mixing colors in a display
EP1754955B1 (en) 2005-08-19 2015-05-06 Aisin Aw Co., Ltd. Navigation method and corresponding system for determining a travel related time
EP1963370A1 (en) 2005-12-06 2008-09-03 Domantis Limited Bispecific ligands with binding specificity to cell surface targets and methods of use therefor
US20100260853A1 (en) 2007-12-13 2010-10-14 Amrik Basran Compositions for pulmonary delivery
AU2009237662A1 (en) 2008-04-17 2009-10-22 Ablynx N.V. Peptides capable of binding to serum proteins and compounds, constructs and polypeptides comprising the same
US8040275B2 (en) * 2009-07-15 2011-10-18 Lockheed Martin Corporation Method and apparatus for geographic positioning

Patent Citations (58)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5891996A (en) * 1972-09-17 1999-04-06 Centro De Inmunologia Molecular Humanized and chimeric monoclonal antibodies that recognize epidermal growth factor receptor (EGF-R); diagnostic and therapeutic use
US4405306A (en) * 1981-12-08 1983-09-20 Beecham Inc. Medicated disposable douche product
US5672347A (en) * 1984-07-05 1997-09-30 Genentech, Inc. Tumor necrosis factor antagonists and their use
US5091513A (en) * 1987-05-21 1992-02-25 Creative Biomolecules, Inc. Biosynthetic antibody binding sites
US5976532A (en) * 1988-05-18 1999-11-02 Centocor, Inc. Method of antithrombotic therapy using anti-GPIIb/IIIa antibodies or fragments thereof, including c7E3
US5196193A (en) * 1989-10-31 1993-03-23 Ophidian Pharmaceuticals, Inc. Antivenoms and methods for making antivenoms
US5843440A (en) * 1990-10-03 1998-12-01 Redcell Canada, Inc. Cellular and serum protein anchors for modulating pharmacokinetics
US5487890A (en) * 1990-10-04 1996-01-30 University Of Virginia Patent Foundation Mammalian primate erythrocyte bound heteropolymerized monoclonal antibodies and methods of use thereof
US6066718A (en) * 1992-09-25 2000-05-23 Novartis Corporation Reshaped monoclonal antibodies against an immunoglobulin isotype
US5656273A (en) * 1994-01-18 1997-08-12 Genentech, Inc. Method of treatment of parasitic infection using IgE antagonists
US20020028204A1 (en) * 1994-11-30 2002-03-07 Ajinomoto Co., Inc. Antithrombotic agent and anti-von willebrand factor monoclonal antibody
US5837243A (en) * 1995-06-07 1998-11-17 Medarex, Inc. Therapeutic compounds comprised of anti-Fc receptor antibodies
US20040197326A1 (en) * 1995-07-27 2004-10-07 Genentech, Inc. Method for treatment of allergic asthma
US7368111B2 (en) * 1995-10-06 2008-05-06 Cambridge Antibody Technology Limited Human antibodies specific for TGFβ2
US5994511A (en) * 1997-07-02 1999-11-30 Genentech, Inc. Anti-IgE antibodies and methods of improving polypeptides
US20020054878A1 (en) * 1997-07-02 2002-05-09 Genentech, Inc. Anti-IgE antibodies
US6670453B2 (en) * 1997-10-27 2003-12-30 Unilever Patent Holdings B.V. Multivalent antigen-binding proteins
US20030092892A1 (en) * 1997-10-27 2003-05-15 Frenken Leon Gerardus Multivalent antigen-binding proteins
US20020076404A1 (en) * 1998-01-29 2002-06-20 Chang Tse Wen Treating atopic dermatitis with IgE antagonists
US6759518B1 (en) * 1998-04-09 2004-07-06 Vectron Therapeutics Ag Single-chain multiple antigen-binding molecule, its preparation and use
US20010024647A1 (en) * 1998-10-23 2001-09-27 Handin Robert I. Conformation-specific anti-von Willebrand factor antibodies
US6251393B1 (en) * 1998-10-23 2001-06-26 The Brigham And Women's Hospital, Inc. Conformation-specific anti-von Willebrand Factor antibodies
US6419934B1 (en) * 1999-02-24 2002-07-16 Edward L. Tobinick TNF modulators for treating neurological disorders associated with viral infection
US6504013B1 (en) * 2000-02-01 2003-01-07 Idexx Laboratories, Inc. Canine allergy therapeutic recombinant chimeric anti-IgE monoclonal antibody
US20040180046A1 (en) * 2000-04-26 2004-09-16 Jeff Himawan Bispecific molecules and uses thereof
US20020009453A1 (en) * 2000-06-16 2002-01-24 Symphogen A/S Polyclonal antibody composition for treating allergy
