WO2007005608A2 - Il-12/p40 binding proteins - Google Patents

Il-12/p40 binding proteins Download PDF

Info

Publication number
WO2007005608A2
WO2007005608A2 PCT/US2006/025584 US2006025584W WO2007005608A2 WO 2007005608 A2 WO2007005608 A2 WO 2007005608A2 US 2006025584 W US2006025584 W US 2006025584W WO 2007005608 A2 WO2007005608 A2 WO 2007005608A2
Authority
WO
WIPO (PCT)
Prior art keywords
seq
residues
cdr
binding protein
antibody
Prior art date
Application number
PCT/US2006/025584
Other languages
French (fr)
Other versions
WO2007005608A3 (en
Inventor
Susan E. Lacy
Emma Fung
Jonathan P. Belk
Richard W. Dixon
Michael Roguska
Paul R. Hinton
Shankar Kumar
Original Assignee
Abbott Laboratories
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to MX2007016401A priority Critical patent/MX2007016401A/en
Priority to KR1020137014441A priority patent/KR20130080058A/en
Priority to JP2008519612A priority patent/JP5396080B2/en
Priority to CA002612239A priority patent/CA2612239A1/en
Priority to EP20060785967 priority patent/EP1907421A4/en
Priority to CN2006800240973A priority patent/CN101379085B/en
Priority to BRPI0611714-7A priority patent/BRPI0611714A2/en
Priority to AU2006265932A priority patent/AU2006265932B2/en
Application filed by Abbott Laboratories filed Critical Abbott Laboratories
Publication of WO2007005608A2 publication Critical patent/WO2007005608A2/en
Priority to IL187691A priority patent/IL187691A/en
Priority to NO20080557A priority patent/NO20080557L/en
Publication of WO2007005608A3 publication Critical patent/WO2007005608A3/en
Priority to IL216803A priority patent/IL216803A0/en
Priority to IL228793A priority patent/IL228793A0/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/24Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against cytokines, lymphokines or interferons
    • C07K16/244Interleukins [IL]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/715Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/14Prodigestives, e.g. acids, enzymes, appetite stimulants, antidyspeptics, tonics, antiflatulents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/04Antipruritics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/14Drugs for dermatological disorders for baldness or alopecia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/04Centrally acting analgesics, e.g. opioids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/18Antipsychotics, i.e. neuroleptics; Drugs for mania or schizophrenia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/24Antidepressants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/30Drugs for disorders of the nervous system for treating abuse or dependence
    • A61P25/32Alcohol-abuse
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/10Antimycotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/02Non-specific cardiovascular stimulants, e.g. drugs for syncope, antihypotensives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/04Inotropic agents, i.e. stimulants of cardiac contraction; Drugs for heart failure
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6863Cytokines, i.e. immune system proteins modifying a biological response such as cell growth proliferation or differentiation, e.g. TNF, CNF, GM-CSF, lymphotoxin, MIF or their receptors
    • G01N33/6869Interleukin
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/567Framework region [FR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value

Definitions

  • the present invention relates to IL-12p40 binding proteins, and specifically to their uses in the prevention and/or treatment of acute and chronic inflammatory diseases.
  • IL-12 Human interleukin-12
  • IL-12 is a cytokine with a unique structure and pleiotropic effects (Kobayashi, et al. (1989) J Exp Med 170:827-845; Seder, et al. (1993) Proc. Natl. Acad. Sd. 90:10188-10192, Ling, et al. (1995) J Exp Med 154:116-127; Podlaski, et al. (1992) Arch. Biochem. Biophys. 294:230-237).
  • IL-12 plays a critical role in the pathology associated with several diseases involving immune and inflammatory responses. A review of IL-12, its biological activities, and its role in disease can be found in Trinchieri, G. (2003) Nat. Rev.
  • IL-12 is a heterodimeric protein (referred to as the "p70 protein") comprising a 35 kDa subunit (p35) and a 40 kDa subunit (p40) which are linked together by a disulfide bridge.
  • the heterodimeric protein is produced primarily by antigen-presenting cells such as monocytes, macrophages and dendritic cells. These cell types also secrete an excess of the p40 subunit relative to p70 subunit.
  • the p40 and p35 subunits are genetically unrelated and neither has been reported to possess biological activity, although the p40 homodimer may function as an IL- 12 antagonist.
  • IL-12 plays a central role in regulating the balance between antigen-specific T helper type 1 (ThI) and type 2 (Th2) lymphocytes.
  • ThI and Th2 cells govern the initiation and progression of autoimmune disorders, and IL-12 is critical in the regulation of ThI- lymphocyte differentiation and maturation.
  • Cytokines released by the ThI cells are inflammatory and include interferon gamma (IFN- ⁇ ), IL-2, and lymphotoxin (LT).
  • Th2 cells secrete IL-4, IL-5, IL-6, IL-10 and IL-13 to facilitate humoral immunity, allergic reactions, and immunosuppression.
  • IL-12 may play a major role in the pathology associated with many autoimmune and inflammatory diseases such as rheumatoid arthritis (RA), multiple sclerosis (MS), psoriasis (PS) and Crohn's disease (CD).
  • RA rheumatoid arthritis
  • MS multiple sclerosis
  • PS psoriasis
  • CD Crohn's disease
  • Cytokine messenger ribonucleic acid (mRNA) expression profile in the RA synovia identified predominantly ThI cytokines (Bucht et al. (1996) Clin. Exp. Immunol. 103:347-367).
  • IL-12 also appears to play a critical role in the pathology associated with Crohn's disease. Increased expression of EMF- ⁇ and IL-12 has been observed in the intestinal mucosa of patients with this disease (Fais et al. (1994) J Interferon Res. 14:235-238; Parronchi et al. (1997) Am. J. Path. 150:823-832; Monteleone et al. (1997) Gastroent. 112:1169- 1178, and Berrebi et al.
  • the cytokine secretion profile of T cells from the lamina intestinal of CD patients is characteristic of a predominantly ThI response, including greatly elevated IFN- ⁇ levels (Fuss, et al. (1996) J Immunol. 157:1261-1270). Moreover, colon tissue sections from CD patients show an abundance of IL- 12 expressing macrophages and IFN- ⁇ expressing T cells (Parronchi et al (1997) Am. J. Path. 150:823-832).
  • IL-23 is also a heterodimeric cytokine and belongs to a family of five such heterodimeric cytokines including IL-12 and IL-27 (Trinchieri et al., (2003) Immunity 19:641-644).
  • IL-23 shares the identical p40 subunit as IL-12, but it is associated with a pl9 subunit via a disulphide- linkage.
  • the pl9 subunit is structurally related to IL-6, granulocyte-colony stimulating factor (G- CSF), and the p35 subunit of IL-12.
  • IL-23 is produced by similar cell types as JLL-12, ana its receptor is expressed on T cells, NK cells, and phagocytic and dendritic hematopoietic cells.
  • IL- 23 mediates signaling by binding to a heterodimeric receptor, comprised of BL-23R and IL- 12betal.
  • the EL-12betal subunit is shared by the 1L-12 receptor, which is composed of IL- 12betal and IL-12beta2.
  • DL-23 does share overlapping functions with IL-12 (by inducing IFN- ⁇ production, ThI cell differentiation and activating the antigen-presenting functions of dendritic cells) however it selectively induces proliferation of memory T cells (Oppmann et al. (2000) Immunity 13:715-725, Parham, et al. (2002) J. Immunol. 168:5699-5708).
  • IL-23 The role of IL-23 in autoimmune inflammation has been dissected in part through studies with pl9 knockout mice (Murphy et al. J Exp Med 198:1951-1957; Cua et al. (2003) Nature 421:744-748). Studies have demonstrated that IL-23 modulates immune response to infection (see, e.g., Pirhonen, et al. (2002) J. Immunol. 169:5673-5678; Broberg, et al. (2002) J. Interferon Cytokine Res. 22:641-651; Elkins, et al. (2002) Infection Immunity 70:1936-1948; Cooper, et al. (2002) J. Immunol. 168:1322-1327). IL-23 is thought to play a role in immune-mediated inflammatory diseases (Langrish et.al. (2004) Immunological Reviews202: 96-105).
  • IL-12 Due to the role of human IL-12 in a variety of human disorders, therapeutic strategies have been designed to inhibit or counteract IL-12 activity.
  • antibodies that bind to, and neutralize, IL-12 have been sought as a means to inhibit IL-12 activity.
  • Some of the earliest antibodies were murine monoclonal antibodies (mAbs), secreted by hybridomas prepared from lymphocytes of mice immunized with IL-12 (see e.g., Strober et al., PCT Publication No. WO 97/15327; Gately et al., WO9937682 A2; Neurath et al., J Exp. Med 182:1281-1290 (1995); Duchmarm et al., J Immunol.
  • mAbs murine monoclonal antibodies
  • murine IL-12 antibodies are limited for their use in vivo due to problems associated with administration of mouse antibodies to humans, such as short serum half life, an inability to trigger certain human effector functions and elicitation of an unwanted immune response against the mouse antibody in a human (the "human anti-mouse antibody” (HAMA) reaction).
  • HAMA human anti-mouse antibody
  • Such chimeric antibodies to IL-12 are also disclosed in Peritt et al. PCT publication No.WO2002097048A2. However, because these chimeric antibodies still retain murine variable chain sequences, they still may elicit an unwanted immune reaction, the human anti-chimeric antibody (HACA) reaction especially when administered for prolonged periods.
  • HACA human anti-chimeric antibody
  • the antibodies capable of binding the p40 subunit of IL-12 (IL-12p40).
  • the antibodies bind IL-12 and /or IL-23.
  • the antibodies are capable of neutralizing IL-12 and /or 1L-23.
  • the present invention provides a novel family of binding proteins, CDR grafted antibodies, humanized antibodies, and fragments thereof, capable binding IL-12p40, binding with high affinity, and binding and neutralizing IL-12 and/or IL-23.
  • This invention pertains to IL-12p40 binding proteins, particularly antibodies capable of binding the p40 subunit of human IL-12 and the p40 subunit of human IL-23. Further, the invention provides methods of making and using IL-12p40 binding proteins.
  • One aspect of this invention pertains to a binding protein comprising an antigen binding domain capable of binding a p40 subunit of IL-12.
  • the antigen binding domain comprises at least one CDR comprising an amino acid sequence selected from the group consisting of:
  • X 1 is D, K, T, or S
  • X 2 is Y, S, or T
  • X 3 is Y, V, G, W, S, or F;
  • X 4 is I, or M
  • X 5 is H, G, E, or V;
  • X 6 is V, or is not present
  • X 7 is S, or is not present
  • X 1 is H, D, G, W, S, Y or R;
  • X 2 is I, or F
  • X 3 is Y, W, L, S, N, D or G;
  • X 4 is W, P, H, T, or S;
  • X 5 is D, G, E, A, or I;
  • X 6 is D, G, S, T, or N;
  • X 7 is D, G, S, or P;
  • X 8 is K, N, S, E, T, or H
  • X 9 is Y, T, P, I, or N;
  • Xio is Y, N, T, H, K, S, or G;
  • Xn is N, or Y;
  • Xi 2 is P, N, A, D, or S;
  • X 13 is S, E, D, or P;
  • X 15 is K, F, V, M, R, or A;
  • Xi 6 is S, K, Q, P, or is not present
  • Xn is D, G, R, or is not present
  • Xi 8 is F, or is not present
  • Xi 9 is Q, or is not present
  • X 20 is D, or is not present; CDR-H3.
  • X 2 is G, T, R, P, or H
  • X 3 is I, R, F, Y, or Q;
  • X 4 is R, V, Y, F, or A;
  • X 5 is S, N, G, A, or R;
  • X 6 is A, Y, L, F, or M;
  • X 7 is M, A, D, L, or F
  • X 8 is D, M, Y, or W
  • X 9 is Y, D, or N
  • X 10 is Y, A, or is not present
  • Xn is M, or is not present
  • Xi 2 is D, or is not present
  • Xi 3 is Y, or is not present; CDR-Ll.
  • X 2 is A
  • X 3 is S
  • X 4 is Q, or E
  • X 5 is S, or N
  • X 6 is V, or I
  • X 7 is S, G, or D
  • X 8 is N, T, or K
  • X 9 is D, N, or Y;
  • X 10 is V, G, or L;
  • Xn is A, I, or H
  • X 12 is S, or is not present
  • Xi 3 is F, or is not present
  • Xi 4 is M, or is not present
  • Xi 5 is N, or is not present; CDR-L2.
  • X 1 -X 2 -X 3 -X 4 -X 5 -X 6 -X 7 -X 8 (SEQ ID NO: 59), wherein;
  • X 1 is Y, or S
  • X 2 is A, or T;
  • X 3 is S, or A
  • X 4 is N, H, S, or Q
  • X 5 is R, N, or S
  • X 6 is Y, Q, or I
  • X 7 is T, S, or G
  • X 8 is S, or is not present; and CDR-L3.
  • X 1 -X 2 -X 3 -X 4 -X 5 -X 6 -X 7 -X 8 -X 9 (SEQ ID NO: 60), wherein;
  • X 3 is D, Y, or S
  • X 4 is Y, N, K, or I
  • X 5 is N, T, S, or E;
  • X 6 is S, Y, V, or W
  • X 7 is P
  • X 8 is W, F, Y, L, or P
  • X 9 is T, or S.
  • the antigen binding domain comprises at least one CDR comprising an amino acid sequence selected from the group consisting of residues 31-37 of SEQ ID NO.:35;residues 52-67 of SEQ ID NO.:35;residues 100-108 of SEQ ID NO.:35;residues 24-34 of SEQ ID NO.:36;residues 50-56 of SEQ ID NO.:36;residues 89-97 of SEQ ID NO.:36;residues 31-37 of SEQ ID NO.:37;residues 52-67 of SEQ ID NO.:37 residues.100-109 of SEQ ID NO.:37;residues 24-34 of SEQ ID NO.:38;residues 50-56 of SEQ ID NO.:38;residues 89-97 of SEQ ID NO.:38;residues 31-35 of SEQ ID NO.:39;residu
  • VH 3G7 CDR-H3 Residues 99-106 of SEQ ID NO. . 41
  • VL 3G7 CDR Set
  • VH 3 All CDR-Hl Residues 31-35 of SEQ ID NO. 49
  • VH 4B4 CDR-Hl Residues 31-37 of SEQ ID NO. : 51
  • VL 4B4 CDR-Ll Residues 24-34 of SEQ ID NO. : 52
  • the binding protein of the invention comprises at least two variable domain CDR sets. More preferably, the two variable domain CDR sets are selected from a group consisting of: VH 1D4 CDR Set & VL 1D4 CDR Set; VH 1A6 CDR Set & VL 1A6 CDR Set; VH 1D8 CDR Set & VL 1D8 CDR Set; VH 3G7 CDR Set & VL 3G7 CDR Set; VH 5E8 CDR Set & VL 5E8 CDR Set; VH 8El CDR Set & VL 8El CDR Set; VH 1H6 CDR Set & VL 1H6 CDR Set; VH 3Al 1 CDR Set & VL 3Al 1 CDR Set; VH 4B4 CDR Set & VL 4B4 CDR Set; and VH 7G3 CDR Set & VL 7G3
  • the binding protein disclosed above further comprises a human acceptor framework.
  • the human acceptor framework comprises a amino acid sequence selected from the group consisting of SEQ ID NO.:6; SEQ ID NO.:7; SEQ ID NO.:8; SEQ ID NO.:9; SEQ ID NO.: 10; SEQ ID NO.: 11; SEQ ID NO.: 12; SEQ ID NO.: 13; SEQ ID NO.: 14; SEQ ID NO.: 15; SEQ ID NO.: 16; SEQ ID NO.: 17; SEQ ID NO.: 18; SEQ ID NO.: 19; SEQ ID NO.:20; SEQ ID NO.:21; SEQ ID NO.:22; SEQ ID NO.:23; SEQ ID NO.:24; SEQ ID NO.:25; SEQ ID NO.:26; SEQ ID NO.:27; SEQ ID NO.:28; SEQ ID NO.:29; SEQ ID NO.:30; SEQ ID NO.:31; SEQ ID NO.:32; SEQ ID NO
  • the binding protein is a CDR grafted antibody or antigen binding portion thereof capable of binding the p40 subunit of IL-12 or IL-23.
  • the CDR grafted antibody or antigen binding portion thereof comprise one or more CDRs disclosed above.
  • the CDR grafted antibody or antigen binding portion thereof comprises at least one variable domain having an amino acid sequence selected from the group consisting of SEQ ID NO.:61; SEQ ID NO.:62; SEQ ID NO.:63; SEQ ID NO.:64; SEQ ID NO.:65; SEQ ID NO.:66; SEQ ID NO.:67; SEQ ID NO.:68; SEQ ID NO.:69; SEQ ID NO.:70; SEQ ID NO.:71; SEQ ID NO.:72; SEQ ID NO.:73; SEQ ID NO.:74; SEQ ID NO.:75; SEQ ID NO.:76; SEQ ID NO.:77; and SEQ TD NO.:78.
  • the CDR grafted antibody or antigen binding portion thereof comprises two variable domains selected from the group disclosed above.
  • the CDR grafted antibody or antigen binding portion thereof comprises a human acceptor framework. More preferably the human acceptor framework is any one of the human acceptor frameworks disclosed above.
  • the binding protein is a humanized antibody or antigen binding portion thereof capable of binding the p40 subunit of IL-12 or IL-23.
  • the humanized antibody or antigen binding portion thereof comprise one or more CDRs disclosed above incorporated into a human antibody variable domain of a human acceptor framework.
  • the human antibody variable domain is a consensus human variable domain.
  • the human acceptor framework comprises at least one Framework Region amino acid substitution at a key residue, wherein the key residue is selected from the group consisting of a residue adjacent to a CDR; a glycosylation site residue; a rare residue; a residue capable of interacting with a p40 subunit of human IL- 12; a residue capable of interacting with a CDR; a canonical residue; a contact residue between heavy chain variable region and light chain variable region; a residue within a Vernier zone; and a residue in a region that overlaps between a Chothia-defined variable heavy chain CDRl and a Kabat-defined first heavy chain framework.
  • the key residue is selected from the group consisting of a residue adjacent to a CDR; a glycosylation site residue; a rare residue; a residue capable of interacting with a p40 subunit of human IL- 12; a residue capable of interacting with a CDR; a canonical residue; a contact residue between heavy chain variable region and light chain variable region
  • the key residue is selected from the group consisting of 3H, 5H, 1OH, HH, 12H, 13H, 15H, 16H, 18H, 19H, 23H, 24H, 25H, 3OH, 41H, 44H, 46H, 49H, 66H, 68H, 71H, 73H, 74H, 75H, 76H, 77H, 78H, 79H, 8 IH, 82H, 82AH, 82BH, 82CH, 83H, 84H, 85H, 86H, 87H, 89H, 93H, 98H, 108H, 109H, IL, 2L, 3L, 7L, 8L, 9L, 1OL, HL, 12L, 13L, 15L, 17L, 19L, 2OL, 21L, 22L, 36L, 41L, 42L, 43L, 45L, 46L, 58L, 6OL, 62L, 63L, 67L, 7OL,
  • the binding protein is a humanized antibody or antigen binding portion thereof capable of binding the p40 subunit of IL- 12 or IL-23.
  • the humanized antibody, or antigen binding portion, thereof comprises one or more CDRs disclosed above. More preferably the humanized antibody, or antigen binding portion, thereof comprises three or more CDRs disclosed above. Most preferably the humanized antibody, or antigen binding portion, thereof comprises six CDRs disclosed above.
  • the humanized antibody or antigen binding portion thereof comprises at least one variable domain having an amino acid sequence selected from the group consisting of SEQ ID NO.:79, SEQ ID NO.:80, SEQ ID NO.:81, SEQ ID NO.:82, SEQ ID NO.:83, SEQ ID NO.:84, SEQ ID NO.:85, SEQ ID NO.:86, SEQ ID NO.:87, SEQ ID NO.:88, SEQ ID NO.:89, SEQ ID NO.:90, SEQ ID NO.:91, SEQ ID NO.:98, SEQ ID NO.:99, SEQ ID NO.: 100, SEQ ID NO.: 101, SEQ ID NO.: 102, AND SEQ ID NO.: 103, SEQ ID NO.: 104, SEQ ED NO.: 105, SEQ DD NO.: 106, SEQ ID NO.: 107, SEQ ID NO.: 108, and SEQ ID NO.: 109.
  • humanized antibody or antigen binding portion thereof comprises two variable domains selected from the group disclosed above.
  • humanized antibody, or antigen binding portion thereof comprises two variable domains, wherein said two variable domains have amino acid sequences selected from the group consisting of SEQ ID NO.:67 & SEQ ID NO.:79, SEQ ID NO.:80 & SEQ ID NO.:81, SEQ ID NO.:82 & SEQ ID NO.:83, SEQ ID NO.:84 & SEQ ID NO.:85, SEQ ID NO.:86 & SEQ ID NO.:87, SEQ ID NO.:88 & SEQ ID NO.:89, SEQ ID NO.:90 & SEQ ID NO.:91, SEQ ID NO.:98 & SEQ ID NO.:99, SEQ ID NO.: 100 & SEQ ID NO.: 101, SEQ ID NO.: 102 & SEQ ID NO.: 103, SEQ ID NO.: 104 & SEQ ID NO.: 105, SEQ ID NO.
  • the binding protein disclosed above comprises a heavy chain immunoglobulin constant domain selected from the group consisting of a human IgM constant domain, a human IgGl constant domain, a human IgG2 constant domain, a human IgG3 constant domain, a human IgG4 constant domain, a human IgE constant domain, and a human IgA constant domain. More preferably, the binding protein comprises SEQ ID NO.:2; SEQ DD NO.:3; SEQ ID NO.:4; and SEQ ID NO.:5.
  • the binding protein of the invention is capable of binding a target selected from the group consisting of IL-12 and IL-23.
  • the binding protein is capable of modulating a biological function of a target selected from the group consisting of IL-12 and IL-23. More preferably the binding protein is capable of neutralizing a target selected from the group consisting of IL-12 and IL-23.
  • the binding protein of the invention has an on rate constant (Kon) to IL-12 or IL-23 of at least about 10 2 M -1 S '1 ; at least about 10 3 M 4 S “1 ; at least about 10 4 M 4 S “1 ; at least about 10 5 M -1 S “1 ; or at least about 10 6 M -1 S “1 , as measured by surface plasmon resonance.
  • Kon on rate constant
  • the binding protein of the invention has an on rate constant (Kon) to IL-12 or IL-23 between 10 2 M- 1 S “1 to 10 3 M- 1 S “1 ; between 10 3 M- 1 S “1 to 10 4 M- 1 S “1 ; between lOVrV 1 to 10 5 M- 1 S “1 ; or between 10 5 M- 1 S “1 to 10 6 M- 1 S “1 , as measured by surface plasmon resonance.
  • Kon on rate constant
  • the binding protein of the invention has an off rate constant (Koff) to IL-12 or IL-23 of at most about 10 "3 S “1 ; at most about 10 "4 S “1 ; at most about 10 "5 S “1 ; or at most about 10 "6 S “1 , as measured by surface plasmon resonance.
  • the binding protein of the invention has an off rate constant (Koff) to IL-12 or IL-23 of 10 " Y 1 to 10 "4 S “1 ; of 10 "4 S “1 to 10 “ 5 S “1 ; or of 10 "5 S “1 to 10 '6 S “1 , as measured by surface plasmon resonance.
  • the binding protein of the invention has a dissociation constant (K D ) to IL-12 or IL-23 of at most about lO '7 M; at most about lO '8 M; at most about 10 "9 M; at most about 10 "10 M; at most about 10 "u M; at most about 10 "12 M; or at most 10 "13 M.
  • K D dissociation constant
  • the binding protein of the invention has a dissociation constant (K D ) to IL-12 or IL-23 of 10 "7 M to 10 "8 M; of 10 "8 M to 10 "9 M; of 10 "9 M to 10 “10 M; of 10 "10 to 10 "11 M; of 10 "11 M to 10 "12 M; or of lO '12 to M 10 '13 M.
  • K D dissociation constant
  • One embodiment of the invention provides an antibody construct comprising any one of the binding proteins disclosed above and a linker polypeptide or an immunoglobulin.
  • the antibody construct is selected from the group consisting of an immunoglobulin molecule, a monoclonal antibody, a chimeric antibody, a CDR- grafted antibody, a humanized antibody, a Fab, a Fab', a F(ab')2, a Fv, a disulfide linked Fv, a scFv, a single domain antibody, a diabody, a multispecific antibody, a dual specific antibody, and a bispecific antibody.
  • an immunoglobulin molecule a monoclonal antibody, a chimeric antibody, a CDR- grafted antibody, a humanized antibody, a Fab, a Fab', a F(ab')2, a Fv, a disulfide linked Fv, a scFv, a single domain antibody, a diabody, a multispecific antibody, a dual specific antibody, and a bispecific antibody.
  • the antibody construct comprises a heavy chain immunoglobulin constant domain selected from the group consisting of a human IgM constant domain, a human IgGl constant domain, a human IgG2 constant domain, a human IgG3 constant domain, a human IgG4 constant domain, a human IgE constant domain, and a human IgA constant domain. More preferably, the antibody construct comprises SEQ ID NO.:2; SEQ ID NO.:3; SEQ ID NO.:4; and SEQ ID NO.:5.
  • the invention provides an antibody conjugate comprising an the antibody construct disclosed above and an agent an agent selected from the group consisting of; an immunoadhension molecule, an imaging agent, a therapeutic agent, and a cytotoxic agent.
  • the imaging agent selected from the group consisting of a radiolabel, an enzyme, a fluorescent label, a luminescent label, a bioluminescent label, a magnetic label, and biotin. More preferably the imaging agent is a radiolabel selected from the group consisting of: 3 H 14 C 35 S, 90 Y, 99 Tc, 111 In, 125 1, 131 1, 177 Lu, 166 Ho, and 153 Sm.
  • the therapeutic or cytotoxic agent is selected from the group consisting of; an anti-metabolite, an alkylating agent, an antibiotic, a growth factor, a cytokine, an anti-angiogenic agent, an anti-mitotic agent, an anthracycline, toxin, and an apoptotic agent.
  • the antibody construct is glycosylated.
  • the glycosylation is a human glycosylation pattern.
  • binding protein, antibody construct or antibody conjugate disclosed above exists as a crystal.
  • the crystal is a carrier-free pharmaceutical controlled release crystal.
  • the crystallized binding protein, crystallized antibody construct or crystallized antibody conjugate has a greater half life in vivo than its soluble counterpart.
  • the crystallized binding protein, crystallized antibody construct or crystallized antibody conjugate retains biological activity after crystallization.
  • One aspect of the invention pertains to an isolated nucleic acid encoding any one of the binding protein, antibody construct or antibody conjugate disclosed above.
  • a further embodiment provides a vector comprising the isolated nucleic acid disclosed above wherein said vector is selected from the group consisting of pcDNA; pTT (Durocher et al., Nucleic Acids Research 2002, VoI 30, No.2); pTT3 (pTT with additional multiple cloning site; pEFBOS (Mizushima, S. and Nagata, S., (1990) Nucleic acids Research VoI 18, No. 17); pBV; pJV; and pBJ.
  • a host cell is transformed with the vector disclosed above.
  • the host cell is a prokaryotic cell. More preferably the host cell is E.Coli.
  • the host cell is an eukaryotic cell.
  • the eukaryotic cell is selected from the group consisting of protist cell, animal cell, plant cell and fungal cell. More preferably the host cell is a mammalian cell including, but not limited to, CHO and COS; or a fungal cell such as Saccharomyces cerevisiae; or an insect cell such as Sf9.
  • Another aspect of the invention provides a method of producing a binding protein that binds the p40 subunit of EL-12, comprising culturing any one of the host cells disclosed above in a culture medium under conditions sufficient to produce a binding protein that binds the p40 subunit of IL-12.
  • Another embodiment provides a binding protein produced according to the method disclosed above.
  • compositions for the release of a binding protein wherein the composition comprises a formulation which in turn comprises a crystallized binding protein, crystallized antibody construct or crystallized antibody conjugate as disclosed above and an ingredient; and at least one polymeric carrier.
  • the polymeric carrier is a polymer selected from one or more of the group consisting of: poly (acrylic acid), poly (cyanoacrylates), poly (amino acids), poly (anhydrides), poly (depsipeptide), poly (esters), poly (lactic acid), poly (lactic-co-glycolic acid) or PLGA, poly (b-hydroxybutryate), poly (caprolactone), poly (dioxanone); poly (ethylene glycol), poly ((hydroxypropyl) methacrylamide, poly [(organo)phosphazene], poly (ortho esters), poly (vinyl alcohol), poly (vinylpyrrolidone), maleic anhydride- alkyl vinyl ether copolymers, pluronic polyols, album
  • the ingredient is selected from the group consisting of albumin, sucrose, trehalose, lactitol, gelatin, hydroxypropyl- ⁇ - cyclodextrin, methoxypolyethylene glycol and polyethylene glycol.
  • Another embodiment provides a method for treating a mammal comprising the step of administering to the mammal an effective amount of the composition disclosed above.
  • the invention also provides a pharmaceutical composition
  • a pharmaceutical composition comprising a binding protein, antibody construct or antibody conjugate as disclosed above and a pharmaceutically acceptable carrier.
  • the pharmaceutical composition comprises at least one additional therapeutic agent for treating a disorder in which JL-12 and/or IL-23 activity is detrimental.
  • the additional agent is selected from the group consisting of: Therapeutic agent, imaging agent, cytotoxic agent, angiogenesis inhibitors (including but not limited to anti-VEGF antibodies or VEGF-trap); kinase inhibitors (including but not limited to KDR and TIE-2 inhibitors); co-stimulation molecule blockers (including but not limited to anti-B7.1, anti-B7.2, CTLA4-Ig, anti-CD20); adhesion molecule blockers (including but not limited to anti-LFA-1 Abs, anti-E/L selectin Abs, small molecule inhibitors); anti-cytokine antibody or functional fragment thereof (including but not limited to anti-IL-18, anti-TNF, anti-IL-6/cytokine receptor antibodies); methotrexate; cyclosporin; rapamycin; FK506; detectable label or reporter; a TNF antagonist; an antirheumatic; a muscle relaxant, a narcotic, a non-steroid anti-inflammatory drug (NSAID), an analgesic, an
  • the invention provides a method for inhibiting human IL-12 and/or human DL-23 activity comprising contacting human IL-12 and/or human IL-23 with a binding protein disclosed above such that human IL-12 and/or human EL-23 activity is inhibited.
  • the invention provides a method for inhibiting human IL-12 and/or human IL-23 activity in a human subject suffering from a disorder in which IL-12 and/or IL-23 activity is detrimental, comprising administering to the human subject a binding protein disclosed above such that human IL-12 and/or human IL-23 activity in the human subject is inhibited and treatment is achieved.
  • the disorder is selected from the group comprising arthritis, osteoarthritis, juvenile chronic arthritis, septic arthritis, Lyme arthritis, psoriatic arthritis, reactive arthritis, spondyloarthropathy, systemic lupus erythematosus, Crohn's disease, ulcerative colitis, inflammatory bowel disease, insulin dependent diabetes mellitus, thyroiditis, asthma, allergic diseases, psoriasis, dermatitis scleroderma, graft versus host disease, organ transplant rejection, acute or chronic immune disease associated with organ transplantation, sarcoidosis, atherosclerosis, disseminated intravascular coagulation, Kawasaki's disease, Grave's disease, nephrotic syndrome, chronic fatigue syndrome, Wegener's granulomatosis, Henoch-Schoenlein purpurea, microscopic vasculitis of the kidneys, chronic active hepatitis, uveitis, septic shock, toxic shock syndrome, sepsis syndrome, cache
  • the invention provides a method of treating a patient suffering from a disorder in which human IL-12 and/or human IL-23 is detrimental comprising the step of administering any one of the binding proteins disclosed above before, concurrent, or after the administration of a second agent, as discussed above.
  • the second agent is selected from the group consisting of budenoside, epidermal growth factor, corticosteroids, cyclosporin, sulfasalazine, aminosalicylates, 6-mercaptopurine, azathioprine, metronidazole, lipoxygenase inhibitors, mesalamine, olsalazine, balsalazide, antioxidants, thromboxane inhibitors, IL-I receptor antagonists, anti-IL-l ⁇ monoclonal antibodies, anti-IL-6 monoclonal antibodies, growth factors, elastase inhibitors, pyridinyl-imidazole compounds, antibodies or agonists of TNF, LT, IL-I, IL-2, IL-6, IL-7, IL-8, IL-15, IL-16, IL-18, EMAP-II, GM-CSF, FGF, and PDGF, antibodies of CD2, CD3, CD4, CD8, CD25, CD28
  • compositions disclosed above are administered to the subject by at least one mode selected from parenteral, subcutaneous, intramuscular, intravenous, intrarticular, intrabronchial, intraabdominal, intracapsular, intracartilaginous, intracavitary, intracelial, intracerebellar, intracerebroventricular, intracolic, intracervical, intragastric, intrahepatic, intramyocardial, intraosteal, intrapelvic, intrapericardiac, intraperitoneal, intrapleural, intraprostatic, intrapulmonary, intrarectal, intrarenal, intraretinal, intraspinal, intrasynovial, intrathoracic, intrauterine, intravesical, bolus, vaginal, rectal, buccal, sublingual, intranasal, and transdermal.
  • parenteral subcutaneous, intramuscular, intravenous, intrarticular, intrabronchial, intraabdominal, intracapsular, intracartilaginous, intracavitary, intracelial, intra
  • the anti-idiotype antibody includes any protein or peptide containing molecule that comprises at least a portion of an immunoglobulin molecule such as, but not limited to, at least one complementarily determining region (CDR) of a heavy or light chain or a ligand binding portion thereof, a heavy chain or light chain variable region, a heavy chain or light chain constant region, a framework region, or; any portion thereof, that can be incorporated into a binding protein of the present invention.
  • CDR complementarily determining region
  • This invention pertains to IL-12p40 binding proteins, particularly anti-IL-12p40 antibodies, or antigen-binding portions thereof, that bind IL-12p40.
  • Various aspects of the invention relate to antibodies and antibody fragments, and pharmaceutical compositions thereof, as well as nucleic acids, recombinant expression vectors and host cells for making such antibodies and fragments.
  • Methods of using the antibodies of the invention to detect human IL- 12p40, human IL-12 and human IL-23; to inhibit human DL-12 and/or human IL-23 activity, either in vitro or in vivo; and to regulate gene expression are also encompassed by the invention.
  • Polypeptide refers to any polymeric chain of amino acids.
  • peptide and protein are used interchangeably with the term polypeptide and also refer to a polymeric chain of amino acids.
  • polypeptide encompasses native or artificial proteins, protein fragments and polypeptide analogs of a protein sequence.
  • a polypeptide may be monomeric or polymeric.
  • isolated protein or "isolated polypeptide” is a protein or polypeptide that by virtue of its origin or source of derivation is not associated with naturally associated components that accompany it in its native state; is substantially free of other proteins from the same species; is expressed by a cell from a different species; or does not occur in nature.
  • a polypeptide that is chemically synthesized or synthesized in a cellular system different from the cell from which it naturally originates will be “isolated” from its naturally associated components.
  • a protein may also be rendered substantially free of naturally associated components by isolation, using protein purification techniques well known in the art.
  • recovery refers to the process of rendering a chemical species such as a polypeptide substantially free of naturally associated components by isolation, e.g., using protein purification techniques well known in the art.
  • human IL-12 (abbreviated herein as hIL-12, or IL-12), as used herein, includes a human cytokine that is secreted primarily by antigen presenting cells such as monocytes macrophages and dendritic cells.
  • the term includes a heterodimeric protein comprising a 35 kD subunit (p35) and a 40 kD subunit (p40) which are both linked together with a disulfide bridge.
  • the heterodimeric protein is referred to as a "p70 protein”.
  • the structure of human IL-12 is described further in, for example, Kobayashi, et al. (1989) J. Exp Med. 170:827- 845; Seder, et al.
  • human IL- 12 is intended to include recombinant human IL-12 (rh IL-12), which can be prepared by standard recombinant expression methods.
  • human IL-23 (abbreviated herein as hIL-23, or IL-23), as used herein, includes a heterodimeric human cytokine belonging to a family of five such heterodimeric cytokines including IL-12 and DL-27 (Trinchieri et al., (2003) Immunity 19:641-644).
  • the term includes a heterodimeric protein comprising a 19 kD subunit (pi 9) and a 40 kD subunit (p40) which are both linked together with a disulfide bridge.
  • human BL-23 is intended to include recombinant human IL-23 (rh IL-23), which can be prepared by standard recombinant expression methods.
  • IL-12p40 includes the 40 kD subunit of the human cytokine IL-12 (p40) and the 40 kD subunit of the human cytokine IL-23.
  • Table 1 shows the amino acid sequence of IL-12p40, SEQ ID No. 1, which is known in the art.
  • Biological activity refers to all inherent biological properties of the cytokine.
  • Biological properties of IL-12 include but are not limited to binding IL-12 receptor; induction of interferon-garnma (IFN- ⁇ ) secretion and regulation of balance between antigen- specific T helper type 1 (ThI) and type 2 (Th2) lymphocytes.
  • Biological properties of IL-23 include but are not limited to binding IL-23 receptor, inducing IFN- ⁇ production, ThI cell differentiation and activating the antigen-presenting functions of dendritic cells, and selectively inducing proliferation of memory T cells.
  • telomere binding in reference to the interaction of an antibody, a protein, or a peptide with a second chemical species, mean that the interaction is dependent upon the presence of a particular structure (e.g., an antigenic determinant or epitope) on the chemical species; for example, an antibody recognizes and binds to a specific protein structure rather than to proteins generally. If an antibody is specific for epitope "A”, the presence of a molecule containing epitope A (or free, unlabeled A), in a reaction containing labeled "A” and the antibody, will reduce the amount of labeled A bound to the antibody.
  • a particular structure e.g., an antigenic determinant or epitope
  • antibody broadly refers to any immunoglobulin (Ig) molecule comprised of four polypeptide chains, two heavy (H) chains and two light (L) chains, or any functional fragment, mutant, variant, or derivation thereof, which retains the essential epitope binding features of an Ig molecule.
  • Ig immunoglobulin
  • Such mutant, variant, or derivative anitbody formats are known in the art. Nonlimiting embodiments of which are discussed below.
  • each heavy chain is comprised of a heavy chain variable region (abbreviated herein as HCVR or VH) and a heavy chain constant region.
  • the heavy chain constant region is comprised of three domains, CHl, CH2 and CH3.
  • Each light chain is comprised of a light chain variable region (abbreviated herein as LCVR or VL) and a light chain constant region.
  • the light chain constant region is comprised of one domain, CL.
  • the VH and VL regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDR), interspersed with regions that are more conserved, termed framework regions (FR).
  • CDR complementarity determining regions
  • Each VH and VL is composed of three CDRs and four FRs, arranged from amino-terminus to carboxy-terminus in the following order: FRl, CDRl, FR2, CDR2, FR3, CDR3, FR4.
  • Immunoglobulin molecules can be of any type (e.g., IgG, IgE, IgM, IgD, IgA and IgY), class (e.g., IgG 1, IgG2, IgG 3, IgG4, IgAl and IgA2) or subclass.
  • antibody portion refers to one or more fragments of an antibody that retain the ability to specifically bind to an antigen ⁇ e.g., bJDL-12). It has been shown that the antigen-binding function of an antibody can be performed by fragments of a full-length antibody. Such antibody embodiments may also be bispecific, dual specific, or multi-specific formats; specifically binding to two or more different antigens.
  • binding fragments encompassed within the term "antigen- binding portion" of an antibody include (i) a Fab fragment, a monovalent fragment consisting of the VL, VH, CL and CHl domains; (ii) a F(ab')2 fragment, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) a Fd fragment consisting of the VH and CHl domains; (iv) a Fv fragment consisting of the VL and VH domains of a single arm of an antibody, (v) a dAb fragment (Ward et al, (1989) Nature 341:544-546, Winter et al., PCT publication WO 90/05144 Al herein incorporated by reference), which comprises a single variable domain; and (vi) an isolated complementarity determining region (CDR).
  • CDR complementarity determining region
  • the two domains of the Fv fragment, VL and VH are coded for by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the VL and VH regions pair to form monovalent molecules (known as single chain Fv (scFv); see e.g., Bird et al. (1988) Science 242:423-426; and Huston et al. (1988) Proc. Natl. Acad. Sd. USA 85:5879-5883).
  • single chain Fv single chain Fv
  • Such single chain antibodies are also intended to be encompassed within the term "antigen-binding portion" of an antibody.
  • Other forms of single chain antibodies, such as diabodies are also encompassed.
  • Diabodies are bivalent, bispecific antibodies in which VH and VL domains are expressed on a single polypeptide chain, but using a linker that is too short to allow for pairing between the two domains on the same chain, thereby forcing the domains to pair with complementary domains of another chain and creating two antigen binding sites (see e.g., Holliger, P., et al. (1993) Proc. Natl. Acad. Sci. USA 90:6444-6448; Poljak, R. J., et al. (1994) Structure 2:1121-1123).
  • Such antibody binding portions are known in the art (Kontermann and Dubel eds., Antibody Engineering (2001) Springer-Verlag. New York. 790 pp. (ISBN 3-540-41354-5).
  • antibody construct refers to a polypeptide comprising one or more the antigen binding portions of the invention linked to a linker polypeptide or an immunoglobulin constant domain.
  • Linker polypeptides comprise two or more amino acid residues joined by peptide bonds and are used to link one or more antigen binding portions.
  • Such linker polypeptides are well known in the art (see e.g., Holliger, P., et al. (1993) Proc. Natl. Acad. ScL USA 90:6444-6448; Poljak, RJ., et al. (1994) Structure 2:1121-1123).
  • An immunoglobulin constant domain refers to a heavy or light chain constant domain. Human IgG heavy chain and light chain constant domain amino acid sequences are known in the art and represented in Table 2. Table 2: Sequence of human IgG heavy chain constant domain and light chain constant domain
  • an antibody or antigen-binding portion thereof may be part of a larger immunoadhesion molecules, formed by covalent or noncovalent association of the antibody or antibody portion with one or more other proteins or peptides.
  • immunoadhesion molecules include use of the streptavidin core region to make a tetrameric scFv molecule (Kipriyanov, S.M., et al. (1995) Human Antibodies and Hybridomas 6:93-101) and use of a cysteine residue, a marker peptide and a C-terminal polyhistidine tag to make bivalent and biotinylated scFv molecules (Kipriyanov, S.M., et al. (1994) MoI.
  • Antibody portions such as Fab and F(ab')2 fragments, can be prepared from whole antibodies using conventional techniques, such as papain or pepsin digestion, respectively, of whole antibodies.
  • antibodies, antibody portions and immunoadhesion molecules can be obtained using standard recombinant DNA techniques, as described herein.
  • an "isolated antibody”, as used herein, is intended to refer to an antibody that is substantially free of other antibodies having different antigenic specificities ⁇ e.g., an isolated antibody that specifically binds hIL-12 is substantially free of antibodies that specificallybind antigens other than hIL-12).
  • An isolated antibody that specifically binds hIL-12 may, however, have cross-reactivity to other antigens, such as IL-12 molecules from other species.
  • an isolated antibody may be substantially free of other cellular material and/or chemicals.
  • human antibody is intended to include antibodies having variable and constant regions derived from human germline immunoglobulin sequences.
  • the human antibodies of the invention may include amino acid residues not encoded by human germline immunoglobulin sequences (e.g., mutations introduced by random or site-specific mutagenesis in vitro or by somatic mutation in vivo), for example in the CDRs and in particular CDR3.
  • human antibody as used herein, is not intended to include antibodies in which CDR sequences derived from the germline of another mammalian species, such as a mouse, have been grafted onto human framework sequences.
  • recombinant human antibody is intended to include all human antibodies that are prepared, expressed, created or isolated by recombinant means, such as antibodies expressed using a recombinant expression vector transfected into a host cell (described further in Section II C, below), antibodies isolated from a recombinant, combinatorial human antibody library (HoogenboomKR., (1997) TIB Tech. 15:62-70; Azzazy H., and Highsmith W.E., (2002) Clin. Biochem. 35:425-445; Gavilondo J.V., and Larrick J.W. (2002) BioTechniques 29:128-145; HoogenboomH., and Chames P.
  • such recombinant human antibodies are subjected to in vitro mutagenesis (or, when an animal transgenic for human Ig sequences is used, in vivo somatic mutagenesis) and thus the amino acid sequences of the VH and VL regions of the recombinant antibodies are sequences that, while derived from and related to human germline VH and VL sequences, may not naturally exist within the human antibody germline repertoire in vivo.
  • chimeric antibody refers to antibodies which comprise heavy and light chain variable region sequences from one species and constant region sequences from another species, such as antibodies having murine heavy and light chain variable regions linked to human constant regions.
  • CDR-grafted antibody refers to antibodies which comprise heavy and light chain variable region sequences from one species but in which the sequences of one or more of the CDR regions of VH and/or VL are replaced with CDR sequences of another species, such as antibodies having murine heavy and light chain variable regions in which one or more of the murine CDRs (e.g., CDR3) has been replaced with human CDR sequences.
  • humanized antibody refers to antibodies which comprise heavy and light chain variable region sequences from a non-human species (e.g., a mouse) but in which at least a portion of the VH and/or VL sequence has been altered to be more "human-like", Le., more similar to human germline variable sequences.
  • a non-human species e.g., a mouse
  • human CDR-grafted antibody in which human CDR sequences are introduced into non-human VH and VL sequences to replace the corresponding nonhuman CDR sequences.
  • Kabat numbering Kabat definitions and “Kabat labeling” are used interchangeably herein. These terms, which are recognized in the art, refer to a system of numbering amino acid residues which are more variable (Le. hypervariable) than other amino acid residues in the heavy and light chain variable regions of an antibody, or an antigen binding portion thereof (Kabat et ⁇ l. (1971) Ann. NYAc ⁇ d, ScL 190:382-391 and , Kabat, E.A., et ⁇ l. (1991) Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No. 91-3242).
  • the hypervariable region ranges from amino acid positions 31 to 35 for CDRl, amino acid positions 50 to 65 for CDR2, and amino acid positions 95 to 102 for CDR3.
  • the hypervariable region ranges from amino acid positions 24 to 34 for CDRl, amino acid positions 50 to 56 for CDR2, and amino acid positions 89 to 97 for CDR3.
  • the terms “acceptor” and “acceptor antibody” refer to the antibody or nucleic acid sequence providing or encoding at least 80%, at least 85%, at least 90%, at least 95%, at least 98% or 100% of the amino acid sequences of one or more of the framework regions.
  • the term “acceptor” refers to the antibody amino acid or nucleic acid sequence providing or encoding the constant region(s).
  • the term “acceptor” refers to the antibody amino acid or nucleic acid sequence providing or encoding one or more of the framework regions and the constant region(s).
  • the term "acceptor” refers to a human antibody amino acid or nucleic acid sequence that provides or encodes at least 80%, preferably, at least 85%, at least 90%, at least 95%, at least 98%, or 100% of the amino acid sequences of one or more of the framework regions.
  • an acceptor may contain at least 1, at least 2, at least 3, least 4, at least 5, or at least 10 amino acid residues that does (do) not occur at one or more specific positions of a human antibody.
  • An acceptor framework region and/or acceptor constant region(s) may be, e.g., derived or obtained from a germline antibody gene, a mature antibody gene, a functional antibody (e.g., antibodies well-known in the art, antibodies in development, or antibodies commercially available).
  • CDR refers to the complementarity determining region within antibody variable sequences. There are three CDRs in each of the variable regions of the heavy chain and the light chain, which are designated CDRl, CDR2 and CDR3, for each of the variable regions.
  • CDR set refers to a group of three CDRs that occur in a single variable region capable of binding the antigen.
  • CDR boundary definitions may not strictly follow one of the above systems, but will nonetheless overlap with the Kabat CDRs, although they may be shortened or lengthened in light of prediction or experimental findings that particular residues or groups of residues or even entire CDRs do not significantly impact antigen binding.
  • the methods used herein may utilize CDRs defined according to any of these systems, although preferred embodiments use Kabat or Chothia defined CDRs.
  • canonical residue refers to a residue in a CDR or framework that defines a particular canonical CDR structure as defined by Chothia et al. (J. MoI. Biol. 196:901-907 (1987); Chothia et al., J. MoI. Biol. 227:799 (1992), both are incorporated herein by reference). According to Chothia et al., critical portions of the CDRs of many antibodies have nearly identical peptide backbone confirmations despite great diversity at the level of amino acid sequence. Each canonical structure specifies primarily a set of peptide backbone torsion angles for a contiguous segment of amino acid residues forming a loop.
  • the terms “donor” and “donor antibody” refer to an antibody providing one or more CDRs.
  • the donor antibody is an antibody from a species different from the antibody from which the framework regions are obtained or derived.
  • the term “donor antibody” refers to a non-human antibody providing one or more CDRs.
  • the term “framework” or “framework sequence” refers to the remaining sequences of a variable region minus the CDRs. Because the exact definition of a CDR sequence can be determined by different systems, the meaning of a framework sequence is subject to correspondingly different interpretations.
  • the six CDRs also divide the framework regions on the light chain and the heavy chain into four sub-regions (FRl, FR2, FR3 and FR4) on each chain, in which CDRl is positioned between FRl and FR2, CDR2 between FR2 and FR3, and CDR3 between FR3 and FR4.
  • a framework region represents the combined FR's within the variable region of a single, naturally occurring immunoglobulin chain.
  • a FR represents one of the four sub- regions, and FRs represents two or more of the four sub- regions constituting a framework region.
  • Human heavy chain and light chain acceptor sequences are known in the art. In one embodiment of the invention the human heavy chain and light chain acceptor sequences are selected from the sequences described in Table 3 and Table 4.
  • the term "germline antibody gene” or “gene fragment” refers to an immunoglobulin sequence encoded by non- lymphoid cells that have not undergone the maturation process that leads to genetic rearrangement and mutation for expression of a particular immunoglobulin. (See, e.g., Shapiro et al., Crit. Rev. Immunol. 22(3): 183-200 (2002); Marchalonis et al., Adv Exp Med Biol. 484:13-30 (2001)).
  • One of the advantages provided by various embodiments of the present invention stems from the recognition that germline antibody genes are more likely than mature antibody genes to conserve essential amino acid sequence structures characteristic of individuals in the species, hence less likely to be recognized as from a foreign source when used therapeutically in that species.
  • key residues refer to certain residues within the variable region that have more impact on the binding specificity and/or affinity of an antibody, in particular a humanized antibody.
  • a key residue includes, but is not limited to, one or more of the following: a residue that is adjacent to a CDR, a potential glycosylation site (can be either N- or O-glycosylation site), a rare residue, a residue capable of interacting with the antigen, a residue capable of interacting with a CDR, a canonical residue, a contact residue between heavy chain variable region and light chain variable region, a residue within the Vernier zone, and a residue in the region that overlaps between the Chothia definition of a variable heavy chain CDRl and the Kabat definition of the first heavy chain framework.
  • humanized antibody is an antibody or a variant, derivative, analog or fragment thereof which immunospecif ⁇ cally binds to an antigen of interest and which comprises a framework (FR) region having substantially the amino acid sequence of a human antibody and a complementary determining region (CDR) having substantially the amino acid sequence of a non-human antibody.
  • FR framework
  • CDR complementary determining region
  • substantially in the context of a CDR refers to a CDR having an amino acid sequence at least 80%, preferably at least 85%, at least 90%, at least 95%, at least 98% or at least 99% identical to the amino acid sequence of a non-human antibody CDR.
  • a humanized antibody comprises substantially all of at least one, and typically two, variable domains (Fab, Fab', F(ab') 2, FabC, Fv) in which all or substantially all of the CDR regions correspond to those of a non-human immunoglobulin (i.e., donor antibody) and all or substantially all of the framework regions are those of a human immunoglobulin consensus sequence.
  • a humanized antibody also comprises at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin.
  • a humanized antibody contains both the light chain as well as at least the variable domain of a heavy chain.
  • the antibody also may include the CHl, hinge, CH2, CH3, and CH4 regions of the heavy chain.
  • a humanized antibody only contains a humanized light chain. In some embodiments, a humanized antibody only contains a humanized heavy chain. In specific embodiments, a humanized antibody only contains a humanized variable domain of a light chain and/or humanized heavy chain.
  • the humanized antibody can be selected from any class of immunoglobulins, including IgM, IgG, IgD, IgA and IgE, and any isotype, including without limitation IgG 1, IgG2, IgG3 and IgG4.
  • the humanized antibody may comprise sequences from more than one class or isotype, and particular constant domains may be selected to optimize desired effector functions using techniques well- known in the art.
  • the framework and CDR regions of a humanized antibody need not correspond precisely to the parental sequences, e.g., the donor antibody CDR or the consensus framework may be mutagenized by substitution, insertion and/or deletion of at least one amino acid residue so that the CDR or framework residue at that site does not correspond to either the donor antibody or the consensus framework. In a preferred embodiment, such mutations, however, will not be extensive. Usually, at least 80%, preferably at least 85%, more preferably at least 90%, and most preferably at least 95% of the humanized antibody residues will correspond to those of the parental FR and CDR sequences.
  • the term "consensus framework" refers to the framework region in the consensus immunoglobulin sequence.
  • the term "consensus immunoglobulin sequence” refers to the sequence formed from the most frequently occurring amino acids (or nucleotides) in a family of related immunoglobulin sequences (See e.g., Winnaker, From Genes to Clones (Verlagsgesellschaft, Weinheim, Germany 1987). In a family of immunoglobulins, each position in the consensus sequence is occupied by the amino acid occurring most frequently at that position in the family. If two amino acids occur equally frequently, either can be included in the consensus sequence.
  • “Vernier” zone refers to a subset of framework residues that may adjust CDR structure and fine-tune the fit to antigen as described by Foote and Winter (1992, J. MoI. Biol. 224:487-499, which is incorporated herein by reference). Vernier zone residues form a layer underlying the CDRs and may impact on the structure of CDRs and the affinity of the antibody.
  • neutralizing refers to neutralization of biological activity of a cytokine when a binding protein specifically binds the cytokine.
  • a neutralizing binding protein is a neutralizing antibody whose binding to hIL-12 and/or bJL-23 results in inhibition of a biological activity of hIL-12 and/or hIL-23.
  • the neutralizing binding protein binds hIL-12 and/or bJL-23 and reduces a biologically activity of IL-12 and/or hIL-23 by at least about 20%, 40%, 60%, 80%, 85% or more.
  • Inhibition of a biological activity of hIL-12 and/or hIL-23 by a neutralizing binding protein can be assessed by measuring one or more indicators of hIL-12 and/or hIL-23 biological activity well known in the art. For example inhibition of human phytohemagglutinin blast proliferation in a PHA blast Interferon-gamma Induction Assay (see Example l.l.C) or inhibition of receptor binding in a human IL-12 receptor binding assay, (also see Salfeld et al., PCT publication No.WO 00/56772 Al).
  • activity includes activities such as the binding specificity/affinity of an antibody for an antigen, for example, an anti-hlL-12 antibody that binds to an EL-12 antigen and/or the neutralizing potency of an antibody, for example, an anti-hIL-12 antibody whose binding to hIL-12 inhibits the biological activity of hIL-12, e.g. inhibition of PHA blast proliferation or inhibition of receptor binding in a human IL-12 receptor binding assay, or or PHA blast Interferon-gamma Induction Assay (see Example l.l.C).
  • epitope includes any polypeptide determinant capable of specific binding to an immunoglobulin or T-cell receptor.
  • epitope determinants include chemically active surface groupings of molecules such as amino acids, sugar side chains, phosphoryl, or sulfonyl, and, in certain embodiments, may have specific three dimensional structural characteristics, and/or specific charge characteristics.
  • An epitope is a region of an antigen that is bound by an antibody.
  • an antibody is said to specifically bind an antigen when it preferentially recognizes its target antigen in a complex mixture of proteins and/or macromolecules.
  • surface plasmon resonance refers to an optical phenomenon that allows for the analysis of real-time biospecific interactions by detection of alterations in protein concentrations within a biosensor matrix, for example using the BIAcore system (Pharmacia Biosensor AB, Uppsala, Sweden and Piscataway, NJ).
  • BIAcore Pharmacia Biosensor AB, Uppsala, Sweden and Piscataway, NJ.
  • K 0n is intended to refer to the on rate constant for association of an antibody to the antigen to form the antibody/antigen complex as is known in the art.
  • K o ff is intended to refer to the off rate constant for dissociation of an antibody from the antibody/antigen complex as is known in the art.
  • Kj is intended to refer to the dissociation constant of a particular antibody-antigen interaction as is known in the art.
  • label binding protein refers to a protein with a label incorporated that provides for the identification of the binding protein.
  • the label is a detectable marker, e.g., incorporation of a radiolabeled amino acid or attachment to a polypeptide of biotinyl moieties that can be detected by marked avidin (e.g., streptavidin containing a fluorescent marker or enzymatic activity that can be detected by optical or colorimetric methods).
  • labels for polypeptides include, but are not limited to, the following: radioisotopes or radionuclides (e.g., 3 H 14 C, 35 S, 90 Y, 99 Tc, 111 In, 125 1, 131 1, 177 Lu, 166 Ho, or 153 Sm); fluorescent labels (e.g., FTTC, rhodamine, lanthanide phosphors), enzymatic labels (e.g., horseradish peroxidase, luciferase, alkaline phosphatase); chemiluminescent markers; biotinyl groups; predetermined polypeptide epitopes recognized by a secondary reporter (e.g., leucine zipper pair sequences, binding sites for secondary antibodies, metal binding domains, epitope tags); and magnetic agents, such as gadolinium chelates.
  • radioisotopes or radionuclides e.g., 3 H 14 C, 35 S, 90 Y, 99 Tc, 111 In,
  • antibody conjugate refers to a binding protein, such as an antibody, chemically linked to a second chemical moiety, such as a therapeutic or cytotoxic agent.
  • agent is used herein to denote a chemical compound, a mixture of chemical compounds, a biological macromolecule, or an extract made from biological materials.
  • the therapeutic or cytotoxic agents include, but are not limited to, pertussis toxin, taxol, cytochalasin B, gramicidin D, ethidium bromide, emetine, mitomycin, etoposide, tenoposide, vincristine, vinblastine, colchicin, doxorubicin, daunorubicin, dihydroxy anthracin dione, mitoxantrone, mithramycin, actinomycin D, 1-dehydrotestosterone, glucocorticoids, procaine, tetracaine, lidocaine, propranolol, and puromycin and analogs or homologs thereof.
  • crystal refers to an antibody, or antigen binding portion thereof, that exists in the form of a crystal.
  • Crystals are one form of the solid state of matter, which is distinct from other forms such as the amorphous solid state or the liquid crystalline state.
  • Crystals are composed of regular, repeating, three-dimensional arrays of atoms, ions, molecules (e.g., proteins such as antibodies), or molecular assemblies (e.g., antigen/antibody complexes). These three-dimensional arrays are arranged according to specific mathematical relationships that are well-understood in the field.
  • the fundamental unit, or building block, that is repeated in a crystal is called the asymmetric unit.
  • polynucleotide as referred to herein means a polymeric form of two or more nucleotides, either ribonucleotides or deoxvnucleotides or a modified form of either type of nucleotide.
  • the term includes single and double stranded forms of DNA but preferably is double-stranded DNA.
  • isolated polynucleotide shall mean a polynucleotide (e.g., of genomic, cDNA, or synthetic origin, or some combination thereof) that, by virtue of its origin , the "isolated polynucleotide”: is not associated with all or a portion of a polynucleotide with which the "isolated polynucleotide” is found in nature; is operably linked to a polynucleotide that it is not linked to in nature; or does not occur in nature as part of a larger sequence.
  • isolated polynucleotide e.g., of genomic, cDNA, or synthetic origin, or some combination thereof
  • vector is intended to refer to a nucleic acid molecule capable of transporting another nucleic acid to which it has been linked.
  • plasmid refers to a circular double stranded DNA loop into which additional DNA segments may be ligated.
  • viral vector Another type of vector is a viral vector, wherein additional DNA segments may be ligated into the viral genome.
  • Certain vectors are capable of autonomous replication in a host cell into which they are introduced (e.g., bacterial vectors having a bacterial origin of replication and episomal mammalian vectors).
  • vectors e.g., non-episomal mammalian vectors
  • vectors can be integrated into the genome of a host cell upon introduction into the host cell, and thereby are replicated along with the host genome.
  • certain vectors are capable of directing the expression of genes to which they are operatively linked.
  • Such vectors are referred to herein as "recombinant expression vectors" (or simply, "expression vectors”).
  • expression vectors of utility in recombinant DNA techniques are often in the form of plasmids.
  • plasmid and vector may be used interchangeably as the plasmid is the most commonly used form of vector.
  • the invention is intended to include such other forms of expression vectors, such as viral vectors (e.g., replication defective retroviruses, adenoviruses and adeno-associated viruses), which serve equivalent functions.
  • operably linked refers to a juxtaposition wherein the components described are in a relationship permitting them to function in their intended manner.
  • a control sequence "operably linked" to a coding sequence is ligated in such a way that expression of the coding sequence is achieved under conditions compatible with the control sequences.
  • "Operably linked” sequences include both expression control sequences that are contiguous with the gene of interest and expression control sequences that act in trans or at a distance to control the gene of interest.
  • expression control sequence refers to polynucleotide sequences which are necessary to effect the expression and processing of coding sequences to which they are ligated.
  • Expression control sequences include appropriate transcription initiation, termination, promoter and enhancer sequences; efficient RNA processing signals such as splicing and polyadenylation signals; sequences that stabilize cytoplasmic mRNA; sequences that enhance translation efficiency (i.e., Kozak consensus sequence); sequences that enhance protein stability; and when desired, sequences that enhance protein secretion.
  • the nature of such control sequences differs depending upon the host organism; in prokaryotes, such control sequences generally include promoter, ribosomal binding site, and transcription termination sequence; in eukaryotes, generally, such control sequences include promoters and transcription termination sequence.
  • control sequences is intended to include components whose presence is essential for expression and processing, and can also include additional components whose presence is advantageous, for example, leader sequences and fusion partner sequences.
  • Transformation refers to any process by which exogenous DNA enters a host cell. Transformation may occur under natural or artificial conditions using various methods well known in the art. Transformation may rely on any known method for the insertion of foreign nucleic acid sequences into a prokaryotic or eukaryotic host cell. The method is selected based on the host cell being transformed and may include, but is not limited to, viral infection, electroporation, lipofection, and particle bombardment.
  • Such "transformed” cells include stably transformed cells in which the inserted DNA is capable of replication either as an autonomously replicating plasmid or as part of the host chromosome. They also include cells which transiently express the inserted DNA or RNA for limited periods of time.
  • host cell is intended to refer to a cell into which exogenous DNA has been introduced. It should be understood that such terms are intended to refer not only to the particular subject cell, but, to the progeny of such a cell. Because certain modifications may occur in succeeding generations due to either mutation or environmental influences, such progeny may not, in fact, be identical to the parent cell, but are still included within the scope of the term "host cell” as used herein.
  • host cells include prokaryotic and eukaryotic cells selected from any of the Kingdoms of life.
  • Preferred eukaryotic cells include protist, fungal, plant and animal cells.
  • host cells include but are not limited to the prokaryotic cell line E.Coli; mammalian cell lines CHO, HEK 293 and COS; the insect cell line Sf9; and the fungal cell Saccharomyces cerevisiae.
  • Standard techniques may be used for recombinant DNA, oligonucleotide synthesis, and tissue culture and transformation (e.g., electroporation, lipofection).
  • Enzymatic reactions and purification techniques may be performed according to manufacturer's specifications or as commonly accomplished in the art or as described herein.
  • the foregoing techniques and procedures may be generally performed according to conventional methods well known in the art and as described in various general and more specific references that are cited and discussed throughout the present specification. See e.g., Sambrook et al. Molecular Cloning: A Laboratory Manual (2d ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (1989)), which is incorporated herein by reference for any purpose.
  • Transgenic organism refers to an organism having cells that contain a transgene, wherein the transgene introduced into the organism (or an ancestor of the organism) expresses a polypeptide not naturally expressed in the organism.
  • a "transgene” is a DNA construct, which is stably and operably integrated into the genome of a cell from which a transgenic organism develops, directing the expression of an encoded gene product in one or more cell types or tissues of the transgenic organism.
  • the term “regulate”and “modulate” are used interchangeably, and, as used herein, refers to a change or an alteration in the activity of a molecule of interest (e.g., the biological activity of hIL-12). Modulation may be an increase or a decrease in the magnitude of a certain activity or function of the molecule of interest. Exemplary activities and functions of a molecule include, but are not limited to, binding characteristics, enzymatic activity, cell receptor activation, and signal transduction.
  • a modulator is a compound capable of changing or altering an activity or function of a molecule of interest (e.g., the biological activity of hIL-12).
  • a modulator may cause an increase or decrease in the magnitude of a certain activity or function of a molecule compared to the magnitude of the activity or function observed in the absence of the modulator.
  • a modulator is an inhibitor, which decreases the magnitude of at least one activity or function of a molecule.
  • Exemplary inhibitors include, but are not limited to, proteins, peptides, antibodies, peptibodies, carbohydrates or small organic molecules. Peptibodies are described, e.g., in WO01/83525.
  • agonist refers to a modulator that, when contacted with a molecule of interest, causes an increase in the magnitude of a certain activity or function of the molecule compared to the magnitude of the activity or function observed in the absence of the agonist.
  • agonists of interest may include, but are not limited to, IL-12 polypeptides or polypeptides, nucleic acids, carbohydrates, or any other molecules that bind to hIL-12.
  • antagonist refers to a modulator that, when contacted with a molecule of interest causes a decrease in the magnitude of a certain activity or function of the molecule compared to the magnitude of the activity or function observed in the absence of the antagonist.
  • Particular antagonists of interest include those that block or modulate the biological or immunological activity of hIL-12 and/or hIL-23.
  • Antagonists and inhibitors of ML-12 and/or ML-23 may include, but are not limited to, proteins, nucleic acids, carbohydrates, or any other molecules, which bind to hDL-12 and/or hIL-23.
  • the term "effective amount” refers to the amount of a therapy which is sufficient to reduce or ameliorate the severity and/or duration of a disorder or one or more symptoms thereof, prevent the advancement of a disorder, cause regression of a disorder, prevent the recurrence, development, onset or progression of one or more symptoms associated with a disorder, detect a disorder, or enhance or improve the prophylactic or therapeutic effect(s) of another therapy (e.g. , prophylactic or therapeutic agent).
  • sample includes, but is not limited to, any quantity of a substance from a living thing or formerly living thing.
  • living things include, but are not limited to, humans, mice, rats, monkeys, dogs, rabbits and other animals.
  • substances include, but are not limited to, blood, serum, urine, synovial fluid, cells, organs, tissues, bone marrow, lymph nodes and spleen.
  • One aspect of the present invention provides isolated murine monoclonal antibodies, or antigen-binding portions thereof, that bind to IL-12p40 with high affinity, a slow off rate and high neutralizing capacity.
  • a second aspect of the invention provides chimeric antibodies that bind IL-12p40.
  • a third aspect of the invention provides CDR grafted antibodies, or antigen- binding portions thereof, that bind IL-12p40.
  • a fourth aspect of the invention provides humanized antibodies, or antigen-binding portions thereof, that bind EL-12p40.
  • the antibodies, or portions thereof are isolated antibodies.
  • the antibodies of the invention are neutralizing human anti-IL-12 and/or human anti-IL-23 antibodies.
  • Antibodies of the present invention may be made by any of a number of techniques known in the art.
  • Monoclonal antibodies can be prepared using a wide variety of techniques known in the art including the use of hybridoma, recombinant, and phage display technologies, or a combination thereof.
  • monoclonal antibodies can be produced using hybridoma techniques including those known in the art and taught, for example, in Harlow et al. , Antibodies: A Laboratory Manual, (Cold Spring Harbor Laboratory Press, 2nd ed. 1988); Hammerling, et al., in: Monoclonal Antibodies and T-CeIl Hybridomas 563-681 (Elsevier, N. Y., 1981) (said references incorporated by reference in their entireties).
  • the term “monoclonal antibody” as used herein is not limited to antibodies produced through hybridoma technology.
  • the term “monoclonal antibody” refers to an antibody that is derived from a single clone, including any eukaryotic, prokaryotic, or phage clone, and not the method by which it is produced.
  • the present invention provides methods of generating monoclonal antibodies as well as antibodies produced by the method comprising culturing a hybridoma cell secreting an antibody of the invention wherein, preferably, the hybridoma is generated by fusing splenocytes isolated from a mouse immunized with an antigen of the invention with myeloma cells and then screening the hybridomas resulting from the fusion for hybridoma clones that secrete an antibody able to bind a polypeptide of the invention ( See Example 1.2). Briefly, mice can be immunized with an IL-12 antigen.
  • the IL-12 antigen is administered with a adjuvant to stimulate the immune response.
  • adjuvants include complete or incomplete Freund's adjuvant, RIBI (muramyl dipeptides) or ISCOM (immunostimulating complexes).
  • RIBI muramyl dipeptides
  • ISCOM immunological complexes
  • Such adjuvants may protect the polypeptide from rapid dispersal by sequestering it in a local deposit, or they may contain substances that stimulate the host to secrete factors that are chemotactic for macrophages and other components of the immune system.
  • the immunization schedule will involve two or more administrations of the polypeptide, spread out over several weeks.
  • antibodies and/or antibody- producing cells may be obtained from the animal.
  • An anti- IL-12 antibody-containing serum is obtained from the animal by bleeding or sacrificing the animal.
  • the serum may be used as it is obtained from the animal, an immunoglobulin fraction may be obtained from the serum, or the anti- EL-12 antibodies may be purified from the serum.
  • Serum or immunoglobulins obtained in this manner are polyclonal, thus having a heterogeneous array of properties.
  • the mouse spleen is harvested and splenocytes isolated.
  • the splenocytes are then fused by well-known techniques to any suitable myeloma cells, for example cells from cell line SP20 available from the ATCC.
  • Hybridomas are selected and cloned by limited dilution.
  • the hybridoma clones are then assayed by methods known in the art for cells that secrete antibodies capable of binding IL-12.
  • Ascites fluid which generally contains high levels of antibodies, can be generated by immunizing mice with positive hybridoma clones.
  • antibody-producing immortalized hybridomas may be prepared from the immunized animal. After immunization, the animal is sacrificed and the splenic B cells are fused to immortalized myeloma cells as is well known in the art. See, e.g., Harlow and Lane, supra. In a preferred embodiment, the myeloma cells do not secrete immunoglobulin polypeptides (a non-secretory cell line). After fusion and antibiotic selection, the hybridomas are screened using IL-12, or a portion thereof, or a cell expressing BL-12.
  • the initial screening is performed using an enzyme-linked immunoassay (ELISA) or a radioimmunoassay (RIA), preferably an ELISA.
  • ELISA enzyme-linked immunoassay
  • RIA radioimmunoassay
  • An example of ELISA screening is provided in WO 00/37504, herein incorporated by reference.
  • Anti- IL-12p40 antibody-producing hybridomas are selected, cloned and further screened for desirable characteristics, including robust hybridoma growth, high antibody production and desirable antibody characteristics, as discussed further below.
  • Hybridomas may be cultured and expanded in vivo in syngeneic animals, in animals that lack an immune system, e.g., nude mice, or in cell culture in vitro. Methods of selecting, cloning and expanding hybridomas are well known to those of ordinary skill in the art.
  • the hybridomas are mouse hybridomas, as described above.
  • the hybridomas are produced in a non-human, non-mouse species such as rats, sheep, pigs, goats, cattle or horses.
  • the hybridomas are human hybridomas, in which a human non-secretory myeloma is fused with a human cell expressing an anti-IL-12 antibody.
  • Antibody fragments that recognize specific epitopes may be generated by known techniques.
  • Fab and F(ab')2 fragments of the invention may be produced by proteolytic cleavage of immunoglobulin molecules, using enzymes such as papain (to produce Fab fragments) or pepsin (to produce F(ab')2 fragments).
  • F(ab')2 fragments contain the variable region, the light chain constant region and the CHI domain of the heavy chain.
  • recombinant antibodies are generated from single, isolated lymphocytes using a procedure referred to in the art as the selected lymphocyte antibody method (SLAM), as described in U.S. Patent No. 5,627,052, PCT Publication WO 92/02551 and Babcock, J.S. et al. (1996) Proc. Natl. Acad. ScL USA 93:7843-7848.
  • SAM selected lymphocyte antibody method
  • single cells secreting antibodies of interest e.g., lymphocytes derived from any one of the immunized animals described in Section 1 are screened using an antigen-specific hemolytic plaque assay, wherein the antigen IL-12, a subunint of IL-12, or a fragment thereof, is coupled to sheep red blood cells using a linker, such as biotin, and used to identify single cells that secrete antibodies with specificity for IL-12.
  • a linker such as biotin
  • variable regions can then be expressed, in the context of appropriate immunoglobulin constant regions (e.g., human constant regions), in mammalian host cells, such as COS or CHO cells.
  • the host cells transfected with the amplified immunoglobulin sequences, derived from in vivo selected lymphocytes, can then undergo further analysis and selection in vitro, for example by panning the transfected cells to isolate cells expressing antibodies to IL-12.
  • the amplified immunoglobulin sequences further can be manipulated in vitro, such as by in vitro affinity maturation methods such as those described in PCT Publication WO 97/29131 and PCT Publication WO 00/56772.
  • antibodies are produced by immunizing a non-human animal comprising some, or all, of the human immunoglobulin locus with an IL- 12 antigen.
  • the non-human animal is a XENOMOUSE transgenic mouse, an engineered mouse strain that comprises large fragments of the human immunoglobulin loci and is deficient in mouse antibody production. See, e.g., Green et al. Nature Genetics 7:13-21 (1994) and United States Patents 5,916,771, 5,939,598, 5,985,615, 5,998,209, 6,075,181, 6,091,001, 6,114,598 and 6,130,364.
  • WO 91/10741 published July 25,1991, WO 94/02602, published February 3, 1994, WO 96/34096 and WO 96/33735, both published October 31, 1996, WO 98/16654, published April 23, 1998, WO 98/24893, published June 11, 1998, WO 98/50433, published November 12, 1998, WO 99/45031, published September 10, 1999, WO 99/53049, published October 21, 1999, WO 0009560, published February 24, 2000 and WO 00/037504, published June 29, 2000.
  • the XENOMOUSE transgenic mouse produces an adult-like human repertoire of fully human antibodies, and generates antigen-specific human Mabs.
  • the XENOMOUSE transgenic mouse contains approximately 80% of the human antibody repertoire through introduction of megabase sized, germline configuration YAC fragments of the human heavy chain loci and x light chain loci. See Mendez et al., Nature Genetics 15:146-156 (1997), Green and Jakobovits J. Exp. Med. 188:483-495 (1998), the disclosures of which are hereby incorporated by reference.
  • In vitro methods also can be used to make the antibodies of the invention, wherein an antibody library is screened to identify an antibody having the desired binding specificity.
  • Methods for such screening of recombinant antibody libraries are well known in the art and include methods described in, for example, Ladner et al. U.S. Patent No. 5,223,409; Kang et al. PCT Publication No. WO 92/18619; Dower et al. PCT Publication No. WO 91/17271; Winter et al. PCT Publication No. WO 92/20791; Markland et al. PCT Publication No. WO 92/15679; Breitling et al. PCT Publication No.
  • the recombinant antibody library may be from a subject immunized with IL-12 or IL-23, or a portion of IL-12 or IL-23.
  • the recombinant antibody library may be from a na ⁇ ve subject, i.e., one who has not been immunized with IL-12 or IL-23, such as a human antibody library from a human subject who has not been immunized with human IL-12 or ⁇ L-Ti.
  • Antibodies of the invention are selected by screening the recombinant antibody library with the peptide comprising human IL-12p40 to thereby select those antibodies that recognize IL-12p40.
  • the invention pertains to an isolated antibody, or an antigen-binding portion thereof, that binds human IL-12 and/or human IL-23.
  • the antibody is a neutralizing antibody.
  • the antibody is a recombinant antibody or a monoclonal antibody.
  • the antibodies of the present invention can also be generated using various phage display methods known in the art.
  • phage display methods functional antibody domains are displayed on the surface of phage particles which carry the polynucleotide sequences encoding them.
  • phage can be utilized to display antigen-binding domains expressed from a repertoire or combinatorial antibody library (e. g., human or murine).
  • Phage expressing an antigen binding domain that binds the antigen of interest can be selected or identified with antigen, e.g., using labeled antigen or antigen bound or captured to a solid surface or bead.
  • Phage used in these methods are typically filamentous phage including fd and M13 binding domains expressed from phage with Fab, Fv or disulfide stabilized Fv antibody domains recombinantly fused to either the phage gene IH or gene VDI protein.
  • Examples of phage display methods that can be used to make the antibodies of the present invention include those disclosed in Brinkman et al., J. Immunol. Methods 182:41-50 (1995); Ames et al., J. Immunol. Methods 184:177-186 (1995); Kettleborough et al., Eur. J. Immunol.
  • the antibody coding regions from the phage can be isolated and used to generate whole antibodies including human antibodies or any other desired antigen binding fragment, and expressed in any desired host, including mammalian cells, insect cells, plant cells, yeast, and bacteria, e.g., as described in detail below.
  • RNA-protein fusions as described in PCT Publication No. WO 98/31700 by Szostak and Roberts, and in Roberts, R.W. and Szostak, J.W. (1997) Proc. Natl. Acad. Sci. USA 94:12297-12302.
  • a covalent fusion is created between an mRNA and the peptide or protein that it encodes by in vitro translation of synthetic mRNAs that carry puromycin, a peptidyl acceptor antibiotic, at their 3' end.
  • a specific mRNA can be enriched from a complex mixture of mRNAs (e.g., a combinatorial library) based on the properties of the encoded peptide or protein, e.g., antibody, or portion thereof, such as binding of the antibody, or portion thereof, to the dual specificity antigen.
  • Nucleic acid sequences encoding antibodies, or portions thereof, recovered from screening of such libraries can be expressed by recombinant means as described above (e.g., in mammalian host cells) and, moreover, can be subjected to further affinity maturation by either additional rounds of screening of mRNA-peptide fusions in which mutations have been introduced into the originally selected sequence(s), or by other methods for affinity maturation in vitro of recombinant antibodies, as described above.
  • the antibodies of the present invention can also be generated using yeast display methods known in the art.
  • yeast display methods genetic methods are used to tether antibody domains to the yeast cell wall and display them on the surface of yeast.
  • yeast can be utilized to display antigen-binding domains expressed from a repertoire or combinatorial antibody library (e. g., human or murine).
  • yeast display methods that can be used to make the antibodies of the present invention include those disclosed Wittrup, et al. U.S. Patent No. 6,699,658 incorporated herein by reference.
  • Antibodies of the present invention may be produced by any of a number of techniques known in the art. For example, expression from host cells, wherein expression vector(s) encoding the heavy and light chains is (are) transfected into a host cell by standard techniques.
  • transfection are intended to encompass a wide variety of techniques commonly used for the introduction of exogenous DNA into a prokaryotic or eukaryotic host cell, e.g., electroporation, calcium-phosphate precipitation, DEAE-dextran transfection and the like.
  • the antibodies of the invention in either prokaryotic or eukaryotic host cells, expression of antibodies in eukaryotic cells is preferable, and most preferable in mammalian host cells, because such eukaryotic cells (and in particular mammalian cells) are more likely than prokaryotic cells to assemble and secrete a properly folded and immunologically active antibody.
  • Preferred mammalian host cells for expressing the recombinant antibodies of the invention include Chinese Hamster Ovary (CHO cells) (including dhfr- CHO cells, described in Urlaub and Chasin, (1980) Proc. Natl. Acad. ScL USA 77:4216-4220, used with a DHFR selectable marker, e.g., as described in RJ. Kaufman and P.A. Sharp (1982) MoI. Biol. 159:601-621), NSO myeloma cells, COS cells and SP2 cells.
  • Chinese Hamster Ovary CHO cells
  • dhfr- CHO cells described in Urlaub and Chasin, (1980) Proc. Natl. Acad. ScL USA 77:4216-4220, used with a DHFR selectable marker, e.g., as described in RJ. Kaufman and P.A. Sharp (1982) MoI. Biol. 159:601-621
  • NSO myeloma cells
  • the antibodies When recombinant expression vectors encoding antibody genes are introduced into mammalian host cells, the antibodies are produced by culturing the host cells for a period of time sufficient to allow for expression of the antibody in the host cells or, more preferably, secretion of the antibody into the culture medium in which the host cells are grown. Antibodies can be recovered from the culture medium using standard protein purification methods.
  • Host cells can also be used to produce functional antibody fragments, such as Fab fragments or scFv molecules. It will be understood that variations on the above procedure are within the scope of the present invention. For example, it may be desirable to transfect a host cell with DNA encoding functional fragments of either the light chain and/or the heavy chain of an antibody of this invention. Recombinant DNA technology may also be used to remove some, or all, of the DNA encoding either or both of the light and heavy chains that is not necessary for binding to the antigens of interest. The molecules expressed from such truncated DNA molecules are also encompassed by the antibodies of the invention.
  • bifunctional antibodies may be produced in which one heavy and one light chain are an antibody of the invention and the other heavy and light chain are specific for an antigen other than the antigens of interest by crosslinking an antibody of the invention to a second antibody by standard chemical crosslinking methods.
  • a recombinant expression vector encoding both the antibody heavy chain and the antibody light chain is introduced into dhfr- CHO cells by calcium phosphate-mediated transfection.
  • the antibody heavy and light chain genes are each operatively linked to CMV enhancer/AdMLP promoter regulatory elements to drive high levels of transcription of the genes.
  • the recombinant expression vector also carries a DHFR gene, which allows for selection of CHO cells that have been transfected with the vector using methotrexate selection/amplification.
  • the selected transformant host cells are cultured to allow for expression of the antibody heavy and light chains and intact antibody is recovered from the culture medium.
  • Standard molecular biology techniques are used to prepare the recombinant expression vector, transfect the host cells, select for transformants, culture the host cells and recover the antibody from the culture medium.
  • the invention provides a method of synthesizing a recombinant antibody of the invention by culturing a host cell of the invention in a suitable culture medium until a recombinant antibody of the invention is synthesized. The method can further comprise isolating the recombinant antibody from the culture medium.
  • Table 5 is a list of amino acid sequences of VH and VL regions of preferred anti-hlL- 12p40 antibodies of the invention.
  • the foregoing isolated anti-IL-12p40 antibody CDR sequences establish a novel family of IL-12p40 binding proteins, isolated in accordance with this invention, and comprising polypeptides that include the CDR sequences listed in Table 6 below.
  • CDR' s of the invention having preferred IL-12p40 binding and/or neutralizing activity with respect to hIL-12 and or hIL-23
  • standard methods known in the art for generating binding proteins of the present invention and assessing the IL- 12 and or IL-23 binding and/or neutralizing characteristics of those binding protein may be used, including but not limited to those specifically described herein.
  • a chimeric antibody is a molecule in which different portions of the antibody are derived from different animal species, such as antibodies having a variable region derived from a murine monoclonal antibody and a human immunoglobulin constant region.
  • Methods for producing chimeric antibodies are known in the art and discussed in detail in Example 2.1. See e.g., Morrison, Science 229:1202 (1985); Oi et al, BioTechniques 4:214 (1986); Gillies et al., (1989) J. Immunol. Methods 125:191-202; U.S. Pat. Nos. 5,807,715; 4,816,567; and 4,816,397, which are incorporated herein by reference in their entireties.
  • the chimeric antibodies of the invention are produced by replacing the heavy chain constant region of the murine monoclonal anti human IL- 12 antibodies described in section 1 with a human IgGl constant region.
  • the chimeric antibody of the invention comprises a heavy chain variable region (V H ) comprising the amino acid sequence of SEQ ID NO: 35; SEQ ID NO: 37; SEQ ID NO: 39; SEQ ID NO: 41; SEQ ID NO: 43; SEQ ID NO: 45; SEQ ID NO: 47; SEQ ID NO: 49; SEQ ID NO: 51; or SEQ ID NO: 53 and a light chain variable region (V L ) comprising the amino acid sequence of SEQ ID NO: 36; SEQ ID NO: 38; SEQ ID NO: 40; SEQ ID NO: 42; SEQ ID NO: 44; SEQ ID NO: 46; SEQ ID NO: 48; SEQ BD NO: 50; SEQ ID NO: 52; or SEQ ID NO: 54.
  • V H heavy chain variable region
  • V L light chain variable region
  • CDR-grafted antibodies of the invention comprise heavy and light chain variable region sequences from a human antibody wherein one or more of the CDR regions of V H and/or V L are replaced with CDR sequences of the murine antibodies of the invention.
  • a framework sequence from any human antibody may serve as the template for CDR grafting.
  • straight chain replacement onto such a framework often leads to some loss of binding affinity to the antigen. The more homologous a human antibody is to the original murine antibody, the less likely the possibility that combining the murine CDRs with the human framework will introduce distortions in the CDRs that could reduce affinity.
  • the human variable framework that is chosen to replace the murine variable framework apart from the CDRs have at least a 65% sequence identity with the murine antibody variable region framework. It is more preferable that the human and murine variable regions apart from the CDRs have at least 70% sequence identify. It is even more preferable that the human and murine variable regions apart from the CDRs have at least 75% sequence identity. It is most preferable that the human and murine variable regions apart from the CDRs have at least 80% sequence identity.
  • Methods for producing chimeric antibodies are known in the art and discussed in detail in Example 2.2. (also see EP 239,400; PCT publication WO 91/09967; U.S. Pat. Nos.
  • the invention provides CDR grafted antibodies with V H and/or V L chains as described in Table 7.
  • Humanized antibodies are antibody molecules from non-human species antibody that binds the desired antigen having one or more complementarity determining regions (CDRs) from the non-human species and framework regions from a human immunoglobulin molecule.
  • CDRs complementarity determining regions
  • Known human Ig sequences are disclosed, e.g., www.ncbi.nlm.nih.gov/entrez- /query.fcgi; www.atcc.org/phage/hdb.html; www.sciquest.com/; www.abcam.com/; www.antibodyresource.com/onlinecomp.html; www.public.iastate.edu/.about.pedro/research_tools.html; www.mgen.uni- heidelberg.de/SD/IT/rr.html; www.whfreeman.com/immunology/CH- 05/kuby05.htm; www.library.thinkquest.org/12429/
  • Framework residues in the human framework regions may be substituted with the corresponding residue from the CDR donor antibody to alter, preferably improve, antigen binding.
  • These framework substitutions are identified by methods well known in the art, e.g., by modeling of the interactions of the CDR and framework residues to identify framework residues important for antigen binding and sequence comparison to identify unusual framework residues at particular positions. (See, e.g., Queen et al., U.S. Pat. No. 5,585,089; Riechmann et al., Nature 332:323 (1988), which are incorporated herein by reference in their entireties.) Three- dimensional immunoglobulin models are commonly available and are familiar to those skilled in the art.
  • Antibodies can be humanized using a variety of techniques known in the art, such as but not limited to those described in Jones et al., Nature 321:522 (1986); Verhoeyen et al., Science 239:1534 (1988)), Sims et al., J. Immunol. 151: 2296 (1993); Chothia and Lesk, J. MoI. Biol. 196:901 (1987), Carter et al., Proc. Natl. Acad. Sci. U.S.A. 89:4285 (1992); Presta et al., J. Immunol.
  • anti-IL-12p40 antibodies of the present invention exhibit a high capacity to reduce or to neutralize IL-12 activity, e.g.,as assessed by any one of several in vitro and in vivo assays known in the art (e.g., see Example l.l.C).
  • these antibodies neutralize JL-12- induced production of interferon gamma by PHA blasts with IC 50 values in the range of at least about 10 '8 M, about 10 '9 M, or about 10 '10 M.
  • anti-IL-12p40 antibodies of the present invention also exhibit a high capacity to reduce or to neutralize IL-23 activity
  • the isolated antibody, or antigen-binding portion thereof binds human BL-12p40, wherein the antibody, or antigen-binding portion thereof, dissociates from human IL-12p40 with a Ic 0Jf rate constant of about 0. Is "1 or less, as determined by surface plasmon resonance, or which inhibits human IL-12 and/or human IL-23 activity with an IC 50 of about 1 x 10 '6 M or less.
  • the antibody, or an antigen-binding portion thereof may dissociate from human IL-12p40 with a k Off rate constant of about 1 x 10 "2 S -1 Or less, as determined by surface plasmon resonance, or may inhibit human IL-12 and/or human IL-23 activity with an IC 50 of about 1 x 10 "7 M or less.
  • the antibody, or an antigen-binding portion thereof may dissociate from human IL-12p40 with a karate constant of about 1 x 10 "3 S "1 or less, as determined by surface plasmon resonance, or may inhibit human IL-12 and/or human IL-23 with an IC 50 of about 1 x 10 "8 M or less.
  • the antibody, or an antigen-binding portion thereof may dissociate from human IL-12p40 with a k off rate constant of about 1 x 10 "4 S "1 or less, as determined by surface plasmon resonance, or may inhibit IL-12 and/or human IL-23 activity with an IC 50 of about 1 x 10 "9 M or less.
  • the antibody, or an antigen-binding portion thereof may dissociate from human IL-12p40 with a k ⁇ rate constant of about 1 x 10 "5 S "1 or less, as determined by surface plasmon resonance, or may inhibit IL-12 and/or human EL-23 activity with an IC 50 of about 1 x 10 "10 M or less.
  • the antibody, or an antigen-binding portion thereof may dissociate from human IL-12p40 with a k off rate constant of about 1 x 10 " V 1 Or less, as determined by surface plasmon resonance, or may inhibit IL-12 and/or human EL-23 activity with an IC 50 of about 1 x 10 "11 M or less.
  • the antibody comprises a heavy chain constant region, such as an IgGl, IgG2, IgG3, IgG4, IgA, IgE, IgM or IgD constant region.
  • the heavy chain constant region is an IgGl heavy chain constant region or an IgG4 heavy chain constant region.
  • the antibody can comprise a light chain constant region, either a kappa light chain constant region or a lambda light chain constant region.
  • the antibody comprises a kappa light chain constant region.
  • the antibody portion can be, for example, a Fab fragment or a single chain Fv fragment.
  • the Fc portion of an antibody mediates several important effector functions e.g. cytokine induction, ADCC, phagocytosis, complement dependent cytotoxicity (CDC) and half-life/ clearance rate of antibody and antigen-antibody complexes. In some cases these effector functions are desirable for therapeutic antibody but in other cases might be unnecessary or even deleterious, depending on the therapeutic objectives.
  • Neonatal Fc receptors are the critical components determining the circulating half-life of antibodies.
  • at least one amino acid residue is replaced in the constant region of the antibody, for example the Fc region of the antibody, such that effector functions of the antibody are altered.
  • a labeled binding protein wherein an antibody or antibody portion of the invention is derivatized or linked to another functional molecule (e.g., another peptide or protein).
  • a labeled binding protein of the invention can be derived by functionally linking an antibody or antibody portion of the invention (by chemical coupling, genetic fusion, noncovalent association or otherwise) to one or more other molecular entities, such as another antibody (e.g., a bispecific antibody or a diabody), a detectable agent, a cytotoxic agent, a pharmaceutical agent, and/or a protein or peptide that can mediate associate of the antibody or antibody portion with another molecule (such as a streptavidin core region or a polyhistidine tag).
  • Useful detectable agents with which an antibody or antibody portion of the invention may be derivatized include fluorescent compounds.
  • Exemplary fluorescent detectable agents include fluorescein, fluorescein isothiocyanate, rhodamine, 5-dimethylamine-l- napthalenesulfonyl chloride, phycoerythrin and the like.
  • An antibody may also be derivatized with detectable enzymes, such as alkaline phosphatase, horseradish peroxidase, glucose oxidase and the like. When an antibody is derivatized with a detectable enzyme, it is detected by adding additional reagents that the enzyme uses to produce a detectable reaction product.
  • the detectable agent horseradish peroxidase when the detectable agent horseradish peroxidase is present, the addition of hydrogen peroxide and diaminobenzidine leads to a colored reaction product, which is detectable.
  • An antibody may also be derivatized with biotin, and detected through indirect measurement of avidin or streptavidin binding.
  • Another embodiment of the invention provides a crystallized binding protein.
  • the invention relates to crystals of whole anti-IL-12p40 antibodies and fragments thereof as disclosed herein, and formulations and compositions comprising such crystals.
  • the crystallized binding protein has a greater half-life in vivo than the soluble counterpart of the binding protein.
  • the binding protein retains biological activity after crystallization.
  • Crystallized binding protein of the invention may be produced according methods known in the art and as disclosed in WO 02072636, incorporated herein by reference.
  • Another embodiment of the invention provides a glycosylated binding protein wherein the antibody or antigen-binding portion thereof comprises one or more carbohydrate residues. Nascent in vivo protein production may undergo further processing, known as post-translational modification. In particular, sugar (glycosyl) residues may be added enzymatically, a process known as glycosylation. The resulting proteins bearing covalently linked oligosaccharide side chains are known as glycosylated proteins or glycoproteins.
  • Antibodies are glycoproteins with one or more carbohydrate residues in the Fc domain, as well as the variable domain.
  • Carbohydrate residues in the Fc domain have important effect on the effector function of the Fc domain, with minimal effect on antigen binding or half-life of the antibody (R. Jefferis, Biotechnol. Prog. 21 (2005), pp. 11-16).
  • glycosylation of the variable domain may have an effect on the antigen binding activity of the antibody.
  • Glycosylation in the variable domain may have a negative effect on antibody binding affinity, likely due to steric hindrance (Co, M.S., et al., MoI. Immunol. (1993) 30:1361- 1367), or result in increased affinity for the antigen (Wallick, S.C., et al., Exp. Med. (1988) 168:1099-1109; Wright, A., et al., EMBO J. (1991) 10:2717 2723).
  • One aspect of the present invention is directed to generating glycosylation site mutants in which the O- or N-linked glycosylation site of the binding protein has been mutated.
  • One skilled in the art can generate such mutants using standard well-known technologies.
  • Glycosylation site mutants that retain the biological activity but have increased or decreased binding activity are another object of the present invention.
  • the glycosylation of the antibody or antigen-binding portion of the invention is modified.
  • an aglycoslated antibody can be made (i.e., the antibody lacks glycosylation).
  • Glycosylation can be altered to, for example, increase the affinity of the antibody for antigen.
  • carbohydrate modifications can be accomplished by, for example, altering one or more sites of glycosylation within the antibody sequence.
  • one or more amino acid substitutions can be made that result in elimination of one or more variable region glycosylation sites to thereby eliminate glycosylation at that site.
  • Such aglycosylation may increase the affinity of the antibody for antigen.
  • Such an approach is described in further detail in PCT Publication WO2003016466A2, and U.S. Pat. Nos. 5,714,350 and 6,350,861, each of which is incorporated herein by reference in its entirety.
  • a modified antibody of the invention can be made that has an altered type of glycosylation, such as a hypofucosylated antibody having reduced amounts of fucosyl residues or an antibody having increased bisecting GIcNAc structures.
  • Such altered glycosylation patterns have been demonstrated to increase the ADCC ability of antibodies.
  • carbohydrate modifications can be accomplished by, for example, expressing the antibody in a host cell with altered glycosylation machinery. Cells with altered glycosylation machinery have been described in the art and can be used as host cells in which to express recombinant antibodies of the invention to thereby produce an antibody with altered glycosylation. See, for example, Shields, R. L. et al. (2002) J. Biol. Chem.