US6902734B2 (en) * 2000-08-07 2005-06-07 Centocor, Inc. Anti-IL-12 antibodies and compositions thereof
US7897151B2 (en) * 2000-08-30 2011-03-01 Pharmacia & Upjohn Company, Llc Anti-IgE vaccines
US20020058033A1 (en) * 2000-10-13 2002-05-16 Raisch Kevin Paul Human anti-epidermal growth factor receptor single-chain antibodies
US20020132275A1 (en) * 2000-11-03 2002-09-19 Board Of Regents, The University Of Texas System Methods for detecting the efficacy of anticancer treatments
US20030118592A1 (en) * 2001-01-17 2003-06-26 Genecraft, Inc. Binding domain-immunoglobulin fusion proteins
US7589180B2 (en) * 2001-05-11 2009-09-15 Abbott Laboratories Inc. Specific binding proteins and uses thereof
US20040219643A1 (en) * 2001-06-28 2004-11-04 Greg Winter Dual-specific ligand
US20070031424A1 (en) * 2001-10-24 2007-02-08 Vlaams Interuniversitair Instituut Voor Biotechnogie Vzw Functional heavy chain antibodies, fragments thereof, library thereof and methods of production thereof
US7300655B2 (en) * 2002-08-01 2007-11-27 Immunomedics, Inc. Alpha-fetoprotein Immu31 antibodies and fusion proteins and methods of use thereof
US20060115470A1 (en) * 2002-11-08 2006-06-01 Ablynx N. V. Camelidae antibodies against imminoglobulin e and use thereof for the treatment of allergic disorders
US20110184145A1 (en) * 2002-11-08 2011-07-28 Ablynx N.V. Method of administering therapeutic polypeptides, and polypeptides therefor
US20070178082A1 (en) * 2002-11-08 2007-08-02 Ablynx N.V. Stabilized single domain antibodies
US20110184151A1 (en) * 2002-11-08 2011-07-28 Ablynx N.V. Single domain antibodies directed against epidermal growth factor receptor and uses therefor
US20110178277A1 (en) * 2002-11-08 2011-07-21 Ablynx N.V. Stabilized single domain antibodies
US20110123529A1 (en) * 2002-11-08 2011-05-26 Ablynx N.V. Single domain antibodies directed against epidermal growth factor receptor and uses therefor
US20090022721A1 (en) * 2002-11-08 2009-01-22 Ablynx N.V. Single domain antibodies directed against tumour necrosis factor-alpha and uses therefor
US20060034845A1 (en) * 2002-11-08 2006-02-16 Karen Silence Single domain antibodies directed against tumor necrosis factor alpha and uses therefor
US20090238829A1 (en) * 2002-11-08 2009-09-24 Ablynx N.V. Stabilized single domain antibodies
US20110184150A1 (en) * 2002-11-08 2011-07-28 Ablynx N.V. Single domain antibodies directed against tumor necrosis factor-alpha and uses therefor
US20090324512A1 (en) * 2002-11-08 2009-12-31 Ablynx N.V. Polypeptide constructs for nasal administration
US20100003248A1 (en) * 2002-11-08 2010-01-07 Ablynx N.V Polypeptide constructs for rectal and/or vaginal administration
US20100003249A1 (en) * 2002-11-08 2010-01-07 Ablynx N.V. Polypeptide constructs for topical administration
US20070077249A1 (en) * 2002-11-08 2007-04-05 Ablynx N.V. Single domain antibodies directed against tumour necrosis factor-alpha and uses therefor
US20100021459A1 (en) * 2002-11-08 2010-01-28 Ablynx N.V. Polypeptide constructs for intracellular delivery
US20100040613A1 (en) * 2002-11-08 2010-02-18 Ablynx N.V. Polypeptide constructs for sublingual administration
US20110027281A1 (en) * 2002-11-08 2011-02-03 Ablynx N.V. Single domain antibodies directed against tumor necrosis factor-alpha and uses therefor
US20060034833A1 (en) * 2002-11-08 2006-02-16 Els Beirnaert Single domain antibodies directed against interferron-gamma and uses therefor
US7054257B2 (en) * 2003-04-29 2006-05-30 International Business Machines Corporation AFM-based data storage and microscopy
US20060228355A1 (en) * 2003-11-07 2006-10-12 Toon Laeremans Camelidae single domain antibodies vhh directed against epidermal growth factor receptor and uses therefor
US7598350B2 (en) * 2004-03-19 2009-10-06 Imclone Llc Human anti-epidermal growth factor receptor antibody
US20070264253A1 (en) * 2004-03-19 2007-11-15 Meilin Liu Human Anti-Epidermal Growth Factor Receptor Antibody
US20100003258A1 (en) * 2008-07-01 2010-01-07 Zhigang Weng Fibroblast growth factor receptor 3 (fgfr3) binding proteins