  • Protein glycosylation depends on the amino acid sequence of the protein of interest, as well as the host cell in which the protein is expressed. Different organisms may produce different glycosylation enzymes (eg., glycosyltransferases and glycosidases), and have different substrates (nucleotide sugars) available. Due to such factors, protein glycosylation pattern, and composition of glycosyl residues, may differ depending on the host system in which the particular protein is expressed. Glycosyl residues useful in the invention may include, but are not limited to, glucose, galactose, mannose, fucose, n-acetylglucosamine and sialic acid.
  • the glycosylated binding protein comprises glycosyl residues such that the glycosylation pattern is human.
  • a therapeutic protein produced in a microorganism host such as yeast
  • glycosylated utilizing the yeast endogenous pathway may be reduced compared to that of the same protein expressed in a mammalian cell, such as a CHO cell line.
  • Such glycoproteins may also be immunogenic in humans and show reduced half-life in vivo after administration.
  • Specific receptors in humans and other animals may recognize specific glycosyl residues and promote the rapid clearance of the protein from the bloodstream.
  • a practitioner may prefer a therapeutic protein with a specific composition and pattern of glycosylation, for example glycosylation composition and pattern identical, or at least similar, to that produced in human cells or in the species-specific cells of the intended subject animal.
  • glycosylated proteins different from that of a host cell may be achieved by genetically modifying the host cell to express heterologous glycosylation enzymes. Using techniques known in the art a practitioner may generate antibodies or antigen-binding portions thereof exhibiting human protein glycosylation. For example, yeast strains have been genetically modified to express non-naturally occurring glycosylation enzymes such that glycosylated proteins (glycoproteins) produced in these yeast strains exhibit protein glycosylation identical to that of animal cells, especially human cells (U.S patent applications 20040018590 and 20020137134 and PCT publication WO2005100584 A2).
  • an anti- idiotypic (anti-Id) antibody specific for such binding proteins of the invention is also directed to an anti- idiotypic (anti-Id) antibody specific for such binding proteins of the invention.
  • An anti-Id antibody is an antibody, which recognizes unique determinants generally associated with the antigen-binding region of another antibody.
  • the anti-Id can be prepared by immunizing an animal with the binding protein or a CDR containing region thereof. The immunized animal will recognize, and respond to the idiotypic determinants of the immunizing antibody and produce an anti-Id antibody.
  • the anti-Id antibody may also be used as an "immunogen" to induce an immune response in yet another animal, producing a so-called anti-anti-Id antibody.
  • a protein of interest may be expressed using a library of host cells genetically engineered to express various glycosylation enzymes, such that member host cells of the library produce the protein of interest with variant glycosylation patterns.
  • a practitioner may then select and isolate the protein of interest with particular novel glycosylation patterns.
  • the protein having a particularly selected novel glycosylation pattern exhibits improved or altered biological properties.
  • the anti-human IL-12p40 antibodies, or portions thereof, of the invention can be used to detect EL- 12 and/or human IL-23 ⁇ e.g., in a biological sample, such as serum or plasma), using a conventional immunoassay, such as an enzyme linked immunosorbent assays (ELISA), an radioimmunoassay (RIA) or tissue immunohistochemistry.
  • a biological sample such as serum or plasma
  • a conventional immunoassay such as an enzyme linked immunosorbent assays (ELISA), an radioimmunoassay (RIA) or tissue immunohistochemistry.
  • the invention provides a method for detecting IL- 12 and/or human IL- 23 in a biological sample comprising contacting a biological sample with an antibody, or antibody portion, of the invention and detecting either the antibody (or antibody portion) bound to IL-12 and/or human IL-23 or unbound antibody (or antibody portion), to thereby detect IL-12 and/or human IL-23 in the biological sample.
  • the antibody is directly or indirectly labeled with a detectable substance to facilitate detection of the bound or unbound antibody. Suitable detectable substances include various enzymes, prosthetic groups, fluorescent materials, luminescent materials and radioactive materials.
  • suitable enzymes include horseradish peroxidase, alkaline phosphatase, ⁇ -galactosidase, or acetylcholinesterase;
  • suitable prosthetic group complexes include streptavidin/biotin and avidin/biotin;
  • suitable fluorescent materials include umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride or phycoerythrin; an example of a luminescent material includes luminol; and examples of suitable radioactive material include 3 H 14 C, 35 S 1 90 Y, 99 Tc, 111 In, 125 1, 131 1, 177 Lu, 166 Ho, or 153 Sm.
  • human IL-12 can be assayed in biological fluids by a competition immunoassay utilizing rhIL-12 standards labeled with a detectable substance and an unlabeled anti- human IL-12p40 antibody.
  • the biological sample, the labeled rhlL- 12 standards and the anti- human IL-12p40 antibody are combined and the amount of labeled rhIL-12 standard bound to the unlabeled antibody is determined.
  • the amount of human IL-12 in the biological sample is inversely proportional to the amount of labeled rhIL-12 standard bound to the anti-EL-12p40 antibody.
  • human EL-23 can also be assayed in biological fluids by a competition immunoassay utilizing rhlL-23 standards labeled with a detectable substance and an unlabeled anti-human IL-12p40 antibody.
  • the antibodies and antibody portions of the invention preferably are capable of neutralizing human IL-12 and/or human IL-23 activity both in vitro and in vivo. Accordingly, such antibodies and antibody portions of the invention can be used to inhibit ML-12 and/or ML- 23 activity, e.g., in a cell culture containing hIL-12 and/or hIL-23, in human subjects or in other mammalian subjects having IL-12 and/or hIL-23 with which an antibody of the invention cross- reacts.
  • the invention provides a method for inhibiting ML-12 and/or hIL-23 activity comprising contacting ML- 12 and/or hIL-23 with an antibody or antibody portion of the invention such that hIL-12 and/or hIL-23 activity is inhibited.
  • an antibody or antibody portion of the invention can be added to the culture medium to inhibit ML- 12 and/or hIL-23 activity in the culture.
  • the invention provides a method for reducing ML-12 and/or ML- 23 activity in a subject, advantageously from a subject suffering from a disease or disorder in which IL-12 or IL-23 activity is detrimental.
  • the invention provides methods for reducing IL-12 and/or IL-23 activity in a subject suffering from such a disease or disorder, which method comprises administering to the subject an antibody or antibody portion of the invention such that IL-12 and/or IL-23 activity in the subject is reduced.
  • the IL-12 is human IL-12
  • the IL-23 is human IL-23
  • the subject is a human subject.
  • the subject can be a mammal expressing an IL-12 and/or IL-23 to which an antibody of the invention is capable of binding. Still further the subject can be a mammal into which IL-12 and/or IL-23 has been introduced (e.g., by administration of IL-12 and/or IL-23 or by expression of an IL-12 and/or IL- 23 transgene).
  • An antibody of the invention can be administered to a human subject for therapeutic purposes.
  • an antibody of the invention can be administered to a non- human mammal expressing an IL-12 and/or IL-23 with which the antibody is capable of binding for veterinary purposes or as an animal model of human disease. Regarding the latter, such animal models may be useful for evaluating the therapeutic efficacy of antibodies of the invention (e.g., testing of dosages and time courses of administration).
  • a disorder in which IL-12 and/or IL-23 activity is detrimental is intended to include diseases and other disorders in wMch the presence of IL-12 and/or IL-23 in a subject suffering from the disorder has been shown to be or is suspected of being either responsible for the pathophysiology of the disorder or a factor that contributes to a worsening of the disorder. Accordingly, a disorder in which IL-12 and/or EL-23 activity is detrimental is a disorder in which reduction of IL-12 and/or JL-23 activity is expected to alleviate the symptoms and/or progression of the disorder.
  • disorders may be evidenced, for example, by an increase in the concentration of IL-12 and/or IL-23 in a biological fluid of a subject suffering from the disorder (e.g., an increase in the concentration of IL-12 and/or IL-23 in serum, plasma, synovial fluid, etc. of the subject), which can be detected, for example, using an anti-EL-12p40 antibody as described above.
  • disorders that can be treated with the antibodies of the invention include those disorders discussed in the section below pertaining to pharmaceutical compositions of the antibodies of the invention.
  • the invention also provides pharmaceutical compositions comprising an antibody, or antigen-binding portion thereof, of the invention and a pharmaceutically acceptable carrier.
  • the pharmaceutical compositions comprising antibodies of the invention are for use in, but not limited to, diagnosing, detecting, or monitoring a disorder, in preventing, treating, managing, or ameliorating of a disorder or one or more symptoms thereof, and/or in research.
  • a composition comprises one or more antibodies of the invention
  • the pharmaceutical composition comprises one or more antibodies of the invention and one or more prophylactic or therapeutic agents other than antibodies of the invention for treating a disorder in which IL-12 and/or IL-23 activity is detrimental.
  • the composition may further comprise of a carrier, diluent or excipient.
  • the antibodies and antibody-portions of the invention can be incorporated into pharmaceutical compositions suitable for administration to a subject.
  • the pharmaceutical composition comprises an antibody or antibody portion of the invention and a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable carrier includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like that are physiologically compatible.
  • pharmaceutically acceptable carriers include one or more of water, saline, phosphate buffered saline, dextrose, glycerol, ethanol and the like, as well as combinations thereof.
  • isotonic agents for example, sugars, polyalcohols such as mannitol, sorbitol, or sodium chloride in the composition.
  • Pharmaceutically acceptable carriers may further comprise minor amounts of auxiliary substances such as wetting or emulsifying agents, preservatives or buffers, which enhance the shelf life or effectiveness of the antibody or antibody portion.
  • Various delivery systems are known and can be used to administer one or more antibodies of the invention or the combination of one or more antibodies of the invention and a prophylactic agent or therapeutic agent useful for preventing, managing, treating, or ameliorating a disorder or one or more symptoms thereof, e.g., encapsulation in liposomes, microparticles, microcapsules, recombinant cells capable of expressing the antibody or antibody fragment, receptor- mediated endocytosis (see, e. g., Wu and Wu, J. Biol. Chem. 262:4429-4432 (1987)), construction of a nucleic acid as part of a retroviral or other vector, etc.
  • a prophylactic agent or therapeutic agent useful for preventing, managing, treating, or ameliorating a disorder or one or more symptoms thereof, e.g., encapsulation in liposomes, microparticles, microcapsules, recombinant cells capable of expressing the antibody or antibody fragment, receptor- mediated endocytosis (
  • Methods of administering a prophylactic or therapeutic agent of the invention include, but are not limited to, parenteral administration (e.g., intradermal, intramuscular, intraperitoneal, intravenous and subcutaneous) , epidurala administration, intratumoral administration, and mucosal adminsitration (e.g., intranasal and oral routes).
  • parenteral administration e.g., intradermal, intramuscular, intraperitoneal, intravenous and subcutaneous
  • epidurala administration e.g., intratumoral administration
  • mucosal adminsitration e.g., intranasal and oral routes.
  • pulmonary administration can be employed, e.g., by use of an inhaler or nebulizer, and formulation with an aerosolizing agent. See, e.g., U.S. Pat. Nos.
  • an antibody of the invention, combination therapy, or a composition of the invention is administered using Alkermes AIR® pulmonary drug delivery technology (Alkermes, Inc., Cambridge, Mass.).
  • prophylactic or therapeutic agents of the invention are administered intramuscularly, intravenously, intratumorally, orally, intranasally, pulmonary, or subcutaneously.
  • the prophylactic or therapeutic agents may be administered by any convenient route, for example by infusion or bolus injection, by absorption through epithelial or mucocutaneous linings (e.g. , oral mucosa, rectal and intestinal mucosa, etc.) and may be administered together with other biologically active agents. Administration can be systemic or local.
  • the prophylactic or therapeutic agents of the invention may be desirable to administer the prophylactic or therapeutic agents of the invention locally to the area in need of treatment; this may be achieved by, for example, and not by way of limitation, local infusion, by injection, or by means of an implant, said implant being of a porous or non-porous material, including membranes and matrices, such as sialastic membranes, polymers, fibrous matrices (e.g., Tissuel®), or collagen matrices.
  • an effective amount of one or more antibodies of the invention antagonists is administered locally to the affected area to a subject to prevent, treat, manage, and/or ameliorate a disorder or a symptom thereof.
  • an effective amount of one or more antibodies of the invention is administered locally to the affected area in combination with an effective amount of one or more therapies (e. g., one or more prophylactic or therapeutic agents) other than an antibody of the invention of a subject to prevent, treat, manage, and/or ameliorate a disorder or one or more symptoms thereof.
  • therapies e. g., one or more prophylactic or therapeutic agents
  • the prophylactic or therapeutic agent can be delivered in a controlled release or sustained release system.
  • a pump may be used to achieve controlled or sustained release (see Langer, supra; Sefton, 1987, CRC Crit. Ref. Biomed. Eng. 14:20; Buchwald et al., 1980, Surgery 88:507; Saudek et al., 1989, N. Engl. J. Med. 321:574).
  • polymeric materials can be used to achieve controlled or sustained release of the therapies of the invention (see e.g., Medical Applications of Controlled Release, Langer and Wise (eds.), CRC Pres., Boca Raton, FIa.
  • polymers used in sustained release formulations include, but are not limited to, poly(2-hydroxy ethyl methacrylate), poly(methyl methacrylate), poly(acrylic acid), poly(ethylene-co-vinyl acetate), poly(methacrylic acid), polyglycolides (PLG), polyanhydrides, poly(N- vinyl pyrrolidone), poly(vinyl alcohol), polyacrylamide, poly(ethylene glycol), polylactides (PLA), poly(lactide-co-glycolides) (PLGA), and polyorthoesters.
  • the polymer used in a sustained release formulation is inert, free of leachable impurities, stable on storage, sterile, and biodegradable.
  • a controlled or sustained release system can be placed in proximity of the prophylactic or therapeutic target, thus requiring only a fraction of the systemic dose (see, e.g., Goodson, in Medical Applications of Controlled Release, supra, vol. 2, pp. 115-138 (1984)).
  • Controlled release systems are discussed in the review by Langer (1990, Science 249:1527-1533). Any technique known to one of skill in the art can be used to produce sustained release formulations comprising one or more therapeutic agents of the invention. See, e.g., U. S. Pat. No. 4,526, 938, PCT publication WO 91/05548, PCT publication WO 96/20698, Ning et al.
  • the composition of the invention is a nucleic acid encoding a prophylactic or therapeutic agent
  • the nucleic acid can be administered in vivo to promote expression of its encoded prophylactic or therapeutic agent, by constructing it as part of an appropriate nucleic acid expression vector and administering it so that it becomes intracellular, e.g., by use of a retroviral vector (see U. S. Pat. No.
  • a nucleic acid can be introduced intracellularly and incorporated within host cell DNA for expression by homologous recombination.
  • a pharmaceutical composition of the invention is formulated to be compatible with its intended route of administration.
  • routes of administration include, but are not limited to, parenteral, e.g., intravenous, intradermal, subcutaneous, oral, intranasal (e.g., inhalation), transdermal (e.g., topical), transmucosal, and rectal administration.
  • the composition is formulated in accordance with routine procedures as a pharmaceutical composition adapted for intravenous, subcutaneous, intramuscular, oral, intranasal, or topical administration to human beings.
  • compositions for intravenous administration are solutions in sterile isotonic aqueous buffer.
  • the composition may also include a solubilizing agent and a local anesthetic such as lignocamne to ease pain at the site of the injection.
  • compositions of the invention are to be administered topically, the compositions can be formulated in the form of an ointment, cream, transdermal patch, lotion, gel, shampoo, spray, aerosol, solution, emulsion, or other form well known to one of skill in the art. See, e.g., Remington's Pharmaceutical Sciences and Introduction to Pharmaceutical Dosage Forms, 19th ed., Mack Pub. Co., Easton, Pa. (1995).
  • viscous to semisolid or solid forms comprising a carrier or one or more excipients compatible with topical application and having a dynamic viscosity preferably greater than water are typically employed.
  • Suitable formulations include, without limitation, solutions, suspensions, emulsions, creams, ointments, powders, liniments, salves, and the like, which are, if desired, sterilized or mixed with auxiliary agents (e.g., preservatives, stabilizers, wetting agents, buffers, or salts) for influencing various properties, such as, for example, osmotic pressure.
  • auxiliary agents e.g., preservatives, stabilizers, wetting agents, buffers, or salts
  • Other suitable topical dosage forms include sprayable aerosol preparations wherein the active ingredient, preferably in combination with a solid or liquid inert carrier, is packaged in a mixture with a pressurized volatile (e.g., a gaseous propellant, such as freon) or in a squeeze bottle.
  • a pressurized volatile e.g., a gaseous propellant, such as freon
  • humectants can also be added to pharmaceutical composition
  • the composition can be formulated in an aerosol form, spray, mist or in the form of drops.
  • prophylactic or therapeutic agents for use according to the present invention can be conveniently delivered in the form of an aerosol spray presentation from pressurized packs or a nebuliser, with the use of a suitable propellant (e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas).
  • a suitable propellant e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • the dosage unit may be determined by providing a valve to deliver a metered amount.
  • Capsules and cartridges for use in an inhaler or insufflator may be formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch.
  • compositions can be formulated orally in the form of tablets, capsules, cachets, gelcaps, solutions, suspensions, and the like.
  • Tablets or capsules can be prepared by conventional means with pharmaceutically acceptable excipients such as binding agents (e.g., pregelatinised maize starch, polyvinylpyrrolidone, or hydroxypropyl methylcellulose); fillers (e.g., lactose, microcrystalline cellulose, or calcium hydrogen phosphate) ; lubricants (e.g., magnesium stearate, talc, or silica); disintegrants (e.g., potato starch or sodium starch glycolate) ; or wetting agents (e.g., sodium lauryl sulphate).
  • binding agents e.g., pregelatinised maize starch, polyvinylpyrrolidone, or hydroxypropyl methylcellulose
  • fillers e.g., lactose, microcrystalline cellulose, or calcium hydrogen phosphate
  • Liquid preparations for oral administration may take the form of, but not limited to, solutions, syrups or suspensions, or they may be presented as a dry product for constitution with water or other suitable vehicle before use.
  • Such liquid preparations may be prepared by conventional means with pharmaceutically acceptable additives such as suspending agents (e.g., sorbitol syrup, cellulose derivatives, or hydrogenated edible fats); emulsifying agents (e.g., lecithin or acacia); non-aqueous vehicles (e.g., almond oil, oily esters, ethyl alcohol, or fractionated vegetable oils); and preservatives (e.g., methyl or propyl-p- hydroxybenzoates or sorbic acid).
  • the preparations may also contain buffer salts, flavoring, coloring, and sweetening agents as appropriate.
  • Preparations for oral administration may be suitably formulated for slow release, controlled release, or sustained release of a prophylactic or therapeutic agent(s).
  • the method of the invention may comprise pulmonary administration, e.g., by use of an inhaler or nebulizer, of a composition formulated with an aerosolizing agent.
  • pulmonary administration e.g., by use of an inhaler or nebulizer, of a composition formulated with an aerosolizing agent.
  • an antibody of the invention, combination therapy, and/or composition of the invention is administered using Alkermes AIR® pulmonary drug delivery technology (Alkermes, Inc., Cambridge, Mass.).
  • the method of the invention may comprise administration of a composition formulated for parenteral administration by injection (e. g., by bolus injection or continuous infusion).
  • Formulations for injection may be presented in unit dosage form (e.g., in ampoules or in multi- dose containers) with an added preservative.
  • the compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • the active ingredient may be in powder form for constitution with a suitable vehicle (e.g., sterile pyrogen- free water) before use.
  • the methods of the invention may additionally comprise of administration of compositions formulated as depot preparations.
  • compositions may be administered by implantation (e.g., subcutaneously or intramuscularly) or by intramuscular injection.
  • the compositions may be formulated with suitable polymeric or hydrophobic materials (e.g., as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives (e.g., as a sparingly soluble salt).
  • compositions formulated as neutral or salt forms include those formed with anions such as those derived from hydrochloric, phosphoric, acetic, oxalic, tartaric acids, etc., and those formed with cations such as those derived from sodium, potassium, ammonium, calcium, ferric hydroxides, isopropylamine, triethylamine, 2- ethylamino ethanol, histidine, procaine, etc.
  • compositions are supplied either separately or mixed together in unit dosage form, for example, as a dry lyophilized powder or water free concentrate in a hermetically sealed container such as an ampoule or sachette indicating the quantity of active agent.
  • a hermetically sealed container such as an ampoule or sachette indicating the quantity of active agent.
  • composition can be dispensed with an infusion bottle containing sterile pharmaceutical grade water or saline.
  • an ampoule of sterile water for injection or saline can be provided so that the ingredients may be mixed prior to administration.
  • the invention also provides that one or more of the prophylactic or therapeutic agents, or pharmaceutical compositions of the invention is packaged in a hermetically sealed container such as an ampoule or sachette indicating the quantity of the agent.
  • a hermetically sealed container such as an ampoule or sachette indicating the quantity of the agent.
  • one or more of the prophylactic or therapeutic agents, or pharmaceutical compositions of the invention is supplied as a dry sterilized lyophilized powder or water free concentrate in a hermetically sealed container and can be reconstituted (e.g., with water or saline) to the appropriate concentration for administration to a subject.
  • one or more of the prophylactic or therapeutic agents or pharmaceutical compositions of the invention is supplied as a dry sterile lyophilized powder in a hermetically sealed container at a unit dosage of at least 5 mg, more preferably at least 10 mg, at least 15 mg, at least 25 mg, at least 35 mg, at least 45 mg, at least 50 mg, at least 75 mg, or at least 100 mg.
  • the lyophilized prophylactic or therapeutic agents or pharmaceutical compositions of the invention should be stored at between 2° C. and 8° C.
  • the prophylactic or therapeutic agents, or pharmaceutical compositions of the invention should be administered within 1 week, preferably within 5 days, within 72 hours, within 48 hours, within 24 hours, within 12 hours, within 6 hours, within 5 hours, within 3 hours, or within 1 hour after being reconstituted.
  • one or more of the prophylactic or therapeutic agents or pharmaceutical compositions of the invention is supplied in liquid form in a hermetically sealed container indicating the quantity and concentration of the agent.
  • the liquid form of the administered composition is supplied in a hermetically sealed container at least 0.25 mg/ml, more preferably at least 0.5 mg/ml, at least 1 mg/ml, at least 2.5 mg/ml, at least 5 mg/ml, at least 8 mg/ml, at least 10 mg/ml, at least 15 mg/kg, at least 25 mg/ml, at least 50 mg/ml, at least 75 mg/ml or at least 100 mg/ml.
  • the liquid form should be stored at between 2° C. and 8° C. in its original container.
  • the antibodies and antibody-portions of the invention can be incorporated into a pharmaceutical composition suitable for parenteral administration.
  • the antibody or antibody-portions will be prepared as an injectable solution containing 0.1-250 mg/ml antibody.
  • the injectable solution can be composed of either a liquid or lyophilized dosage form in a flint or amber vial, ampule or pre-filled syringe.
  • the buffer can be L-histidine (1-50 mM), optimally 5- 1OmM, at pH 5.0 to 7.0 (optimally pH 6.0).
  • Other suitable buffers include but are not limited to, sodium succinate, sodium citrate, sodium phosphate or potassium phosphate.
  • Sodium chloride can be used to modify the toxicity of the solution at a concentration of 0-300 mM (optimally 150 mM for a liquid dosage form).
  • Cryoprotectants can be included for a lyophilized dosage form, principally 0-10% sucrose (optimally 0.5-1.0%).
  • Other suitable cryoprotectants include trehalose and lactose.
  • Bulking agents can be included for a lyophilized dosage form, principally 1-10% mannitol (optimally 2-4%).
  • Stabilizers can be used in both liquid and lyophilized dosage forms, principally 1-50 mM L-Methionine (optimally 5-10 mM).
  • compositions comprising the antibodies and antibody-portions of the invention prepared as an injectable solution for parenteral administration, can further comprise an agent useful as an adjuvant, such as those used to increase the absorption, or dispersion of a therapeutic protein (e.g., antibody).
  • an agent useful as an adjuvant such as those used to increase the absorption, or dispersion of a therapeutic protein (e.g., antibody).
  • a particularly useful adjuvant is hyaluronidase, such as Hylenex® (recombinant human hyaluronidase).
  • Hylenex® recombinant human hyaluronidase
  • Additional hyaluronidase in the injectable solution improves human bioavailability following parenteral administration, particularly subcutaneous administration. It also allows for greater injection site volumes (i.e. greater than 1 ml) with less pain and discomfort, and minimum incidence of injection site reactions, (see WO2004078140, US2006104968 incorporated herein by reference).
  • compositions of this invention may be in a variety of forms. These include, for example, liquid, semi-solid and solid dosage forms, such as liquid solutions (e.g., injectable and infusible solutions), dispersions or suspensions, tablets, pills, powders, liposomes and suppositories.
  • liquid solutions e.g., injectable and infusible solutions
  • dispersions or suspensions tablets, pills, powders, liposomes and suppositories.
  • the preferred form depends on the intended mode of administration and therapeutic application. Typical preferred compositions are in the form of injectable or infusible solutions, such as compositions similar to those used for passive immunization of humans with other antibodies.
  • the preferred mode of administration is parenteral (e.g., intravenous, subcutaneous, intraperitoneal, intramuscular).
  • the antibody is administered by intravenous infusion or injection.
  • the antibody is administered by intramuscular or subcutaneous injection.
  • compositions typically must be sterile and stable under the conditions of manufacture and storage.
  • the composition can be formulated as a solution, microemulsion, dispersion, liposome, or other ordered structure suitable to high drug concentration.
  • Sterile injectable solutions can be prepared by incorporating the active compound (i.e., antibody or antibody portion) in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization.
  • dispersions are prepared by incorporating the active compound into a sterile vehicle that contains a basic dispersion medium and the required other ingredients from those enumerated above.
  • the preferred methods of preparation are vacuum drying and spray-drying that yields a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • the proper fluidity of a solution can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
  • Prolonged absorption of injectable compositions can be brought about by including, in the composition, an agent that delays absorption, for example, monostearate salts and gelatin.
  • the antibodies and antibody-portions of the present invention can be administered by a variety of methods known in the art, although for many therapeutic applications, the preferred route/mode of administration is subcutaneous injection, intravenous injection or infusion. As will be appreciated by the skilled artisan, the route and/or mode of administration will vary depending upon the desired results.
  • the active compound may be prepared with a carrier that will protect the compound against rapid release, such as a controlled release formulation, including implants, transdermal patches, and microencapsulated delivery systems.
  • a carrier such as a controlled release formulation, including implants, transdermal patches, and microencapsulated delivery systems.
  • Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid.
  • an antibody or antibody portion of the invention may be orally administered, for example, with an inert diluent or an assimilable edible carrier.
  • the compound (and other ingredients, if desired) may also be enclosed in a hard or soft shell gelatin capsule, compressed into tablets, or incorporated directly into the subject's diet.
  • the compounds may be incorporated with excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, and the like.
  • To administer a compound of the invention by other than parenteral administration it may be necessary to coat the compound with, or co-administer the compound with, a material to prevent its inactivation.
  • an antibody or antibody portion of the invention is coformulated with and/or coadministered with one or more additional therapeutic agents that are useful for treating disorders in which IL-12 activity is detrimental.
  • an anti-hIL-12 antibody or antibody portion of the invention may be coformulated and/or coadministered with one or more additional antibodies that bind other targets ⁇ e.g., antibodies that bind other cytokines or that bind cell surface molecules).
  • one or more antibodies of the invention may be used in combination with two or more of the foregoing therapeutic agents.
  • Such combination therapies may advantageously utilize lower dosages of the administered therapeutic agents, thus avoiding possible toxicities or complications associated with the various monotherapies.
  • an antibody to IL-12 or fragment thereof is linked to a half-life extending vehicle known in the art.
  • vehicles include, but are not limited to, the Fc domain, polyethylene glycol, and dextran.
  • Such vehicles are described, e.g., in U.S. Application Serial No. 09/428,082 and published PCT Application No. WO 99/25044, which are hereby incorporated by reference for any purpose.
  • nucleic acid sequences comprising nucleotide sequences encoding an antibody of the invention or another prophylactic or therapeutic agent of the invention are administered to treat, prevent, manage, or ameliorate a disorder or one or more symptoms thereof by way of gene therapy.
  • Gene therapy refers to therapy performed by the administration to a subject of an expressed or expressible nucleic acid.
  • the nucleic acids produce their encoded antibody or prophylactic or therapeutic agent of the invention that mediates a prophylactic or therapeutic effect.
  • Interleukin 12 plays a critical role in the pathology associated with a variety of diseases involving immune and inflammatory elements. These diseases include, but are not limited to, rheumatoid arthritis, osteoarthritis, juvenile chronic arthritis, septic arthritis, Lyme arthritis, psoriatic arthritis, reactive arthritis, spondyloarthropathy, systemic lupus erythematosus, Crohn's disease, ulcerative colitis, inflammatory bowel disease, insulin dependent diabetes mellitus, thyroiditis, asthma, allergic diseases, psoriasis, dermatitis scleroderma, graft versus host disease, organ transplant rejection, acute or chronic immune disease associated with organ transplantation, sarcoidosis, atherosclerosis, disseminated intravascular coagulation, Kawasaki's disease, Grave's disease, nephrotic syndrome, chronic fatigue syndrome, Wegener's granulomatosis, Henoch-Schoenlein purpurea, microscopic vascu
  • the antibodies, and antibody portions of the invention can be used to treat humans suffering from autoimmune diseases, in particular those associated with inflammation, including, rheumatoid spondylitis, allergy, autoimmune diabetes, autoimmune uveitis.
  • autoimmune diseases in particular those associated with inflammation, including, rheumatoid spondylitis, allergy, autoimmune diabetes, autoimmune uveitis.
  • the antibodies of the invention or antigen-binding portions thereof are used to treat rheumatoid arthritis, Crohn's disease, multiple sclerosis, insulin dependent diabetes mellitus and psoriasis.
  • An antibody, or antibody portion, of the invention also can be administered with one or more additional therapeutic agents useful in the treatment of autoimmune and inflammatory diseases.
  • Antibodies of the invention, or antigen binding portions thereof can be used alone or in combination to treat such diseases. It should be understood that the antibodies of the invention or antigen binding portion thereof can be used alone or in combination with an additional agent, e.g., a therapeutic agent, said additional agent being selected by the skilled artisan for its intended purpose.
  • the additional agent can be a therapeutic agent art-recognized as being useful to treat the disease or condition being treated by the antibody of the present invention.
  • the additional agent also can be an agent that imparts a beneficial attribute to the therapeutic composition e.g., an agent which effects the viscosity of the composition.
  • the combinations which are to be included within this invention are those combinations useful for their intended purpose.
  • the agents set forth below are illustrative for purposes and not intended to be limited.
  • the combinations, which are part of this invention can be the antibodies of the present invention and at least one additional agent selected from the lists below.
  • the combination can also include more than one additional agent, e.g., two or three additional agents if the combination is such that the formed composition can perform its intended function.
  • Binding proteins described herein may be used in combination with additional therapeutic agents such as a Disease Modifying Anti-Rheumatic Drug (DMARD) or a Nonsteroidal Antiinflammatory Drug (NSAID) or a steroid or any combination thereof.
  • DMARD Disease Modifying Anti-Rheumatic Drug
  • NSAID Nonsteroidal Antiinflammatory Drug
  • Preferred examples of a DMARD are hydroxychloroquine, leflunomide, methotrexate, parenteral gold, oral gold and sulfasalazine.
  • Preferred examples of non-steroidal anti-inflammatory drug(s) also referred to as NSAIDS include drugs like ibuprofen.
  • Non-limiting examples of therapeutic agents for rheumatoid arthritis with which an antibody, or antibody portion, of the invention can be combined include the following: cytokine suppressive anti-inflammatory drug(s) (CSAIDs); antibodies to or antagonists of other human cytokines or growth factors, for example, TNF, LT, IL-I, IL-2, IL-3, IL-4, IL-5, IL-6, IL-I, IL-S, IL-15, IL-16, IL-18, IL-21, IL-23, interferons, EMAP-II, GM-CSF, FGF, and PDGF.
  • CSAIDs cytokine suppressive anti-inflammatory drug
  • Antibodies of the invention, or antigen binding portions thereof, can be combined with antibodies to cell surface molecules such as CD2, CD3, CD4, CD8, CD25, CD28, CD30, CD40, CD45, CD69, CD80 (B7.1), CD86 (B7.2), CD90, CTLA or their ligands including CD154 (gp39 or CD40L).
  • cell surface molecules such as CD2, CD3, CD4, CD8, CD25, CD28, CD30, CD40, CD45, CD69, CD80 (B7.1), CD86 (B7.2), CD90, CTLA or their ligands including CD154 (gp39 or CD40L).
  • TNF antagonists such as soluble p55 or p75 TNF receptors, derivatives, thereof, ( ⁇ 75TNFRlgG (EnbrelTM) or p55TNFRlgG (Lenercept), chimeric, humanized or human TNF antibodies, or a fragment thereof, including infliximab (Remicade ® , Johnson and Johnson; described in U.S. Patent No.
  • CDP571 a humanized monoclonal anti-TNF-alpha IgG4 antibody
  • CDP 870 a humanized monoclonal anti-TNF-alpha antibody fragment
  • an anti- TNF dAb Peptech
  • CNTO 148 golimumab; Medarex and Centocor, see WO 02/12502
  • adalimumab Human anti-TNF mAb, described in US 6,090,382 as D2E7
  • TNF ⁇ converting enzyme (TACE) inhibitors may be effective for the same reason.
  • Other preferred combinations include Merleukin 11.
  • Yet another preferred combination are other key players of the autoimmune response which may act parallel to, dependent on or in concert with IL-12 function; especially preferred are IL-18 antagonists including IL-18 antibodies or soluble IL-18 receptors, or IL-18 binding proteins. It has been shown that IL-12 and IL-18 have overlapping but distinct functions and a combination of antagonists to both may be most effective.
  • Yet another preferred combination are non-depleting anti-CD4 inhibitors.
  • Yet other preferred combinations include antagonists of the co-stimulatory pathway CD80 (B7.1) or CD86 (B7.2) including antibodies, soluble receptors or antagonistic ligands.
  • the antibodies of the invention, or antigen binding portions thereof, may also be combined with agents, such as methotrexate, 6-MP, azathioprine sulphasalazine, mesalazine, olsalazine chloroquinine/hydroxychloroquine, pencillamine, aurothiomalate (intramuscular and oral), azathioprine, cochicine, corticosteroids (oral, inhaled and local injection), beta-2 adrenoreceptor agonists (salbutamol, terbutaline, salmeteral), xanthines (theophylline, aminophylline), cromoglycate, nedocromil, ketotifen, ipratropium and oxitropium, cyclosporin, FK506, rapamycin, mycophenolate mofetil, leflunomide, NSAIDs, for example, ibuprofen, corticosteroids such as prednisolone,
  • IRAK, NTK, IKK , p38 or MAP kinase inhibitors IL-l ⁇ converting enzyme inhibitors
  • TACE TNFq converting enzyme
  • T-cell signalling inhibitors such as kinase inhibitors, metalloproteinase inhibitors, sulfasalazine, azathioprine, 6-mercaptopurines, angiotensin converting enzyme inhibitors, soluble cytokine receptors and derivatives thereof (e.g.
  • soluble p55 or p75 TNF receptors and the derivatives p75TNFRIgG EnbrelTM and p55TNFRIgG (Lenercept)
  • sIL-lRI e.g., IL-lRIL sIL-6R
  • antiinflammatory cytokines e.g.
  • IL-4, IL-10, IL-Il, IL-13 and TGF ⁇ celecoxib, folic acid, hydroxychloroquine sulfate, rofecoxib, etanercept, infliximab, naproxen, valdecoxib, sulfasalazine, methylprednisolone, meloxicam, methylprednisolone acetate, gold sodium thiomalate, aspirin, triamcinolone acetonide, propoxyphene napsylate/apap, folate, nabumetone, diclofenac, piroxicam, etodolac, diclofenac sodium, oxaprozin, oxycodone hcl, hydrocodone bitartrate/apap, diclofenac sodium/misoprostol, fentanyl, anakinra, human recombinant, tramadol hc
  • Nonlimiting additional agents which can also be used in combination with an IL-12 or IL-23 antibody, or antigen-binding portion thereof, to treat rheumatoid arthritis include, but are not limited to, the following: non-steroidal anti-inflammatory drug(s) (NSAIDs); cytokine suppressive anti-inflammatory drug(s) (CSADDs); CDP-571/BAY-10-3356 (humanized anti- TNF ⁇ antibody; Celltech/Bayer); cA2/infliximab (chimeric anti-TNF ⁇ antibody; Centocor); 75 kdTNFR-IgG/etanercept (75 kD TNF receptor-IgG fusion protein; Immunex; see e.g., Arthritis & Rheumatism (1994) Vol.
  • NSAIDs non-steroidal anti-inflammatory drug(s)
  • CSADDs cytokine suppressive anti-inflammatory drug(s)
  • CDP-571/BAY-10-3356 humanized anti- TNF ⁇ antibody
  • Anti-Tac humanized anti-IL-2R ⁇ ; Protein Design Labs/Roche
  • EL-4 anti-inflammatory cytokine; DNAX/Schering
  • IL-10 SCH 52000; recombinant IL-10, anti-inflammatory cytokine; DNAX/Schering
  • IL-4 EL-IO and/or IL-4 agonists (e.g., agonist antibodies);
  • IL-IRA IL-I receptor antagonist; Synergen/Amgen); anakinra (Kineret ® /Amgen); TNF-bp/s-TNF (soluble TNF binding protein; see e.g., Arthritis & Rheumatism (1996) Vol. 39, No.
  • thalidomide see e.g., Arthritis & Rheumatism (1996) Vol. 39, No. 9 (supplement), S282) and thalidomide- related drugs (e.g., Celgen); lefiunomide (anti-inflammatory and cytokine inhibitor; see e.g., Arthritis & Rheumatism (1996) Vol. 39, No. 9 (supplement), S131; Inflammation Research (1996) Vol. 45, pp. 103-107); tranexamic acid (inhibitor of plasminogen activation; see e.g., Arthritis & Rheumatism (1996) Vol. 39, No.
  • Meloxicam non-steroidal anti-inflammatory drug
  • Ibuprofen non-steroidal anti-inflammatory drug
  • Piroxicam non-steroidal anti-inflammatory drug
  • Diclofenac non-steroidal anti-inflammatory drug
  • Indomethacin non-steroidal antiinflammatory drug
  • Sulfasalazine see e.g., Arthritis & Rheumatism (1996) Vol. 39, No. 9 (supplement), S281)
  • Azathioprine see e.g., Arthritis & Rheumatism (1996) Vol. 39, No.
  • ICE inhibitor inhibitor of the enzyme interleukin-l ⁇ converting enzyme
  • zap-70 and/or lck inhibitor inhibitor of the tyrosine kinase zap-70 or lck
  • VEGF inhibitor and/or VEGF-R inhibitor inhibitors of vascular endothelial cell growth factor or vascular endothelial cell growth factor receptor; inhibitors of angiogenesis
  • corticosteroid anti-inflammatory drugs e.g., SB203580
  • TNF-convertase inhibitors anti-IL-12 antibodies; anti-IL-18 antibodies; interleukin-11 (see e.g., Arthritis & Rheumatism (1996) Vol. 39, No.
  • the IL-12 antibody, or antigen-binding portion thereof is administered in combination with one of the following agents for the treatment of rheumatoid arthritis: small molecule inhibitor of KDR (ABT-123), small molecule inhibitor of Tie-2; methotrexate; prednisone; celecoxib; folic acid; hydroxychloroquine sulfate; rofecoxib; etanercept; infliximab; leflunomide; naproxen; valdecoxib; sulfasalazine; methylprednisolone; ibuprofen; meloxicam; methylprednisolone acetate; gold sodium thiomalate; aspirin; azathioprine; triamcinolone acetonide; propxyphene napsylate/apap; folate; nabumetone; diclofenac; piroxicam; etodolac; diclo
  • Non-limiting examples of therapeutic agents for inflammatory bowel disease with which an antibody, or antibody portion, of the invention can be combined include the following: budenoside; epidermal growth factor; corticosteroids; cyclosporin, sulfasalazine; aminosalicylates; 6-mercaptopurine; azathioprine; metronidazole; lipoxygenase inhibitors; mesalamine; olsalazine; balsalazide; antioxidants; thromboxane inhibitors; IL-I receptor antagonists; anti-IL-l ⁇ monoclonal antibodies; anti-IL-6 monoclonal antibodies; growth factors; elastase inhibitors; pyridinyl-imidazole compounds; antibodies to or antagonists of other human cytokines or growth factors, for example, TNF, LT, IL-I, IL-2, IL-6, EL-7, IL-8, IL-15, DL-16, TL- 17, IL-18, E
  • Antibodies of the invention, or antigen binding portions thereof can be combined with antibodies to cell surface molecules such as CD2, CD3, CD4, CD8, CD25, CD28, CD30, CD40, CD45, CD69, CD90 or their ligands.
  • the antibodies of the invention, or antigen binding portions thereof may also be combined with agents, such as methotrexate, cyclosporin, FK506, rapamycin, mycophenolate mofetil, leflunomide, NSABDs, for example, ibuprofen, corticosteroids such as prednisolone, phosphodiesterase inhibitors, adenosine agonists, antithrombotic agents, complement inhibitors, adrenergic agents, agents which interfere with signalling by proinflammatory cytokines such as TNF ⁇ or IL-I (e.g.
  • IL-l ⁇ converting enzyme inhibitors such as kinase inhibitors, metalloproteinase inhibitors, sulfasalazine, azathioprine, 6-mercaptopurmes, angiotensin converting enzyme inhibitors, soluble cytokine receptors and derivatives thereof (e.g. soluble p55 or p75 TNF receptors, sIL-lRI, sIL-lRIL sIL-6R) and antiinflammatory cytokines (e.g. IL-4, IL-10, IL-Il, IL- 13 and TGF ⁇ ).
  • TNF ⁇ converting enzyme inhibitors such as kinase inhibitors, metalloproteinase inhibitors, sulfasalazine, azathioprine, 6-mercaptopurmes, angiotensin converting enzyme inhibitors, soluble cytokine receptors and derivatives thereof (e.g. soluble p55 or p75 TNF receptors, sIL-l
  • TNF antagonists for example, anti-TNF antibodies, D2E7 (PCT Publication No. WO 97/29131; HUMIRA), CA2 (REMICADE), CDP 571, TNFR-Ig constructs, (p75TNFRIgG (ENBREL) and p55TNFRIgG (LENERCEPT)) inhibitors and PDE4 inhibitors.
  • Antibodies of the invention, or antigen binding portions thereof, can be combined with corticosteroids, for example, budenoside and dexamethasone.
  • Antibodies of the invention or antigen binding portions thereof may also be combined with agents such as sulfasalazine, 5-aminosalicylic acid and olsalazine, and agents which interfere with synthesis or action of proinflammatory cytokines such as IL-I, for example, IL-l ⁇ converting enzyme inhibitors and IL-lra.
  • agents such as sulfasalazine, 5-aminosalicylic acid and olsalazine
  • agents which interfere with synthesis or action of proinflammatory cytokines such as IL-I, for example, IL-l ⁇ converting enzyme inhibitors and IL-lra.
  • Antibodies of the invention or antigen binding portion thereof may also be used with T cell signaling inhibitors, for example, tyrosine kinase inhibitors 6-mercaptopurines.
  • Antibodies of the invention, or antigen binding portions thereof can be combined with IL-Il.
  • Antibodies of the invention, or antigen binding portions thereof, can be combined with mesalamine, prednisone, azathioprine, mercaptopurine, infliximab, methylprednisolone sodium succinate, diphenoxylate/atrop sulfate, loperamide hydrochloride, methotrexate, omeprazole, folate, ciprofloxacin/dextrose-water, hydrocodone bitartrate/apap, tetracycline hydrochloride, fluocinonide, metronidazole, thimerosal/boric acid, cholestyramine/sucrose, ciprofloxacin hydrochloride, hyoscyamine sulfate, meperidine hydrochloride, midazolam hydrochloride, oxycodone hcl/acetaminophen, promethazine hydrochloride, sodium phosphate, sulfamethoxazole
  • Non-limiting examples of therapeutic agents for multiple sclerosis with which an antibody, or antibody portion, of the invention can be combined include the following: corticosteroids; prednisolone; methylprednisolone; azathioprine; cyclophosphamide; cyclosporine; methotrexate; 4-ammopyridine; tizanidine; interferon- ⁇ la (AVONEX; Biogen); interferon- ⁇ lb (BETASERON; Chiron/Berlex); interferon ⁇ -n3) (Interferon Sciences/Fujimoto), interferon- ⁇ (Alfa Wassermann/J&J), interferon ⁇ IA-IF (Serono/Inhale Therapeutics), Peginterferon ⁇ 2b (Enzon/Schering-Plough), Copolymer 1 (Cop-1; COPAXONE; Teva Pharmaceutical Industries, Inc.); hyperbaric oxygen; intravenous immunoglobulin; clabribine; antibodies to or antagonists
  • Antibodies of the invention, or antigen binding portions thereof, can be combined with antibodies to cell surface molecules such as CD2, CD3, CD4, CD8, CD19, CD20, CD25, CD28, CD30, CD40, CD45, CD69, CD80, CD86, CD90 or their ligands.
  • cell surface molecules such as CD2, CD3, CD4, CD8, CD19, CD20, CD25, CD28, CD30, CD40, CD45, CD69, CD80, CD86, CD90 or their ligands.
  • the antibodies of the invention, or antigen binding portions thereof, may also be combined with agents, such as methotrexate, cyclosporine, FK506, rapamycin, mycophenolate mofetil, leflunomide, NSAIDs, for example, ibuprofen, corticosteroids such as prednisolone, phosphodiesterase inhibitors, adensosine agonists, antithrombotic agents, complement inhibitors, adrenergic agents, agents which interfere with signalling by proinflammatory cytokines such as TNF ⁇ or IL-I (e.g.
  • agents such as methotrexate, cyclosporine, FK506, rapamycin, mycophenolate mofetil, leflunomide, NSAIDs, for example, ibuprofen, corticosteroids such as prednisolone, phosphodiesterase inhibitors, adensosine agonists, antithrombotic agents, complement inhibitors, adrenergic agents
  • IL-4, IL-10, IL-13 and TGF ⁇ IL-4, IL-10, IL-13 and TGF ⁇ .
  • interferon- ⁇ for example, IFN ⁇ la and IFN ⁇ lb
  • Copaxone corticosteroids
  • caspase inhibitors for example inhibitors of caspase-1, IL-I inhibitors, TNF inhibitors, and antibodies to CD40 ligand and CD80.
  • the antibodies of the invention, or antigen binding portions thereof, may also be combined with agents, such as alemtuzumab, dronabinol, Unimed, daclizumab, mitoxantrone, xaliproden hydrochloride, fampridine, glatiramer acetate, natalizumab, sinnabidol, a-immunokine NNSO3, ABR-215062, AnergiX.MS, chemokine receptor antagonists, BBR-2778, calagualine, CPI-1189, LEM (liposome encapsulated mitoxantrone), THCCBD (cannabinoid agonist) MBP- 8298, mesopram (PDE4 inhibitor), MNA-715, anti-IL-6 receptor antibody, neurovax, pirfenidone allotrap 1258 (RDP-1258), sTNF-Rl, talampanel, teriflunomide,TGF-beta2, tiplimo
  • Non-limiting examples of therapeutic agents for Angina with which an antibody, or antibody portion, of the invention can be combined include the following: aspirin, nitroglycerin, isosorbide mononitrate, metoprolol succinate, atenolol, metoprolol tartrate, amlodipine besylate, diltiazem hydrochloride, isosorbide dinitrate, clopidogrel bisulfate, nifedipine, atorvastatin calcium, potassium chloride, furosemide, simvastatin, verapamil hcl, digoxin, propranolol hydrochloride, carvedilol, lisinopril, spironolactone, hydrochlorothiazide, enalapril maleate, nadolol, ramipril, enoxaparin sodium, heparin sodium, valsartan, sotalol hydrochloride, fenofib
  • Non-limiting examples of therapeutic agents for Ankylosing Spondylitis with which an antibody, or antibody portion, of the invention can be combined include the following: ibuprofen, diclofenac and misoprostol, naproxen, meloxicam, indomethacin, diclofenac, celecoxib, rofecoxib, Sulfasalazine, Methotrexate, azathioprine, minocyclin, prednisone, etanercept, infliximab.