Cited By (58)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040253638A1 (en) * 1992-08-21 2004-12-16 Vrije Universiteit Brussel Immunoglobulins devoid of light chains
US20060121026A1 (en) * 1992-08-21 2006-06-08 Vrije Universiteit Brussel Immunoglobulins devoid of light chains
US20060134107A1 (en) * 1992-08-21 2006-06-22 Vrije Universiteit Brussel Immunoglobulins devoid of light chains
US20070025985A1 (en) * 1992-08-21 2007-02-01 Vrije Universiteit Brussel Immunoglobulins devoid of light chains
US20070077240A1 (en) * 1992-08-21 2007-04-05 Vrije Universiteit Brussel Immunoglobulins devoid of light chains
US20070238133A1 (en) * 1992-08-21 2007-10-11 Vrije Universiteit Brussel Immunoglobulins devoid of light chains
US7722871B2 (en) 1992-08-21 2010-05-25 Vrije Universiteit Brussel Immunoglobulins devoid of light chains
US7786047B2 (en) 1992-08-21 2010-08-31 Vrije Universiteit Brussel Immunoglobulins devoid of light chains
US7790367B2 (en) 1992-08-21 2010-09-07 Vrije Universiteit Brussel Method for screening a library of VHH polypeptides
US20050054001A1 (en) * 2001-10-24 2005-03-10 Serge Muyldermans Functional heavy chain antibodies, fragments thereof, library thereof and methods of production thereof
US8097251B2 (en) 2001-10-24 2012-01-17 Vlaams Interuniversitair Instituut Voor Biotechnologie Vzw Functional heavy chain antibodies, fragments thereof, library thereof and methods of production thereof
US9156905B2 (en) 2001-10-24 2015-10-13 Vib Vzw Functional heavy chain antibodies, fragments thereof, library thereof and methods of production thereof
US20070031424A1 (en) * 2001-10-24 2007-02-08 Vlaams Interuniversitair Instituut Voor Biotechnogie Vzw Functional heavy chain antibodies, fragments thereof, library thereof and methods of production thereof
US20100047241A1 (en) * 2001-10-24 2010-02-25 Vlaams Interuniversitair Instituut Voor Biotechnologies Vzw Functional heavy chain antibodies, fragments thereof, library thereof and methods of production thereof
US10041102B2 (en) 2002-10-08 2018-08-07 Pfenex Inc. Expression of mammalian proteins in Pseudomonas fluorescens
US20110178277A1 (en) * 2002-11-08 2011-07-21 Ablynx N.V. Stabilized single domain antibodies
US20090022721A1 (en) * 2002-11-08 2009-01-22 Ablynx N.V. Single domain antibodies directed against tumour necrosis factor-alpha and uses therefor
US20100021459A1 (en) * 2002-11-08 2010-01-28 Ablynx N.V. Polypeptide constructs for intracellular delivery
US20100040613A1 (en) * 2002-11-08 2010-02-18 Ablynx N.V. Polypeptide constructs for sublingual administration
US20100003248A1 (en) * 2002-11-08 2010-01-07 Ablynx N.V Polypeptide constructs for rectal and/or vaginal administration
US20100003249A1 (en) * 2002-11-08 2010-01-07 Ablynx N.V. Polypeptide constructs for topical administration
US9243065B2 (en) 2002-11-08 2016-01-26 Ablynx N.V. Polypeptide constructs including VHH directed against EGFR for intracellular delivery
US20060034845A1 (en) * 2002-11-08 2006-02-16 Karen Silence Single domain antibodies directed against tumor necrosis factor alpha and uses therefor
US20090324512A1 (en) * 2002-11-08 2009-12-31 Ablynx N.V. Polypeptide constructs for nasal administration
US20090238829A1 (en) * 2002-11-08 2009-09-24 Ablynx N.V. Stabilized single domain antibodies
US9725522B2 (en) 2002-11-08 2017-08-08 Ablynx N.V. Pulmonary administration of immunoglobulin single variable domains and constructs thereof
US20110027281A1 (en) * 2002-11-08 2011-02-03 Ablynx N.V. Single domain antibodies directed against tumor necrosis factor-alpha and uses therefor
US20110123529A1 (en) * 2002-11-08 2011-05-26 Ablynx N.V. Single domain antibodies directed against epidermal growth factor receptor and uses therefor
US9320792B2 (en) 2002-11-08 2016-04-26 Ablynx N.V. Pulmonary administration of immunoglobulin single variable domains and constructs thereof
US20110184145A1 (en) * 2002-11-08 2011-07-28 Ablynx N.V. Method of administering therapeutic polypeptides, and polypeptides therefor
US20110184150A1 (en) * 2002-11-08 2011-07-28 Ablynx N.V. Single domain antibodies directed against tumor necrosis factor-alpha and uses therefor
US20110184151A1 (en) * 2002-11-08 2011-07-28 Ablynx N.V. Single domain antibodies directed against epidermal growth factor receptor and uses therefor
US20070178082A1 (en) * 2002-11-08 2007-08-02 Ablynx N.V. Stabilized single domain antibodies
US20070077249A1 (en) * 2002-11-08 2007-04-05 Ablynx N.V. Single domain antibodies directed against tumour necrosis factor-alpha and uses therefor
US20100003253A1 (en) * 2002-11-08 2010-01-07 Ablynx N.V. Single domain antibodies directed against epidermal growth factor receptor and uses therefor
US9371381B2 (en) 2002-11-08 2016-06-21 Ablynx, N.V. Single domain antibodies directed against tumor necrosis factor-alpha and uses therefor
US8603824B2 (en) 2004-07-26 2013-12-10 Pfenex, Inc. Process for improved protein expression by strain engineering
US9109229B2 (en) 2004-07-26 2015-08-18 Pfenex Inc. Process for improved protein expression by strain engineering
US9067991B2 (en) 2005-05-18 2015-06-30 Ablynx N.V. Nanobodies against tumor necrosis factor-alpha
US8188223B2 (en) 2005-05-18 2012-05-29 Ablynx N.V. Serum albumin binding proteins
US11472871B2 (en) 2005-05-18 2022-10-18 Ablynx N.V. Nanobodies against tumor necrosis factor-alpha
US8703131B2 (en) 2005-05-21 2014-04-22 Ablynx N.V. Nanobodies against tumor necrosis factor-alpha
US20100297111A1 (en) * 2005-05-21 2010-11-25 Els Anna Alice Beirnaert Nanobodies against tumor necrosis factor-alpha
US20070269422A1 (en) * 2006-05-17 2007-11-22 Ablynx N.V. Serum albumin binding proteins with long half-lives
US9580719B2 (en) 2007-04-27 2017-02-28 Pfenex, Inc. Method for rapidly screening microbial hosts to identify certain strains with improved yield and/or quality in the expression of heterologous proteins
US9394571B2 (en) 2007-04-27 2016-07-19 Pfenex Inc. Method for rapidly screening microbial hosts to identify certain strains with improved yield and/or quality in the expression of heterologous proteins
US10689640B2 (en) 2007-04-27 2020-06-23 Pfenex Inc. Method for rapidly screening microbial hosts to identify certain strains with improved yield and/or quality in the expression of heterologous proteins
US9393304B2 (en) 2008-10-29 2016-07-19 Ablynx N.V. Formulations of single domain antigen binding molecules
US9993552B2 (en) 2008-10-29 2018-06-12 Ablynx N.V. Formulations of single domain antigen binding molecules
US20100172894A1 (en) * 2008-10-29 2010-07-08 Wyeth Methods for purification of single domain antigen binding molecules
US10118962B2 (en) 2008-10-29 2018-11-06 Ablynx N.V. Methods for purification of single domain antigen binding molecules
US11370835B2 (en) 2008-10-29 2022-06-28 Ablynx N.V. Methods for purification of single domain antigen binding molecules
WO2014033313A1 (en) 2012-09-03 2014-03-06 Vib Vzw Protective anti-etec antibody
US10633438B2 (en) 2015-03-31 2020-04-28 Vhsquared Limited Polypeptides
WO2016156474A1 (en) * 2015-03-31 2016-10-06 Vhsquared Limited Polypeptide comprising an immunoglobulin chain variable domain which binds to clostridium difficile toxin a
US11684677B2 (en) 2016-09-30 2023-06-27 Sorriso Pharmaceuticals, Inc. Compositions
US11623952B2 (en) 2019-06-21 2023-04-11 Sorriso Pharmaceuticals, Inc. IL-23 and TNF-alpha binding bi-specific heavy chain polypeptides
US11667719B2 (en) 2019-06-21 2023-06-06 Sorriso Pharmaceuticals, Inc. VHH immunoglobulin chain variable domain that binds to IL-7R and methods of use thereof for treating autoimmune and/or inflammatory diseases