  • Non-limiting examples of therapeutic agents for Asthma with which an antibody, or antibody portion, of the invention can be combined include the following: albuterol, salmeterol/fluticasone, montelukast sodium, fluticasone propionate, budesonide, prednisone, salmeterol xinafoate, levalbuterol hcl, albuterol sulfate/ipratropium, prednisolone sodium phosphate, triamcinolone acetonide, beclomethasone dipropionate, ipratropium bromide, azithromycin, pirbuterol acetate, prednisolone, theophylline anhydrous, methylprednisolone sodium succinate, clarithromycin, zafirlukast, formoterol fumarate, influenza virus vaccine, methylprednisolone, amoxicillin trihydrate, flunisolide, allergy injection, cromolyn sodium, fexofen
  • Non-limiting examples of therapeutic agents for COPD with which an antibody, or antibody portion, of the invention can be combined include the following: albuterol sulfate/ipratropium, ipratropium bromide, salmeterol/fluticasone, albuterol, salmeterol xinafoate, fluticasone propionate, prednisone, theophylline anhydrous, methylprednisolone sodium succinate, montelukast sodium, budesonide, formoterol fumarate, triamcinolone acetonide, levofloxacin, guaifenesin, azithromycin, beclomethasone dipropionate, levalbuterol hcl, flunisolide, ceftriaxone sodium, amoxicillin trihydrate, gatifloxacin, zafirlukast, amoxicillin/clavulanate, flunisolide/menthol, chlorpheniramine/hydrocodone,
  • Non-limiting examples of therapeutic agents for HCV with which an antibody, or antibody portion, of the invention can be combined include the following: Interferon-alpha-2a, Interferon-alpha-2b, Lnterferon-alpha conl, Interferon-alpha-nl, Pegylated interferon-alpha-2a, Pegylated interferon-alpha-2b, ribavirin, Peginterferon alfa-2b + ribavirin, Ursodeoxycholic Acid, Glycyrrhizic Acid, Thymalfasin, Maxamine, VX-497 and any compounds that are used to treat HCV through intervention with the following targets:HCV polymerase, HCV protease, HCV helicase, HCV IRES (internal ribosome entry site).
  • Non-limiting examples of therapeutic agents for Idiopathic Pulmonary Fibrosis with which an antibody, or antibody portion, of the invention can be combined include the following: prednisone, azathioprine, albuterol, colchicine, albuterol sulfate, digoxin, gamma interferon, methylprednisolone sod succ, lorazepam, furosemide, lisinopril, nitroglycerin, spironolactone, cyclophosphamide, ipratropium bromide, actinomycin d, alteplase, fluticasone propionate, levofloxacin, metaproterenol sulfate, morphine sulfate, oxycodone hcl, potassium chloride, triamcinolone acetonide, tacrolimus anhydrous, calcium, interferon-alpha, methotrexate, mycophenolate mofetil, Interferon-gam
  • Non-limiting examples of therapeutic agents for Myocardial Infarction with which an antibody, or antibody portion, of the invention can be combined include the following: aspirin, nitroglycerin, metoprolol tartrate, enoxaparin sodium, heparin sodium, clopidogrel bisulfate, carvedilol, atenolol, morphine sulfate, metoprolol succinate, warfarin sodium, lisinopril, isosorbide mononitrate, digoxin, furosemide, simvastatin, ramipril, tenecteplase, enalapril maleate, torsemide, retavase, losartan potassium, quinapril hcl/mag carb, bumetanide, alteplase, enalaprilat, amiodarone hydrochloride, tirofiban hcl m-hydrate, diltiazem hydrochloride,
  • Non-limiting examples of therapeutic agents for Psoriasis with which an antibody, or antibody portion, of the invention can be combined include the following: small molecule inhibitor of KDR (ABT-123), small molecule inhibitor of Tie-2, calcipotriene, clobetasol propionate, triamcinolone acetonide, halobetasol propionate, tazarotene, methotrexate, fluocinonide, betamethasone diprop augmented, fluocinolone acetonide, acitretin, tar shampoo, betamethasone valerate, mometasone furoate, ketoconazole, pramoxine/fluocinolone, hydrocortisone valerate, flurandrenolide, urea, betamethasone, clobetasol propionate/emoll, fluticasone propionate, azithromycin, hydrocortisone, moisturizing formula, folic acid, desonide, pimecrolimus
  • Non-limiting examples of therapeutic agents for Psoriatic Arthritis with which an antibody, or antibody portion, of the invention can be combined include the following: methotrexate, etanercept, rofecoxib, celecoxib, folic acid, sulfasalazine, naproxen, leflunomide, methylprednisolone acetate, indomethacin, hydroxychloroquine sulfate, prednisone, sulindac, betamethasone diprop augmented, infliximab, methotrexate, folate, triamcinolone acetonide, diclofenac, dimethylsulfoxide, piroxicam, diclofenac sodium, ketoprofen, meloxicam, methylprednisolone, nabumetone, tolmetin sodium, calcipotriene, cyclosporine, diclofenac sodium/misoprostol, fluocinon
  • Non-limiting examples of therapeutic agents for Restenosis with which an antibody, or antibody portion, of the invention can be combined include the following: sirolimus, paclitaxel, everolimus, tacrolimus, ABT-578, acetaminophen.
  • Non-limiting examples of therapeutic agents for Sciatica with which an antibody, or antibody portion, of the invention can be combined include the following: hydrocodone bitartrate/apap, rofecoxib, cyclobenzaprine hcl, methylprednisolone, naproxen, ibuprofen, oxycodone hcl/acetaminophen, celecoxib, valdecoxib, methylprednisolone acetate, prednisone, codeine phosphate/apap, tramadol hcl/acetaminophen, metaxalone, meloxicam, methocarbamol, lidocaine hydrochloride, diclofenac sodium, gabapentin, dexamethasone, carisoprodol, ketorolac tromethamine, indomethacin, acetaminophen, diazepam, nabumetone, oxycodone hcl,
  • Preferred examples of therapeutic agents for SLE (Lupus) in which an antibody or an antigen binding portion can be combined include the following: NSAIDS, for example, diclofenac, naproxen, ibuprofen, piroxicam, indomethacin; COX2 inhibitors, for example, Celecoxib, rofecoxib, valdecoxib; anti-malarials, for example, hydroxychloroquine; Steroids, for example, prednisone, prednisolone, budenoside, dexamethasone; Cytotoxics, for example, azathioprine, cyclophosphamide, mycophenolate mofetil, methotrexate; inhibitors of PDE4 or purine synthesis inhibitor, for example Cellcept.
  • NSAIDS for example, diclofenac, naproxen, ibuprofen, piroxicam, indomethacin
  • COX2 inhibitors for example, Celecoxib,
  • Antibodies of the invention or antigen binding portions thereof may also be combined with agents such as sulfasalazine, 5-aminosalicylic acid, olsalazine, Imuran and agents which interfere with synthesis, production or action of proinflammatory cytokines such as IL-I, for example, caspase inhibitors like IL-l ⁇ converting enzyme inhibitors and IL- Ira.
  • Antibodies of the invention or antigen binding portion thereof may also be used with T cell signaling inhibitors, for example, tyrosine kinase inhibitors; or molecules that target T cell activation molecules, for example, CTLA-4-IgG or anti-B7 family antibodies, anti-PD-1 family antibodies.
  • Antibodies of the invention, or antigen binding portions thereof can be combined with IL-Il or anti-cytokine antibodies, for example, fonotolizumab (anti-IFNg antibody), or anti-receptor receptor antibodies, for example, anti-IL-6 receptor antibody and antibodies to B-cell surface molecules.
  • Antibodies of the invention or antigen binding portion thereof may also be used with LJP 394 (abetimus), agents that deplete or inactivate B-cells, for example, Rituximab (anti-CD20 antibody), lymphostat-B (anti-BlyS antibody), TNF antagonists, for example, anti-TNF antibodies, D2E7 (PCT Publication No. WO 97/29131; HUMIRA), CA2 (REMICADE), CDP 571, TNFR-Ig constructs, (p75TNFRIgG (ENBREI ⁇ and p55TNFRIgG (LENERCEPT)).
  • compositions of the invention may include a "therapeutically effective amount” or a “prophylactically effective amount” of an antibody or antibody portion of the invention.
  • a “therapeutically effective amount” refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic result.
  • a therapeutically effective amount of the antibody or antibody portion may be determined by a person skilled in the art and may vary according to factors such as the disease state, age, sex, and weight of the individual, and the ability of the antibody or antibody portion to elicit a desired response in the individual.
  • a therapeutically effective amount is also one in which any toxic or detrimental effects of the antibody, or antibody portion, are outweighed by the therapeutically beneficial effects.
  • prophylactically effective amount refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired prophylactic result. Typically, since a prophylactic dose is used in subjects prior to or at an earlier stage of disease, the prophylactically effective amount will be less than the therapeutically effective amount.
  • Dosage regimens may be adjusted to provide the optimum desired response (e.g., a therapeutic or prophylactic response). For example, a single bolus may be administered, several divided doses may be administered over time or the dose may be proportionally reduced or increased as indicated by the exigencies of the therapeutic situation. It is especially advantageous to formulate parenteral compositions in dosage unit form for ease of administration and uniformity of dosage.
  • Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the mammalian subjects to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier.
  • An exemplary, non-limiting range for a therapeutically or prophylactically effective amount of an antibody or antibody portion of the invention is 0.1-20 mg/kg, more preferably 1-10 mg/kg. It is to be noted that dosage values may vary with the type and severity of the condition to be alleviated. It is to be further understood that for any particular subject, specific dosage regimens should be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the compositions, and that dosage ranges set forth herein are exemplary only and are not intended to limit the scope or practice of the claimed composition.
  • Example 1 Generation and isolation of anti human IL-12 monoclonal antibodies
  • Example 1.1 Assays to identify anti human IL-12 antibodies
  • Example 1 the following assays were used to identify and characterize anti human IL-12 antibodies unless otherwise stated.
  • Example 1.1 A: ELISA
  • Enzyme Linked Immunosorbent Assays to screen for antibodies that bind human BL-12 were performed as follows.
  • Example 1.1 A.I: ELISA to detect binding of anti human IL-12 antibodies to IL-12 p70
  • ELISA plates (Corning Costar, Acton, MA) were coated with 50 ⁇ L/well of 5 ⁇ g/ml goat anti-mouse IgG Fc specific (Pierce # 31170, Rockford, IL.) in Phosphate Buffered Saline (PBS) overnight at 4 degrees Celsius. Plates were washed once with PBS containing 0.05% Tween-20. Plates were blocked by addition of 200 ⁇ L/well blocking solution diluted to 2% in PBS (BioRad #170-6404, Hercules, CA.) for 1 hour at room temperature. Plates were washed once after blocking with PBS containing 0.05% Tween-20.
  • PBS Phosphate Buffered Saline
  • Streptavidin HRP (Pierce # 21126, Rockland, IL.) was diluted 1:20000 in PBS containing 0.1% BSA; 50 ⁇ L/well was added and the plates incubated for 1 hour at room temperature. Plates were washed 3 times with PBS containing 0.05% Tween-20. Fifty microliters of TMB solution (Sigma # T0440, St. Louis, MO.) was added to each well and incubated for 10 minutes at room temperature. The reaction was stopped by addition of IN sulphuric acid. Plates were read spectrophotmetrically at a wavelength of 450 nm.
  • Example 1.1.A.2 ELISA to assess ability of IL-12 p70 or IL-12 p40 to compete with binding of anti human IL-12 antibodies to IL-12 p70
  • ELISA plates (Corning Costar, Acton, MA) were coated with 50 ⁇ L/well of 5 ⁇ g/rnl goat anti-mouse IgG Fc specific (Pierce # 31170, Rockford, IL.) in PBS overnight at 4 degrees Celsius. Plates were washed once with PBS+ 0.05% Tween-20. Plates were blocked by addition of PBS + 10% powdered milk for 1 hour at room temperature. Plates were washed three times after blocking with PBS+ 0.05% Tween-20.
  • Biotinylated recombinant purified human IL-12 p70 was prepared as a three times concentrated (3x) stock at O.l ⁇ g/ml in PBS containing 0.1% BSA. Recombinant purified human IL-12 p70 was prepared at various concentrations ranging from 0.1 to lO ⁇ g/ml in PBS containing 0.1% BSA.
  • Equal volumes (75 ⁇ L) of each of the following solutions were mixed: diluted mouse sera or hybridoma supernatant, biotinylated recombinant purified human IL-12 p70, and recombinant purified human IL-12 p70. Fifty microliters of this mixture was added to the coated ELISA plates described above and were incubated for 1 hour at room temperature. Wells were washed three times with PBS containing 0.05% Tween-20. Streptavidin HRP (Pierce # 21126, Rockland, IL.) was diluted 1:20000 in PBS containing 0.1% BSA; 50 ⁇ L/well was added and the plates incubated for 1 hour at room temperature.
  • Biotinylated recombinant purified human IL-12 p70 was prepared as a three times concentrated (3x) stock at O.l ⁇ g/ml in PBS containing 0.1% BSA.
  • Recombinant purified human IL-12 p40 was prepared at various concentrations ranging from 0.1 to lO ⁇ g/ml in PBS containing 0.1% BSA.
  • Equal volumes (75 ⁇ L) of each of the following solutions were mixed: diluted mouse sera or hybridoma supernatant, biotinylated recombinant purified human EL- 12 p70, and recombinant purified human IL- 12 p40. Fifty microliters of this mixture was added to the coated ELISA plates and incubated for 1 hour at room temperature. Wells were washed three times with PBS containing 0.05% Tween-20. Streptavidin HRP (Pierce # 21126, Rockland, IL.) was diluted 1:20000 in PBS containing 0.1% BSA; 50 ⁇ L/well was added and the plates incubated for 1 hour at room temperature.
  • Example 1.1.B Affinity Determinations using BIACORE technology
  • the BIACORE assay (Biacore, Inc, Piscataway, NJ) determines the affinity of antibodies with kinetic measurements of on-, off-rate constants. Binding of antibodies to recombinant purified human IL-12 p70 or recombinant purified human IL-12 p40 were determined by surface plasmon resonance-based measurements with a Biacore® 3000 instrument (Biacore® AB, Uppsala, Sweden) using running HBS-EP (10 mM HEPES [pH 7.4], 150 mM NaCl, 3 mM EDTA, and 0.005% surfactant P20) at 25° C.
  • HBS-EP 10 mM HEPES [pH 7.4], 150 mM NaCl, 3 mM EDTA, and 0.005% surfactant P20
  • AU chemicals were obtained from Biacore® AB (Uppsala, Sweden) or otherwise from a different source as described in the text. Approximately 5000 RU of goat anti-mouse IgG, (Fc ⁇ ), fragment specific polyclonal antibody (Pierce Biotechnology Inc, Rockford, IL) diluted in 10 mM sodium acetate (pH 4.5) was directly immobilized across a CM5 research grade biosensor chip using a standard amine coupling kit according to manufacturer's instructions and procedures at 25 ⁇ g/ml. Unreacted moieties on the biosensor surface were blocked with ethanolamine. Modified carboxymethyl dextran surface in flowcell 2 and 4 was used as a reaction surface.
  • Unmodified carboxymethyl dextran without goat anti-mouse IgG in flow cell 1 and 3 was used as the reference surface.
  • rate equations derived from the 1:1 Langmuir binding model were fitted simultaneously to association and dissociation phases of all eight injections (using global fit analysis) with the use of Biaevaluation 4.0.1 software.
  • Purified antibodies were diluted in HEPES-buffered saline for capture across goat anti-mouse IgG specific reaction surfaces.
  • Mouse antibodies to be captured as a ligand 25 ⁇ g/ml were injected over reaction matrices at a flow rate of 5 ⁇ l/min.
  • the association and dissociation rate constants, Ic 0n (unit MT 1 S “1 ) and k off (unit s '1 ) were determined under a continuous flow rate of 25 ⁇ l/min. Rate constants were derived by making kinetic binding measurements at ten different antigen concentrations ranging from 10 - 200 nM.
  • Example l.l.C Functional Activity of anti human IL-12 antibodies
  • the antibodies were used in the following assays that measure the ability of an antibody to inhibit IL-12 activity.
  • Example l.l.C 1 Preparation of Human PHA-activated Lymphoblasts
  • PBMCs Human peripheral blood mononuclear cells
  • the PBMC were then activated to form lymphoblasts as described in Current Protocols in Immunology, Unit 6.16.
  • the washed PBMC were resuspended at 0.5-lxIO 6 cells/mL in RPMI complete medium (RPMI 1640 medium, 10% fetal bovine serum (FBS), 100 U/ml penicillin, 100 tg/ml streptomycin), supplemented with O.Olmg/mL PHA-P (Sigma #L8754, St. Louis, MO) and cultured for 4 days at 37 0 C in a 5% CO 2 atmosphere.
  • RPMI complete medium RPMI 1640 medium, 10% fetal bovine serum (FBS), 100 U/ml penicillin, 100 tg/ml streptomycin
  • O.Olmg/mL PHA-P Sigma #L8754, St. Louis, MO
  • cell cultures were then re- seeded at IxIO 6 cells/mL in culture media with 0.01mg/mL PHA-P and 50U/mL recombinant human IL-2 (R&D Systems #202-EL, Minneapolis, MN.). Cells were incubated at 37°C for 24 hours, washed with RPMI complete medium, then frozen in 95% FBS, 5% DMSO at IxIO 7 cells/ml.
  • Example l.l.C 2 PHA blast IFN- ⁇ induction assay: Inhibition of human IL-12 activity
  • anti-human IL-12 antibodies to inhibit the human IL-12 induced production of IFN- ⁇ by PHA blasts was analyzed as follows. Various concentrations of immunized mouse serum, murine hybridoma supernatant or purified anti-human IL-12 antibodies were preincubated for one hour at 37 degrees C with 400 pg/ml recombinant purified human IL-12 p70 in lOO ⁇ L RPMI complete medium in a microtiter plate (U-bottom, 96-well, Costar). PHA blasts isolated as described above, were washed once and resuspended in RPMI complete medium to a cell density of IXlO 7 cells/ml.
  • PHA blasts (lOO ⁇ L of IXlO 6 cells/ml) were added to the antibody plus recombinant purified human IL-12 p70 mixture (final IL-12 p70 concentration was 200 pg/ml) and incubated for 18 hours at 37 deg C. After incubation, 150 ⁇ L of cell-free supernatant was withdrawn from each well and the level of human IFN- ⁇ produced was measured using a human IFN- ⁇ ELISA (R&D Systems Cat#DIF50).
  • Example l.l.C 3 PHA blast IFN-Y induction assay: Inhibition of cynomolgus monkey (cyno) IL-12 activity
  • anti-human IL-12 antibodies to inhibit the cynomolgus monkey IL-12 induced production of IFN- ⁇ by PHA blasts was analyzed as follows. Various concentrations of immunized mouse serum, murine hybridoma supernatant or purified anti-human IL-12 antibodies were preincubated for one hour at 37 degrees C with 150 pg/mL recombinant purified cyno IL-12 p70 in lOO ⁇ L RPMI complete medium in a microliter plate (U-bottom, 96-well, Costar). PHA blasts isolated as described above, were washed once and resuspended in RPMI complete medium to a cell density of IXlO 7 cells/ml.
  • PHA blasts (lOO ⁇ L of IXlO 7 cells/mL) were added to the antibody plus recombinant purified cyno IL-12 ⁇ 70 mixture (final cyno IL-12 p70 concentration was 75 ⁇ g/ml) and incubated for 18 hours at 37 deg C. After incubation, 150 ⁇ L of cell-free supernatant was withdrawn from each well and the level of human IFN- ⁇ produced was measured using a human IFN- ⁇ ELISA (R&D Systems Cat#D]F50).
  • Example 1.2 Generation of Anti human IL-12 monoclonal antibodies
  • Anti human IL-12 mouse monoclonal antibodies were obtained as follows:
  • Example 1.2.A Immunization of mice with human IL-12 antigen
  • mice were injected intravenously with 10 ug recombinant purified human IL-12 p70 or 2ug recombinant purified human IL-12 p40 (R & D Systems, Minneapolis, MN).
  • Splenocytes obtained from the immunized mice described in Example 1.2. A were fused with SP2/O-Ag-14 cells at a ratio of 5: 1 according to the established method described in Kohler, G. and Milstein 1975, Nature, 256:495 to generate hybridomas. Fusion products were plated in selection media containing azaserine and hypoxanthine in 96-well plates at a density of 2.5xlO 6 spleen cells per well. Seven to ten days post fusion, macroscopic hybridoma colonies were observed. Supernatant from each well containing hybridoma colonies was tested by ELISA for the presence of antibody to IL-12 p70 (as described in Example l.l.A.l).
  • Table 8 Fusion and screening data following immunizations of mice with human IL-12
  • Example 1.2.C Identification and characterization of anti human IL-12p40 monoclonal antibodies
  • Hybridomas producing antibodies that bound IL-12 generated according to Examples 1.2.B and 1.2.C, and capable of binding IL-12 p40 specifically and particularly those with IC 50 values in the PHA blast assay of 12nM or less than 12nM were scaled up and cloned by limiting dilution.
  • Hybridoma cells were expanded into media containing 10% low IgG fetal bovine serum (Hyclone #SH30151, Logan, UT.). On average, 250 mL of each hybridoma supernatant (derived from a clonal population) was harvested, concentrated and purified by protein A affinity chromatography, as described in Harlow, E. and Lane, D. 1988 "Antibodies: A Laboratory Manual". The ability of purified mAbs to inhibit IL-12 activity was determined using the PHA blast assay as described in Examples l.l.C 2 and 1.1.C3. Table 9 shows IC 50 values from the PHA blast assays for ten monoclonal antibodies. Table 9: Neutralization of IL-12 by anti IL-12p40 Murine Monoclonal Antibodies
  • the binding affinities of the monoclonal antibodies to recombinant purified human IL-12 p70 were determined using surface plasmon resonance (Biacore®) measurement as described in Example l.l.B. Table 10 shows the affinity of the ten monoclonal antibodies described above for human IL-12 p70.
  • Example 1.2.C.1 Species Specificity of murine monoclonal anti-human IL-12p40 antibodies
  • a direct ELISA was set up by directly coating ELISA plates with 5 ug/ml of recombinant purified mouse IL-12 (Peprotech).
  • Murine-anti-human IL-12 p40 mAbs were prepared at various concentrations ranging from 3 to 200 ng/ml in PBS containing 0.1% BSA (Sigma, StLouis, MO). 50 ⁇ l of each antibody dilution was added to the coated ELISA plate and incubated for 1 hour at room temperature. Wells were washed 3 times with PBS containing 0.05% Tween-20.
  • Anti-mouse IgG-HRP antibody (R&D #HAF007, Minneapolis, MN) was diluted 1:2000 in PBS containing 0.1% BSA; 50 ul/well was added and the plates incubated for 1 hour at room temperature. Fifty microliters of TMB solution (Sigma # T0440, St. Louis, MO.) was added to each well and incubated for 10 minutes at room temperature. The reaction was stopped by addition of 2N sulphuric acid. Plates were read spectrophotmetrically at a wavelength of 450 nm.
  • an indirect ELISA was set up by coating ELISA plates with 5ug/ml of goat anti- mouse IgG, Fc fragment specific antibody (Pierce # 31170, Rockland, IL).
  • Murine anti-human IL-12 p40 mAbs were prepared at various concentrations ranging from 0.1 to 100 ng/ml in PBS containing 0.1% BSA; 50 ul of each antibody dilution was added to the coated ELISA plate and incubated for 1 hour at room temperature. Wells were washed 3 times with PBS containing 0.05% Tween-20.
  • Recombinant purified mouse IL-12 (Preprotech) was diluted at 0.2 ug/ml in PBS containing 0.1% BSA; 50 ul/well was added and the plates incubated for 1 hour at room temperature. Wells were washed 3 times with PBS containing 0.05% Tween-20.
  • Biotinylated anti-mouse IL-12 antibody (R&D # BAF419) was diluted at 0.2 ug/ml in PBS containing 0.1% BSA; 50 ul/well was added and the plates incubated for 1 hour at room temperature. Wells were washed 3 times with PBS containing 0.05% Tween-20.
  • Streptavidin HRP (Pierce # 21126, Rockland, IL.) was diluted 1:20000 in PBS containing 0.1% BSA; 50 ⁇ L/well was added and the plates incubated for 1 hour at room temperature. Plates were washed 3 times with PBScontaining 0.05% Tween-20. Fifty microliters of TMB solution was added to each well and incubated for 10 minutes at room temperature. The reaction was stopped by addition of 2N sulphuric acid. Plates were read spectrophotmetrically at a wavelength of 450 nm. Results from the direct and indirect ELISAs performed with the ten monoclonal antibodies are shown in Table 11. Table 11: Binding of anti IL-12 Murine Monoclonal Antibodies to mouse IL-12 p40
  • Example 1.2.D Determination of the amino acid sequence of the variable region for each murine anti-human IL-12 p40 mAb
  • RNA was subjected to first strand DNA synthesis using the Superscript First-Strand Synthesis System (hivitrogen, Carlsbad, CA) per the manufacturers instructions. Oligo(dT) was used to prime first-strand synthesis to select for poly(A) + RNA. The first-strand cDNA product was then amplified by PCR with primers designed for amplification of murine immunoglobulin variable regions (Ig-Primer Sets, Novagen, Madison, WI).
  • Trizol Gibco BRL/Invitrogen, Carlsbad, CA.
  • Oligo(dT) was used to prime first-strand synthesis to select for poly(A) + RNA.
  • the first-strand cDNA product was then amplified by PCR with primers designed for amplification of murine immunoglobulin variable regions (Ig-Primer Sets, Novagen, Madison, WI).
  • PCR products were resolved on an agarose gel, excised, purified, and then subcloned with the TOPO Cloning kit into pCR2.1-TOPO vector (hwitrogen, Carlsbad, CA) and transformed into TOPlO chemically competent E. coli (Invitrogen, Carlsbad, CA). Colony PCR was performed on the transformants to identify clones containing insert. Plasmid DNA was isolated from clones containing insert using a QIAprep Miniprep kit (Qiagen, Valencia, CA). Inserts in the plasmids were sequenced on both strands to determine the variable heavy or variable light chain DNA sequences using M13 forward and M13 reverse primers ( Fermentas Life Sciences, Hanover MD). Variable heavy and variable light chain sequences of the ten monoclonal antibodies described in Example 1.2.C are described in Table 1.
  • Example 2 Recombinant anti human IL-12p40 antibodies
  • Example 2.1 Construction and expression of recombinant chimeric anti human IL-12p40 antibodies
  • the DNA encoding the heavy chain constant region of murine anti-human IL ⁇ 12p40 monoclonal antibodies 3G7, 8El, 1A6, and 1D4 was replaced by a cDNA fragment encoding the human IgGl constant region containing 2 hinge-region amino acid mutations by homologous recombination in bacteria. These mutations are a leucine to alanine change at position 234 (EU numbering) and a leucine to alanine change at position 235 (Lund et al., 1991, J. Immunol., 147:2657).
  • the light chain constant region of each of these antibodies was replaced by a human kappa constant region.
  • chimeric antibodies were transiently expressed in COS cells by co-transfection of chimeric heavy and light chain cDNAs ligated into the pBOS expression plasmid (Mizushima and Nagata, Nucleic Acids Research 1990, VoI 18, pg 5322) comprising a heavy chain signal sequence MEFGLSWLFLVAILKGVQC (SEQ ID NO. 110), and a light chain signal sequence MDMRVPAQLLGLLLLWFPGSRC ((SEQ ID NO. 111).
  • Cell supernatants containing recombinant chimeric antibody were purified by Protein A Sepharose chromatography and bound antibody was eluted by addition of acid buffer. Antibodies were neutralized and dialyzed into PBS.
  • the heavy chain cDNA encoding chimeric 3G7 was co-transfected with the 1D4 chimeric light chain cDNA (both ligated in the pBOS vector) into COS cells.
  • Cell supernatant containing recombinant chimeric antibody was purified by Protein A Sepharose chromatography and bound antibody was eluted by addition of acid buffer. Antibodies were neutralized and dialyzed into PBS.
  • the purified chimeric anti-human IL-12 monoclonal antibodies were then tested for their ability to inhibit the IL-12 induced production of IFN- ⁇ by PHA blasts as described in Examples l.l.C 2 and 1.1.C3.
  • Table 12 shows IC 50 values from the PHA blast assays for five chimeric antibodies.
  • Example 2.2 Construction and expression of CDR grafted anti human IL-12p40 antibodies
  • CDR-grafted anti-human IL- 12 antibodies were generated as follows.
  • Each murine variable heavy and variable light chain gene sequence (as described in Table 3) was separately aligned against 44 human immunoglobulin germline variable heavy chain or 46 germline variable light chain sequences (derived from NCBI Ig Blast website at http://www.ncbi.nlm.nih.gov/igblast/retrieveig.html. ' ) using Vector NTI software.
  • Human variable domain sequences having the highest overall homology to the original murine sequences (as well as the highest homology at positions known to be important for antigen binding) (Welschof, M. and Krauss, J.
  • VoI 207 were selected for each heavy chain and light chain sequence to provide the framework (FW) 1, 2 and 3 sequences for CDR-grafting purposes.
  • Identification of a suitable human variable heavy and light chain FW4 region was accomplished by separately aligning each murine heavy chain and light chain FW4 region with 6 human immunoglobulin germline joining heavy chain and 5 germline joining light chain sequences in the NCBI database.
  • In silico construction of complete CDR grafted antibodies was accomplished by substitution of human variable domain CDR sequences (derived from the NCBI website) with murine CDR sequences (derived from the hybridomas) with addition of a FW4 region (derived from the NCBI website) to each 3' end.
  • oligonucleotides were constructed de novo using oligonucleotides.
  • 6 oligonucleotides of 60-80 nucleotides each were designed to overlap each other by 20 nucleotides at the 5' and/or 3' end of each oligonucleotide.
  • all 6 oligos were combined, boiled, and annealed in the presence of dNTPs.
  • DNA polymerase I, Large (Klenow) fragment was added to fill-in the approximately 40bp gaps between the overlapping oligonucleotides.
  • PCR was then performed to amplify the entire variable region gene using two outermost primers containing overhanging sequences complementary to the multiple cloning site in a modified pBOS vector (Mizushima, S. and Nagata, S., (1990) Nucleic acids Research VoI 18, No. 17)).
  • variable heavy region was inserted in-frame onto a cDNA fragment encoding the human IgGl constant region containing 2 hinge-region amino acid mutations (SEQ ID NO. 3) by homologous recombination in bacteria. These mutations are a leucine to alanine change at position 234 (EU numbering) and a leucine to alanine change at position 235 (Lund et al., 1991, J. Immunol., 147:2657).
  • the variable light chain region was inserted in-frame with the human kappa constant region (SEQ ID NO.
  • Cell supernatants containing recombinant chimeric antibody were purified by Protein A Sepharose chromatography and bound antibody was eluted by addition of acid buffer. Antibodies were neutralized and dialyzed into PBS. Nine CDR grafted antibodies are described in Table 5.
  • the ability of purified CDR grafted antibodies to inhibit IL-12 activity was determined using the PHA blast assay as described in Examples l.l.C 2 and 1.1.C3.
  • the binding affinities of the purified CDR grafted antibodies to recombinant purified human IL-12p70 were determined using surface plasmon resonance (Biacore®) measurement as described in Example 1.1.
  • Biacore® surface plasmon resonance
  • Table 13 shows shows IC 50 values from the PHA blast assays and the affinity of the nine CDR grafted antibodies described in Table 7 for human EL-12p70 and cynomolgous IL-12p70.
  • Table 13 Neutralization of IL-12 by anti IL-12p40 CDR grafted Antibodies and Affinity of anti IL-12p40 CDR grafted Antibodies for human and cynomolgous IL-12p70 and
  • Homology modeling is a computational method whereby approximate three dimensional coordinates are generated for a protein.
  • the source of initial coordinates and guidance for their further refinement is a second protein, the reference protein, for which the three dimensional coordinates are known and the sequence of which is related to the sequence of the first protein.
  • the relationship among the sequences of the two proteins is used to generate a correspondence between the reference protein and the protein for which coordinates are desired, the target protein.
  • the primary sequences of the reference and target proteins are aligned with coordinates of identical portions of the two proteins transferred directly from the reference protein to the target protein. Coordinates for mismatched portions of the two proteins, e.g.
  • the reference structures chosen for 1A6 and 1D4 was the PDB entry IJRH.
  • the heavy chain reference structure was PDB entry 1FL3 and the light chain reference was IMEX. Sequence identity of 25% between the reference and target amino acid sequences is considered the minimum necessary to attempt a homology modeling exercise.
  • the primary sequences of the murine and human framework regions of the selected antibodies share significant identity. Residue positions that differ are candidates for inclusion of the murine residue in the humanized sequence in order to retain the observed binding potency of the murine antibody. A list of framework residues that differ between the human and murine sequences was constructed manually.
  • hybrid antibodies were constructed by pairing a CDR-grafted H or L chain with an appropriate chimeric murine H or L chain followed by co-transfection into COS cells.
  • Table 14 shows the VH and VL amino acid sequences of the hybrid antibodies 1A6.3, 1A6.4, 1A6.7, 1A6.8, 1D4.4, and 1D4.5.
  • Hybrid antibodies were purified by protein A affinity chromatography (Example 1.2.C) and tested for potency in the PHA blast assay as in Examples 1.1.C2 and 1.1.C3. Kinetic measurements were determined using BIAcore as in Example 1.1. B. Table 15 shows the K D and IC 50 values of the hybrid antibodies.
  • the potency and affinity data derived with the hybrid mAbs was compared to data generated with the appropriate CDR-grafted mAbs (Example 2.3.1) to identify changes in potency and affinity attributed to a particular VH or VL chain. Whether or not, a humanized VH or VL chain was the optimum chain, was assessed using the methods described in Example 2.3.1. In some cases residues that did not fall within 4.5 A 0 of any CDR atom were additionally targeted for back mutations.
  • Table 15 Neutralization of IL-12 by anti-IL-12p40 hybrid antibodies and affinity of anti- IL-12p40 hybrid antibodies
  • 1A6.1 light chain VKB3 variant 1 (Ll-D- ⁇ S, L2-I->V, L36-Y-3-F, L67-S ⁇ Y), 1D4.1 VKB3 variant 2 (Ll-D ⁇ S, L36-Y->F, L67-S ⁇ Y), 1D4.1 VKB3 variant 3 (Ll-D ⁇ S, L67-S->Y), 1D4.1 VKB3 variant 4 (L2-I ⁇ V, L67-S ⁇ Y), and 1D4.1 VKB3 variant 5 (L67-S ⁇ Y).
  • 1D4.1 heavy chain VH2-70 variant 1 (Hl-E ⁇ Q, H93-A ⁇ T)
  • 1D4.1 VH2-70 variant 2 (H93-A->T).
  • PBOS expression vectors (see Example 2.1 and 2.2.2) harboring heavy and light chains containing framework back mutations were co-transfected into COS cells to transiently produce full-length humanized antibodies.
  • the amino acid sequences of the VH and VL regions of the humanized antibodies are disclosed in Table 16.
  • amino acid positions back mutated in the VH and VL of the CDR-grafted antibodies are listed in Table 17.
  • Example I.2.C Cell supernatants containing humanized antibodies were purified as described in Example I.2.C.
  • the ability of the purified humanized antibodies to neutralize IL-12 in vitro was determined using the PHA blast assay as described in Examples 1.1. C2 and 1.1. C3.
  • the binding affinities of the purified antibodies to recombinant purified human IL-12 ⁇ 70 were determined using BIAcore as described in Example 1.1.
  • Table 18 shows the IC 50 values from the PHA blast assay and the KpS from BIAcore.
  • Table 18 Neutralization of IL-12 by anti-IL-12p40 humanized antibodies and affinity of anti-IL-12p40 humanized antibodies
  • Humanization of the variable regions of the murine monoclonal antibodies 8El and 1A6 were carried out essentially according to the procedure of Queen, C, et al., Proc. Natl. Acad. Sci. USA 86: 10029-10033 (1989). First, human V segments with high homology to the murine monoclonal antibody VH or VL amino acid sequences were identified. Next, the complementarity-determining region (CDR) sequences together with framework amino acids important for maintaining the structures of the CDRs were grafted into the selected human framework sequences, hi addition, human framework amino acids that were found to be rare in the corresponding V region subgroup were substituted with consensus amino acids to reduce potential immunogenicity. The resulting humanized monoclonal antibodies were expressed in cells, purified and characterized as described below.
  • Humanized monoclonal antibodies 8E1.4, 8E1.5, 8E1.6, 1A6.10, 1A6.11, and 1A6.12 were generated as follows.
  • Example 2.4.1 Generation of Humanized monoclonal antibodies 8E1.4, 8E1.5, 8E1.6
  • human V region frameworks used as acceptors for the CDRs of 8El were chosen based on sequence homology.
  • ABMOD and ENCAD Levitt, M., J. MoI. Biol. 168: 595-620 (1983)
  • VH segment HA3D1 Olet, T., et al., J. Exp. Med.
  • Example 2.4.2 Construction of 8E1.4, 8E1.5 and 8E1.6 antibodies.
  • the heavy and light chain variable region genes were constructed and amplified using approximately 30 overlapping synthetic oligonucleotides ranging in length from approximately 20 to 40 bases following a published method (Rouillard, J.-M., et al, Nucleic Acids Res. 32: W176-W180 (2004)).
  • the oligonucleotides were annealed and assembled with the Expand High Fidelity PCR System (Roche Diagnostics Corporation, Indianapolis, IN), yielding a full-length product.
  • the resulting product was amplified by the polymerase chain reaction (PCR) using the Expand High Fidelity PCR System.
  • PCR-amplified fragments were gel-purified, digested with MIuI and Xbal, gel-purified, and subcloned, respectively, into a modified form of pVgl.D.Tt (Cole, M.S., et al., J. Immunol. 159: 3613-3621 (1997); and see below) and pVk (Co, M.S., et al., J. Immunol. 148: 1149-1154 (1992)).
  • Site-directed mutagenesis of the synthetic V-genes was done using the QuikChange II Site-Directed Mutagenesis Kit (Stratagene, La Jolla, CA), following the manufacturer's recommendations. Specific mutations in the 8El.6 VH gene were created using mutagenesis oligos and PCR methods well known in the art.
  • the PCR step was done using PfuUltra HF DNA Polymerase (Stratagene), following the manufacturer's recommendations, by incubating at 95°C for 30 sec, followed by 18 cycles of 95°C for 30 sec, 55°C for 1 min and 68°C for 1 min, followed by incubating at 68°C for 7 min. Following digestion with Dpnl, E.
  • the 8E1.4, 8E1.5 and 8E1.6 VH and VL genes were constructed by assembly of overlapping synthetic oligonucleotides and PCR methods well known in the art.
  • the identity of the framework amino acids between 1A6 VH and the acceptor human M60 and JH4 segments was 74%, while the identity between 1A6 VL and the acceptor human UI-3R and JK4 segments was 71%.
  • the antibody sequences were generated as disclosed in Example 2.3.3.
  • the allotype of the human gamma-1 constant region gene in the expression plasmid pVgl.D.Tt was modified from GIm (z,a) to the z, non-a allotype.
  • the overlap-extension PCR method (Higuchi, R., in “PCR Technology: Principles and Applications for DNA Amplification", Stockton Press, New York (1989), pp. 61-70) was used to generate the amino acid substitutions D356E and L358M (numbered according to the EU index of Kabat, E.A., et al., "Sequences of Proteins of Immunological Interest", 5 th ed., National Institutes of Health, Bethesda, MD (1991)), using mutagenesis" primers.
  • the PCR step was done using the QuikChange II Site-Directed Mutagenesis Kit (Stratagene). Following digestion or PCR generated product with Sfil and Eagl, the resulting restriction fragment was subcloned into a modified form of the pVgl.D.Tt expression vector containing an Nhel restriction site in the intron between the hinge and CH2 exons.
  • Mutations to the lower hinge region of the gamma-1 constant region gene were also generated by site-directed mutagenesis. Specifically amino acid substitutions L234A and L235A (numbered according to the EU index of Kabat, E.A., et al.) were generated using mutagenesis oligos. The PCR step was done using the QuikChange II Site-Directed Mutagenesis Kit (Stratagene) as described above. Following digestion of PCR generated product with Nhel and Eagl, the resulting restriction fragment was subcloned into the modified pVgl.D.Tt expression vector described above containing the D356E and L358M mutations and an Nhel site in the intron between the hinge and CH2 exons. Mutations were verified by nucleotide sequencing.
  • the amino acid sequences of the humanized VH and VL mini-exons are shown in Table 21.
  • the resulting V gene fragments were cloned, respectively, into a modified form of pVgLD.Tt and pVk.
  • Human kidney cell line 293T/17 (American Type Culture Collection, Manassus, VA) was maintained in DMEM (BioWhittaker, Walkersville, MD) containing 10% Fetal Bovine Serum (FBS) (HyClone, Logan, UT), 0.1 mM MEM non-essential amino acids (Invitrogen Corporation) and 2 mM L-glutamine (Invitrogen Corporation), hereinafter referred to as 293 medium, at 37°C in a 7.5% CO 2 incubator.
  • FBS Fetal Bovine Serum
  • 293 medium 293 medium
  • 293T/17 cells were incubated in DMEM containing 10% low-IgG FBS (HyClone), 0.1 mM MEM non-essential amino acids and 2 mM L-glutamine, hereinafter referred to as low-IgG 293 medium.
  • Transient transfection of 293T/17 cells was carried out using Lipofectamine 2000 (Invitrogen Corporation) following the manufacturer's recommendations. Approximately 2 x 10 7 cells per transfection were plated in a T-175 flask in 50 ml of 293 medium and grown overnight to confluence. The next day, 35 ⁇ g of light chain plasmid and 35 ⁇ g of heavy chain plasmid were combined with 3.75 ml of Hybridoma-SFM (HSFM) (Life Technologies, Rockville, MD). In a separate tube, 175 ⁇ l of Lipofectamine 2000 reagent and 3.75 ml of HSFM were combined and incubated for 5 min at room temperature.
  • HSFM Hybridoma-SFM
  • the 3.75 ml Lipofectamine 2000-HSFM mixture was mixed gently with the 3.75ml DNA-HSFM mixture and incubated at room temperature for 20 min.
  • the medium covering the 293T/17 cells was aspirated and replaced with low-IgG 293 medium, then the lipofectamine-DNA complexes were added dropwise to the cells, mixed gently by swirling, and the cells were incubated for 7 days at 37°C in a 7.5% CO 2 incubator before harvesting the supernatants.
  • Transient transfectants producing 8E1.4, 8E1.5 and 8E1.6 were generated as described above. Expression of humanized 8El.4, 8El.5 and 8El.6 antibodies was measured by sandwich ELISA.
  • IgGl/ ⁇ monoclonal antibodies were purified from exhausted culture supernatant with a protein A Sepharose column as follows. Culture supernatants from transient transfections were harvested by centrifugation, and sterile filtered. The pH of the filtered supernatants was adjusted by addition of 1/50 volume of 1 M sodium citrate, pH 7.0. Supernatants were run over a 1 ml HiTrap Protein A HP column (GE Healthcare Bio-Sciences Corporation, Piscataway, NJ) that was pre-equilibrated with 20 mM sodium citrate, 150 mM NaCl, pH 7.0.
  • HiTrap Protein A HP column GE Healthcare Bio-Sciences Corporation, Piscataway, NJ
  • the values represent IC50 ( ⁇ g/ml) required to compete 0.8 ( ⁇ g/ml) biotinylated 8El antibody.
  • Antibodies 1A6.10, 1A6.11, 1A6.12, 8E1.4 and 8El.5 were also generated using methods described in Example 2.3. Antibodies were expressed in COS cells and purified by Protein A affinity chromatography as described in examples 2.2.2 and 1.2.C, respectively. These purified mAbs were characterized for IC 50 and K D according to example 1.1. B andl.l.C 2. Table 24 shows the binding properties of 1A6.10, 1A6.11, 1A6.12, 8E1.4 and 8E1.5.
  • WO 03/035835 WO 90/02809, PCT/US91/05939, PCT7US91/09630, PCT/US94/01234, PCT/US96/18978, PCT/US98/16280, WO 92/20791, WO 90/05144 Al WO 00 09560, WO 00/037504 * WO 02072636, WO 91/10737, WO 91/10741 WO 92/01047, WO 92/18619, WO 93/11236, WO 94/02602, WO 95/15982, WO 95/20401, WO 96/33735 O 97/32572, O 97/32572, WO 97/44013, O 97/44013, WO 98/16654 WO 98/24893, WO 98/31346, WO 98/31346, WO 98/50433, WO 99/45031, WO 99/53049, WO 99/543428, WO 99/6

Abstract

The present invention encompasses IL-12p40 binding proteins, particularly antibodies that bind human interleukin-12 (hIL-12) and/or human IL-23 (hIL-23). Specifically, the invention relates to antibodies that are chimeric, CDR grafted and humanized antibodies. Preferred antibodies have high affinity for hIL-12 and/or hIL-23 and neutralize h IL-12 and/or hIL-23 activity in vitro and in vivo. An antibody of the invention can be a full-length antibody or an antigen-binding portion thereof. Method of making and method of using the antibodies of the invention are also provided. The antibodies, or antibody portions, of the invention are useful for detecting hIL-12 and/or hIL-23 and for inhibiting hIL-12 and/or hIL-23 activity, e.g., in a human subject suffering from a disorder in which hIL-12 and/or hIL-23 activity is detrimental.

Description

IL-12/P40 BINDING PROTEINS
Cross-reference to Related Application
This application claims the benefit of priority to US provisional application no. 60/695,679 filed June 30, 2005.
This application is related to U.S. Patent Application No. 09/534717 filed on March 24, 2000 (issued as U.S. Patent Number 6,914,128) entitled "Human antibodies that bind human IL- 12 and methods for producing." The entire contents of this patent application, and patents issued therefrom, are hereby incorporated herein by reference.
Reference to joint research agreement
Contents of this application are under a joint research agreement entered into by and between Protein Design Labs, Inc. and Abbott Laboratories on December 14, 2005, and directed to recombinantly engineered antibodies.
Field of the Invention
The present invention relates to IL-12p40 binding proteins, and specifically to their uses in the prevention and/or treatment of acute and chronic inflammatory diseases.
Background of the Invention
Human interleukin-12 (IL-12) is a cytokine with a unique structure and pleiotropic effects (Kobayashi, et al. (1989) J Exp Med 170:827-845; Seder, et al. (1993) Proc. Natl. Acad. Sd. 90:10188-10192, Ling, et al. (1995) J Exp Med 154:116-127; Podlaski, et al. (1992) Arch. Biochem. Biophys. 294:230-237). IL-12 plays a critical role in the pathology associated with several diseases involving immune and inflammatory responses. A review of IL-12, its biological activities, and its role in disease can be found in Trinchieri, G. (2003) Nat. Rev. Immun. 3:133- 146. Structurally, IL-12 is a heterodimeric protein (referred to as the "p70 protein") comprising a 35 kDa subunit (p35) and a 40 kDa subunit (p40) which are linked together by a disulfide bridge. The heterodimeric protein is produced primarily by antigen-presenting cells such as monocytes, macrophages and dendritic cells. These cell types also secrete an excess of the p40 subunit relative to p70 subunit. The p40 and p35 subunits are genetically unrelated and neither has been reported to possess biological activity, although the p40 homodimer may function as an IL- 12 antagonist.
Functionally, IL-12 plays a central role in regulating the balance between antigen- specific T helper type 1 (ThI) and type 2 (Th2) lymphocytes. The ThI and Th2 cells govern the initiation and progression of autoimmune disorders, and IL-12 is critical in the regulation of ThI- lymphocyte differentiation and maturation. Cytokines released by the ThI cells are inflammatory and include interferon gamma (IFN-γ), IL-2, and lymphotoxin (LT). Th2 cells secrete IL-4, IL-5, IL-6, IL-10 and IL-13 to facilitate humoral immunity, allergic reactions, and immunosuppression. Consistent with the preponderance of ThI responses in autoimmune diseases and the proinflammatory activities of IFN-γ, IL-12 may play a major role in the pathology associated with many autoimmune and inflammatory diseases such as rheumatoid arthritis (RA), multiple sclerosis (MS), psoriasis (PS) and Crohn's disease (CD).
Human patients with MS have demonstrated an increase in IL-12 expression as documented by p40 mRNA levels in acute MS plaques (Windhagen et al., (1995) J Exp. Med. 182:1985-1996). In addition, ex vivo stimulation of antigen-presenting cells with CD40L expressing T cells from MS patients resulted in increased IL-12 production compared with control T cells, consistent with the observation that CD40/CD40L interactions are potent inducers of IL-12. Elevated levels of IL-12 p70 have been detected in the synovia of RA patients compared with healthy controls (Morita et al. (1998) Arth. and Rheumat. 41:306-314). Cytokine messenger ribonucleic acid (mRNA) expression profile in the RA synovia identified predominantly ThI cytokines (Bucht et al. (1996) Clin. Exp. Immunol. 103:347-367). IL-12 also appears to play a critical role in the pathology associated with Crohn's disease. Increased expression of EMF-γ and IL-12 has been observed in the intestinal mucosa of patients with this disease (Fais et al. (1994) J Interferon Res. 14:235-238; Parronchi et al. (1997) Am. J. Path. 150:823-832; Monteleone et al. (1997) Gastroent. 112:1169- 1178, and Berrebi et al. (1998) Am. J Path 152:667-672). The cytokine secretion profile of T cells from the lamina propria of CD patients is characteristic of a predominantly ThI response, including greatly elevated IFN-γ levels (Fuss, et al. (1996) J Immunol. 157:1261-1270). Moreover, colon tissue sections from CD patients show an abundance of IL- 12 expressing macrophages and IFN-γ expressing T cells (Parronchi et al (1997) Am. J. Path. 150:823-832).
IL-23 is also a heterodimeric cytokine and belongs to a family of five such heterodimeric cytokines including IL-12 and IL-27 (Trinchieri et al., (2003) Immunity 19:641-644). IL-23 shares the identical p40 subunit as IL-12, but it is associated with a pl9 subunit via a disulphide- linkage. The pl9 subunit is structurally related to IL-6, granulocyte-colony stimulating factor (G- CSF), and the p35 subunit of IL-12. IL-23 is produced by similar cell types as JLL-12, ana its receptor is expressed on T cells, NK cells, and phagocytic and dendritic hematopoietic cells. IL- 23 mediates signaling by binding to a heterodimeric receptor, comprised of BL-23R and IL- 12betal. The EL-12betal subunit is shared by the 1L-12 receptor, which is composed of IL- 12betal and IL-12beta2. DL-23 does share overlapping functions with IL-12 (by inducing IFN-γ production, ThI cell differentiation and activating the antigen-presenting functions of dendritic cells) however it selectively induces proliferation of memory T cells (Oppmann et al. (2000) Immunity 13:715-725, Parham, et al. (2002) J. Immunol. 168:5699-5708).
The role of IL-23 in autoimmune inflammation has been dissected in part through studies with pl9 knockout mice (Murphy et al. J Exp Med 198:1951-1957; Cua et al. (2003) Nature 421:744-748). Studies have demonstrated that IL-23 modulates immune response to infection (see, e.g., Pirhonen, et al. (2002) J. Immunol. 169:5673-5678; Broberg, et al. (2002) J. Interferon Cytokine Res. 22:641-651; Elkins, et al. (2002) Infection Immunity 70:1936-1948; Cooper, et al. (2002) J. Immunol. 168:1322-1327). IL-23 is thought to play a role in immune-mediated inflammatory diseases (Langrish et.al. (2004) Immunological Reviews202: 96-105).
Due to the role of human IL-12 in a variety of human disorders, therapeutic strategies have been designed to inhibit or counteract IL-12 activity. In particular, antibodies that bind to, and neutralize, IL-12 have been sought as a means to inhibit IL-12 activity. Some of the earliest antibodies were murine monoclonal antibodies (mAbs), secreted by hybridomas prepared from lymphocytes of mice immunized with IL-12 (see e.g., Strober et al., PCT Publication No. WO 97/15327; Gately et al., WO9937682 A2; Neurath et al., J Exp. Med 182:1281-1290 (1995); Duchmarm et al., J Immunol. 26:934- 938(1996)). These murine IL-12 antibodies are limited for their use in vivo due to problems associated with administration of mouse antibodies to humans, such as short serum half life, an inability to trigger certain human effector functions and elicitation of an unwanted immune response against the mouse antibody in a human (the "human anti-mouse antibody" (HAMA) reaction).
One approach to overcome the problem problems associated with use of fully murine antibodies in humans is to generate fully human antibodies such as those disclosed in Salfeld et al., PCT publication No.WO 00/56772 Al. Other approaches to overcome the problems associated with use of fully murine antibodies in humans have involved genetically engineering the antibodies to be more "human-like." For example, chimeric antibodies, in which the variable regions of the antibody chains are murine-derived and the constant regions of the antibody chains are human-derived, have been prepared (Junghans, et al. (1990) Cancer Res. 50:1495-1502; Brown et al. (1991) Proc. Natl. Acad. Sci. 88:2663-2667; Kettleborough et al. (1991) Prot. Engineer. 4:773-783). Such chimeric antibodies to IL-12 are also disclosed in Peritt et al. PCT publication No.WO2002097048A2. However, because these chimeric antibodies still retain murine variable chain sequences, they still may elicit an unwanted immune reaction, the human anti-chimeric antibody (HACA) reaction especially when administered for prolonged periods.
There is a need in the art for improved antibodies capable of binding the p40 subunit of IL-12 (IL-12p40). Preferably the antibodies bind IL-12 and /or IL-23. Preferably the antibodies are capable of neutralizing IL-12 and /or 1L-23. The present invention provides a novel family of binding proteins, CDR grafted antibodies, humanized antibodies, and fragments thereof, capable binding IL-12p40, binding with high affinity, and binding and neutralizing IL-12 and/or IL-23.
Summary of the Invention
This invention pertains to IL-12p40 binding proteins, particularly antibodies capable of binding the p40 subunit of human IL-12 and the p40 subunit of human IL-23. Further, the invention provides methods of making and using IL-12p40 binding proteins.
One aspect of this invention pertains to a binding protein comprising an antigen binding domain capable of binding a p40 subunit of IL-12. In one embodiment the antigen binding domain comprises at least one CDR comprising an amino acid sequence selected from the group consisting of:
CDR-Hl. X1-X2-X3-X4-X5-X6-X? (SEQ ID NO: 55), wherein;
X1 is D, K, T, or S;
X2 is Y, S, or T;
X3 is Y, V, G, W, S, or F;
X4 is I, or M;
X5 is H, G, E, or V;
X6 is V, or is not present; and
X7 is S, or is not present;
CDR-H2. Xi- X2- X3- X4- X5— X6- X7- Xg- X9— Xio— Xii— Xi2- X13— X14— X15— Xi6— Xn Xi8~ X19-X20 (SEQ ID NO: 56), wherein;
X1 is H, D, G, W, S, Y or R;
X2 is I, or F;
X3 is Y, W, L, S, N, D or G;
X4 is W, P, H, T, or S;
X5 is D, G, E, A, or I;
X6 is D, G, S, T, or N;
X7 is D, G, S, or P;
X8 is K, N, S, E, T, or H;
X9 is Y, T, P, I, or N;
Xio is Y, N, T, H, K, S, or G; Xn is N, or Y;
Xi2 is P, N, A, D, or S;
X13 is S, E, D, or P;
Figure imgf000006_0001
X15 is K, F, V, M, R, or A;
Xi6 is S, K, Q, P, or is not present;
Xn is D, G, R, or is not present;
Xi8 is F, or is not present;
Xi9 is Q, or is not present; and
X20 is D, or is not present; CDR-H3. XI-X2-X3-X4-X5-X6-XT-XS-X9-XIO-XII-XI2-XI3 (SEQ ID NO: 57), wherein;
Figure imgf000006_0002
X2 is G, T, R, P, or H;
X3 is I, R, F, Y, or Q;
X4 is R, V, Y, F, or A;
X5 is S, N, G, A, or R;
X6 is A, Y, L, F, or M;
X7 is M, A, D, L, or F;
X8 is D, M, Y, or W;
X9 is Y, D, or N;
X10 is Y, A, or is not present;
Xn is M, or is not present;
Xi2 is D, or is not present; and
Xi3 is Y, or is not present; CDR-Ll. Xi-X2-X3-X4-X5-X6-X7-X8-X9-XiO-XiI-XIa-XiS-XM-X^ (SEQ ID NO:
58), wherein;
Figure imgf000006_0003
X2 is A;
X3 is S;
X4 is Q, or E;
X5 is S, or N;
X6 is V, or I;
X7 is S, G, or D;
X8 is N, T, or K;
X9 is D, N, or Y; X10 is V, G, or L;
Xn is A, I, or H;
X12 is S, or is not present;
Xi3 is F, or is not present;
Xi4 is M, or is not present; and
Xi5 is N, or is not present; CDR-L2. X1-X2-X3-X4-X5-X6-X7-X8 (SEQ ID NO: 59), wherein;
X1 is Y, or S;
X2 is A, or T;
X3 is S, or A;
X4 is N, H, S, or Q;
X5 is R, N, or S;
X6 is Y, Q, or I;
X7 is T, S, or G; and
X8 is S, or is not present; and CDR-L3. X1-X2-X3-X4-X5-X6-X7-X8-X9 (SEQ ID NO: 60), wherein;
Figure imgf000007_0001
X2 Is Q;
X3 is D, Y, or S;
X4 is Y, N, K, or I;
X5 is N, T, S, or E;
X6 is S, Y, V, or W;
X7 is P;
X8 is W, F, Y, L, or P; and
X9 is T, or S.
Preferably, the antigen binding domain comprises at least one CDR comprising an amino acid sequence selected from the group consisting of residues 31-37 of SEQ ID NO.:35;residues 52-67 of SEQ ID NO.:35;residues 100-108 of SEQ ID NO.:35;residues 24-34 of SEQ ID NO.:36;residues 50-56 of SEQ ID NO.:36;residues 89-97 of SEQ ID NO.:36;residues 31-37 of SEQ ID NO.:37;residues 52-67 of SEQ ID NO.:37 residues.100-109 of SEQ ID NO.:37;residues 24-34 of SEQ ID NO.:38;residues 50-56 of SEQ ID NO.:38;residues 89-97 of SEQ ID NO.:38;residues 31-35 of SEQ ID NO.:39;residues 50-66 of SEQ ID NO.:39;residues 99-106 of SEQ ID NO.:39;residues 24-34 of SEQ ID NO.:40;residues 50-56 of SEQ ID NO.:40;residues 89- 97 of SEQ ID NO.:40;residues 31-35 of SEQ ID NO.:41;residues 50-66 of SEQ ID NO.:41;residues 99-106 of SEQ ID NO.:41;residues 24-34 of SEQ ID NO.:42;residues 50-56 of SEQ E) NO.:42;residues 89-97 of SEQ ID NO.:42;residues 31-35 of SEQ ID JNU.:4i;resiaues DU- 66 of SEQ ID NO.:43;residues 99-106 of SEQ ID NO.:43;residues 24-34 of SEQ E) NO.:44;residues 50-56 of SEQ E) NO.:44;residues 89-97 of SEQ E) NO.:44;residues 31-35 of SEQ E) NO.:45;residues 50-66 of SEQ E) NO.:45;residues 99-101 of SEQ E) NO.:45;residues 24-34 of SEQ E) NO.:46;residues 50-56 of SEQ E) NO.:46;residues 89-97 of SEQ E) NO.:46;residues 31-35 of SEQ E) NO.:47;residues 50-66 of SEQ E) NO.:47;residues 99-106 of SEQ E) NO.:47;residues 24-34 of SEQ E) NO.:48;residues 50-56 of SEQ E) NO.:48;residues 89- 97 of SEQ E) NO.:48;residues 31-35 of SEQ JD NO.:49;residues 50-66 of SEQ E) NO.:49;residues 99-111 of SEQ E) NO.:49;residues 24-38 of SEQ E) NO.:50;residues 53-60 of SEQ E) NO.:50;residues 93-101 of SEQ E) NO.:50;residues 31-37 of SEQ E) NO.:51;residues 52-67 of SEQ E) NO.:51;residues 100-109 of SEQ E) NO.:51;residues 24-34 of SEQ E) NO.:52;residues 50-56 of SEQ E) NO.:52;residues 89-97 of SEQ E) NO.:52;residues 31-35 of SEQ E) NO.:53;residues 47-66 of SEQ E) NO.:53;residues 99-107 of SEQ E) NO.:53;residues 24-34 of SEQ E) NO.:54;residues 50-56 of SEQ E) NO.:54;and residues 89-97 of SEQ E) NO.:54. In a preferred embodiment, the binding protein comprises at least 3 CDRs selected from the group consisting of the sequences disclosed above. More preferably the 3 CDRs selected are from sets of variable domain CDRs selected from the group consisting of:
VH 1D4 CDR Set
VH 1D4 CDR-Hl Residues 31-37 of SEQ ID NO. :35
VH 1D4 CDR-H2 Residues 52-67 of SEQ ID NO. : 35
VH 1D4 CDR-H3 Residues 100-108 of SEQ ID NO. 35
VL 1D4 CDR Set
VL 1D4 CDR-Ll Residues24-34 of SEQ ID NO :36
VL 1D4 CDR-L2 Residues50-56 of SEQ ID NO. 36
VL 1D4 CDR-L3 Residues89-97 of SEQ ID NO. 36
VH 1A6 CDR Set
VH 1A6 CDR-Hl Residues31-37 of SEQ ID NO. 37
VH 1A6 CDR-H2 Residues52-67 of SEQ ID NO. 37
VH 1A6 CDR-H3 ResidueslOO-109 of SEQ ID NO. 37
VL 1A6 CDR Set
VL 1A6 CDR-Ll Residues24-34 of SEQ ID NO. 38
VL 1A6 CDR-L2 Residues50-56 of SEQ ID NO. 38
VL 1A6 CDR-L3 Residues89-97 of SEQ ID NO. 38
VH 1D8 CDR Set
VH 1D8 CDR-Hl Residues 31-35 of SEQ ID NO. 39
VH 1D8 CDR-H2 Residues 50-66 of SEQ ID NO. 39
VH 1D8 CDR-H3 Residues 99-106 of SEQ ID NO. 39
VL 1D8 CDR Set
VL 1D8 CDR-Ll Residues 24-34 of SEQ ID NO :40
VL 1D8 CDR-L2 Residues 50-56 of SEQ ID NO. 40
VL 1D8 CDR-L3 Residues 89-97 of SEQ ID NO. 40
VH 3G7 CDR Set
VH 3G7 CDR-Hl Residues 31-35 of SEQ ID NO. 41
VH 3G7 CDR-H2 Residues 50-66 of SEQ ID NO. . 41
VH 3G7 CDR-H3 Residues 99-106 of SEQ ID NO. . 41 VL 3G7 CDR Set
VL 3G7 CDR-Ll Residues 24-34 of SEQ ID NO. : 42
VL 3G7 CDR-L2 Residues 50-56 of SEQ ID NO. : 42
VL 3G7 CDR-L3 Residues 89-97 of SEQ ID NO. : 42
VH 5E8 CDR Set
VH 5E8 CDR-Hl Residues 31-35 of SEQ ID NO. : 43
VH 5E8 CDR-H2 Residues 50-66 of SEQ ID NO. : 43
VH 5E8 CDR-H3 Residues 99-106 of SEQ ID NO. 43
VL 5E8 CDR Sθt
VL 5E8 CDR-Ll Residues 24-34 of SEQ ID NO. 44
VL 5E8 CDR-L2 Residues 50-56 of SEQ ID NO. 44
VL 5E8 CDR-L3 Residues 89-97 of SEQ ID NO. 44
VH 8El CDR Set
VH 8El CDR-Hl Residues 31-35 of SEQ ID NO. 45
VH 8El CDR-H2 Residues 50-66 of SEQ ID NO :45
VH 8El CDR-H3 Residues 99-101 of SEQ ID NO. 45
VL 8El CDR Set
VL 8El CDR-Ll Residues 24-34 of SEQ ID NO. 46
VL 8El CDR-L2 Residues 50-56 of SEQ ID NO. 46
VL 8El CDR-L3 Residues 89-97 of SEQ ID NO. 46
VH 1H6 CDR Sθt
VH 1H6 CDR-Hl Residues 31-35 of SEQ ID NO. 47
VH 1H6 CDR-H2 Residues 50-66 of SEQ ID NO. 47
VH 1H6 CDR-H3 Residues 99-106 of SEQ ID NO. 47
VL 1H6 CDR Set
VL 1H6 CDR-Ll Residues 24-34 of SEQ ID NO. 48
VL 1H6 CDR-L2 Residues 50-56 of SEQ ID NO. 48
VL 1H6 CDR-L3 Residues 89-97 of SEQ ID NO. 48
VH 3A11 CDR Sθt
VH 3All CDR-Hl Residues 31-35 of SEQ ID NO. 49
VH 3Al1 CDR-H2 Residues 50-66 of SEQ ID NO. : 49
VH 3Al1 CDR-H3 Residues 99-111 of SEQ ID NO. 49
VL 3Al1 CDR Sθt
VL 3Al1 CDR-Ll Residues 24-38 of SEQ ID NO. : 50
VL 3A11 CDR-L2 Residues 53-60 of SEQ ID NO. : 50
VL 3Al1 CDR-L3 Residues 93-101 of SEQ ID NO. : 50
VH 4B4 CDR Sθt
VH 4B4 CDR-Hl Residues 31-37 of SEQ ID NO. : 51
VH 4B4 CDR-H2 Residues 52-67 of SEQ ID NO. : 51
VH 4B4 CDR-H3 Residues 100-109 of SEQ ID NO. : 51
VL 4B4 CDR Set
VL 4B4 CDR-Ll Residues 24-34 of SEQ ID NO. : 52
VL 4B4 CDR-L2 Residues 50-56 of SEQ ID NO. : 52
VL 4B4 CDR-L3 Residues 89-97 of SEQ ID NO. : 52
VH 7G3 CDR Set
VH 7G3 CDR-Hl Residues 31-35 of SEQ ID NO. : 53
VH 7G3 CDR-H2 Residues 47-66 of SEQ ID NO. : 53
VH 7G3 CDR-H3 Residues 99-107 of SEQ ID NO. : 53
And
VL 7G3 CDR . Set
VL 7G3 CDR-Ll Residues 24-34 of SEQ ID NO. : 54
VL 7G3 CDR-L2 Residues 50-56 of SEQ ID NO. : 54
VL 7G3 CDR-L3 Residues 89-97 of SEQ ID NO. : 54 In one embodiment the binding protein of the invention comprises at least two variable domain CDR sets. More preferably, the two variable domain CDR sets are selected from a group consisting of: VH 1D4 CDR Set & VL 1D4 CDR Set; VH 1A6 CDR Set & VL 1A6 CDR Set; VH 1D8 CDR Set & VL 1D8 CDR Set; VH 3G7 CDR Set & VL 3G7 CDR Set; VH 5E8 CDR Set & VL 5E8 CDR Set; VH 8El CDR Set & VL 8El CDR Set; VH 1H6 CDR Set & VL 1H6 CDR Set; VH 3Al 1 CDR Set & VL 3Al 1 CDR Set; VH 4B4 CDR Set & VL 4B4 CDR Set; and VH 7G3 CDR Set & VL 7G3 CDR Set.
In another embodiment the binding protein disclosed above further comprises a human acceptor framework. Preferably the human acceptor framework comprises a amino acid sequence selected from the group consisting of SEQ ID NO.:6; SEQ ID NO.:7; SEQ ID NO.:8; SEQ ID NO.:9; SEQ ID NO.: 10; SEQ ID NO.: 11; SEQ ID NO.: 12; SEQ ID NO.: 13; SEQ ID NO.: 14; SEQ ID NO.: 15; SEQ ID NO.: 16; SEQ ID NO.: 17; SEQ ID NO.: 18; SEQ ID NO.: 19; SEQ ID NO.:20; SEQ ID NO.:21; SEQ ID NO.:22; SEQ ID NO.:23; SEQ ID NO.:24; SEQ ID NO.:25; SEQ ID NO.:26; SEQ ID NO.:27; SEQ ID NO.:28; SEQ ID NO.:29; SEQ ID NO.:30; SEQ ID NO.:31; SEQ ID NO.:32; SEQ ID NO.:33; SEQ ID NO.:34, SEQ ID NO.:92, SEQ ID NO.:93, SEQ ID NO.:94, SEQ ID NO.:95, SEQ ID NO.:96, AND SEQ ID NO.-.97.
In a preferred embodiment the binding protein is a CDR grafted antibody or antigen binding portion thereof capable of binding the p40 subunit of IL-12 or IL-23. Preferably the CDR grafted antibody or antigen binding portion thereof comprise one or more CDRs disclosed above. More preferably the CDR grafted antibody or antigen binding portion thereof comprises at least one variable domain having an amino acid sequence selected from the group consisting of SEQ ID NO.:61; SEQ ID NO.:62; SEQ ID NO.:63; SEQ ID NO.:64; SEQ ID NO.:65; SEQ ID NO.:66; SEQ ID NO.:67; SEQ ID NO.:68; SEQ ID NO.:69; SEQ ID NO.:70; SEQ ID NO.:71; SEQ ID NO.:72; SEQ ID NO.:73; SEQ ID NO.:74; SEQ ID NO.:75; SEQ ID NO.:76; SEQ ID NO.:77; and SEQ TD NO.:78. Most preferably the CDR grafted antibody or antigen binding portion thereof comprises two variable domains selected from the group disclosed above. Preferably the CDR grafted antibody or antigen binding portion thereof comprises a human acceptor framework. More preferably the human acceptor framework is any one of the human acceptor frameworks disclosed above.
In a preferred embodiment the binding protein is a humanized antibody or antigen binding portion thereof capable of binding the p40 subunit of IL-12 or IL-23. Preferably the humanized antibody or antigen binding portion thereof comprise one or more CDRs disclosed above incorporated into a human antibody variable domain of a human acceptor framework. Preferably the human antibody variable domain is a consensus human variable domain. More preferably the human acceptor framework comprises at least one Framework Region amino acid substitution at a key residue, wherein the key residue is selected from the group consisting of a residue adjacent to a CDR; a glycosylation site residue; a rare residue; a residue capable of interacting with a p40 subunit of human IL- 12; a residue capable of interacting with a CDR; a canonical residue; a contact residue between heavy chain variable region and light chain variable region; a residue within a Vernier zone; and a residue in a region that overlaps between a Chothia-defined variable heavy chain CDRl and a Kabat-defined first heavy chain framework. Preferably the key residue is selected from the group consisting of 3H, 5H, 1OH, HH, 12H, 13H, 15H, 16H, 18H, 19H, 23H, 24H, 25H, 3OH, 41H, 44H, 46H, 49H, 66H, 68H, 71H, 73H, 74H, 75H, 76H, 77H, 78H, 79H, 8 IH, 82H, 82AH, 82BH, 82CH, 83H, 84H, 85H, 86H, 87H, 89H, 93H, 98H, 108H, 109H, IL, 2L, 3L, 7L, 8L, 9L, 1OL, HL, 12L, 13L, 15L, 17L, 19L, 2OL, 21L, 22L, 36L, 41L, 42L, 43L, 45L, 46L, 58L, 6OL, 62L, 63L, 67L, 7OL, 73L, 74L, 77L, 78L, 79L, 80L, 83L, 85L, 87L, 104L, and 106L. Preferably the human acceptor framework human acceptor framework comprises at least one Framework Region amino acid substitution, wherein the amino acid sequence of the framework is at least 65% identical to the sequence of said human acceptor framework and comprises at least 70 amino acid residues identical to said human acceptor framework.
In a preferred embodiment the binding protein is a humanized antibody or antigen binding portion thereof capable of binding the p40 subunit of IL- 12 or IL-23. Preferably the humanized antibody, or antigen binding portion, thereof comprises one or more CDRs disclosed above. More preferably the humanized antibody, or antigen binding portion, thereof comprises three or more CDRs disclosed above. Most preferably the humanized antibody, or antigen binding portion, thereof comprises six CDRs disclosed above.
In another embodiment of the claimed invention, the humanized antibody or antigen binding portion thereof comprises at least one variable domain having an amino acid sequence selected from the group consisting of SEQ ID NO.:79, SEQ ID NO.:80, SEQ ID NO.:81, SEQ ID NO.:82, SEQ ID NO.:83, SEQ ID NO.:84, SEQ ID NO.:85, SEQ ID NO.:86, SEQ ID NO.:87, SEQ ID NO.:88, SEQ ID NO.:89, SEQ ID NO.:90, SEQ ID NO.:91, SEQ ID NO.:98, SEQ ID NO.:99, SEQ ID NO.: 100, SEQ ID NO.: 101, SEQ ID NO.: 102, AND SEQ ID NO.: 103, SEQ ID NO.: 104, SEQ ED NO.: 105, SEQ DD NO.: 106, SEQ ID NO.: 107, SEQ ID NO.: 108, and SEQ ID NO.: 109. More preferably the humanized antibody or antigen binding portion thereof comprises two variable domains selected from the group disclosed above. Most preferably humanized antibody, or antigen binding portion thereof comprises two variable domains, wherein said two variable domains have amino acid sequences selected from the group consisting of SEQ ID NO.:67 & SEQ ID NO.:79, SEQ ID NO.:80 & SEQ ID NO.:81, SEQ ID NO.:82 & SEQ ID NO.:83, SEQ ID NO.:84 & SEQ ID NO.:85, SEQ ID NO.:86 & SEQ ID NO.:87, SEQ ID NO.:88 & SEQ ID NO.:89, SEQ ID NO.:90 & SEQ ID NO.:91, SEQ ID NO.:98 & SEQ ID NO.:99, SEQ ID NO.: 100 & SEQ ID NO.: 101, SEQ ID NO.: 102 & SEQ ID NO.: 103, SEQ ID NO.: 104 & SEQ ID NO.: 105, SEQ ID NO.: 106 & SEQ ID NO.: 107, and SEQ ID NO.: 108 & SEQ ID NO.: 109.
In a preferred embodiment the binding protein disclosed above comprises a heavy chain immunoglobulin constant domain selected from the group consisting of a human IgM constant domain, a human IgGl constant domain, a human IgG2 constant domain, a human IgG3 constant domain, a human IgG4 constant domain, a human IgE constant domain, and a human IgA constant domain. More preferably, the binding protein comprises SEQ ID NO.:2; SEQ DD NO.:3; SEQ ID NO.:4; and SEQ ID NO.:5.
The binding protein of the invention is capable of binding a target selected from the group consisting of IL-12 and IL-23. Preferably the binding protein is capable of modulating a biological function of a target selected from the group consisting of IL-12 and IL-23. More preferably the binding protein is capable of neutralizing a target selected from the group consisting of IL-12 and IL-23.
In one embodiment, the binding protein of the invention has an on rate constant (Kon) to IL-12 or IL-23 of at least about 102M-1S'1; at least about 103M4S"1; at least about 104M4S"1; at least about 105M-1S"1; or at least about 106M-1S"1, as measured by surface plasmon resonance. Preferably, the binding protein of the invention has an on rate constant (Kon) to IL-12 or IL-23 between 102M-1S"1 to 103M-1S"1; between 103M-1S"1 to 104M-1S"1; between lOVrV1 to 105M-1S"1; or between 105M-1S"1 to 106M-1S"1, as measured by surface plasmon resonance.
In another embodiment, the binding protein of the invention has an off rate constant (Koff) to IL-12 or IL-23 of at most about 10"3S"1; at most about 10"4S"1; at most about 10"5S"1; or at most about 10"6S"1, as measured by surface plasmon resonance. Preferably, the binding protein of the invention has an off rate constant (Koff) to IL-12 or IL-23 of 10"Y1 to 10"4S"1; of 10"4S"1 to 10" 5S"1; or of 10"5S"1 to 10'6S"1, as measured by surface plasmon resonance.
In another embodiment, the binding protein of the invention has a dissociation constant (KD) to IL-12 or IL-23 of at most about lO'7 M; at most about lO'8 M; at most about 10"9 M; at most about 10"10 M; at most about 10"u M; at most about 10"12 M; or at most 10"13M. Preferably, the binding protein of the invention has a dissociation constant (KD) to IL-12 or IL-23 of 10"7 M to 10"8 M; of 10"8 M to 10"9 M; of 10"9 M to 10"10 M; of 10"10 to 10"11 M; of 10"11 M to 10"12 M; or of lO'12 to M 10'13M.One embodiment of the invention provides an antibody construct comprising any one of the binding proteins disclosed above and a linker polypeptide or an immunoglobulin. In a preferred embodiment the antibody construct is selected from the group consisting of an immunoglobulin molecule, a monoclonal antibody, a chimeric antibody, a CDR- grafted antibody, a humanized antibody, a Fab, a Fab', a F(ab')2, a Fv, a disulfide linked Fv, a scFv, a single domain antibody, a diabody, a multispecific antibody, a dual specific antibody, and a bispecific antibody. In a preferred embodiment the antibody construct comprises a heavy chain immunoglobulin constant domain selected from the group consisting of a human IgM constant domain, a human IgGl constant domain, a human IgG2 constant domain, a human IgG3 constant domain, a human IgG4 constant domain, a human IgE constant domain, and a human IgA constant domain. More preferably, the antibody construct comprises SEQ ID NO.:2; SEQ ID NO.:3; SEQ ID NO.:4; and SEQ ID NO.:5. In another embodiment the invention provides an antibody conjugate comprising an the antibody construct disclosed above and an agent an agent selected from the group consisting of; an immunoadhension molecule, an imaging agent, a therapeutic agent, and a cytotoxic agent. In a preferred embodiment the imaging agent selected from the group consisting of a radiolabel, an enzyme, a fluorescent label, a luminescent label, a bioluminescent label, a magnetic label, and biotin. More preferably the imaging agent is a radiolabel selected from the group consisting of: 3H 14C 35S, 90Y, 99Tc, 111In, 1251, 1311, 177Lu, 166Ho, and 153Sm. In a preferred embodiment the therapeutic or cytotoxic agent is selected from the group consisting of; an anti-metabolite, an alkylating agent, an antibiotic, a growth factor, a cytokine, an anti-angiogenic agent, an anti-mitotic agent, an anthracycline, toxin, and an apoptotic agent.
In another embodiment the antibody construct is glycosylated. Preferably the glycosylation is a human glycosylation pattern.
In another embodiment binding protein, antibody construct or antibody conjugate disclosed above exists as a crystal. Preferably the crystal is a carrier-free pharmaceutical controlled release crystal. In a preferred embodiment the crystallized binding protein, crystallized antibody construct or crystallized antibody conjugate has a greater half life in vivo than its soluble counterpart. In another preferred embodiment the crystallized binding protein, crystallized antibody construct or crystallized antibody conjugate retains biological activity after crystallization.
One aspect of the invention pertains to an isolated nucleic acid encoding any one of the binding protein, antibody construct or antibody conjugate disclosed above. A further embodiment provides a vector comprising the isolated nucleic acid disclosed above wherein said vector is selected from the group consisting of pcDNA; pTT (Durocher et al., Nucleic Acids Research 2002, VoI 30, No.2); pTT3 (pTT with additional multiple cloning site; pEFBOS (Mizushima, S. and Nagata, S., (1990) Nucleic acids Research VoI 18, No. 17); pBV; pJV; and pBJ.
In another aspect a host cell is transformed with the vector disclosed above. Preferably the host cell is a prokaryotic cell. More preferably the host cell is E.Coli. In a related embodiment the host cell is an eukaryotic cell. Preferably the eukaryotic cell is selected from the group consisting of protist cell, animal cell, plant cell and fungal cell. More preferably the host cell is a mammalian cell including, but not limited to, CHO and COS; or a fungal cell such as Saccharomyces cerevisiae; or an insect cell such as Sf9. Another aspect of the invention provides a method of producing a binding protein that binds the p40 subunit of EL-12, comprising culturing any one of the host cells disclosed above in a culture medium under conditions sufficient to produce a binding protein that binds the p40 subunit of IL-12. Another embodiment provides a binding protein produced according to the method disclosed above.
One embodiment provides a composition for the release of a binding protein wherein the composition comprises a formulation which in turn comprises a crystallized binding protein, crystallized antibody construct or crystallized antibody conjugate as disclosed above and an ingredient; and at least one polymeric carrier. Preferably the polymeric carrier is a polymer selected from one or more of the group consisting of: poly (acrylic acid), poly (cyanoacrylates), poly (amino acids), poly (anhydrides), poly (depsipeptide), poly (esters), poly (lactic acid), poly (lactic-co-glycolic acid) or PLGA, poly (b-hydroxybutryate), poly (caprolactone), poly (dioxanone); poly (ethylene glycol), poly ((hydroxypropyl) methacrylamide, poly [(organo)phosphazene], poly (ortho esters), poly (vinyl alcohol), poly (vinylpyrrolidone), maleic anhydride- alkyl vinyl ether copolymers, pluronic polyols, albumin, alginate, cellulose and cellulose derivatives, collagen, fibrin, gelatin, hyaluronic acid, oligosaccharides, glycaminoglycans, sulfated polyeaccharides, blends and copolymers thereof. Preferably the ingredient is selected from the group consisting of albumin, sucrose, trehalose, lactitol, gelatin, hydroxypropyl-β- cyclodextrin, methoxypolyethylene glycol and polyethylene glycol. Another embodiment provides a method for treating a mammal comprising the step of administering to the mammal an effective amount of the composition disclosed above.
The invention also provides a pharmaceutical composition comprising a binding protein, antibody construct or antibody conjugate as disclosed above and a pharmaceutically acceptable carrier. In a further embodiment the pharmaceutical composition comprises at least one additional therapeutic agent for treating a disorder in which JL-12 and/or IL-23 activity is detrimental. Preferably the additional agent is selected from the group consisting of: Therapeutic agent, imaging agent, cytotoxic agent, angiogenesis inhibitors (including but not limited to anti-VEGF antibodies or VEGF-trap); kinase inhibitors (including but not limited to KDR and TIE-2 inhibitors); co-stimulation molecule blockers (including but not limited to anti-B7.1, anti-B7.2, CTLA4-Ig, anti-CD20); adhesion molecule blockers (including but not limited to anti-LFA-1 Abs, anti-E/L selectin Abs, small molecule inhibitors); anti-cytokine antibody or functional fragment thereof (including but not limited to anti-IL-18, anti-TNF, anti-IL-6/cytokine receptor antibodies); methotrexate; cyclosporin; rapamycin; FK506; detectable label or reporter; a TNF antagonist; an antirheumatic; a muscle relaxant, a narcotic, a non-steroid anti-inflammatory drug (NSAID), an analgesic, an anesthetic, a sedative, a local anesthetic, a neuromuscular blocker, an antimicrobial, an antipsoriatic, a corticosteriod, an anabolic steroid, an erythropoietin, an immunization, an immunoglobulin, an immunosuppressive, a growth hormone, a hormone replacement drug, a radiopharmaceutical, an antidepressant, an antipsychotic, a stimulant, an asthma medication, a beta agonist, an inhaled steroid, an epinephrine or analog, a cytokine, and a cytokine antagonist.
In another aspect, the invention provides a method for inhibiting human IL-12 and/or human DL-23 activity comprising contacting human IL-12 and/or human IL-23 with a binding protein disclosed above such that human IL-12 and/or human EL-23 activity is inhibited. In a related aspect the invention provides a method for inhibiting human IL-12 and/or human IL-23 activity in a human subject suffering from a disorder in which IL-12 and/or IL-23 activity is detrimental, comprising administering to the human subject a binding protein disclosed above such that human IL-12 and/or human IL-23 activity in the human subject is inhibited and treatment is achieved. Preferably the disorder is selected from the group comprising arthritis, osteoarthritis, juvenile chronic arthritis, septic arthritis, Lyme arthritis, psoriatic arthritis, reactive arthritis, spondyloarthropathy, systemic lupus erythematosus, Crohn's disease, ulcerative colitis, inflammatory bowel disease, insulin dependent diabetes mellitus, thyroiditis, asthma, allergic diseases, psoriasis, dermatitis scleroderma, graft versus host disease, organ transplant rejection, acute or chronic immune disease associated with organ transplantation, sarcoidosis, atherosclerosis, disseminated intravascular coagulation, Kawasaki's disease, Grave's disease, nephrotic syndrome, chronic fatigue syndrome, Wegener's granulomatosis, Henoch-Schoenlein purpurea, microscopic vasculitis of the kidneys, chronic active hepatitis, uveitis, septic shock, toxic shock syndrome, sepsis syndrome, cachexia, infectious diseases, parasitic diseases, acquired immunodeficiency syndrome, acute transverse myelitis, Huntington's chorea, Parkinson's disease, Alzheimer's disease, stroke, primary biliary cirrhosis, hemolytic anemia, malignancies, heart failure, myocardial infarction, Addison's disease, sporadic, polyglandular deficiency type I and polyglandular deficiency type π, Schmidt's syndrome, adult (acute) respiratory distress syndrome, alopecia, alopecia areata, seronegative arthopathy, arthropathy, Reiter's disease, psoriatic arthropathy, ulcerative colitic arthropathy, enteropathic synovitis, chlamydia, yersinia and salmonella associated arthropathy, spondyloarthopathy, atheromatous disease/arteriosclerosis, atopic allergy, autoimmune bullous disease, pemphigus vulgaris, pemphigus foliaceus, pemphigoid, linear IgA disease, autoimmune haemolytic anaemia, Coombs positive haemolytic anaemia, acquired pernicious anaemia, juvenile pernicious anaemia, myalgic encephalitis/Royal Free Disease, chronic mucocutaneous candidiasis, giant cell arteritis, primary sclerosing hepatitis, cryptogenic autoimmune hepatitis, Acquired Immunodeficiency Disease Syndrome, Acquired Immunodeficiency Related Diseases, Hepatitis B, Hepatitis C, common varied immunodeficiency (common variable hypogammaglobulinaemia), dilated cardiomyopathy, female infertility, ovarian failure, premature ovarian failure, fibrotic lung disease, cryptogenic fibrosing alveolitis, post-inflammatory interstitial lung disease, interstitial pneumonitis, connective tissue disease associated interstitial lung disease, mixed connective tissue disease associated lung disease, systemic sclerosis associated interstitial lung disease, rheumatoid arthritis associated interstitial lung disease, systemic lupus erythematosus associated lung disease, dermatomyositis/polymyositis associated lung disease, Sjogren's disease associated lung disease, ankylosing spondylitis associated lung disease, vasculitic diffuse lung disease, haemosiderosis associated lung disease, drug-induced interstitial lung disease, fibrosis, radiation fibrosis, bronchiolitis obliterans, chronic eosinophilic pneumonia, lymphocytic infiltrative lung disease, postinfectious interstitial lung disease, gouty arthritis, autoimmune hepatitis, type-1 autoimmune hepatitis (classical autoimmune or lupoid hepatitis), type-2 autoimmune hepatitis (anti-LKM antibody hepatitis), autoimmune mediated hypoglycaemia, type B insulin resistance with acanthosis nigricans, hypoparathyroidism, acute immune disease associated with organ transplantation, chronic immune disease associated with organ transplantation, osteoarthrosis, primary sclerosing cholangitis, psoriasis type 1, psoriasis type 2, idiopathic leucopaenia, autoimmune neutropaenia, renal disease NOS, glomerulonephritides, microscopic vasulitis of the kidneys, lyme disease, discoid lupus erythematosus, male infertility idiopathic or NOS, sperm autoimmunity, multiple sclerosis (all subtypes), sympathetic ophthalmia, pulmonary hypertension secondary to connective tissue disease, Goodpasture's syndrome, pulmonary manifestation of polyarteritis nodosa, acute rheumatic fever, rheumatoid spondylitis, Still's disease, systemic sclerosis, Sjorgren's syndrome, Takayasu's disease/arteritis, autoimmune thrombocytopaenia, idiopathic thrombocytopaenia, autoimmune thyroid disease, hyperthyroidism, goitrous autoimmune hypothyroidism (Hashimoto's disease), atrophic autoimmune hypothyroidism, primary myxoedema, phacogenic uveitis, primary vasculitis, vitiligo acute liver disease, chronic liver diseases, alcoholic cirrhosis, alcohol-induced liver injury, choleosatatis, idiosyncratic liver disease, Drug-Induced hepatitis, Non-alcoholic Steatohepatitis, allergy and asthma, group B streptococci (GBS) infection, mental disorders (e.g., depression and schizophrenia), Th2 Type and ThI Type mediated diseases, acute and chronic pain (different forms of pain), and cancers such as lung, breast, stomach, bladder, colon, pancreas, ovarian, prostate and rectal cancer and hematopoietic malignancies (leukemia and lymphoma), Abetalipoprotemia, Acrocyanosis, acute and chronic parasitic or infectious processes, acute leukemia, acute lymphoblastic leukemia (ALL), acute myeloid leukemia (AML), acute or chronic bacterial infection, acute pancreatitis, acute renal failure, adenocarcinomas, aerial ectopic beats, AIDS dementia complex, alcohol- induced hepatitis, allergic conjunctivitis, allergic contact dermatitis, allergic rhinitis, allograft rejection, alpha-1- antitrypsin deficiency, amyotrophic lateral sclerosis, anemia, angina pectoris, anterior horn cell degeneration, anti cd3 therapy, antiphospholipid syndrome, anti-receptor hypersensitivity reactions, aordic and peripheral aneuryisms, aortic dissection, arterial hypertension, arteriosclerosis, arteriovenous fistula, ataxia, atrial fibrillation (sustained or paroxysmal), atrial flutter, atrioventricular block, B cell lymphoma, bone graft rejection, bone marrow transplant (BMT) rejection, bundle branch block, Burkitt's lymphoma, Burns, cardiac arrhythmias, cardiac stun syndrome, cardiac tumors, cardiomyopathy, cardiopulmonary bypass inflammation response, cartilage transplant rejection, cerebellar cortical degenerations, cerebellar disorders, chaotic or multifocal atrial tachycardia, chemotherapy associated disorders, chromic myelocytic leukemia (CML), chronic alcoholism, chronic inflammatory pathologies, chronic lymphocytic leukemia (CLL), chronic obstructive pulmonary disease (COPD), chronic salicylate intoxication, colorectal carcinoma, congestive heart failure, conjunctivitis, contact dermatitis, cor pulmonale, coronary artery disease, Creutzfeldt- Jakob disease, culture negative sepsis, cystic fibrosis, cytokine therapy associated disorders, Dementia pugilistica, demyelinating diseases, dengue hemorrhagic fever, dermatitis, dermatologic conditions, diabetes, diabetes mellitus, diabetic ateriosclerotic disease, Diffuse Lewy body disease, dilated congestive cardiomyopathy, disorders of the basal ganglia, Down's Syndrome in middle age, drug- induced movement disorders induced by drugs which block CNS dopamine receptors, drug sensitivity, eczema, encephalomyelitis, endocarditis, endocrinopathy, epiglottitis, epstein-barr virus infection, erythromelalgia, extrapyramidal and cerebellar disorders, familial hematophagocytic lymphohistiocytosis, fetal thymus implant rejection, Friedreich's ataxia, functional peripheral arterial disorders, fungal sepsis, gas gangrene, gastric ulcer, glomerular nephritis, graft rejection of any organ or tissue, gram negative sepsis, gram positive sepsis, granulomas due to intracellular organisms, hairy cell leukemia, Hallerrorden-Spatz disease, hashimoto's thyroiditis, hay fever, heart transplant rejection, hemachromatosis, hemodialysis, hemolytic uremic syndrome/thrombolytic thrombocytopenic purpura, hemorrhage, hepatitis (A), His bundle arrythmias, EQV infection/HIV neuropathy, Hodgkin's disease, hyperkinetic movement disorders, hypersensitity reactions, hypersensitivity pneumonitis, hypertension, hypokinetic movement disorders, hypothalamic-pituitary-adrenal axis evaluation, idiopathic Addison's disease, idiopathic pulmonary fibrosis, antibody mediated cytotoxicity, Asthenia, infantile spinal muscular atrophy, inflammation of the aorta, influenza a, ionizing radiation exposure, iridocyclitis/uveitis/optic neuritis, ischemia- reperfusion injury, ischemic stroke, juvenile rheumatoid arthritis, juvenile spinal muscular atrophy, Kaposi's sarcoma, kidney transplant rejection, legionella, leishmaniasis, leprosy, lesions of the corticospinal system, lipedema, liver transplant rejection, lymphedema, malaria, malignamt Lymphoma, malignant histiocytosis, malignant melanoma, meningitis, meningococcemia, metabolic/idiopathic, migraine headache, mitochondrial multi. system disorder, mixed connective tissue disease, monoclonal gammopathy, multiple myeloma, multiple systems degenerations (Mencel Dejerine- Thomas Shi-Drager and Machado-Joseph), myasthenia gravis, mycobacterium avium intracellulare, mycobacterium tuberculosis, myelodyplastic syndrome, myocardial infarction, myocardial ischemic disorders, nasopharyngeal carcinoma, neonatal chronic lung disease, nephritis, nephrosis, neurodegenerative diseases, neurogenic I muscular atrophies , neutropenic fever, non- hodgkins lymphoma, occlusion of the abdominal aorta and its branches, occulsive arterial disorders, okt3 therapy, orchitis/epidydimitis, orchitis/vasectomy reversal procedures, organomegaly, osteoporosis, pancreas transplant rejection, pancreatic carcinoma, paraneoplastic syndrome/hypercalcemia of malignancy, parathyroid transplant rejection, pelvic inflammatory disease, perennial rhinitis, pericardial disease, peripheral atherlosclerotic disease, peripheral vascular disorders, peritonitis, pernicious anemia, Pneumocystis carinii pneumonia, pneumonia, POEMS syndrome (polyneuropathy, organomegaly, endocrinopathy, monoclonal gammopathy, and skin changes syndrome), post perfusion syndrome, post pump syndrome, post-MI cardiotomy syndrome, preeclampsia, Progressive supranucleo Palsy, primary pulmonary hypertension, radiation therapy, Raynaud's phenomenon and disease, Raynoud's disease, Refsum's disease, regular narrow QRS tachycardia, renovascular hypertension, reperfusion injury, restrictive cardiomyopathy, sarcomas, scleroderma, senile chorea, Senile Dementia of Lewy body type, seronegative arthropathies, shock, sickle cell anemia, skin allograft rejection, skin changes syndrome, small bowel transplant rejection, solid tumors, specific arrythmias, spinal ataxia, spinocerebellar degenerations, streptococcal myositis, structural lesions of the cerebellum, Subacute sclerosing panencephalitis, Syncope, syphilis of the cardiovascular system, systemic anaphalaxis, systemic inflammatory response syndrome, systemic onset juvenile rheumatoid arthritis, T-cell or FAB ALL, Telangiectasia, thromboangitis obliterans, thrombocytopenia, toxicity, transplants, trauma/hemorrhage, type m hypersensitivity reactions, type IV hypersensitivity, unstable angina, uremia, urosepsis, urticaria, valvular heart diseases, varicose veins, vasculitis, venous diseases, venous thrombosis, ventricular fibrillation, viral and fungal infections, vital encephalitis/aseptic meningitis, vital-associated hemaphagocytic syndrome, Wernicke- Korsakoff syndrome, Wilson's disease, xenograft rejection of any organ or tissue.