Also Published As

Publication number Publication date
US20070077249A1 (en) 2007-04-05
US20090022721A1 (en) 2009-01-22
US9371381B2 (en) 2016-06-21
US20120202977A1 (en) 2012-08-09
US20110027281A1 (en) 2011-02-03
US20150110782A1 (en) 2015-04-23
US20170096481A1 (en) 2017-04-06
US20110184150A1 (en) 2011-07-28
US20060034845A1 (en) 2006-02-16

Similar Documents

Publication Publication Date Title
US9371381B2 (en) Single domain antibodies directed against tumor necrosis factor-alpha and uses therefor
EP1558647B1 (en) Single domain antibodies directed against tumour necrosis factor-alpha and uses therefor
EP2390268B1 (en) Single domain antibodies directed against tumour necrosis factor-alpha and uses therefor
CA2608770C (en) Improved nanobodies against tumor necrosis factor-alpha
ES2551682T3 (en) Single domain antibodies directed against tumor necrosis factor-alpha and uses for them
US20110184151A1 (en) Single domain antibodies directed against epidermal growth factor receptor and uses therefor
US20060034833A1 (en) Single domain antibodies directed against interferron-gamma and uses therefor
CN1735629B (en) Single domain antibodies directed against tumor necrosis factor alpha and uses therefor

Legal Events

Date Code Title Description
AS Assignment

Owner name: ABLYNX N.V., BELGIUM

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:SILENCE, KAREN;LAUWEREYS, MARC;DE HAARD, HANS;REEL/FRAME:019272/0365

Effective date: 20060807

STCB Information on status: application discontinuation

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