In another aspect the invention provides a method of treating a patient suffering from a disorder in which human IL-12 and/or human IL-23 is detrimental comprising the step of administering any one of the binding proteins disclosed above before, concurrent, or after the administration of a second agent, as discussed above. In a preferred embodiment the second agent is selected from the group consisting of budenoside, epidermal growth factor, corticosteroids, cyclosporin, sulfasalazine, aminosalicylates, 6-mercaptopurine, azathioprine, metronidazole, lipoxygenase inhibitors, mesalamine, olsalazine, balsalazide, antioxidants, thromboxane inhibitors, IL-I receptor antagonists, anti-IL-lβ monoclonal antibodies, anti-IL-6 monoclonal antibodies, growth factors, elastase inhibitors, pyridinyl-imidazole compounds, antibodies or agonists of TNF, LT, IL-I, IL-2, IL-6, IL-7, IL-8, IL-15, IL-16, IL-18, EMAP-II, GM-CSF, FGF, and PDGF, antibodies of CD2, CD3, CD4, CD8, CD25, CD28, CD30, CD40, CD45, CD69, CD90 or their ligands, methotrexate, cyclosporin, FK506, rapamycin, mycophenolate mofetil, leflunomide, NSAIDs, ibuprofen, corticosteroids, prednisolone, phosphodiesterase inhibitors, adenβosine agonists, antithrombotic agents, complement inhibitors, adrenergic agents, IRAK, NIK, IKK, p38, MAP kinase inhibitors, IL-I β converting enzyme inhibitors, TNFαconverting enzyme inhibitors, T-cell signalling inhibitors, metalloproteinase inhibitors, sulfasalazine, azathioprine, 6-mercaptopurines, angiotensin converting enzyme inhibitors, soluble cytokine receptors, soluble p55 TNF receptor, soluble p75 TNF receptor, sDL- IRI, sIL-lRπ, SIL-6R, antiinflammatory cytokines, IL-4, IL-10, IL-Il, DL-13 and TGFβ. In a preferred embodiment the pharmaceutical compositions disclosed above are administered to the subject by at least one mode selected from parenteral, subcutaneous, intramuscular, intravenous, intrarticular, intrabronchial, intraabdominal, intracapsular, intracartilaginous, intracavitary, intracelial, intracerebellar, intracerebroventricular, intracolic, intracervical, intragastric, intrahepatic, intramyocardial, intraosteal, intrapelvic, intrapericardiac, intraperitoneal, intrapleural, intraprostatic, intrapulmonary, intrarectal, intrarenal, intraretinal, intraspinal, intrasynovial, intrathoracic, intrauterine, intravesical, bolus, vaginal, rectal, buccal, sublingual, intranasal, and transdermal.
One aspect of the invention provides at least one IL- 12 anti-idiotype antibody to at least one IL- 12 binding protein of the present invention. The anti-idiotype antibody includes any protein or peptide containing molecule that comprises at least a portion of an immunoglobulin molecule such as, but not limited to, at least one complementarily determining region (CDR) of a heavy or light chain or a ligand binding portion thereof, a heavy chain or light chain variable region, a heavy chain or light chain constant region, a framework region, or; any portion thereof, that can be incorporated into a binding protein of the present invention.
Detailed Description of the Invention
This invention pertains to IL-12p40 binding proteins, particularly anti-IL-12p40 antibodies, or antigen-binding portions thereof, that bind IL-12p40. Various aspects of the invention relate to antibodies and antibody fragments, and pharmaceutical compositions thereof, as well as nucleic acids, recombinant expression vectors and host cells for making such antibodies and fragments. Methods of using the antibodies of the invention to detect human IL- 12p40, human IL-12 and human IL-23; to inhibit human DL-12 and/or human IL-23 activity, either in vitro or in vivo; and to regulate gene expression are also encompassed by the invention.
Unless otherwise defined herein, scientific and technical terms used in connection with the present invention shall have the meanings that are commonly understood by those of ordinary skill in the art. The meaning and scope of the terms should be clear, however, in the event of any latent ambiguiy, definitions provided herein take precedent over any dictionary or extrinsic definition. Further, unless otherwise required by context, singular terms shall include pluralities and plural terms shall include the singular. In this application, the use of "or" means "and/or" unless stated otherwise. Furthermore, the use of the term "including", as well as other forms, such as "includes" and "included", is not limiting. Also, terms such as "element" or "component" encompass both elements and components comprising one unit and elements and components that comprise more than one subunit unless specifically stated otherwise.
Generally, nomenclatures used in connection with, and techniques of, cell and tissue culture, molecular biology, immunology, microbiology, genetics and protein and nucleic acid chemistry and hybridization described herein are those well known and commonly used in the art. The methods and techniques of the present invention are generally performed according to conventional methods well known in the art and as described in various general and more specific references that are cited and discussed throughout the present specification unless otherwise indicated. Enzymatic reactions and purification techniques are performed according to manufacturer's specifications, as commonly accomplished in the art or as described herein. The nomenclatures used in connection with, and the laboratory procedures and techniques of, analytical chemistry, synthetic organic chemistry, and medicinal and pharmaceutical chemistry described herein are those well known and commonly used in the art. Standard techniques are used for chemical syntheses, chemical analyses, pharmaceutical preparation, formulation, and delivery, and treatment of patients.
That the present invention may be more readily understood, select terms are defined below.
The term "Polypeptide" as used herein, refers to any polymeric chain of amino acids. The terms "peptide" and "protein" are used interchangeably with the term polypeptide and also refer to a polymeric chain of amino acids. The term "polypeptide" encompasses native or artificial proteins, protein fragments and polypeptide analogs of a protein sequence. A polypeptide may be monomeric or polymeric.
The term "isolated protein" or "isolated polypeptide" is a protein or polypeptide that by virtue of its origin or source of derivation is not associated with naturally associated components that accompany it in its native state; is substantially free of other proteins from the same species; is expressed by a cell from a different species; or does not occur in nature. Thus, a polypeptide that is chemically synthesized or synthesized in a cellular system different from the cell from which it naturally originates will be "isolated" from its naturally associated components. A protein may also be rendered substantially free of naturally associated components by isolation, using protein purification techniques well known in the art. The term "recovering" as used herein, refers to the process of rendering a chemical species such as a polypeptide substantially free of naturally associated components by isolation, e.g., using protein purification techniques well known in the art.
The term "human IL-12" (abbreviated herein as hIL-12, or IL-12), as used herein, includes a human cytokine that is secreted primarily by antigen presenting cells such as monocytes macrophages and dendritic cells. The term includes a heterodimeric protein comprising a 35 kD subunit (p35) and a 40 kD subunit (p40) which are both linked together with a disulfide bridge. The heterodimeric protein is referred to as a "p70 protein". The structure of human IL-12 is described further in, for example, Kobayashi, et al. (1989) J. Exp Med. 170:827- 845; Seder, et al. (1993) Proc. Natl. Acad. Sci. 90:10188-10192; Ling, et al. (1995) J. Exp Med. 154: 116-127; Podlaski, et al. (1992) Arch. Biochem. Biophys. 294:230-237. The term human IL- 12 is intended to include recombinant human IL-12 (rh IL-12), which can be prepared by standard recombinant expression methods.
The term "human IL-23" (abbreviated herein as hIL-23, or IL-23), as used herein, includes a heterodimeric human cytokine belonging to a family of five such heterodimeric cytokines including IL-12 and DL-27 (Trinchieri et al., (2003) Immunity 19:641-644). The term includes a heterodimeric protein comprising a 19 kD subunit (pi 9) and a 40 kD subunit (p40) which are both linked together with a disulfide bridge. The term human BL-23 is intended to include recombinant human IL-23 (rh IL-23), which can be prepared by standard recombinant expression methods.
The term "IL-12p40", identicial to "IL-23p40", and also referred to simply as "p40", as used herein, includes the 40 kD subunit of the human cytokine IL-12 (p40) and the 40 kD subunit of the human cytokine IL-23. Table 1 shows the amino acid sequence of IL-12p40, SEQ ID No. 1, which is known in the art.
Table 1: Sequence of p40 subunit of IL-12 and IL-23
Figure imgf000021_0001
"Biological activity " as used herein, refers to all inherent biological properties of the cytokine. Biological properties of IL-12 include but are not limited to binding IL-12 receptor; induction of interferon-garnma (IFN-γ ) secretion and regulation of balance between antigen- specific T helper type 1 (ThI) and type 2 (Th2) lymphocytes. Biological properties of IL-23 include but are not limited to binding IL-23 receptor, inducing IFN-γ production, ThI cell differentiation and activating the antigen-presenting functions of dendritic cells, and selectively inducing proliferation of memory T cells.
The terms "specific binding" or "specifically binding", as used herein, in reference to the interaction of an antibody, a protein, or a peptide with a second chemical species, mean that the interaction is dependent upon the presence of a particular structure (e.g., an antigenic determinant or epitope) on the chemical species; for example, an antibody recognizes and binds to a specific protein structure rather than to proteins generally. If an antibody is specific for epitope "A", the presence of a molecule containing epitope A (or free, unlabeled A), in a reaction containing labeled "A" and the antibody, will reduce the amount of labeled A bound to the antibody.
The term "antibody", as used herein, broadly refers to any immunoglobulin (Ig) molecule comprised of four polypeptide chains, two heavy (H) chains and two light (L) chains, or any functional fragment, mutant, variant, or derivation thereof, which retains the essential epitope binding features of an Ig molecule. Such mutant, variant, or derivative anitbody formats are known in the art. Nonlimiting embodiments of which are discussed below.
In a full-length antibody, each heavy chain is comprised of a heavy chain variable region (abbreviated herein as HCVR or VH) and a heavy chain constant region. The heavy chain constant region is comprised of three domains, CHl, CH2 and CH3. Each light chain is comprised of a light chain variable region (abbreviated herein as LCVR or VL) and a light chain constant region. The light chain constant region is comprised of one domain, CL. The VH and VL regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDR), interspersed with regions that are more conserved, termed framework regions (FR). Each VH and VL is composed of three CDRs and four FRs, arranged from amino-terminus to carboxy-terminus in the following order: FRl, CDRl, FR2, CDR2, FR3, CDR3, FR4. Immunoglobulin molecules can be of any type (e.g., IgG, IgE, IgM, IgD, IgA and IgY), class (e.g., IgG 1, IgG2, IgG 3, IgG4, IgAl and IgA2) or subclass.
The term "antigen-binding portion" of an antibody (or simply "antibody portion"), as used herein, refers to one or more fragments of an antibody that retain the ability to specifically bind to an antigen {e.g., bJDL-12). It has been shown that the antigen-binding function of an antibody can be performed by fragments of a full-length antibody. Such antibody embodiments may also be bispecific, dual specific, or multi-specific formats; specifically binding to two or more different antigens. Examples of binding fragments encompassed within the term "antigen- binding portion" of an antibody include (i) a Fab fragment, a monovalent fragment consisting of the VL, VH, CL and CHl domains; (ii) a F(ab')2 fragment, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) a Fd fragment consisting of the VH and CHl domains; (iv) a Fv fragment consisting of the VL and VH domains of a single arm of an antibody, (v) a dAb fragment (Ward et al, (1989) Nature 341:544-546, Winter et al., PCT publication WO 90/05144 Al herein incorporated by reference), which comprises a single variable domain; and (vi) an isolated complementarity determining region (CDR). Furthermore, although the two domains of the Fv fragment, VL and VH, are coded for by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the VL and VH regions pair to form monovalent molecules (known as single chain Fv (scFv); see e.g., Bird et al. (1988) Science 242:423-426; and Huston et al. (1988) Proc. Natl. Acad. Sd. USA 85:5879-5883). Such single chain antibodies are also intended to be encompassed within the term "antigen-binding portion" of an antibody. Other forms of single chain antibodies, such as diabodies are also encompassed. Diabodies are bivalent, bispecific antibodies in which VH and VL domains are expressed on a single polypeptide chain, but using a linker that is too short to allow for pairing between the two domains on the same chain, thereby forcing the domains to pair with complementary domains of another chain and creating two antigen binding sites (see e.g., Holliger, P., et al. (1993) Proc. Natl. Acad. Sci. USA 90:6444-6448; Poljak, R. J., et al. (1994) Structure 2:1121-1123). Such antibody binding portions are known in the art (Kontermann and Dubel eds., Antibody Engineering (2001) Springer-Verlag. New York. 790 pp. (ISBN 3-540-41354-5).
The term "antibody construct" as used herein refers to a polypeptide comprising one or more the antigen binding portions of the invention linked to a linker polypeptide or an immunoglobulin constant domain. Linker polypeptides comprise two or more amino acid residues joined by peptide bonds and are used to link one or more antigen binding portions. Such linker polypeptides are well known in the art (see e.g., Holliger, P., et al. (1993) Proc. Natl. Acad. ScL USA 90:6444-6448; Poljak, RJ., et al. (1994) Structure 2:1121-1123). An immunoglobulin constant domain refers to a heavy or light chain constant domain. Human IgG heavy chain and light chain constant domain amino acid sequences are known in the art and represented in Table 2. Table 2: Sequence of human IgG heavy chain constant domain and light chain constant domain
Figure imgf000024_0001
Still further, an antibody or antigen-binding portion thereof may be part of a larger immunoadhesion molecules, formed by covalent or noncovalent association of the antibody or antibody portion with one or more other proteins or peptides. Examples of such immunoadhesion molecules include use of the streptavidin core region to make a tetrameric scFv molecule (Kipriyanov, S.M., et al. (1995) Human Antibodies and Hybridomas 6:93-101) and use of a cysteine residue, a marker peptide and a C-terminal polyhistidine tag to make bivalent and biotinylated scFv molecules (Kipriyanov, S.M., et al. (1994) MoI. Immunol. 31:1047-1058). Antibody portions, such as Fab and F(ab')2 fragments, can be prepared from whole antibodies using conventional techniques, such as papain or pepsin digestion, respectively, of whole antibodies. Moreover, antibodies, antibody portions and immunoadhesion molecules can be obtained using standard recombinant DNA techniques, as described herein.
An "isolated antibody", as used herein, is intended to refer to an antibody that is substantially free of other antibodies having different antigenic specificities {e.g., an isolated antibody that specifically binds hIL-12 is substantially free of antibodies that specificallybind antigens other than hIL-12). An isolated antibody that specifically binds hIL-12 may, however, have cross-reactivity to other antigens, such as IL-12 molecules from other species. Moreover, an isolated antibody may be substantially free of other cellular material and/or chemicals.
The term "human antibody", as used herein, is intended to include antibodies having variable and constant regions derived from human germline immunoglobulin sequences. The human antibodies of the invention may include amino acid residues not encoded by human germline immunoglobulin sequences (e.g., mutations introduced by random or site-specific mutagenesis in vitro or by somatic mutation in vivo), for example in the CDRs and in particular CDR3. However, the term "human antibody", as used herein, is not intended to include antibodies in which CDR sequences derived from the germline of another mammalian species, such as a mouse, have been grafted onto human framework sequences.
The term "recombinant human antibody", as used herein, is intended to include all human antibodies that are prepared, expressed, created or isolated by recombinant means, such as antibodies expressed using a recombinant expression vector transfected into a host cell (described further in Section II C, below), antibodies isolated from a recombinant, combinatorial human antibody library (HoogenboomKR., (1997) TIB Tech. 15:62-70; Azzazy H., and Highsmith W.E., (2002) Clin. Biochem. 35:425-445; Gavilondo J.V., and Larrick J.W. (2002) BioTechniques 29:128-145; HoogenboomH., and Chames P. (2000) Immunology Today 21:371- 378 ), antibodies isolated from an animal (e.g., a mouse) that is transgenic for human immunoglobulin genes (see e.g., Taylor, L. D., et al. (1992) Nucl. Acids Res. 20:6287-6295; Kellermann S-A., and Green L.L. (2002) Current Opinion in Biotechnology 13:593-597; Little M. et al (2000) Immunology Today 21:364-370) or antibodies prepared, expressed, created or isolated by any other means that involves splicing of human immunoglobulin gene sequences to other DNA sequences. Such recombinant human antibodies have variable and constant regions derived from human germline immunoglobulin sequences. In certain embodiments, however, such recombinant human antibodies are subjected to in vitro mutagenesis (or, when an animal transgenic for human Ig sequences is used, in vivo somatic mutagenesis) and thus the amino acid sequences of the VH and VL regions of the recombinant antibodies are sequences that, while derived from and related to human germline VH and VL sequences, may not naturally exist within the human antibody germline repertoire in vivo.
The term "chimeric antibody" refers to antibodies which comprise heavy and light chain variable region sequences from one species and constant region sequences from another species, such as antibodies having murine heavy and light chain variable regions linked to human constant regions.
The term "CDR-grafted antibody" refers to antibodies which comprise heavy and light chain variable region sequences from one species but in which the sequences of one or more of the CDR regions of VH and/or VL are replaced with CDR sequences of another species, such as antibodies having murine heavy and light chain variable regions in which one or more of the murine CDRs (e.g., CDR3) has been replaced with human CDR sequences.
The term "humanized antibody" refers to antibodies which comprise heavy and light chain variable region sequences from a non-human species (e.g., a mouse) but in which at least a portion of the VH and/or VL sequence has been altered to be more "human-like", Le., more similar to human germline variable sequences. One type of humanized antibody is a CDR-grafted antibody, in which human CDR sequences are introduced into non-human VH and VL sequences to replace the corresponding nonhuman CDR sequences.
The terms "Kabat numbering", "Kabat definitions and "Kabat labeling" are used interchangeably herein. These terms, which are recognized in the art, refer to a system of numbering amino acid residues which are more variable (Le. hypervariable) than other amino acid residues in the heavy and light chain variable regions of an antibody, or an antigen binding portion thereof (Kabat et αl. (1971) Ann. NYAcαd, ScL 190:382-391 and , Kabat, E.A., et αl. (1991) Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No. 91-3242). For the heavy chain variable region, the hypervariable region ranges from amino acid positions 31 to 35 for CDRl, amino acid positions 50 to 65 for CDR2, and amino acid positions 95 to 102 for CDR3. For the light chain variable region, the hypervariable region ranges from amino acid positions 24 to 34 for CDRl, amino acid positions 50 to 56 for CDR2, and amino acid positions 89 to 97 for CDR3.
As used herein, the terms "acceptor" and "acceptor antibody" refer to the antibody or nucleic acid sequence providing or encoding at least 80%, at least 85%, at least 90%, at least 95%, at least 98% or 100% of the amino acid sequences of one or more of the framework regions. In some embodiments, the term "acceptor" refers to the antibody amino acid or nucleic acid sequence providing or encoding the constant region(s). In yet another embodiment, the term "acceptor" refers to the antibody amino acid or nucleic acid sequence providing or encoding one or more of the framework regions and the constant region(s). In a specific embodiment, the term "acceptor" refers to a human antibody amino acid or nucleic acid sequence that provides or encodes at least 80%, preferably, at least 85%, at least 90%, at least 95%, at least 98%, or 100% of the amino acid sequences of one or more of the framework regions. In accordance with this embodiment, an acceptor may contain at least 1, at least 2, at least 3, least 4, at least 5, or at least 10 amino acid residues that does (do) not occur at one or more specific positions of a human antibody. An acceptor framework region and/or acceptor constant region(s) may be, e.g., derived or obtained from a germline antibody gene, a mature antibody gene, a functional antibody (e.g., antibodies well-known in the art, antibodies in development, or antibodies commercially available). As used herein, the term "CDR" refers to the complementarity determining region within antibody variable sequences. There are three CDRs in each of the variable regions of the heavy chain and the light chain, which are designated CDRl, CDR2 and CDR3, for each of the variable regions. The term "CDR set" as used herein refers to a group of three CDRs that occur in a single variable region capable of binding the antigen. The exact boundaries of these CDRs have been defined differently according to different systems. The system described by Kabat (Kabat et al., Sequences of Proteins of Immunological Interest (National Institutes of Health, Bethesda, Md. (1987) and (1991)) not only provides an unambiguous residue numbering system applicable to any variable region of an antibody, but also provides precise residue boundaries defining the three CDRs. These CDRs may be referred to as Kabat CDRs. Chothia and coworkers (Chothia &Lesk, J. MoI. Biol. 196:901-917 (1987) and Chothia et al., Nature 342:877-883 (1989)) found that certain sub- portions within Kabat CDRs adopt nearly identical peptide backbone conformations, despite having great diversity at the level of amino acid sequence. These sub- portions were designated as Ll, L2 and L3 or Hl, H2 and H3 where the "L" and the "H" designates the light chain and the heavy chains regions, respectively. These regions may be referred to as Chothia CDRs, which have boundaries that overlap with Kabat CDRs. Other boundaries defining CDRs overlapping with the Kabat CDRs have been described by Padlan (FASEB J. 9:133-139 (1995)) and MacCallum (J MoI Biol 262(5):732-45 (1996)). Still other CDR boundary definitions may not strictly follow one of the above systems, but will nonetheless overlap with the Kabat CDRs, although they may be shortened or lengthened in light of prediction or experimental findings that particular residues or groups of residues or even entire CDRs do not significantly impact antigen binding. The methods used herein may utilize CDRs defined according to any of these systems, although preferred embodiments use Kabat or Chothia defined CDRs.
As used herein, the term "canonical" residue refers to a residue in a CDR or framework that defines a particular canonical CDR structure as defined by Chothia et al. (J. MoI. Biol. 196:901-907 (1987); Chothia et al., J. MoI. Biol. 227:799 (1992), both are incorporated herein by reference). According to Chothia et al., critical portions of the CDRs of many antibodies have nearly identical peptide backbone confirmations despite great diversity at the level of amino acid sequence. Each canonical structure specifies primarily a set of peptide backbone torsion angles for a contiguous segment of amino acid residues forming a loop.
As used herein, the terms "donor" and "donor antibody" refer to an antibody providing one or more CDRs. In a preferred embodiment, the donor antibody is an antibody from a species different from the antibody from which the framework regions are obtained or derived. In the context of a humanized antibody, the term "donor antibody" refers to a non-human antibody providing one or more CDRs. As used herein, the term "framework" or "framework sequence" refers to the remaining sequences of a variable region minus the CDRs. Because the exact definition of a CDR sequence can be determined by different systems, the meaning of a framework sequence is subject to correspondingly different interpretations. The six CDRs (CDR-Ll, -L2, and -L3 of light chain and CDR-Hl, -H2, and -H3 of heavy chain) also divide the framework regions on the light chain and the heavy chain into four sub-regions (FRl, FR2, FR3 and FR4) on each chain, in which CDRl is positioned between FRl and FR2, CDR2 between FR2 and FR3, and CDR3 between FR3 and FR4. Without specifying the particular sub-regions as FRl, FR2, FR3 or FR4, a framework region, as referred by others, represents the combined FR's within the variable region of a single, naturally occurring immunoglobulin chain. As used herein, a FR represents one of the four sub- regions, and FRs represents two or more of the four sub- regions constituting a framework region.
Human heavy chain and light chain acceptor sequences are known in the art. In one embodiment of the invention the human heavy chain and light chain acceptor sequences are selected from the sequences described in Table 3 and Table 4.
TABLE 3: HEAVY CHAIN ACCEPTOR SEQUENCES
Figure imgf000028_0001
TABLE 4: LIGHT CHAIN ACCEPTOR SEQUENCES
Figure imgf000029_0001
As used herein, the term "germline antibody gene" or "gene fragment" refers to an immunoglobulin sequence encoded by non- lymphoid cells that have not undergone the maturation process that leads to genetic rearrangement and mutation for expression of a particular immunoglobulin. (See, e.g., Shapiro et al., Crit. Rev. Immunol. 22(3): 183-200 (2002); Marchalonis et al., Adv Exp Med Biol. 484:13-30 (2001)). One of the advantages provided by various embodiments of the present invention stems from the recognition that germline antibody genes are more likely than mature antibody genes to conserve essential amino acid sequence structures characteristic of individuals in the species, hence less likely to be recognized as from a foreign source when used therapeutically in that species.
As used herein, the term "key" residues refer to certain residues within the variable region that have more impact on the binding specificity and/or affinity of an antibody, in particular a humanized antibody. A key residue includes, but is not limited to, one or more of the following: a residue that is adjacent to a CDR, a potential glycosylation site (can be either N- or O-glycosylation site), a rare residue, a residue capable of interacting with the antigen, a residue capable of interacting with a CDR, a canonical residue, a contact residue between heavy chain variable region and light chain variable region, a residue within the Vernier zone, and a residue in the region that overlaps between the Chothia definition of a variable heavy chain CDRl and the Kabat definition of the first heavy chain framework.
As used herein, the term "humanized antibody" is an antibody or a variant, derivative, analog or fragment thereof which immunospecifϊcally binds to an antigen of interest and which comprises a framework (FR) region having substantially the amino acid sequence of a human antibody and a complementary determining region (CDR) having substantially the amino acid sequence of a non-human antibody. As used herein, the term "substantially" in the context of a CDR refers to a CDR having an amino acid sequence at least 80%, preferably at least 85%, at least 90%, at least 95%, at least 98% or at least 99% identical to the amino acid sequence of a non-human antibody CDR. A humanized antibody comprises substantially all of at least one, and typically two, variable domains (Fab, Fab', F(ab') 2, FabC, Fv) in which all or substantially all of the CDR regions correspond to those of a non-human immunoglobulin (i.e., donor antibody) and all or substantially all of the framework regions are those of a human immunoglobulin consensus sequence. Preferably, a humanized antibody also comprises at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin. In some embodiments, a humanized antibody contains both the light chain as well as at least the variable domain of a heavy chain. The antibody also may include the CHl, hinge, CH2, CH3, and CH4 regions of the heavy chain. In some embodiments, a humanized antibody only contains a humanized light chain. In some embodiments, a humanized antibody only contains a humanized heavy chain. In specific embodiments, a humanized antibody only contains a humanized variable domain of a light chain and/or humanized heavy chain.
The humanized antibody can be selected from any class of immunoglobulins, including IgM, IgG, IgD, IgA and IgE, and any isotype, including without limitation IgG 1, IgG2, IgG3 and IgG4. The humanized antibody may comprise sequences from more than one class or isotype, and particular constant domains may be selected to optimize desired effector functions using techniques well- known in the art.
The framework and CDR regions of a humanized antibody need not correspond precisely to the parental sequences, e.g., the donor antibody CDR or the consensus framework may be mutagenized by substitution, insertion and/or deletion of at least one amino acid residue so that the CDR or framework residue at that site does not correspond to either the donor antibody or the consensus framework. In a preferred embodiment, such mutations, however, will not be extensive. Usually, at least 80%, preferably at least 85%, more preferably at least 90%, and most preferably at least 95% of the humanized antibody residues will correspond to those of the parental FR and CDR sequences. As used herein, the term "consensus framework" refers to the framework region in the consensus immunoglobulin sequence. As used herein, the term "consensus immunoglobulin sequence" refers to the sequence formed from the most frequently occurring amino acids (or nucleotides) in a family of related immunoglobulin sequences (See e.g., Winnaker, From Genes to Clones (Verlagsgesellschaft, Weinheim, Germany 1987). In a family of immunoglobulins, each position in the consensus sequence is occupied by the amino acid occurring most frequently at that position in the family. If two amino acids occur equally frequently, either can be included in the consensus sequence. As used herein, "Vernier" zone refers to a subset of framework residues that may adjust CDR structure and fine-tune the fit to antigen as described by Foote and Winter (1992, J. MoI. Biol. 224:487-499, which is incorporated herein by reference). Vernier zone residues form a layer underlying the CDRs and may impact on the structure of CDRs and the affinity of the antibody.
As used herein, the term "neutralizing" refers to neutralization of biological activity of a cytokine when a binding protein specifically binds the cytokine. Preferably a neutralizing binding protein is a neutralizing antibody whose binding to hIL-12 and/or bJL-23 results in inhibition of a biological activity of hIL-12 and/or hIL-23. Preferably the neutralizing binding protein binds hIL-12 and/or bJL-23 and reduces a biologically activity of IL-12 and/or hIL-23 by at least about 20%, 40%, 60%, 80%, 85% or more. Inhibition of a biological activity of hIL-12 and/or hIL-23 by a neutralizing binding protein can be assessed by measuring one or more indicators of hIL-12 and/or hIL-23 biological activity well known in the art. For example inhibition of human phytohemagglutinin blast proliferation in a PHA blast Interferon-gamma Induction Assay (see Example l.l.C) or inhibition of receptor binding in a human IL-12 receptor binding assay, (also see Salfeld et al., PCT publication No.WO 00/56772 Al).
The term "activity" includes activities such as the binding specificity/affinity of an antibody for an antigen, for example, an anti-hlL-12 antibody that binds to an EL-12 antigen and/or the neutralizing potency of an antibody, for example, an anti-hIL-12 antibody whose binding to hIL-12 inhibits the biological activity of hIL-12, e.g. inhibition of PHA blast proliferation or inhibition of receptor binding in a human IL-12 receptor binding assay, or or PHA blast Interferon-gamma Induction Assay (see Example l.l.C).
The term "epitope" includes any polypeptide determinant capable of specific binding to an immunoglobulin or T-cell receptor. In certain embodiments, epitope determinants include chemically active surface groupings of molecules such as amino acids, sugar side chains, phosphoryl, or sulfonyl, and, in certain embodiments, may have specific three dimensional structural characteristics, and/or specific charge characteristics. An epitope is a region of an antigen that is bound by an antibody. In certain embodiments, an antibody is said to specifically bind an antigen when it preferentially recognizes its target antigen in a complex mixture of proteins and/or macromolecules.
The term "surface plasmon resonance", as used herein, refers to an optical phenomenon that allows for the analysis of real-time biospecific interactions by detection of alterations in protein concentrations within a biosensor matrix, for example using the BIAcore system (Pharmacia Biosensor AB, Uppsala, Sweden and Piscataway, NJ). For further descriptions, see Jδnsson, U., etal. (1993) Ann. Biol. Clin. 51:19-26; Jδnsson, U., et al (1991) Biotechniques 11:620-627; Johnsson, B., et al (1995) J. MoI Recognit. 8:125-131; and Johnnson, B., et al. (1991) Anal. Biochem. 198:268-277. The term "K0n", as used herein, is intended to refer to the on rate constant for association of an antibody to the antigen to form the antibody/antigen complex as is known in the art. The term "Koff", as used herein, is intended to refer to the off rate constant for dissociation of an antibody from the antibody/antigen complex as is known in the art.
The term "Kj", as used herein, is intended to refer to the dissociation constant of a particular antibody-antigen interaction as is known in the art.
The term "labeled binding protein" as used herein, refers to a protein with a label incorporated that provides for the identification of the binding protein. Preferably, the label is a detectable marker, e.g., incorporation of a radiolabeled amino acid or attachment to a polypeptide of biotinyl moieties that can be detected by marked avidin (e.g., streptavidin containing a fluorescent marker or enzymatic activity that can be detected by optical or colorimetric methods). Examples of labels for polypeptides include, but are not limited to, the following: radioisotopes or radionuclides (e.g., 3H 14C, 35S, 90Y, 99Tc, 111In, 1251, 1311, 177Lu, 166Ho, or 153Sm); fluorescent labels (e.g., FTTC, rhodamine, lanthanide phosphors), enzymatic labels (e.g., horseradish peroxidase, luciferase, alkaline phosphatase); chemiluminescent markers; biotinyl groups; predetermined polypeptide epitopes recognized by a secondary reporter (e.g., leucine zipper pair sequences, binding sites for secondary antibodies, metal binding domains, epitope tags); and magnetic agents, such as gadolinium chelates.
The term "antibody conjugate" refers to a binding protein, such as an antibody, chemically linked to a second chemical moiety, such as a therapeutic or cytotoxic agent. The term "agent" is used herein to denote a chemical compound, a mixture of chemical compounds, a biological macromolecule, or an extract made from biological materials. Preferably the therapeutic or cytotoxic agents include, but are not limited to, pertussis toxin, taxol, cytochalasin B, gramicidin D, ethidium bromide, emetine, mitomycin, etoposide, tenoposide, vincristine, vinblastine, colchicin, doxorubicin, daunorubicin, dihydroxy anthracin dione, mitoxantrone, mithramycin, actinomycin D, 1-dehydrotestosterone, glucocorticoids, procaine, tetracaine, lidocaine, propranolol, and puromycin and analogs or homologs thereof.
The terms "crystal", and "crystallized" as used herein, refer to an antibody, or antigen binding portion thereof, that exists in the form of a crystal. Crystals are one form of the solid state of matter, which is distinct from other forms such as the amorphous solid state or the liquid crystalline state. Crystals are composed of regular, repeating, three-dimensional arrays of atoms, ions, molecules (e.g., proteins such as antibodies), or molecular assemblies (e.g., antigen/antibody complexes). These three-dimensional arrays are arranged according to specific mathematical relationships that are well-understood in the field. The fundamental unit, or building block, that is repeated in a crystal is called the asymmetric unit. Repetition of the asymmetric unit in an arrangement that conforms to a given, well-defined crystallographic symmetry provides the "unit cell" of the crystal. Repetition of the unit cell by regular translations in all three dimensions provides the crystal. See Giege, R. and Ducruix, A. Barrett, Crystallization of Nucleic Acids and Proteins, a Practical Approach, 2nd ea., pp. 20 1-16, Oxford University Press, New York, New York, (1999)."
The term "polynucleotide" as referred to herein means a polymeric form of two or more nucleotides, either ribonucleotides or deoxvnucleotides or a modified form of either type of nucleotide. The term includes single and double stranded forms of DNA but preferably is double-stranded DNA.
The term "isolated polynucleotide" as used herein shall mean a polynucleotide (e.g., of genomic, cDNA, or synthetic origin, or some combination thereof) that, by virtue of its origin , the "isolated polynucleotide": is not associated with all or a portion of a polynucleotide with which the "isolated polynucleotide" is found in nature; is operably linked to a polynucleotide that it is not linked to in nature; or does not occur in nature as part of a larger sequence.
The term "vector", as used herein, is intended to refer to a nucleic acid molecule capable of transporting another nucleic acid to which it has been linked. One type of vector is a "plasmid", which refers to a circular double stranded DNA loop into which additional DNA segments may be ligated. Another type of vector is a viral vector, wherein additional DNA segments may be ligated into the viral genome. Certain vectors are capable of autonomous replication in a host cell into which they are introduced (e.g., bacterial vectors having a bacterial origin of replication and episomal mammalian vectors). Other vectors (e.g., non-episomal mammalian vectors) can be integrated into the genome of a host cell upon introduction into the host cell, and thereby are replicated along with the host genome. Moreover, certain vectors are capable of directing the expression of genes to which they are operatively linked. Such vectors are referred to herein as "recombinant expression vectors" (or simply, "expression vectors"). In general, expression vectors of utility in recombinant DNA techniques are often in the form of plasmids. In the present specification, "plasmid" and "vector" may be used interchangeably as the plasmid is the most commonly used form of vector. However, the invention is intended to include such other forms of expression vectors, such as viral vectors (e.g., replication defective retroviruses, adenoviruses and adeno-associated viruses), which serve equivalent functions.
The term "operably linked" refers to a juxtaposition wherein the components described are in a relationship permitting them to function in their intended manner. A control sequence "operably linked" to a coding sequence is ligated in such a way that expression of the coding sequence is achieved under conditions compatible with the control sequences. "Operably linked" sequences include both expression control sequences that are contiguous with the gene of interest and expression control sequences that act in trans or at a distance to control the gene of interest. The term "expression control sequence" as used herein refers to polynucleotide sequences which are necessary to effect the expression and processing of coding sequences to which they are ligated. Expression control sequences include appropriate transcription initiation, termination, promoter and enhancer sequences; efficient RNA processing signals such as splicing and polyadenylation signals; sequences that stabilize cytoplasmic mRNA; sequences that enhance translation efficiency (i.e., Kozak consensus sequence); sequences that enhance protein stability; and when desired, sequences that enhance protein secretion. The nature of such control sequences differs depending upon the host organism; in prokaryotes, such control sequences generally include promoter, ribosomal binding site, and transcription termination sequence; in eukaryotes, generally, such control sequences include promoters and transcription termination sequence. The term "control sequences" is intended to include components whose presence is essential for expression and processing, and can also include additional components whose presence is advantageous, for example, leader sequences and fusion partner sequences.
'Transformation", as defined herein, refers to any process by which exogenous DNA enters a host cell. Transformation may occur under natural or artificial conditions using various methods well known in the art. Transformation may rely on any known method for the insertion of foreign nucleic acid sequences into a prokaryotic or eukaryotic host cell. The method is selected based on the host cell being transformed and may include, but is not limited to, viral infection, electroporation, lipofection, and particle bombardment. Such "transformed" cells include stably transformed cells in which the inserted DNA is capable of replication either as an autonomously replicating plasmid or as part of the host chromosome. They also include cells which transiently express the inserted DNA or RNA for limited periods of time.
The term "recombinant host cell" (or simply "host cell"), as used herein, is intended to refer to a cell into which exogenous DNA has been introduced. It should be understood that such terms are intended to refer not only to the particular subject cell, but, to the progeny of such a cell. Because certain modifications may occur in succeeding generations due to either mutation or environmental influences, such progeny may not, in fact, be identical to the parent cell, but are still included within the scope of the term "host cell" as used herein. Preferably host cells include prokaryotic and eukaryotic cells selected from any of the Kingdoms of life. Preferred eukaryotic cells include protist, fungal, plant and animal cells. Most preferably host cells include but are not limited to the prokaryotic cell line E.Coli; mammalian cell lines CHO, HEK 293 and COS; the insect cell line Sf9; and the fungal cell Saccharomyces cerevisiae.
Standard techniques may be used for recombinant DNA, oligonucleotide synthesis, and tissue culture and transformation (e.g., electroporation, lipofection). Enzymatic reactions and purification techniques may be performed according to manufacturer's specifications or as commonly accomplished in the art or as described herein. The foregoing techniques and procedures may be generally performed according to conventional methods well known in the art and as described in various general and more specific references that are cited and discussed throughout the present specification. See e.g., Sambrook et al. Molecular Cloning: A Laboratory Manual (2d ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (1989)), which is incorporated herein by reference for any purpose.
"Transgenic organism", as known in the art and as used herein, refers to an organism having cells that contain a transgene, wherein the transgene introduced into the organism (or an ancestor of the organism) expresses a polypeptide not naturally expressed in the organism. A "transgene" is a DNA construct, which is stably and operably integrated into the genome of a cell from which a transgenic organism develops, directing the expression of an encoded gene product in one or more cell types or tissues of the transgenic organism.
The term "regulate"and "modulate" are used interchangeably, and, as used herein, refers to a change or an alteration in the activity of a molecule of interest (e.g., the biological activity of hIL-12). Modulation may be an increase or a decrease in the magnitude of a certain activity or function of the molecule of interest. Exemplary activities and functions of a molecule include, but are not limited to, binding characteristics, enzymatic activity, cell receptor activation, and signal transduction.
Correspondingly, the term "modulator," as used herein, is a compound capable of changing or altering an activity or function of a molecule of interest (e.g., the biological activity of hIL-12). For example, a modulator may cause an increase or decrease in the magnitude of a certain activity or function of a molecule compared to the magnitude of the activity or function observed in the absence of the modulator. In certain embodiments, a modulator is an inhibitor, which decreases the magnitude of at least one activity or function of a molecule. Exemplary inhibitors include, but are not limited to, proteins, peptides, antibodies, peptibodies, carbohydrates or small organic molecules. Peptibodies are described, e.g., in WO01/83525.
The term "agonist", as used herein, refers to a modulator that, when contacted with a molecule of interest, causes an increase in the magnitude of a certain activity or function of the molecule compared to the magnitude of the activity or function observed in the absence of the agonist. Particular agonists of interest may include, but are not limited to, IL-12 polypeptides or polypeptides, nucleic acids, carbohydrates, or any other molecules that bind to hIL-12.
The term "antagonist" or "inhibitor", as used herein, refer to a modulator that, when contacted with a molecule of interest causes a decrease in the magnitude of a certain activity or function of the molecule compared to the magnitude of the activity or function observed in the absence of the antagonist. Particular antagonists of interest include those that block or modulate the biological or immunological activity of hIL-12 and/or hIL-23. Antagonists and inhibitors of ML-12 and/or ML-23 may include, but are not limited to, proteins, nucleic acids, carbohydrates, or any other molecules, which bind to hDL-12 and/or hIL-23.
As used herein, the term "effective amount" refers to the amount of a therapy which is sufficient to reduce or ameliorate the severity and/or duration of a disorder or one or more symptoms thereof, prevent the advancement of a disorder, cause regression of a disorder, prevent the recurrence, development, onset or progression of one or more symptoms associated with a disorder, detect a disorder, or enhance or improve the prophylactic or therapeutic effect(s) of another therapy (e.g. , prophylactic or therapeutic agent).
The term "sample", as used herein, is used in its broadest sense. A "biological sample", as used herein, includes, but is not limited to, any quantity of a substance from a living thing or formerly living thing. Such living things include, but are not limited to, humans, mice, rats, monkeys, dogs, rabbits and other animals. Such substances include, but are not limited to, blood, serum, urine, synovial fluid, cells, organs, tissues, bone marrow, lymph nodes and spleen.
I. Antibodies that Bind Human IL-12p40.
One aspect of the present invention provides isolated murine monoclonal antibodies, or antigen-binding portions thereof, that bind to IL-12p40 with high affinity, a slow off rate and high neutralizing capacity. A second aspect of the invention provides chimeric antibodies that bind IL-12p40. A third aspect of the invention provides CDR grafted antibodies, or antigen- binding portions thereof, that bind IL-12p40. A fourth aspect of the invention provides humanized antibodies, or antigen-binding portions thereof, that bind EL-12p40. Preferably, the antibodies, or portions thereof, are isolated antibodies. Preferably, the antibodies of the invention are neutralizing human anti-IL-12 and/or human anti-IL-23 antibodies.
A. Method of making anti IL-12p40 antibodies
Antibodies of the present invention may be made by any of a number of techniques known in the art.
1. Anti-IL-12 p40 monoclonal antibodies using Hybridoma technology
Monoclonal antibodies can be prepared using a wide variety of techniques known in the art including the use of hybridoma, recombinant, and phage display technologies, or a combination thereof. For example, monoclonal antibodies can be produced using hybridoma techniques including those known in the art and taught, for example, in Harlow et al. , Antibodies: A Laboratory Manual, (Cold Spring Harbor Laboratory Press, 2nd ed. 1988); Hammerling, et al., in: Monoclonal Antibodies and T-CeIl Hybridomas 563-681 (Elsevier, N. Y., 1981) (said references incorporated by reference in their entireties). The term "monoclonal antibody" as used herein is not limited to antibodies produced through hybridoma technology. The term "monoclonal antibody" refers to an antibody that is derived from a single clone, including any eukaryotic, prokaryotic, or phage clone, and not the method by which it is produced.
Methods for producing and screening for specific antibodies using hybridoma technology are routine and well known in the art. In one embodiment, the present invention provides methods of generating monoclonal antibodies as well as antibodies produced by the method comprising culturing a hybridoma cell secreting an antibody of the invention wherein, preferably, the hybridoma is generated by fusing splenocytes isolated from a mouse immunized with an antigen of the invention with myeloma cells and then screening the hybridomas resulting from the fusion for hybridoma clones that secrete an antibody able to bind a polypeptide of the invention ( See Example 1.2). Briefly, mice can be immunized with an IL-12 antigen. In a preferred embodiment, the IL-12 antigen is administered with a adjuvant to stimulate the immune response. Such adjuvants include complete or incomplete Freund's adjuvant, RIBI (muramyl dipeptides) or ISCOM (immunostimulating complexes). Such adjuvants may protect the polypeptide from rapid dispersal by sequestering it in a local deposit, or they may contain substances that stimulate the host to secrete factors that are chemotactic for macrophages and other components of the immune system. Preferably, if a polypeptide is being administered, the immunization schedule will involve two or more administrations of the polypeptide, spread out over several weeks.
After immunization of an animal with an IL-12 antigen, antibodies and/or antibody- producing cells may be obtained from the animal. An anti- IL-12 antibody-containing serum is obtained from the animal by bleeding or sacrificing the animal. The serum may be used as it is obtained from the animal, an immunoglobulin fraction may be obtained from the serum, or the anti- EL-12 antibodies may be purified from the serum. Serum or immunoglobulins obtained in this manner are polyclonal, thus having a heterogeneous array of properties.
Once an immune response is detected, e.g., antibodies specific for the antigen IL-12 are detected in the mouse serum, the mouse spleen is harvested and splenocytes isolated. The splenocytes are then fused by well-known techniques to any suitable myeloma cells, for example cells from cell line SP20 available from the ATCC. Hybridomas are selected and cloned by limited dilution. The hybridoma clones are then assayed by methods known in the art for cells that secrete antibodies capable of binding IL-12. Ascites fluid, which generally contains high levels of antibodies, can be generated by immunizing mice with positive hybridoma clones.
In another embodiment, antibody-producing immortalized hybridomas may be prepared from the immunized animal. After immunization, the animal is sacrificed and the splenic B cells are fused to immortalized myeloma cells as is well known in the art. See, e.g., Harlow and Lane, supra. In a preferred embodiment, the myeloma cells do not secrete immunoglobulin polypeptides (a non-secretory cell line). After fusion and antibiotic selection, the hybridomas are screened using IL-12, or a portion thereof, or a cell expressing BL-12. In a preferred embodiment, the initial screening is performed using an enzyme-linked immunoassay (ELISA) or a radioimmunoassay (RIA), preferably an ELISA. An example of ELISA screening is provided in WO 00/37504, herein incorporated by reference.
Anti- IL-12p40 antibody-producing hybridomas are selected, cloned and further screened for desirable characteristics, including robust hybridoma growth, high antibody production and desirable antibody characteristics, as discussed further below. Hybridomas may be cultured and expanded in vivo in syngeneic animals, in animals that lack an immune system, e.g., nude mice, or in cell culture in vitro. Methods of selecting, cloning and expanding hybridomas are well known to those of ordinary skill in the art.
In a preferred embodiment, the hybridomas are mouse hybridomas, as described above. In another preferred embodiment, the hybridomas are produced in a non-human, non-mouse species such as rats, sheep, pigs, goats, cattle or horses. In another embodiment, the hybridomas are human hybridomas, in which a human non-secretory myeloma is fused with a human cell expressing an anti-IL-12 antibody.
Antibody fragments that recognize specific epitopes may be generated by known techniques. For example, Fab and F(ab')2 fragments of the invention may be produced by proteolytic cleavage of immunoglobulin molecules, using enzymes such as papain (to produce Fab fragments) or pepsin (to produce F(ab')2 fragments). F(ab')2 fragments contain the variable region, the light chain constant region and the CHI domain of the heavy chain.
2. Anti-IL-12p40 monoclonal antibodies using SLAM
In another aspect of the invention, recombinant antibodies are generated from single, isolated lymphocytes using a procedure referred to in the art as the selected lymphocyte antibody method (SLAM), as described in U.S. Patent No. 5,627,052, PCT Publication WO 92/02551 and Babcock, J.S. et al. (1996) Proc. Natl. Acad. ScL USA 93:7843-7848. In this method, single cells secreting antibodies of interest, e.g., lymphocytes derived from any one of the immunized animals described in Section 1, are screened using an antigen-specific hemolytic plaque assay, wherein the antigen IL-12, a subunint of IL-12, or a fragment thereof, is coupled to sheep red blood cells using a linker, such as biotin, and used to identify single cells that secrete antibodies with specificity for IL-12. Following identification of antibody-secreting cells of interest, heavy- and light-chain variable region cDNAs are rescued from the cells by reverse transcriptase-PCR and these variable regions can then be expressed, in the context of appropriate immunoglobulin constant regions (e.g., human constant regions), in mammalian host cells, such as COS or CHO cells. The host cells transfected with the amplified immunoglobulin sequences, derived from in vivo selected lymphocytes, can then undergo further analysis and selection in vitro, for example by panning the transfected cells to isolate cells expressing antibodies to IL-12. The amplified immunoglobulin sequences further can be manipulated in vitro, such as by in vitro affinity maturation methods such as those described in PCT Publication WO 97/29131 and PCT Publication WO 00/56772.
3. Anti-IL-12p40 monoclonal antibodies using transgenic animals
In another embodiment of the instant invention, antibodies are produced by immunizing a non-human animal comprising some, or all, of the human immunoglobulin locus with an IL- 12 antigen. In a preferred embodiment, the non-human animal is a XENOMOUSE transgenic mouse, an engineered mouse strain that comprises large fragments of the human immunoglobulin loci and is deficient in mouse antibody production. See, e.g., Green et al. Nature Genetics 7:13-21 (1994) and United States Patents 5,916,771, 5,939,598, 5,985,615, 5,998,209, 6,075,181, 6,091,001, 6,114,598 and 6,130,364. See also WO 91/10741, published July 25,1991, WO 94/02602, published February 3, 1994, WO 96/34096 and WO 96/33735, both published October 31, 1996, WO 98/16654, published April 23, 1998, WO 98/24893, published June 11, 1998, WO 98/50433, published November 12, 1998, WO 99/45031, published September 10, 1999, WO 99/53049, published October 21, 1999, WO 0009560, published February 24, 2000 and WO 00/037504, published June 29, 2000. The XENOMOUSE transgenic mouse produces an adult-like human repertoire of fully human antibodies, and generates antigen-specific human Mabs. The XENOMOUSE transgenic mouse contains approximately 80% of the human antibody repertoire through introduction of megabase sized, germline configuration YAC fragments of the human heavy chain loci and x light chain loci. See Mendez et al., Nature Genetics 15:146-156 (1997), Green and Jakobovits J. Exp. Med. 188:483-495 (1998), the disclosures of which are hereby incorporated by reference.
4. Anti-IL-12 monoclonal antibodies using recombinant antibody libraries
In vitro methods also can be used to make the antibodies of the invention, wherein an antibody library is screened to identify an antibody having the desired binding specificity. Methods for such screening of recombinant antibody libraries are well known in the art and include methods described in, for example, Ladner et al. U.S. Patent No. 5,223,409; Kang et al. PCT Publication No. WO 92/18619; Dower et al. PCT Publication No. WO 91/17271; Winter et al. PCT Publication No. WO 92/20791; Markland et al. PCT Publication No. WO 92/15679; Breitling et al. PCT Publication No. WO 93/01288; McCafferty et al. PCT Publication No. WO 92/01047; Garrard et al. PCT Publication No. WO 92/09690; Fuchs et al. (1991) Bio/Technology 9: 1370-1372; Hay et al. (1992) Hum Antibod Hyhridomas 3:81-85; Huse et al. (1989) Science 246:1275-1281; McCafferty et al, Nature (1990) 348:552-554; Griffiths et al. (1993) EMBO J 12:725-734; Hawkins et al. (1992) JMoI Biol 226:889-896; Clackson et al. (1991) Nature 352:624-628; Gram et al. (1992) PNAS 89:3576-3580; Garrad et al. (1991) Bio/Technology 9:1373-1377; Hoogenboom et al. (1991) Nuc Acid Res 19:4133-4137; and Barbas et al. (1991) PNAS 88:7978-7982, US patent application publication 20030186374, and PCT Publication No. WO 97/29131, the contents of each of which are incorporated herein by reference.
The recombinant antibody library may be from a subject immunized with IL-12 or IL-23, or a portion of IL-12 or IL-23. Alternatively, the recombinant antibody library may be from a naϊve subject, i.e., one who has not been immunized with IL-12 or IL-23, such as a human antibody library from a human subject who has not been immunized with human IL-12 or ^L-Ti. Antibodies of the invention are selected by screening the recombinant antibody library with the peptide comprising human IL-12p40 to thereby select those antibodies that recognize IL-12p40. Methods for conducting such screening and selection are well known in the art, such as described in the references in the preceding paragraph. To select antibodies of the invention having particular binding affinities for hIL-12, such as those that dissociate from human IL-12 with a particular koff rate constant, the art-known method of surface plasmon resonance can be used to select antibodies having the desired koff rate constant. To select antibodies of the invention having a particular neutralizing activity for ML- 12, such as those with a particular an IC50, standard methods known in the art for assessing the inhibition of hIL-12 activity may be used.
In one aspect, the invention pertains to an isolated antibody, or an antigen-binding portion thereof, that binds human IL-12 and/or human IL-23. Preferably, the antibody is a neutralizing antibody. In various embodiments, the antibody is a recombinant antibody or a monoclonal antibody.
For example, the antibodies of the present invention can also be generated using various phage display methods known in the art. In phage display methods, functional antibody domains are displayed on the surface of phage particles which carry the polynucleotide sequences encoding them. In a particular, such phage can be utilized to display antigen-binding domains expressed from a repertoire or combinatorial antibody library (e. g., human or murine). Phage expressing an antigen binding domain that binds the antigen of interest can be selected or identified with antigen, e.g., using labeled antigen or antigen bound or captured to a solid surface or bead. Phage used in these methods are typically filamentous phage including fd and M13 binding domains expressed from phage with Fab, Fv or disulfide stabilized Fv antibody domains recombinantly fused to either the phage gene IH or gene VDI protein. Examples of phage display methods that can be used to make the antibodies of the present invention include those disclosed in Brinkman et al., J. Immunol. Methods 182:41-50 (1995); Ames et al., J. Immunol. Methods 184:177-186 (1995); Kettleborough et al., Eur. J. Immunol. 24:952-958 (1994); Persic et al., Gene 187 9-18 (1997); Burton et al., Advances in Immunology 57:191-280 (1994); PCT application No. PCT/GB91/01134; PCT publications WO 90/02809; WO 91/10737; WO 92/01047; WO 92/18619; WO 93/11236; WO 95/15982; WO 95/20401; and U.S. Pat. Nos. 5,698,426; 5,223,409; 5,403,484; 5,580,717; 5,427,908; 5,750,753; 5,821,047; 5,571,698; 5,427,908; 5,516,637; 5,780, 225; 5,658,727; 5,733,743 and 5,969,108; each of which is incorporated herein by reference in its entirety.
As described in the above references, after phage selection, the antibody coding regions from the phage can be isolated and used to generate whole antibodies including human antibodies or any other desired antigen binding fragment, and expressed in any desired host, including mammalian cells, insect cells, plant cells, yeast, and bacteria, e.g., as described in detail below. For example, techniques to recombinantly produce Fab, Fab' and F(ab')2 fragments can also be employed using methods known in the art such as those disclosed in PCT publication WO 92/22324; Mullinax et al., BioTechniques 12(6): 864-869 (1992); and Sawai et al., AJRI 34:26-34 (1995); and Better et al., Science 240:1041-1043 (1988) (said references incorporated by reference in their entireties). Examples of techniques which can be used to produce single-chain Fvs and antibodies include those described in U.S. Pat. 4,946,778 and 5,258, 498; Huston et al., Methods in Enzymology 203:46-88 (1991); Shu et al., PNAS 90:7995-7999 (1993); and Skerra et al., Science 240:1038-1040 (1988).
Alternative to screening of recombinant antibody libraries by phage display, other methodologies known in the art for screening large combinatorial libraries can be applied to the identification of dual specificity antibodies of the invention. One type of alternative expression system is one in which the recombinant antibody library is expressed as RNA-protein fusions, as described in PCT Publication No. WO 98/31700 by Szostak and Roberts, and in Roberts, R.W. and Szostak, J.W. (1997) Proc. Natl. Acad. Sci. USA 94:12297-12302. In this system, a covalent fusion is created between an mRNA and the peptide or protein that it encodes by in vitro translation of synthetic mRNAs that carry puromycin, a peptidyl acceptor antibiotic, at their 3' end. Thus, a specific mRNA can be enriched from a complex mixture of mRNAs (e.g., a combinatorial library) based on the properties of the encoded peptide or protein, e.g., antibody, or portion thereof, such as binding of the antibody, or portion thereof, to the dual specificity antigen. Nucleic acid sequences encoding antibodies, or portions thereof, recovered from screening of such libraries can be expressed by recombinant means as described above (e.g., in mammalian host cells) and, moreover, can be subjected to further affinity maturation by either additional rounds of screening of mRNA-peptide fusions in which mutations have been introduced into the originally selected sequence(s), or by other methods for affinity maturation in vitro of recombinant antibodies, as described above.
In another approach the antibodies of the present invention can also be generated using yeast display methods known in the art. In yeast display methods, genetic methods are used to tether antibody domains to the yeast cell wall and display them on the surface of yeast. In particular, such yeast can be utilized to display antigen-binding domains expressed from a repertoire or combinatorial antibody library (e. g., human or murine). Examples of yeast display methods that can be used to make the antibodies of the present invention include those disclosed Wittrup, et al. U.S. Patent No. 6,699,658 incorporated herein by reference.
B. Production of recombinant IL-12p40 antibodies
Antibodies of the present invention may be produced by any of a number of techniques known in the art. For example, expression from host cells, wherein expression vector(s) encoding the heavy and light chains is (are) transfected into a host cell by standard techniques. The various forms of the term "transfection" are intended to encompass a wide variety of techniques commonly used for the introduction of exogenous DNA into a prokaryotic or eukaryotic host cell, e.g., electroporation, calcium-phosphate precipitation, DEAE-dextran transfection and the like. Although it is possible to express the antibodies of the invention in either prokaryotic or eukaryotic host cells, expression of antibodies in eukaryotic cells is preferable, and most preferable in mammalian host cells, because such eukaryotic cells (and in particular mammalian cells) are more likely than prokaryotic cells to assemble and secrete a properly folded and immunologically active antibody.
Preferred mammalian host cells for expressing the recombinant antibodies of the invention include Chinese Hamster Ovary (CHO cells) (including dhfr- CHO cells, described in Urlaub and Chasin, (1980) Proc. Natl. Acad. ScL USA 77:4216-4220, used with a DHFR selectable marker, e.g., as described in RJ. Kaufman and P.A. Sharp (1982) MoI. Biol. 159:601-621), NSO myeloma cells, COS cells and SP2 cells. When recombinant expression vectors encoding antibody genes are introduced into mammalian host cells, the antibodies are produced by culturing the host cells for a period of time sufficient to allow for expression of the antibody in the host cells or, more preferably, secretion of the antibody into the culture medium in which the host cells are grown. Antibodies can be recovered from the culture medium using standard protein purification methods.
Host cells can also be used to produce functional antibody fragments, such as Fab fragments or scFv molecules. It will be understood that variations on the above procedure are within the scope of the present invention. For example, it may be desirable to transfect a host cell with DNA encoding functional fragments of either the light chain and/or the heavy chain of an antibody of this invention. Recombinant DNA technology may also be used to remove some, or all, of the DNA encoding either or both of the light and heavy chains that is not necessary for binding to the antigens of interest. The molecules expressed from such truncated DNA molecules are also encompassed by the antibodies of the invention. In addition, bifunctional antibodies may be produced in which one heavy and one light chain are an antibody of the invention and the other heavy and light chain are specific for an antigen other than the antigens of interest by crosslinking an antibody of the invention to a second antibody by standard chemical crosslinking methods.
In a preferred system for recombinant expression of an antibody, or antigen-binding portion thereof, of the invention, a recombinant expression vector encoding both the antibody heavy chain and the antibody light chain is introduced into dhfr- CHO cells by calcium phosphate-mediated transfection. Within the recombinant expression vector, the antibody heavy and light chain genes are each operatively linked to CMV enhancer/AdMLP promoter regulatory elements to drive high levels of transcription of the genes. The recombinant expression vector also carries a DHFR gene, which allows for selection of CHO cells that have been transfected with the vector using methotrexate selection/amplification. The selected transformant host cells are cultured to allow for expression of the antibody heavy and light chains and intact antibody is recovered from the culture medium. Standard molecular biology techniques are used to prepare the recombinant expression vector, transfect the host cells, select for transformants, culture the host cells and recover the antibody from the culture medium. Still further the invention provides a method of synthesizing a recombinant antibody of the invention by culturing a host cell of the invention in a suitable culture medium until a recombinant antibody of the invention is synthesized. The method can further comprise isolating the recombinant antibody from the culture medium.
1. Anti IL-12p40 antibodies
Table 5 is a list of amino acid sequences of VH and VL regions of preferred anti-hlL- 12p40 antibodies of the invention.
Table 5: List of Amino Acid Sequences of VH and VL regions
Figure imgf000043_0001
Figure imgf000044_0001
Figure imgf000045_0001
Figure imgf000046_0001
Figure imgf000047_0001
Figure imgf000048_0001
The foregoing isolated anti-IL-12p40 antibody CDR sequences establish a novel family of IL-12p40 binding proteins, isolated in accordance with this invention, and comprising polypeptides that include the CDR sequences listed in Table 6 below. To generate and to select CDR' s of the invention having preferred IL-12p40 binding and/or neutralizing activity with respect to hIL-12 and or hIL-23, standard methods known in the art for generating binding proteins of the present invention and assessing the IL- 12 and or IL-23 binding and/or neutralizing characteristics of those binding protein may be used, including but not limited to those specifically described herein.
Table 6: Consensus IL-12p40 CDR affinity ligands (alternative residues are listed below each amino acid position; - indicates residue may be absent).
CDR Sequence Consensus region Identifier Sequence
CDR-Hl SEQ ID X1 X2 X3 X4 X5 X6 X7
NO.:55 D Y Y I H - -
K S V M G V S
T T G E
S W V
S
F
CDR-H2 SEQ ID X1 X2 X3 X4 X5 χδ X7 X8 X9 Xio Xn Xi2 Xi3 Xl4 Xl5 X16 X17 X18 X19 X2O
NO.:56 H I Y W D D D K Y Y N P S L K _ _ _ _ _
D F W P G G G N T N Y N E K F S D F Q D
G L H E S S S P T A D D V K G
W S T A T P E I H D P T M Q R
S N S I N T N K S Y R P
Y D H S A
R G G
CDR-H3 SEQ ID X1 X2 X3 X4 X5 X6 X7 X8 X9 Xio Xn Xi2 Xi3
NO.:57 R G I R S A M D Y - - - -
N T R V N Y A M D Y M D Y
W R F Y G L D Y N A
P Y F A F L W
H Q A R M F
CDR-Ll SEQ ID X1 X2 X3 X4 X5 X5 X7 X8 X9 Xio Xn Xi2 X13 Xl 4 X15
NO.:58 K A S Q S V S N D V A - - - -
R E N I G T N G I S F M N
D K Y L H
CDR-L2 SEQ ID X1 X2 X3 X4 X5 X6 X7 X8
NO.:59 Y A S N R Y T -
S T A H N Q S S
S S I G
Q
CDR-L3 SEQ ID X1 X2 X3 X4 X5 X6 X7 X8 X9
NO.:60 Q Q D Y N S P W T
Y N T Y F S
S K S V Y
I E W L
P
2. Anti IL-12p40 Chimeric antibodies
A chimeric antibody is a molecule in which different portions of the antibody are derived from different animal species, such as antibodies having a variable region derived from a murine monoclonal antibody and a human immunoglobulin constant region. Methods for producing chimeric antibodies are known in the art and discussed in detail in Example 2.1. See e.g., Morrison, Science 229:1202 (1985); Oi et al, BioTechniques 4:214 (1986); Gillies et al., (1989) J. Immunol. Methods 125:191-202; U.S. Pat. Nos. 5,807,715; 4,816,567; and 4,816,397, which are incorporated herein by reference in their entireties. In addition, techniques developed for the production of "chimeric antibodies" (Morrison et al., 1984, Proc. Natl. Acad. Sci. 81:851-855; Neuberger et al., 1984, Nature 312:604-608; Takeda et al., 1985, Nature 314:452-454 which are incorporated herein by reference in their entireties) by splicing genes from a mouse antibody molecule of appropriate antigen specificity together with genes from a human antibody molecule of appropriate biological activity can be used.
In one embodiment, the chimeric antibodies of the invention are produced by replacing the heavy chain constant region of the murine monoclonal anti human IL- 12 antibodies described in section 1 with a human IgGl constant region. In a specific embodiment the chimeric antibody of the invention comprises a heavy chain variable region (VH ) comprising the amino acid sequence of SEQ ID NO: 35; SEQ ID NO: 37; SEQ ID NO: 39; SEQ ID NO: 41; SEQ ID NO: 43; SEQ ID NO: 45; SEQ ID NO: 47; SEQ ID NO: 49; SEQ ID NO: 51; or SEQ ID NO: 53 and a light chain variable region (VL ) comprising the amino acid sequence of SEQ ID NO: 36; SEQ ID NO: 38; SEQ ID NO: 40; SEQ ID NO: 42; SEQ ID NO: 44; SEQ ID NO: 46; SEQ ID NO: 48; SEQ BD NO: 50; SEQ ID NO: 52; or SEQ ID NO: 54.
3. Anti IL-12p40 CDR grafted antibodies
CDR-grafted antibodies of the invention comprise heavy and light chain variable region sequences from a human antibody wherein one or more of the CDR regions of VH and/or VL are replaced with CDR sequences of the murine antibodies of the invention. A framework sequence from any human antibody may serve as the template for CDR grafting. However, straight chain replacement onto such a framework often leads to some loss of binding affinity to the antigen. The more homologous a human antibody is to the original murine antibody, the less likely the possibility that combining the murine CDRs with the human framework will introduce distortions in the CDRs that could reduce affinity. Therefore, it is preferable that the human variable framework that is chosen to replace the murine variable framework apart from the CDRs have at least a 65% sequence identity with the murine antibody variable region framework. It is more preferable that the human and murine variable regions apart from the CDRs have at least 70% sequence identify. It is even more preferable that the human and murine variable regions apart from the CDRs have at least 75% sequence identity. It is most preferable that the human and murine variable regions apart from the CDRs have at least 80% sequence identity. Methods for producing chimeric antibodies are known in the art and discussed in detail in Example 2.2. (also see EP 239,400; PCT publication WO 91/09967; U.S. Pat. Nos. 5,225,539; 5,530,101; and 5,585,089), veneering or resurfacing (EP 592,106; EP 519,596; Padlan, Molecular Immunology 28(4/5):489-498 (1991); Studnicka et al., Protein Engineering 7(6):805-814 (1994); Roguska et al., PNAS 91:969-973 (1994)), and chain shuffling (U.S. Pat. No. 5,565,352).
In a specific embodiment the invention provides CDR grafted antibodies with VH and/or VL chains as described in Table 7.
TABLE 7: CDR Grafted antibodies
Figure imgf000051_0001
Figure imgf000052_0001
Figure imgf000053_0001
4. Anti IL-12p40 Humanized antibodies
Humanized antibodies are antibody molecules from non-human species antibody that binds the desired antigen having one or more complementarity determining regions (CDRs) from the non-human species and framework regions from a human immunoglobulin molecule. Known human Ig sequences are disclosed, e.g., www.ncbi.nlm.nih.gov/entrez- /query.fcgi; www.atcc.org/phage/hdb.html; www.sciquest.com/; www.abcam.com/; www.antibodyresource.com/onlinecomp.html; www.public.iastate.edu/.about.pedro/research_tools.html; www.mgen.uni- heidelberg.de/SD/IT/rr.html; www.whfreeman.com/immunology/CH- 05/kuby05.htm; www.library.thinkquest.org/12429/Immune/Antibody.html; www.hhmi.org/grants/lectures/1996/vlab/; www.path.cam.ac.uk/.about.mrcT/m- ikeimages.html; www.antibodyresource.com/; mcb.harvard.edu/BioLinks/Immuno- logy.html.www.immunologylink.com/; pathbox.wustl.edu/.about.hcenter/index.- html; www.biotech.ufl.edu/.about.hcl/; www.pebio.com/pa/340913/340913.html- ; www.nal.usda.gov/awic/pubs/antibody/; www.m.ehime-u.acjp/.about.yasuhito-/Elisa.html; www.biodesign.com/table.asp; www.icnet.uk/axp/facs/davies/lin- ks.html; www.biotech.ufl.edu/.about.fccl/protocol.html; www.isac-net.org/sites_geo.html; aximtl.imt.uni- marburg.de/.about.rek/AEP- Start.html; baserv.uci.kun.nl/.about.jraats/linksl.html; www.recab.uni-hd.de/immuno.bme.nwu.edu/; www.mrc-cpe.cam.ac.uk/imt-doc/pu- blic/INTRO.html; www.ibt.unam.mx/vir/V_mice.html; imgt.cnusc.fr:8104/; www.biochem.ucl.ac.uk/.about.martin/abs/index.html; antibody.bath.ac.uk/; abgen.cvm.tamu.edu/lab/wwwabgen.html; www.unizh.ch/.about.honegger/AHOsem- inar/SlideOl .html; www.cryst.bbk.ac.Uk/.about.ubcg07s/; www.nimr.mrc.ac.uk/CC/ccaewg/ccaewg.htm; www.path.cam.ac.uk/.about.mrcV/h- umanisation/TAHHP.html; www.ibt.unam.mx/vir/structure/stat_aim.html; www.biosci.missouri.edu/smithgp/index.html; www.cryst.bioc.cam.ac.uk/.abo- ut.fmolina/Web- pages/Peρt/spottech.html; www.jerini.de/fr roducts.htm; www.patents.ibm.com/ibm.html.Kabat et al., Sequences of Proteins of Immunological Interest, U.S. Dept. Health (1983), each entirely incorporated herein by reference. Such imported sequences can be used to reduce immunogenicity or reduce, enhance or modify binding, affinity, on-rate, off-rate, avidity, specificity, half -life, or any other suitable characteristic, as known in the art.
Framework residues in the human framework regions may be substituted with the corresponding residue from the CDR donor antibody to alter, preferably improve, antigen binding. These framework substitutions are identified by methods well known in the art, e.g., by modeling of the interactions of the CDR and framework residues to identify framework residues important for antigen binding and sequence comparison to identify unusual framework residues at particular positions. (See, e.g., Queen et al., U.S. Pat. No. 5,585,089; Riechmann et al., Nature 332:323 (1988), which are incorporated herein by reference in their entireties.) Three- dimensional immunoglobulin models are commonly available and are familiar to those skilled in the art. Computer programs are available which illustrate and display probable three-dimensional conformational structures of selected candidate immunoglobulin sequences. Inspection of these displays permits analysis of the likely role of the residues in the functioning of the candidate immunoglobulin sequence, i.e., the analysis of residues that influence the ability of the candidate immunoglobulin to bind its antigen. In this way, FR residues can be selected and combined from the consensus and import sequences so that the desired antibody characteristic, such as increased affinity for the target antigen(s), is achieved. In general, the CDR residues are directly and most substantially involved in influencing antigen binding. Antibodies can be humanized using a variety of techniques known in the art, such as but not limited to those described in Jones et al., Nature 321:522 (1986); Verhoeyen et al., Science 239:1534 (1988)), Sims et al., J. Immunol. 151: 2296 (1993); Chothia and Lesk, J. MoI. Biol. 196:901 (1987), Carter et al., Proc. Natl. Acad. Sci. U.S.A. 89:4285 (1992); Presta et al., J. Immunol. 151:2623 (1993), Padlan, Molecular Immunology 28(4/5):489-498 (1991); Studnicka et al., Protein Engineering 7(6):805-814 (1994); Roguska. et al. , PNAS 91:969-973 (1994); PCT publication WO 91/09967, PCT/: US98/16280, US96/18978, US91/09630, US91/05939, US94/01234, GB89/01334, GB91/01134, GB92/01755; WO90/14443, WO90/14424, WO90/14430, EP 229246, EP 592,106; EP 519,596, EP 239,400, U.S. Pat. Nos. 5,565,332, 5,723,323, 5,976,862, 5,824,514, 5,817,483, 5814476, 5763192, 5723323, 5,766886, 5,714,352, 6,204,023, 6,180,370, 5,693,762, 5,530,101, 5,585,089, 5,225,539; 4,816,567, each entirely incorporated herein by reference, included references cited therein.
C. Production of Antibodies and Antibody-Producing Cell Lines
Preferrably, anti-IL-12p40 antibodies of the present invention, exhibit a high capacity to reduce or to neutralize IL-12 activity, e.g.,as assessed by any one of several in vitro and in vivo assays known in the art (e.g., see Example l.l.C). For example, these antibodies neutralize JL-12- induced production of interferon gamma by PHA blasts with IC50 values in the range of at least about 10'8 M, about 10'9 M, or about 10'10 M. Preferrably, anti-IL-12p40 antibodies of the present invention, also exhibit a high capacity to reduce or to neutralize IL-23 activity
In prefered embodiments, the isolated antibody, or antigen-binding portion thereof, binds human BL-12p40, wherein the antibody, or antigen-binding portion thereof, dissociates from human IL-12p40 with a Ic0Jf rate constant of about 0. Is"1 or less, as determined by surface plasmon resonance, or which inhibits human IL-12 and/or human IL-23 activity with an IC50 of about 1 x 10'6M or less. Alternatively, the antibody, or an antigen-binding portion thereof, may dissociate from human IL-12p40 with a kOff rate constant of about 1 x 10"2S-1Or less, as determined by surface plasmon resonance, or may inhibit human IL-12 and/or human IL-23 activity with an IC50 of about 1 x 10"7M or less. Alternatively, the antibody, or an antigen-binding portion thereof, may dissociate from human IL-12p40 with a karate constant of about 1 x 10"3S"1 or less, as determined by surface plasmon resonance, or may inhibit human IL-12 and/or human IL-23 with an IC50 of about 1 x 10"8M or less. Alternatively, the antibody, or an antigen-binding portion thereof, may dissociate from human IL-12p40 with a koff rate constant of about 1 x 10"4S"1 or less, as determined by surface plasmon resonance, or may inhibit IL-12 and/or human IL-23 activity with an IC50 of about 1 x 10"9M or less. Alternatively, the antibody, or an antigen-binding portion thereof, may dissociate from human IL-12p40 with a k^ rate constant of about 1 x 10"5S"1 or less, as determined by surface plasmon resonance, or may inhibit IL-12 and/or human EL-23 activity with an IC50 of about 1 x 10"10M or less. Alternatively, the antibody, or an antigen-binding portion thereof, may dissociate from human IL-12p40 with a koff rate constant of about 1 x 10"V 1Or less, as determined by surface plasmon resonance, or may inhibit IL-12 and/or human EL-23 activity with an IC50 of about 1 x 10"11M or less.
In certain embodiments, the antibody comprises a heavy chain constant region, such as an IgGl, IgG2, IgG3, IgG4, IgA, IgE, IgM or IgD constant region. Preferably, the heavy chain constant region is an IgGl heavy chain constant region or an IgG4 heavy chain constant region. Furthermore, the antibody can comprise a light chain constant region, either a kappa light chain constant region or a lambda light chain constant region. Preferably, the antibody comprises a kappa light chain constant region. Alternatively, the antibody portion can be, for example, a Fab fragment or a single chain Fv fragment.
Replacements of amino acid residues in the Fc portion to alter antibody effector function are known in the art (Winter, et al. US PAT NOS 5,648,260; 5624821). The Fc portion of an antibody mediates several important effector functions e.g. cytokine induction, ADCC, phagocytosis, complement dependent cytotoxicity (CDC) and half-life/ clearance rate of antibody and antigen-antibody complexes. In some cases these effector functions are desirable for therapeutic antibody but in other cases might be unnecessary or even deleterious, depending on the therapeutic objectives. Certain human IgG isotypes, particularly IgGl and IgG3, mediate ADCC and CDC via binding to FcγRs and complement CIq, respectively. Neonatal Fc receptors (FcRn) are the critical components determining the circulating half-life of antibodies. In still another embodiment at least one amino acid residue is replaced in the constant region of the antibody, for example the Fc region of the antibody, such that effector functions of the antibody are altered.
One embodiment provides a labeled binding protein wherein an antibody or antibody portion of the invention is derivatized or linked to another functional molecule (e.g., another peptide or protein). For example, a labeled binding protein of the invention can be derived by functionally linking an antibody or antibody portion of the invention (by chemical coupling, genetic fusion, noncovalent association or otherwise) to one or more other molecular entities, such as another antibody (e.g., a bispecific antibody or a diabody), a detectable agent, a cytotoxic agent, a pharmaceutical agent, and/or a protein or peptide that can mediate associate of the antibody or antibody portion with another molecule (such as a streptavidin core region or a polyhistidine tag).
Useful detectable agents with which an antibody or antibody portion of the invention may be derivatized include fluorescent compounds. Exemplary fluorescent detectable agents include fluorescein, fluorescein isothiocyanate, rhodamine, 5-dimethylamine-l- napthalenesulfonyl chloride, phycoerythrin and the like. An antibody may also be derivatized with detectable enzymes, such as alkaline phosphatase, horseradish peroxidase, glucose oxidase and the like. When an antibody is derivatized with a detectable enzyme, it is detected by adding additional reagents that the enzyme uses to produce a detectable reaction product. For example, when the detectable agent horseradish peroxidase is present, the addition of hydrogen peroxide and diaminobenzidine leads to a colored reaction product, which is detectable. An antibody may also be derivatized with biotin, and detected through indirect measurement of avidin or streptavidin binding.
Another embodiment of the invention provides a crystallized binding protein. Preferably the invention relates to crystals of whole anti-IL-12p40 antibodies and fragments thereof as disclosed herein, and formulations and compositions comprising such crystals. In one embodiment the crystallized binding protein has a greater half-life in vivo than the soluble counterpart of the binding protein. In another embodiment the binding protein retains biological activity after crystallization.
Crystallized binding protein of the invention may be produced according methods known in the art and as disclosed in WO 02072636, incorporated herein by reference. Another embodiment of the invention provides a glycosylated binding protein wherein the antibody or antigen-binding portion thereof comprises one or more carbohydrate residues. Nascent in vivo protein production may undergo further processing, known as post-translational modification. In particular, sugar (glycosyl) residues may be added enzymatically, a process known as glycosylation. The resulting proteins bearing covalently linked oligosaccharide side chains are known as glycosylated proteins or glycoproteins. Antibodies are glycoproteins with one or more carbohydrate residues in the Fc domain, as well as the variable domain. Carbohydrate residues in the Fc domain have important effect on the effector function of the Fc domain, with minimal effect on antigen binding or half-life of the antibody (R. Jefferis, Biotechnol. Prog. 21 (2005), pp. 11-16). In contrast, glycosylation of the variable domain may have an effect on the antigen binding activity of the antibody. Glycosylation in the variable domain may have a negative effect on antibody binding affinity, likely due to steric hindrance (Co, M.S., et al., MoI. Immunol. (1993) 30:1361- 1367), or result in increased affinity for the antigen (Wallick, S.C., et al., Exp. Med. (1988) 168:1099-1109; Wright, A., et al., EMBO J. (1991) 10:2717 2723).
One aspect of the present invention is directed to generating glycosylation site mutants in which the O- or N-linked glycosylation site of the binding protein has been mutated. One skilled in the art can generate such mutants using standard well-known technologies. Glycosylation site mutants that retain the biological activity but have increased or decreased binding activity are another object of the present invention.
In still another embodiment, the glycosylation of the antibody or antigen-binding portion of the invention is modified. For example, an aglycoslated antibody can be made (i.e., the antibody lacks glycosylation). Glycosylation can be altered to, for example, increase the affinity of the antibody for antigen. Such carbohydrate modifications can be accomplished by, for example, altering one or more sites of glycosylation within the antibody sequence. For example, one or more amino acid substitutions can be made that result in elimination of one or more variable region glycosylation sites to thereby eliminate glycosylation at that site. Such aglycosylation may increase the affinity of the antibody for antigen. Such an approach is described in further detail in PCT Publication WO2003016466A2, and U.S. Pat. Nos. 5,714,350 and 6,350,861, each of which is incorporated herein by reference in its entirety.
Additionally or alternatively, a modified antibody of the invention can be made that has an altered type of glycosylation, such as a hypofucosylated antibody having reduced amounts of fucosyl residues or an antibody having increased bisecting GIcNAc structures. Such altered glycosylation patterns have been demonstrated to increase the ADCC ability of antibodies. Such carbohydrate modifications can be accomplished by, for example, expressing the antibody in a host cell with altered glycosylation machinery. Cells with altered glycosylation machinery have been described in the art and can be used as host cells in which to express recombinant antibodies of the invention to thereby produce an antibody with altered glycosylation. See, for example, Shields, R. L. et al. (2002) J. Biol. Chem. 277:26733-26740; Umana et al. (1999) Nat. Biotech. 17:176-1, as well as, European Patent No: EP 1,176,195; PCT Publications WO 03/035835; WO 99/54342 80, each of which is incorporated herein by reference in its entirety.
Protein glycosylation depends on the amino acid sequence of the protein of interest, as well as the host cell in which the protein is expressed. Different organisms may produce different glycosylation enzymes (eg., glycosyltransferases and glycosidases), and have different substrates (nucleotide sugars) available. Due to such factors, protein glycosylation pattern, and composition of glycosyl residues, may differ depending on the host system in which the particular protein is expressed. Glycosyl residues useful in the invention may include, but are not limited to, glucose, galactose, mannose, fucose, n-acetylglucosamine and sialic acid. Preferably the glycosylated binding protein comprises glycosyl residues such that the glycosylation pattern is human.
It is known to those skilled in the art that differing protein glycosylation may result in differing protein characteristics. For instance, the efficacy of a therapeutic protein produced in a microorganism host, such as yeast, and glycosylated utilizing the yeast endogenous pathway may be reduced compared to that of the same protein expressed in a mammalian cell, such as a CHO cell line. Such glycoproteins may also be immunogenic in humans and show reduced half-life in vivo after administration. Specific receptors in humans and other animals may recognize specific glycosyl residues and promote the rapid clearance of the protein from the bloodstream. Other adverse effects may include changes in protein folding, solubility, susceptibility to proteases, trafficking, transport, compartmentalization, secretion, recognition by other proteins or factors, antigenicity, or allergenicity. Accordingly, a practitioner may prefer a therapeutic protein with a specific composition and pattern of glycosylation, for example glycosylation composition and pattern identical, or at least similar, to that produced in human cells or in the species-specific cells of the intended subject animal.
Expressing glycosylated proteins different from that of a host cell may be achieved by genetically modifying the host cell to express heterologous glycosylation enzymes. Using techniques known in the art a practitioner may generate antibodies or antigen-binding portions thereof exhibiting human protein glycosylation. For example, yeast strains have been genetically modified to express non-naturally occurring glycosylation enzymes such that glycosylated proteins (glycoproteins) produced in these yeast strains exhibit protein glycosylation identical to that of animal cells, especially human cells (U.S patent applications 20040018590 and 20020137134 and PCT publication WO2005100584 A2). Ih addition to the binding proteins, the present invention is also directed to an anti- idiotypic (anti-Id) antibody specific for such binding proteins of the invention. An anti-Id antibody is an antibody, which recognizes unique determinants generally associated with the antigen-binding region of another antibody. The anti-Id can be prepared by immunizing an animal with the binding protein or a CDR containing region thereof. The immunized animal will recognize, and respond to the idiotypic determinants of the immunizing antibody and produce an anti-Id antibody. The anti-Id antibody may also be used as an "immunogen" to induce an immune response in yet another animal, producing a so-called anti-anti-Id antibody.
Further, it will be appreciated by one skilled in the art that a protein of interest may be expressed using a library of host cells genetically engineered to express various glycosylation enzymes, such that member host cells of the library produce the protein of interest with variant glycosylation patterns. A practitioner may then select and isolate the protein of interest with particular novel glycosylation patterns. Preferably, the protein having a particularly selected novel glycosylation pattern exhibits improved or altered biological properties.
D. Uses of Anti-IL-12p40 Antibodies
Given their ability to bind to human IL-12p40, the anti-human IL-12p40 antibodies, or portions thereof, of the invention can be used to detect EL- 12 and/or human IL-23 {e.g., in a biological sample, such as serum or plasma), using a conventional immunoassay, such as an enzyme linked immunosorbent assays (ELISA), an radioimmunoassay (RIA) or tissue immunohistochemistry. The invention provides a method for detecting IL- 12 and/or human IL- 23 in a biological sample comprising contacting a biological sample with an antibody, or antibody portion, of the invention and detecting either the antibody (or antibody portion) bound to IL-12 and/or human IL-23 or unbound antibody (or antibody portion), to thereby detect IL-12 and/or human IL-23 in the biological sample. The antibody is directly or indirectly labeled with a detectable substance to facilitate detection of the bound or unbound antibody. Suitable detectable substances include various enzymes, prosthetic groups, fluorescent materials, luminescent materials and radioactive materials. Examples of suitable enzymes include horseradish peroxidase, alkaline phosphatase, β-galactosidase, or acetylcholinesterase; examples of suitable prosthetic group complexes include streptavidin/biotin and avidin/biotin; examples of suitable fluorescent materials include umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride or phycoerythrin; an example of a luminescent material includes luminol; and examples of suitable radioactive material include 3H 14C, 35S1 90Y, 99Tc, 111In, 1251, 1311, 177Lu, 166Ho, or 153Sm.
Alternative to labeling the antibody, human IL-12 can be assayed in biological fluids by a competition immunoassay utilizing rhIL-12 standards labeled with a detectable substance and an unlabeled anti- human IL-12p40 antibody. In this assay, the biological sample, the labeled rhlL- 12 standards and the anti- human IL-12p40 antibody are combined and the amount of labeled rhIL-12 standard bound to the unlabeled antibody is determined. The amount of human IL-12 in the biological sample is inversely proportional to the amount of labeled rhIL-12 standard bound to the anti-EL-12p40 antibody. Similarly, human EL-23 can also be assayed in biological fluids by a competition immunoassay utilizing rhlL-23 standards labeled with a detectable substance and an unlabeled anti-human IL-12p40 antibody.
The antibodies and antibody portions of the invention preferably are capable of neutralizing human IL-12 and/or human IL-23 activity both in vitro and in vivo. Accordingly, such antibodies and antibody portions of the invention can be used to inhibit ML-12 and/or ML- 23 activity, e.g., in a cell culture containing hIL-12 and/or hIL-23, in human subjects or in other mammalian subjects having IL-12 and/or hIL-23 with which an antibody of the invention cross- reacts. In one embodiment, the invention provides a method for inhibiting ML-12 and/or hIL-23 activity comprising contacting ML- 12 and/or hIL-23 with an antibody or antibody portion of the invention such that hIL-12 and/or hIL-23 activity is inhibited. For example, in a cell culture containing, or suspected of containing hIL-12 and/or hIL-23, an antibody or antibody portion of the invention can be added to the culture medium to inhibit ML- 12 and/or hIL-23 activity in the culture.
In another embodiment, the invention provides a method for reducing ML-12 and/or ML- 23 activity in a subject, advantageously from a subject suffering from a disease or disorder in which IL-12 or IL-23 activity is detrimental. The invention provides methods for reducing IL-12 and/or IL-23 activity in a subject suffering from such a disease or disorder, which method comprises administering to the subject an antibody or antibody portion of the invention such that IL-12 and/or IL-23 activity in the subject is reduced. Preferably, the IL-12 is human IL-12, the IL-23 is human IL-23, and the subject is a human subject. Alternatively, the subject can be a mammal expressing an IL-12 and/or IL-23 to which an antibody of the invention is capable of binding. Still further the subject can be a mammal into which IL-12 and/or IL-23 has been introduced (e.g., by administration of IL-12 and/or IL-23 or by expression of an IL-12 and/or IL- 23 transgene). An antibody of the invention can be administered to a human subject for therapeutic purposes. Moreover, an antibody of the invention can be administered to a non- human mammal expressing an IL-12 and/or IL-23 with which the antibody is capable of binding for veterinary purposes or as an animal model of human disease. Regarding the latter, such animal models may be useful for evaluating the therapeutic efficacy of antibodies of the invention (e.g., testing of dosages and time courses of administration).
As used herein, the term "a disorder in which IL-12 and/or IL-23 activity is detrimental" is intended to include diseases and other disorders in wMch the presence of IL-12 and/or IL-23 in a subject suffering from the disorder has been shown to be or is suspected of being either responsible for the pathophysiology of the disorder or a factor that contributes to a worsening of the disorder. Accordingly, a disorder in which IL-12 and/or EL-23 activity is detrimental is a disorder in which reduction of IL-12 and/or JL-23 activity is expected to alleviate the symptoms and/or progression of the disorder. Such disorders may be evidenced, for example, by an increase in the concentration of IL-12 and/or IL-23 in a biological fluid of a subject suffering from the disorder (e.g., an increase in the concentration of IL-12 and/or IL-23 in serum, plasma, synovial fluid, etc. of the subject), which can be detected, for example, using an anti-EL-12p40 antibody as described above. Non-limiting examples of disorders that can be treated with the antibodies of the invention include those disorders discussed in the section below pertaining to pharmaceutical compositions of the antibodies of the invention.
D. Pharmaceutical Composition
The invention also provides pharmaceutical compositions comprising an antibody, or antigen-binding portion thereof, of the invention and a pharmaceutically acceptable carrier. The pharmaceutical compositions comprising antibodies of the invention are for use in, but not limited to, diagnosing, detecting, or monitoring a disorder, in preventing, treating, managing, or ameliorating of a disorder or one or more symptoms thereof, and/or in research. In a specific embodiment, a composition comprises one or more antibodies of the invention, hi another embodiment, the pharmaceutical composition comprises one or more antibodies of the invention and one or more prophylactic or therapeutic agents other than antibodies of the invention for treating a disorder in which IL-12 and/or IL-23 activity is detrimental. Preferably, the prophylactic or therapeutic agents known to be useful for or having been or currently being used in the prevention, treatment, management, or amelioration of a disorder or one or more symptoms thereof. In accordance with these embodiments, the composition may further comprise of a carrier, diluent or excipient.
The antibodies and antibody-portions of the invention can be incorporated into pharmaceutical compositions suitable for administration to a subject. Typically, the pharmaceutical composition comprises an antibody or antibody portion of the invention and a pharmaceutically acceptable carrier. As used herein, "pharmaceutically acceptable carrier" includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like that are physiologically compatible. Examples of pharmaceutically acceptable carriers include one or more of water, saline, phosphate buffered saline, dextrose, glycerol, ethanol and the like, as well as combinations thereof. In many cases, it will be preferable to include isotonic agents, for example, sugars, polyalcohols such as mannitol, sorbitol, or sodium chloride in the composition. Pharmaceutically acceptable carriers may further comprise minor amounts of auxiliary substances such as wetting or emulsifying agents, preservatives or buffers, which enhance the shelf life or effectiveness of the antibody or antibody portion.
Various delivery systems are known and can be used to administer one or more antibodies of the invention or the combination of one or more antibodies of the invention and a prophylactic agent or therapeutic agent useful for preventing, managing, treating, or ameliorating a disorder or one or more symptoms thereof, e.g., encapsulation in liposomes, microparticles, microcapsules, recombinant cells capable of expressing the antibody or antibody fragment, receptor- mediated endocytosis (see, e. g., Wu and Wu, J. Biol. Chem. 262:4429-4432 (1987)), construction of a nucleic acid as part of a retroviral or other vector, etc. Methods of administering a prophylactic or therapeutic agent of the invention include, but are not limited to, parenteral administration (e.g., intradermal, intramuscular, intraperitoneal, intravenous and subcutaneous) , epidurala administration, intratumoral administration, and mucosal adminsitration (e.g., intranasal and oral routes). In addition, pulmonary administration can be employed, e.g., by use of an inhaler or nebulizer, and formulation with an aerosolizing agent. See, e.g., U.S. Pat. Nos. 6, 019,968, 5,985, 320, 5,985,309, 5,934, 272, 5,874,064, 5,855,913, 5,290, 540, and 4,880,078; and PCT Publication Nos. WO 92/19244, WO 97/32572, WO 97/44013, WO 98/31346, and WO 99/66903, each of which is incorporated herein by reference their entireties. In one embodiment, an antibody of the invention, combination therapy, or a composition of the invention is administered using Alkermes AIR® pulmonary drug delivery technology (Alkermes, Inc., Cambridge, Mass.). In a specific embodiment, prophylactic or therapeutic agents of the invention are administered intramuscularly, intravenously, intratumorally, orally, intranasally, pulmonary, or subcutaneously. The prophylactic or therapeutic agents may be administered by any convenient route, for example by infusion or bolus injection, by absorption through epithelial or mucocutaneous linings (e.g. , oral mucosa, rectal and intestinal mucosa, etc.) and may be administered together with other biologically active agents. Administration can be systemic or local.
In a specific embodiment, it may be desirable to administer the prophylactic or therapeutic agents of the invention locally to the area in need of treatment; this may be achieved by, for example, and not by way of limitation, local infusion, by injection, or by means of an implant, said implant being of a porous or non-porous material, including membranes and matrices, such as sialastic membranes, polymers, fibrous matrices (e.g., Tissuel®), or collagen matrices. In one embodiment, an effective amount of one or more antibodies of the invention antagonists is administered locally to the affected area to a subject to prevent, treat, manage, and/or ameliorate a disorder or a symptom thereof. In another embodiment, an effective amount of one or more antibodies of the invention is administered locally to the affected area in combination with an effective amount of one or more therapies (e. g., one or more prophylactic or therapeutic agents) other than an antibody of the invention of a subject to prevent, treat, manage, and/or ameliorate a disorder or one or more symptoms thereof.
In another embodiment, the prophylactic or therapeutic agent can be delivered in a controlled release or sustained release system. In one embodiment, a pump may be used to achieve controlled or sustained release (see Langer, supra; Sefton, 1987, CRC Crit. Ref. Biomed. Eng. 14:20; Buchwald et al., 1980, Surgery 88:507; Saudek et al., 1989, N. Engl. J. Med. 321:574). In another embodiment, polymeric materials can be used to achieve controlled or sustained release of the therapies of the invention (see e.g., Medical Applications of Controlled Release, Langer and Wise (eds.), CRC Pres., Boca Raton, FIa. (1974); Controlled Drug Bioavailability, Drug Product Design and Performance, Smolen and Ball (eds.), Wiley, New York (1984); Ranger and Peppas, 1983, J., Macromol. Sci. Rev. Macromol. Chem. 23:61; see also Levy et al., 1985, Science 228:190; During et al., 1989, Ann. Neurol. 25:351; Howard et al., 1989, J. Neurosurg. 7 1:105); U.S. Pat. No. 5,679,377; U.S. Pat. No. 5, 916,597; U. S. Pat. No. 5,912,015; U.S. Pat. No. 5,989,463; U.S. Pat. No. 5,128,326; PCT Publication No. WO 99/15154; and PCT Publication No. WO 99/20253. Examples of polymers used in sustained release formulations include, but are not limited to, poly(2-hydroxy ethyl methacrylate), poly(methyl methacrylate), poly(acrylic acid), poly(ethylene-co-vinyl acetate), poly(methacrylic acid), polyglycolides (PLG), polyanhydrides, poly(N- vinyl pyrrolidone), poly(vinyl alcohol), polyacrylamide, poly(ethylene glycol), polylactides (PLA), poly(lactide-co-glycolides) (PLGA), and polyorthoesters. In a preferred embodiment, the polymer used in a sustained release formulation is inert, free of leachable impurities, stable on storage, sterile, and biodegradable. Ia yet another embodiment, a controlled or sustained release system can be placed in proximity of the prophylactic or therapeutic target, thus requiring only a fraction of the systemic dose (see, e.g., Goodson, in Medical Applications of Controlled Release, supra, vol. 2, pp. 115-138 (1984)).
Controlled release systems are discussed in the review by Langer (1990, Science 249:1527-1533). Any technique known to one of skill in the art can be used to produce sustained release formulations comprising one or more therapeutic agents of the invention. See, e.g., U. S. Pat. No. 4,526, 938, PCT publication WO 91/05548, PCT publication WO 96/20698, Ning et al. , 1996, "Intratumoral Radioimmunotheraphy of a Human Colon Cancer Xenograft Using a Sustained-Release Gel," Radiotherapy &Oncology 39:179-189, Song et al., 1995, "Antibody Mediated Lung Targeting of Long- Circulating Emulsions," PDA Journal of Pharmaceutical Science &Technology 50:372-397, Cleek et al., 1997, "Biodegradable Polymeric Carriers for a bFGF Antibody for Cardiovascular Application," Pro. Intl. Symp. Control. ReI. Bioact. Mater. 24:853-854, and Lam et al., 1997, "Microencapsulation of Recombinant Humanized Monoclonal Antibody for Local Delivery," Proc. Int'l. Symp. Control ReI. Bioact. Mater. 24:759- 760, each of which is incorporated herein by reference in their entireties.
In a specific embodiment, where the composition of the invention is a nucleic acid encoding a prophylactic or therapeutic agent, the nucleic acid can be administered in vivo to promote expression of its encoded prophylactic or therapeutic agent, by constructing it as part of an appropriate nucleic acid expression vector and administering it so that it becomes intracellular, e.g., by use of a retroviral vector (see U. S. Pat. No. 4,980,286), or by direct injection, or by use of microparticle bombardment (e.g., a gene gun; Biolistic, Dupont), or coating with lipids or cell- surface receptors or transfecting agents, or by administering it in linkage to a homeobox-like peptide which is known to enter the nucleus (see, e.g., Joliot et al., 1991, Proc. Natl. Acad. Sci. USA 88:1864-1868). Alternatively, a nucleic acid can be introduced intracellularly and incorporated within host cell DNA for expression by homologous recombination.
A pharmaceutical composition of the invention is formulated to be compatible with its intended route of administration. Examples of routes of administration include, but are not limited to, parenteral, e.g., intravenous, intradermal, subcutaneous, oral, intranasal (e.g., inhalation), transdermal (e.g., topical), transmucosal, and rectal administration. In a specific embodiment, the composition is formulated in accordance with routine procedures as a pharmaceutical composition adapted for intravenous, subcutaneous, intramuscular, oral, intranasal, or topical administration to human beings. Typically, compositions for intravenous administration are solutions in sterile isotonic aqueous buffer. Where necessary, the composition may also include a solubilizing agent and a local anesthetic such as lignocamne to ease pain at the site of the injection.
If the compositions of the invention are to be administered topically, the compositions can be formulated in the form of an ointment, cream, transdermal patch, lotion, gel, shampoo, spray, aerosol, solution, emulsion, or other form well known to one of skill in the art. See, e.g., Remington's Pharmaceutical Sciences and Introduction to Pharmaceutical Dosage Forms, 19th ed., Mack Pub. Co., Easton, Pa. (1995). For non- sprayable topical dosage forms, viscous to semisolid or solid forms comprising a carrier or one or more excipients compatible with topical application and having a dynamic viscosity preferably greater than water are typically employed. Suitable formulations include, without limitation, solutions, suspensions, emulsions, creams, ointments, powders, liniments, salves, and the like, which are, if desired, sterilized or mixed with auxiliary agents (e.g., preservatives, stabilizers, wetting agents, buffers, or salts) for influencing various properties, such as, for example, osmotic pressure. Other suitable topical dosage forms include sprayable aerosol preparations wherein the active ingredient, preferably in combination with a solid or liquid inert carrier, is packaged in a mixture with a pressurized volatile (e.g., a gaseous propellant, such as freon) or in a squeeze bottle. Moisturizers or humectants can also be added to pharmaceutical compositions and dosage forms if desired. Examples of such additional ingredients are well known in the art.
If the method of the invention comprises intranasal administration of a composition, the composition can be formulated in an aerosol form, spray, mist or in the form of drops. In particular, prophylactic or therapeutic agents for use according to the present invention can be conveniently delivered in the form of an aerosol spray presentation from pressurized packs or a nebuliser, with the use of a suitable propellant (e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas). In the case of a pressurized aerosol the dosage unit may be determined by providing a valve to deliver a metered amount. Capsules and cartridges (composed of, e.g., gelatin) for use in an inhaler or insufflator may be formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch.
If the method of the invention comprises oral administration, compositions can be formulated orally in the form of tablets, capsules, cachets, gelcaps, solutions, suspensions, and the like. Tablets or capsules can be prepared by conventional means with pharmaceutically acceptable excipients such as binding agents (e.g., pregelatinised maize starch, polyvinylpyrrolidone, or hydroxypropyl methylcellulose); fillers (e.g., lactose, microcrystalline cellulose, or calcium hydrogen phosphate) ; lubricants (e.g., magnesium stearate, talc, or silica); disintegrants (e.g., potato starch or sodium starch glycolate) ; or wetting agents (e.g., sodium lauryl sulphate). The tablets may be coated by methods well-known in the art. Liquid preparations for oral administration may take the form of, but not limited to, solutions, syrups or suspensions, or they may be presented as a dry product for constitution with water or other suitable vehicle before use. Such liquid preparations may be prepared by conventional means with pharmaceutically acceptable additives such as suspending agents (e.g., sorbitol syrup, cellulose derivatives, or hydrogenated edible fats); emulsifying agents (e.g., lecithin or acacia); non-aqueous vehicles (e.g., almond oil, oily esters, ethyl alcohol, or fractionated vegetable oils); and preservatives (e.g., methyl or propyl-p- hydroxybenzoates or sorbic acid). The preparations may also contain buffer salts, flavoring, coloring, and sweetening agents as appropriate. Preparations for oral administration may be suitably formulated for slow release, controlled release, or sustained release of a prophylactic or therapeutic agent(s).
The method of the invention may comprise pulmonary administration, e.g., by use of an inhaler or nebulizer, of a composition formulated with an aerosolizing agent. See, e.g., U.S. Pat. Nos. 6,019, 968, 5,985, 320, 5, 985,309, 5,934,272, 5,874,064, 5,855,913, 5,290,540, and 4,880,078; and PCT Publication Nos. WO 92/19244, WO 97/32572, WO 97/44013, WO 98/31346, and WO 99/66903, each of which is incorporated herein by reference their entireties. In a specific embodiment, an antibody of the invention, combination therapy, and/or composition of the invention is administered using Alkermes AIR® pulmonary drug delivery technology (Alkermes, Inc., Cambridge, Mass.).
The method of the invention may comprise administration of a composition formulated for parenteral administration by injection (e. g., by bolus injection or continuous infusion). Formulations for injection may be presented in unit dosage form (e.g., in ampoules or in multi- dose containers) with an added preservative. The compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents. Alternatively, the active ingredient may be in powder form for constitution with a suitable vehicle (e.g., sterile pyrogen- free water) before use. The methods of the invention may additionally comprise of administration of compositions formulated as depot preparations. Such long acting formulations may be administered by implantation (e.g., subcutaneously or intramuscularly) or by intramuscular injection. Thus, for example, the compositions may be formulated with suitable polymeric or hydrophobic materials (e.g., as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives (e.g., as a sparingly soluble salt).
The methods of the invention encompasse administration of compositions formulated as neutral or salt forms. Pharmaceutically acceptable salts include those formed with anions such as those derived from hydrochloric, phosphoric, acetic, oxalic, tartaric acids, etc., and those formed with cations such as those derived from sodium, potassium, ammonium, calcium, ferric hydroxides, isopropylamine, triethylamine, 2- ethylamino ethanol, histidine, procaine, etc. [0262] Generally, the ingredients of compositions are supplied either separately or mixed together in unit dosage form, for example, as a dry lyophilized powder or water free concentrate in a hermetically sealed container such as an ampoule or sachette indicating the quantity of active agent. Where the mode of administration is infusion, composition can be dispensed with an infusion bottle containing sterile pharmaceutical grade water or saline. Where the mode of administration is by injection, an ampoule of sterile water for injection or saline can be provided so that the ingredients may be mixed prior to administration.
In particular, the invention also provides that one or more of the prophylactic or therapeutic agents, or pharmaceutical compositions of the invention is packaged in a hermetically sealed container such as an ampoule or sachette indicating the quantity of the agent. In one embodiment, one or more of the prophylactic or therapeutic agents, or pharmaceutical compositions of the invention is supplied as a dry sterilized lyophilized powder or water free concentrate in a hermetically sealed container and can be reconstituted (e.g., with water or saline) to the appropriate concentration for administration to a subject. Preferably, one or more of the prophylactic or therapeutic agents or pharmaceutical compositions of the invention is supplied as a dry sterile lyophilized powder in a hermetically sealed container at a unit dosage of at least 5 mg, more preferably at least 10 mg, at least 15 mg, at least 25 mg, at least 35 mg, at least 45 mg, at least 50 mg, at least 75 mg, or at least 100 mg. The lyophilized prophylactic or therapeutic agents or pharmaceutical compositions of the invention should be stored at between 2° C. and 8° C. in its original container and the prophylactic or therapeutic agents, or pharmaceutical compositions of the invention should be administered within 1 week, preferably within 5 days, within 72 hours, within 48 hours, within 24 hours, within 12 hours, within 6 hours, within 5 hours, within 3 hours, or within 1 hour after being reconstituted. In an alternative embodiment, one or more of the prophylactic or therapeutic agents or pharmaceutical compositions of the invention is supplied in liquid form in a hermetically sealed container indicating the quantity and concentration of the agent. Preferably, the liquid form of the administered composition is supplied in a hermetically sealed container at least 0.25 mg/ml, more preferably at least 0.5 mg/ml, at least 1 mg/ml, at least 2.5 mg/ml, at least 5 mg/ml, at least 8 mg/ml, at least 10 mg/ml, at least 15 mg/kg, at least 25 mg/ml, at least 50 mg/ml, at least 75 mg/ml or at least 100 mg/ml. The liquid form should be stored at between 2° C. and 8° C. in its original container.
The antibodies and antibody-portions of the invention can be incorporated into a pharmaceutical composition suitable for parenteral administration. Preferably, the antibody or antibody-portions will be prepared as an injectable solution containing 0.1-250 mg/ml antibody. The injectable solution can be composed of either a liquid or lyophilized dosage form in a flint or amber vial, ampule or pre-filled syringe. The buffer can be L-histidine (1-50 mM), optimally 5- 1OmM, at pH 5.0 to 7.0 (optimally pH 6.0). Other suitable buffers include but are not limited to, sodium succinate, sodium citrate, sodium phosphate or potassium phosphate. Sodium chloride can be used to modify the toxicity of the solution at a concentration of 0-300 mM (optimally 150 mM for a liquid dosage form). Cryoprotectants can be included for a lyophilized dosage form, principally 0-10% sucrose (optimally 0.5-1.0%). Other suitable cryoprotectants include trehalose and lactose. Bulking agents can be included for a lyophilized dosage form, principally 1-10% mannitol (optimally 2-4%). Stabilizers can be used in both liquid and lyophilized dosage forms, principally 1-50 mM L-Methionine (optimally 5-10 mM). Other suitable bulking agents include glycine, arginine, can be included as 0-0.05% polysorbate-80 (optimally 0.005-0.01%). Additional surfactants include but are not limited to polysorbate 20 and BRIJ surfactants. The pharmaceutical composition comprising the antibodies and antibody-portions of the invention prepared as an injectable solution for parenteral administration, can further comprise an agent useful as an adjuvant, such as those used to increase the absorption, or dispersion of a therapeutic protein (e.g., antibody). A particularly useful adjuvant is hyaluronidase, such as Hylenex® (recombinant human hyaluronidase).Addition of hyaluronidase in the injectable solution improves human bioavailability following parenteral administration, particularly subcutaneous administration. It also allows for greater injection site volumes (i.e. greater than 1 ml) with less pain and discomfort, and minimum incidence of injection site reactions, (see WO2004078140, US2006104968 incorporated herein by reference).
The compositions of this invention may be in a variety of forms. These include, for example, liquid, semi-solid and solid dosage forms, such as liquid solutions (e.g., injectable and infusible solutions), dispersions or suspensions, tablets, pills, powders, liposomes and suppositories. The preferred form depends on the intended mode of administration and therapeutic application. Typical preferred compositions are in the form of injectable or infusible solutions, such as compositions similar to those used for passive immunization of humans with other antibodies. The preferred mode of administration is parenteral (e.g., intravenous, subcutaneous, intraperitoneal, intramuscular). In a preferred embodiment, the antibody is administered by intravenous infusion or injection. In another preferred embodiment, the antibody is administered by intramuscular or subcutaneous injection.
Therapeutic compositions typically must be sterile and stable under the conditions of manufacture and storage. The composition can be formulated as a solution, microemulsion, dispersion, liposome, or other ordered structure suitable to high drug concentration. Sterile injectable solutions can be prepared by incorporating the active compound (i.e., antibody or antibody portion) in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the active compound into a sterile vehicle that contains a basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile, lyophilized powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum drying and spray-drying that yields a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof. The proper fluidity of a solution can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. Prolonged absorption of injectable compositions can be brought about by including, in the composition, an agent that delays absorption, for example, monostearate salts and gelatin.
The antibodies and antibody-portions of the present invention can be administered by a variety of methods known in the art, although for many therapeutic applications, the preferred route/mode of administration is subcutaneous injection, intravenous injection or infusion. As will be appreciated by the skilled artisan, the route and/or mode of administration will vary depending upon the desired results. In certain embodiments, the active compound may be prepared with a carrier that will protect the compound against rapid release, such as a controlled release formulation, including implants, transdermal patches, and microencapsulated delivery systems. Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Many methods for the preparation of such formulations are patented or generally known to those skilled in the art. See, e.g., Sustained and Controlled Release Drug Delivery Systems, J.R. Robinson, ed., Marcel Dekker, Inc., New York, 1978.
In certain embodiments, an antibody or antibody portion of the invention may be orally administered, for example, with an inert diluent or an assimilable edible carrier. The compound (and other ingredients, if desired) may also be enclosed in a hard or soft shell gelatin capsule, compressed into tablets, or incorporated directly into the subject's diet. For oral therapeutic administration, the compounds may be incorporated with excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, and the like. To administer a compound of the invention by other than parenteral administration, it may be necessary to coat the compound with, or co-administer the compound with, a material to prevent its inactivation.
Supplementary active compounds can also be incorporated into the compositions. In certain embodiments, an antibody or antibody portion of the invention is coformulated with and/or coadministered with one or more additional therapeutic agents that are useful for treating disorders in which IL-12 activity is detrimental. For example, an anti-hIL-12 antibody or antibody portion of the invention may be coformulated and/or coadministered with one or more additional antibodies that bind other targets {e.g., antibodies that bind other cytokines or that bind cell surface molecules). Furthermore, one or more antibodies of the invention may be used in combination with two or more of the foregoing therapeutic agents. Such combination therapies may advantageously utilize lower dosages of the administered therapeutic agents, thus avoiding possible toxicities or complications associated with the various monotherapies.
In certain embodiments, an antibody to IL-12 or fragment thereof is linked to a half-life extending vehicle known in the art. Such vehicles include, but are not limited to, the Fc domain, polyethylene glycol, and dextran. Such vehicles are described, e.g., in U.S. Application Serial No. 09/428,082 and published PCT Application No. WO 99/25044, which are hereby incorporated by reference for any purpose.
In a specific embodiment, nucleic acid sequences comprising nucleotide sequences encoding an antibody of the invention or another prophylactic or therapeutic agent of the invention are administered to treat, prevent, manage, or ameliorate a disorder or one or more symptoms thereof by way of gene therapy. Gene therapy refers to therapy performed by the administration to a subject of an expressed or expressible nucleic acid. In this embodiment of the invention, the nucleic acids produce their encoded antibody or prophylactic or therapeutic agent of the invention that mediates a prophylactic or therapeutic effect.
Any of the methods for gene therapy available in the art can be used according to the present invention. For general reviews of the methods of gene therapy, see Goldspiel et al., 1993, Clinical Pharmacy 12:488-505; Wu and Wu, 1991, Biotherapy 3:87-95; Tolstoshev, 1993, Ann. Rev. Pharmacol. Toxicol. 32:573-596; Mulligan, Science 260:926- 932 (1993); and Morgan and Anderson, 1993, Ann. Rev. Biochem. 62:191-217; May, 1993, TIBTECH 11(5): 155-215. Methods commonly known in the art of recombinant DNA technology which can be used are described in Ausubel et al. (eds.), Current Protocols in Molecular Biology, John Wiley &Sons, NY (1993); and Rriegler, Gene Transfer and Expression, A Laboratory Manual, Stockton Press, NY (1990). Detailed description of various methods of gene therapy are disclosed in US20050042664 Al which is incorporated herein by reference.
Interleukin 12 plays a critical role in the pathology associated with a variety of diseases involving immune and inflammatory elements. These diseases include, but are not limited to, rheumatoid arthritis, osteoarthritis, juvenile chronic arthritis, septic arthritis, Lyme arthritis, psoriatic arthritis, reactive arthritis, spondyloarthropathy, systemic lupus erythematosus, Crohn's disease, ulcerative colitis, inflammatory bowel disease, insulin dependent diabetes mellitus, thyroiditis, asthma, allergic diseases, psoriasis, dermatitis scleroderma, graft versus host disease, organ transplant rejection, acute or chronic immune disease associated with organ transplantation, sarcoidosis, atherosclerosis, disseminated intravascular coagulation, Kawasaki's disease, Grave's disease, nephrotic syndrome, chronic fatigue syndrome, Wegener's granulomatosis, Henoch-Schoenlein purpurea, microscopic vasculitis of the kidneys, chronic active hepatitis, uveitis, septic shock, toxic shock syndrome, sepsis syndrome, cachexia, infectious diseases, parasitic diseases, acquired immunodeficiency syndrome, acute transverse myelitis, Huntington's chorea, Parkinson's disease, Alzheimer's disease, stroke, primary biliary cirrhosis, hemolytic anemia, malignancies, heart failure, myocardial infarction, Addison's disease, sporadic, polyglandular deficiency type I and polyglandular deficiency type π, Schmidt's syndrome, adult (acute) respiratory distress syndrome, alopecia, alopecia areata, seronegative arthopathy, arthropathy, Reiter's disease, psoriatic arthropathy, ulcerative colitic arthropathy, enteropathic synovitis, chlamydia, yersinia and salmonella associated arthropathy, spondyloarthopathy, atheromatous disease/arteriosclerosis, atopic allergy, autoimmune bullous disease, pemphigus vulgaris, pemphigus foliaceus, pemphigoid, linear IgA disease, autoimmune haemolytic anaemia, Coombs positive haemolytic anaemia, acquired pernicious anaemia, juvenile pernicious anaemia, myalgic encephalitis/Royal Free Disease, chronic mucocutaneous candidiasis, giant cell arteritis, primary sclerosing hepatitis, cryptogenic autoimmune hepatitis, Acquired Immunodeficiency Disease Syndrome, Acquired Immunodeficiency Related Diseases, Hepatitis B, Hepatitis C, common varied immunodeficiency (common variable hypogammaglobulinaemia), dilated cardiomyopathy, female infertility, ovarian failure, premature ovarian failure, fibrotic lung disease, cryptogenic fibrosing alveolitis, post-inflammatory interstitial lung disease, interstitial pneumonitis, connective tissue disease associated interstitial lung disease, mixed connective tissue disease associated lung disease, systemic sclerosis associated interstitial lung disease, rheumatoid arthritis associated interstitial lung disease, systemic lupus erythematosus associated lung disease, dermatomyositis/polymyositis associated lung disease, Sjogren's disease associated lung disease, ankylosing spondylitis associated lung disease, vasculitic diffuse lung disease, haemosiderosis associated lung disease, drug-induced interstitial lung disease, fibrosis, radiation fibrosis, bronchiolitis obliterans, chronic eosinophilic pneumonia, lymphocytic infiltrative lung disease, postinfectious interstitial lung disease, gouty arthritis, autoimmune hepatitis, type-1 autoimmune hepatitis (classical autoimmune or lupoid hepatitis), type-2 autoimmune hepatitis (anti-LKM antibody hepatitis), autoimmune mediated hypoglycaemia, type B insulin resistance with acanthosis nigricans, hypoparathyroidism, acute immune disease associated with organ transplantation, chronic immune disease associated with organ transplantation, osteoarthrosis, primary sclerosing cholangitis, psoriasis type 1, psoriasis type 2, idiopathic leucopaenia, autoimmune neutropaenia, renal disease NOS, glomerulonephritides, microscopic vasulitis of the kidneys, lyme disease, discoid lupus erythematosus, male infertility idiopathic or NOS, sperm autoimmunity, multiple sclerosis (all subtypes), sympathetic ophthalmia, pulmonary hypertension secondary to connective tissue disease, Goodpasture's syndrome, pulmonary manifestation of polyarteritis nodosa, acute rheumatic fever, rheumatoid spondylitis, Still's disease, systemic sclerosis, Sjorgren's syndrome, Takayasu's disease/arteritis, autoimmune thrombocytopaenia, idiopathic thrombocytopaenia, autoimmune thyroid disease, hyperthyroidism, goitrous autoimmune hypothyroidism (Hashimoto's disease), atrophic autoimmune hypothyroidism, primary myxoedema, phacogenic uveitis, primary vasculitis, vitiligo acute liver disease, chronic liver diseases, alcoholic cirrhosis, alcohol-induced liver injury, choleosatatis, idiosyncratic liver disease, Drug-Induced hepatitis, Non-alcoholic Steatohepatitis, allergy and asthma, group B streptococci (GBS) infection, mental disorders (e.g., depression and schizophrenia), Th2 Type and ThI Type mediated diseases, acute and chronic pain (different forms of pain), and cancers such as lung, breast, stomach, bladder, colon, pancreas, ovarian, prostate and rectal cancer and hematopoietic malignancies (leukemia and lymphoma), Abetalipoprotemia, Acrocyanosis, acute and chronic parasitic or infectious processes, acute leukemia, acute lymphoblastic leukemia (ALL), acute myeloid leukemia (AML), acute or chronic bacterial infection, acute pancreatitis, acute renal failure, adenocarcinomas, aerial ectopic beats, ADDS dementia complex, alcohol-induced hepatitis, allergic conjunctivitis, allergic contact dermatitis, allergic rhinitis, allograft rejection, alpha-1- antitrypsin deficiency, amyotrophic lateral sclerosis, anemia, angina pectoris, anterior horn cell degeneration, anti cd3 therapy, antiphospholipid syndrome, anti-receptor hypersensitivity reactions, aordic and peripheral aneuryisms, aortic dissection, arterial hypertension, arteriosclerosis, arteriovenous fistula, ataxia, atrial fibrillation (sustained or paroxysmal), atrial flutter, atrioventricular block, B cell lymphoma, bone graft rejection, bone marrow transplant (BMT) rejection, bundle branch block, Burkitt's lymphoma, Burns, cardiac arrhythmias, cardiac stun syndrome, cardiac tumors, cardiomyopathy, cardiopulmonary bypass inflammation response, cartilage transplant rejection, cerebellar cortical degenerations, cerebellar disorders, chaotic or multifocal atrial tachycardia, chemotherapy associated disorders, chromic myelocytic leukemia (CML), chronic alcoholism, chronic inflammatory pathologies, chronic lymphocytic leukemia (CLL), chronic obstructive pulmonary disease (COPD), chronic salicylate intoxication, colorectal carcinoma, congestive heart failure, conjunctivitis, contact dermatitis, cor pulmonale, coronary artery disease, Creutzfeldt-Jakob disease, culture negative sepsis, cystic fibrosis, cytokine therapy associated disorders, Dementia pugilistica, demyelinating diseases, dengue hemorrhagic fever, dermatitis, dermatologic conditions, diabetes, diabetes mellitus, diabetic ateriosclerotic disease, Diffuse Lewy body disease, dilated congestive cardiomyopathy, disorders of the basal ganglia, Down's Syndrome in middle age, drug- induced movement disorders induced by drugs which block CNS dopamine receptors, drug sensitivity, eczema, encephalomyelitis, endocarditis, endocrinopathy, epiglottitis, epstein-barr virus infection, erythromelalgia, extrapyramidal and cerebellar disorders, familial hematophagocytic lymphohistiocytosis, fetal thymus implant rejection, Friedreich's ataxia, functional peripheral arterial disorders, fungal sepsis, gas gangrene, gastric ulcer, glomerular nephritis, graft rejection of any organ or tissue, gram negative sepsis, gram positive sepsis, granulomas due to intracellular organisms, hairy cell leukemia, Hallerrorden-Spatz disease, hashimoto's thyroiditis, hay fever, heart transplant rejection, hemachromatosis, hemodialysis, hemolytic uremic syndrome/thrombolytic thrombocytopenic purpura, hemorrhage, hepatitis (A), His bundle arrythmias, HTV infection/HIV neuropathy, Hodgkin's disease, hyperkinetic movement disorders, hypersensitity reactions, hypersensitivity pneumonitis, hypertension, hypokinetic movement disorders, hypothalamic-pituitary-adrenal axis evaluation, idiopathic Addison's disease, idiopathic pulmonary fibrosis, antibody mediated cytotoxicity, Asthenia, infantile spinal muscular atrophy, inflammation of the aorta, influenza a, ionizing radiation exposure, iridocyclitis/uveitis/optic neuritis, ischemia- reperfusion injury, ischemic stroke, juvenile rheumatoid arthritis, juvenile spinal muscular atrophy, Kaposi's sarcoma, kidney transplant rejection, legionella, leishmaniasis, leprosy, lesions of the corticospinal system, lipedema, liver transplant rejection, lymphederma, malaria, malignamt Lymphoma, malignant histiocytosis, malignant melanoma, meningitis, meningococcemia, metabolic/idiopathic, migraine headache, mitochondrial multi.system disorder, mixed connective tissue disease, monoclonal gammopathy, multiple myeloma, multiple systems degenerations (Mencel Dejerine- Thomas Shi- Drager and Machado- Joseph), myasthenia gravis, mycobacterium avium intracellulare, mycobacterium tuberculosis, myelodyplastic syndrome, myocardial infarction, myocardial ischemic disorders, nasopharyngeal carcinoma, neonatal chronic lung disease, nephritis, nephrosis, neurodegenerative diseases, neurogenic I muscular atrophies , neutropenic fever, non- hodgkins lymphoma, occlusion of the abdominal aorta and its branches, occulsive arterial disorders, okt3 therapy, orchitis/epidydimitis, orchitis/vasectomy reversal procedures, organomegaly, osteoporosis, pancreas transplant rejection, pancreatic carcinoma, paraneoplastic syndrome/hypercalcemia of malignancy, parathyroid transplant rejection, pelvic inflammatory disease, perennial rhinitis, pericardial disease, peripheral atherlosclerotic disease, peripheral vascular disorders, peritonitis, pernicious anemia, Pneumocystis carinii pneumonia, pneumonia, POEMS syndrome (polyneuropathy, organomegaly, endocrinopathy, monoclonal gammopathy, and skin changes syndrome), post perfusion syndrome, post pump syndrome, post-MI cardiotomy syndrome, preeclampsia, Progressive supranucleo Palsy, primary pulmonary hypertension, radiation therapy, Raynaud's phenomenon and disease, Raynoud's disease, Ref sum's disease, regular narrow QRS tachycardia, renovascular hypertension, reperfusion injury, restrictive cardiomyopathy, sarcomas, scleroderma, senile chorea, Senile Dementia of Lewy body type, seronegative arthropathies, shock, sickle cell anemia, skin allograft rejection, skin changes syndrome, small bowel transplant rejection, solid tumors, specific arrythmias, spinal ataxia, spinocerebellar degenerations, streptococcal myositis, structural lesions of the cerebellum, Subacute sclerosing panencephalitis, Syncope, syphilis of the cardiovascular system, systemic anaphalaxis, systemic inflammatory response syndrome, systemic onset juvenile rheumatoid arthritis, T-cell or FAB ALL, Telangiectasia, thromboangitis obliterans, thrombocytopenia, toxicity, transplants, trauma/hemorrhage, type III hypersensitivity reactions, type IV hypersensitivity, unstable angina, uremia, urosepsis, urticaria, valvular heart diseases, varicose veins, vasculitis, venous diseases, venous thrombosis, ventricular fibrillation, viral and fungal infections, vital encephalitis/aseptic meningitis, vital-associated hemaphagocytic syndrome, Wernicke- Korsakoff syndrome, Wilson's disease, xenograft rejection of any organ or tissue, (see Peritt et al. PCT publication No. WO2002097048A2, Leonard et al., PCT publication No. WO9524918 Al, and Salfeld et al., PCT publication No. WO00/56772A1).
The antibodies, and antibody portions of the invention can be used to treat humans suffering from autoimmune diseases, in particular those associated with inflammation, including, rheumatoid spondylitis, allergy, autoimmune diabetes, autoimmune uveitis. Preferably, the antibodies of the invention or antigen-binding portions thereof, are used to treat rheumatoid arthritis, Crohn's disease, multiple sclerosis, insulin dependent diabetes mellitus and psoriasis.
An antibody, or antibody portion, of the invention also can be administered with one or more additional therapeutic agents useful in the treatment of autoimmune and inflammatory diseases.
Antibodies of the invention, or antigen binding portions thereof can be used alone or in combination to treat such diseases. It should be understood that the antibodies of the invention or antigen binding portion thereof can be used alone or in combination with an additional agent, e.g., a therapeutic agent, said additional agent being selected by the skilled artisan for its intended purpose. For example, the additional agent can be a therapeutic agent art-recognized as being useful to treat the disease or condition being treated by the antibody of the present invention. The additional agent also can be an agent that imparts a beneficial attribute to the therapeutic composition e.g., an agent which effects the viscosity of the composition.
It should further be understood that the combinations which are to be included within this invention are those combinations useful for their intended purpose. The agents set forth below are illustrative for purposes and not intended to be limited. The combinations, which are part of this invention, can be the antibodies of the present invention and at least one additional agent selected from the lists below. The combination can also include more than one additional agent, e.g., two or three additional agents if the combination is such that the formed composition can perform its intended function.
Binding proteins described herein may be used in combination with additional therapeutic agents such as a Disease Modifying Anti-Rheumatic Drug (DMARD) or a Nonsteroidal Antiinflammatory Drug (NSAID) or a steroid or any combination thereof. Preferred examples of a DMARD are hydroxychloroquine, leflunomide, methotrexate, parenteral gold, oral gold and sulfasalazine. Preferred examples of non-steroidal anti-inflammatory drug(s) also referred to as NSAIDS include drugs like ibuprofen. Other preferred combinations are corticosteroids including prednisolone; the well known side effects of steroid use can be reduced or even eliminated by tapering the steroid dose required when treating patients in combination with the anti-IL-12 antibodies of this invention. Non-limiting examples of therapeutic agents for rheumatoid arthritis with which an antibody, or antibody portion, of the invention can be combined include the following: cytokine suppressive anti-inflammatory drug(s) (CSAIDs); antibodies to or antagonists of other human cytokines or growth factors, for example, TNF, LT, IL-I, IL-2, IL-3, IL-4, IL-5, IL-6, IL-I, IL-S, IL-15, IL-16, IL-18, IL-21, IL-23, interferons, EMAP-II, GM-CSF, FGF, and PDGF. Antibodies of the invention, or antigen binding portions thereof, can be combined with antibodies to cell surface molecules such as CD2, CD3, CD4, CD8, CD25, CD28, CD30, CD40, CD45, CD69, CD80 (B7.1), CD86 (B7.2), CD90, CTLA or their ligands including CD154 (gp39 or CD40L).
Preferred combinations of therapeutic agents may interfere at different points in the autoimmune and subsequent inflammatory cascade; preferred examples include TNF antagonists such as soluble p55 or p75 TNF receptors, derivatives, thereof, (ρ75TNFRlgG (Enbrel™) or p55TNFRlgG (Lenercept), chimeric, humanized or human TNF antibodies, or a fragment thereof, including infliximab (Remicade®, Johnson and Johnson; described in U.S. Patent No. 5,656,272, incorporated by reference herein), CDP571 (a humanized monoclonal anti-TNF-alpha IgG4 antibody), CDP 870 (a humanized monoclonal anti-TNF-alpha antibody fragment), an anti- TNF dAb (Peptech), CNTO 148 (golimumab; Medarex and Centocor, see WO 02/12502), and adalimumab (Humira® Abbott Laboratories, a human anti-TNF mAb, described in US 6,090,382 as D2E7), . Additional TNF antibodies which can be used in the invention are described in U.S. Patent Nos. 6,593,458; 6,498,237; 6,451,983; and 6,448,380, each of which is incorporated by reference herein. Other combinations including TNFα converting enzyme (TACE) inhibitors; IL- 1 inhibitors (Interleukin-1 -converting enzyme inhibitors, 1L-1RA etc.) may be effective for the same reason. Other preferred combinations include Merleukin 11. Yet another preferred combination are other key players of the autoimmune response which may act parallel to, dependent on or in concert with IL-12 function; especially preferred are IL-18 antagonists including IL-18 antibodies or soluble IL-18 receptors, or IL-18 binding proteins. It has been shown that IL-12 and IL-18 have overlapping but distinct functions and a combination of antagonists to both may be most effective. Yet another preferred combination are non-depleting anti-CD4 inhibitors. Yet other preferred combinations include antagonists of the co-stimulatory pathway CD80 (B7.1) or CD86 (B7.2) including antibodies, soluble receptors or antagonistic ligands.
The antibodies of the invention, or antigen binding portions thereof, may also be combined with agents, such as methotrexate, 6-MP, azathioprine sulphasalazine, mesalazine, olsalazine chloroquinine/hydroxychloroquine, pencillamine, aurothiomalate (intramuscular and oral), azathioprine, cochicine, corticosteroids (oral, inhaled and local injection), beta-2 adrenoreceptor agonists (salbutamol, terbutaline, salmeteral), xanthines (theophylline, aminophylline), cromoglycate, nedocromil, ketotifen, ipratropium and oxitropium, cyclosporin, FK506, rapamycin, mycophenolate mofetil, leflunomide, NSAIDs, for example, ibuprofen, corticosteroids such as prednisolone, phosphodiesterase inhibitors, adensosine agonists, antithrombotic agents, complement inhibitors, adrenergic agents, agents which interfere with signalling by proinflammatory cytokines such as TNFα or IL-I (e.g. IRAK, NTK, IKK , p38 or MAP kinase inhibitors), IL-lβ converting enzyme inhibitors, TNFq converting enzyme (TACE) inhibitors, T-cell signalling inhibitors such as kinase inhibitors, metalloproteinase inhibitors, sulfasalazine, azathioprine, 6-mercaptopurines, angiotensin converting enzyme inhibitors, soluble cytokine receptors and derivatives thereof (e.g. soluble p55 or p75 TNF receptors and the derivatives p75TNFRIgG (Enbrel™ and p55TNFRIgG (Lenercept)), sIL-lRI, sIL-lRIL sIL-6R), antiinflammatory cytokines (e.g. IL-4, IL-10, IL-Il, IL-13 and TGFβ), celecoxib, folic acid, hydroxychloroquine sulfate, rofecoxib, etanercept, infliximab, naproxen, valdecoxib, sulfasalazine, methylprednisolone, meloxicam, methylprednisolone acetate, gold sodium thiomalate, aspirin, triamcinolone acetonide, propoxyphene napsylate/apap, folate, nabumetone, diclofenac, piroxicam, etodolac, diclofenac sodium, oxaprozin, oxycodone hcl, hydrocodone bitartrate/apap, diclofenac sodium/misoprostol, fentanyl, anakinra, human recombinant, tramadol hcl, salsalate, sulindac, cyanocobalamin/fa/pyridoxine, acetaminophen, alendronate sodium, prednisolone, morphine sulfate, lidocaine hydrochloride, indomethacin, glucosamine sulf/chondroitin, amitriptyline hcl, sulfadiazine, oxycodone hcl/acetaminophen, olopatadine hcl, misoprostol, naproxen sodium, omeprazole, cyclophosphamide, rituximab, IL-I TRAP, MRA, CTLA4-IG, IL-18 BP, anti-IL-18, Anti-IL15, BIRB-796, SCIO-469, VX-702, AMG-548, VX- 740, Roflumilast, IC-485, CDC-801, and Mesopram. Preferred combinations include methotrexate or lefiunomide and in moderate or severe rheumatoid arthritis cases, cyclosporine.
Nonlimiting additional agents which can also be used in combination with an IL-12 or IL-23 antibody, or antigen-binding portion thereof, to treat rheumatoid arthritis include, but are not limited to, the following: non-steroidal anti-inflammatory drug(s) (NSAIDs); cytokine suppressive anti-inflammatory drug(s) (CSADDs); CDP-571/BAY-10-3356 (humanized anti- TNFα antibody; Celltech/Bayer); cA2/infliximab (chimeric anti-TNFα antibody; Centocor); 75 kdTNFR-IgG/etanercept (75 kD TNF receptor-IgG fusion protein; Immunex; see e.g., Arthritis & Rheumatism (1994) Vol. 37, S295; /. Invest. Med. (1996) Vol. 44, 235A); 55 kdTNF-IgG (55 kD TNF receptor-IgG fusion protein; Hoffmann-LaRoche); IDEC-CE9.1/SB 210396 (non-depleting primatized anti-CD4 antibody; IDEC/SmithKline; see e.g., Arthritis & Rheumatism (1995) Vol. 38, S185); DAB 486-IL-2 and/or DAB 389-1L-2 (IL-2 fusion proteins; Seragen; see e.g., Arthritis & Rheumatism (1993) Vol. 36, 1223); Anti-Tac (humanized anti-IL-2Rα; Protein Design Labs/Roche); EL-4 (anti-inflammatory cytokine; DNAX/Schering); IL-10 (SCH 52000; recombinant IL-10, anti-inflammatory cytokine; DNAX/Schering); IL-4; EL-IO and/or IL-4 agonists (e.g., agonist antibodies); IL-IRA (IL-I receptor antagonist; Synergen/Amgen); anakinra (Kineret®/Amgen); TNF-bp/s-TNF (soluble TNF binding protein; see e.g., Arthritis & Rheumatism (1996) Vol. 39, No. 9 (supplement), S284; Amer. J. Physiol. - Heart and Circulatory Physiology (1995) Vol. 268, pp. 37-42); R973401 (phosphodiesterase Type IV inhibitor; see e.g., Arthritis & Rheumatism (1996) Vol. 39, No. 9 (supplement), S282); MK-966 (COX-2 Inhibitor; see e.g., Arthritis & Rheumatism (1996) Vol. 39, No. 9 (supplement), S81); Iloprost (see e.g., Arthritis & Rheumatism (1996) Vol. 39, No. 9 (supplement), S82); methotrexate; thalidomide (see e.g., Arthritis & Rheumatism (1996) Vol. 39, No. 9 (supplement), S282) and thalidomide- related drugs (e.g., Celgen); lefiunomide (anti-inflammatory and cytokine inhibitor; see e.g., Arthritis & Rheumatism (1996) Vol. 39, No. 9 (supplement), S131; Inflammation Research (1996) Vol. 45, pp. 103-107); tranexamic acid (inhibitor of plasminogen activation; see e.g., Arthritis & Rheumatism (1996) Vol. 39, No. 9 (supplement), S284); T-614 (cytokine inhibitor; see e.g., Arthritis & Rheumatism (1996) Vol. 39, No. 9 (supplement), S282); prostaglandin El (see e.g., Arthritis & Rheumatism (1996) Vol. 39, No. 9 (supplement), S282); Tenidap (nonsteroidal anti-inflammatory drug; see e.g., Arthritis & Rheumatism (1996) Vol. 39, No. 9 (supplement), S280); Naproxen (non-steroidal anti-inflammatory drug; see e.g., Neuro Report (1996) Vol. 7, pp. 1209-1213); Meloxicam (non-steroidal anti-inflammatory drug); Ibuprofen (non-steroidal anti-inflammatory drug); Piroxicam (non-steroidal anti-inflammatory drug); Diclofenac (non-steroidal anti-inflammatory drug); Indomethacin (non-steroidal antiinflammatory drug); Sulfasalazine (see e.g., Arthritis & Rheumatism (1996) Vol. 39, No. 9 (supplement), S281); Azathioprine (see e.g., Arthritis & Rheumatism (1996) Vol. 39, No. 9 (supplement), S281); ICE inhibitor (inhibitor of the enzyme interleukin-lβ converting enzyme); zap-70 and/or lck inhibitor (inhibitor of the tyrosine kinase zap-70 or lck); VEGF inhibitor and/or VEGF-R inhibitor (inhibitors of vascular endothelial cell growth factor or vascular endothelial cell growth factor receptor; inhibitors of angiogenesis); corticosteroid anti-inflammatory drugs (e.g., SB203580); TNF-convertase inhibitors; anti-IL-12 antibodies; anti-IL-18 antibodies; interleukin-11 (see e.g., Arthritis & Rheumatism (1996) Vol. 39, No. 9 (supplement), S296); interleukin-13 (see e.g., Arthritis & Rheumatism (1996) Vol. 39, No. 9 (supplement), S308); interleukin -17 inhibitors (see e.g., Arthritis & Rheumatism (1996) Vol. 39, No. 9 (supplement), S 120); gold; penicillamine; chloroquine; chlorambucil; hydroxychloroquine; cyclosporine; cyclophosphamide; total lymphoid irradiation; anti-thymocyte globulin; anti-CD4 antibodies; CD5-toxins; orally-administered peptides and collagen; lobenzarit disodium; Cytokine Regulating Agents (CRAs) HP228 and HP466 (Houghten Pharmaceuticals, Inc.); ICAM-I antisense phosphorothioate oligo-deoxynucleotides (ISIS 2302; Isis Pharmaceuticals, Inc.); soluble complement receptor 1 (TPlO; T Cell Sciences, Inc.); prednisone; orgotein; glycosaminoglycan polysulphate; minocycline; anti-IL2R antibodies; marine and botanical lipids (fish and plant seed fatty acids; see e.g., DeLuca et al. (1995) Rheum. Dis. Clin. North Am. 21:759-777); auranofm; phenylbutazone; meclofenamic acid; flufenamic acid; intravenous immune globulin; zileuton; azaribine; mycophenolic acid (RS-61443); tacrolimus (FK-506); sirolimus (rapamycin); amiprilose (therafectin); cladribine (2-chlorodeoxyadenosine); methotrexate; antivirals; and immune modulating agents.
In one embodiment, the IL-12 antibody, or antigen-binding portion thereof, is administered in combination with one of the following agents for the treatment of rheumatoid arthritis: small molecule inhibitor of KDR (ABT-123), small molecule inhibitor of Tie-2; methotrexate; prednisone; celecoxib; folic acid; hydroxychloroquine sulfate; rofecoxib; etanercept; infliximab; leflunomide; naproxen; valdecoxib; sulfasalazine; methylprednisolone; ibuprofen; meloxicam; methylprednisolone acetate; gold sodium thiomalate; aspirin; azathioprine; triamcinolone acetonide; propxyphene napsylate/apap; folate; nabumetone; diclofenac; piroxicam; etodolac; diclofenac sodium; oxaprozin; oxycodone hcl; hydrocodone bitartrate/apap; diclofenac sodium/misoprostol; fentanyl; anakinra, human recombinant; tramadol hcl; salsalate; sulindac; cyanocobalamin/fa/pyridoxine; acetaminophen; alendronate sodium; prednisolone; morphine sulfate; lidocaine hydrochloride; indomethacin; glucosamine sulfate/chondroitin; cyclosporine; amitriptyline hcl; sulfadiazine; oxycodone hcl/acetaminophen; olopatadine hcl; misoprostol; naproxen sodium; omeprazole; mycophenolate mofetil; cyclophosphamide; rituximab; IL-I TRAP; MRA; CTLA4-IG; IL-18 BP; ABT-874; ABT-325 (anti-IL 18); anti-IL 15; BIRB-796; SCIO-469; VX-702; AMG-548; VX-740; Roflumilast; IC- 485; CDC-801; and mesopram. In another embodiment, an IL-12 or IL-23 antibody, or antigen- binding portion thereof, is administered for the treatment of an IL-12 or IL-23 related disorder in combination with one of the above mentioned agents for the treatment of rheumatoid arthritis.
Non-limiting examples of therapeutic agents for inflammatory bowel disease with which an antibody, or antibody portion, of the invention can be combined include the following: budenoside; epidermal growth factor; corticosteroids; cyclosporin, sulfasalazine; aminosalicylates; 6-mercaptopurine; azathioprine; metronidazole; lipoxygenase inhibitors; mesalamine; olsalazine; balsalazide; antioxidants; thromboxane inhibitors; IL-I receptor antagonists; anti-IL-lβ monoclonal antibodies; anti-IL-6 monoclonal antibodies; growth factors; elastase inhibitors; pyridinyl-imidazole compounds; antibodies to or antagonists of other human cytokines or growth factors, for example, TNF, LT, IL-I, IL-2, IL-6, EL-7, IL-8, IL-15, DL-16, TL- 17, IL-18, EMAP-π, GM-CSF, FGF, and PDGF. Antibodies of the invention, or antigen binding portions thereof, can be combined with antibodies to cell surface molecules such as CD2, CD3, CD4, CD8, CD25, CD28, CD30, CD40, CD45, CD69, CD90 or their ligands. The antibodies of the invention, or antigen binding portions thereof, may also be combined with agents, such as methotrexate, cyclosporin, FK506, rapamycin, mycophenolate mofetil, leflunomide, NSABDs, for example, ibuprofen, corticosteroids such as prednisolone, phosphodiesterase inhibitors, adenosine agonists, antithrombotic agents, complement inhibitors, adrenergic agents, agents which interfere with signalling by proinflammatory cytokines such as TNFα or IL-I (e.g. IRAK, NIK, IKK, p38 or MAP kinase inhibitors), IL-lβ converting enzyme inhibitors, TNFα converting enzyme inhibitors, T-cell signalling inhibitors such as kinase inhibitors, metalloproteinase inhibitors, sulfasalazine, azathioprine, 6-mercaptopurmes, angiotensin converting enzyme inhibitors, soluble cytokine receptors and derivatives thereof (e.g. soluble p55 or p75 TNF receptors, sIL-lRI, sIL-lRIL sIL-6R) and antiinflammatory cytokines (e.g. IL-4, IL-10, IL-Il, IL- 13 and TGFβ).
Preferred examples of therapeutic agents for Crohn's disease in which an antibody or an antigen binding portion can be combined include the following: TNF antagonists, for example, anti-TNF antibodies, D2E7 (PCT Publication No. WO 97/29131; HUMIRA), CA2 (REMICADE), CDP 571, TNFR-Ig constructs, (p75TNFRIgG (ENBREL) and p55TNFRIgG (LENERCEPT)) inhibitors and PDE4 inhibitors. Antibodies of the invention, or antigen binding portions thereof, can be combined with corticosteroids, for example, budenoside and dexamethasone. Antibodies of the invention or antigen binding portions thereof, may also be combined with agents such as sulfasalazine, 5-aminosalicylic acid and olsalazine, and agents which interfere with synthesis or action of proinflammatory cytokines such as IL-I, for example, IL-lβ converting enzyme inhibitors and IL-lra. Antibodies of the invention or antigen binding portion thereof may also be used with T cell signaling inhibitors, for example, tyrosine kinase inhibitors 6-mercaptopurines. Antibodies of the invention, or antigen binding portions thereof, can be combined with IL-Il. Antibodies of the invention, or antigen binding portions thereof, can be combined with mesalamine, prednisone, azathioprine, mercaptopurine, infliximab, methylprednisolone sodium succinate, diphenoxylate/atrop sulfate, loperamide hydrochloride, methotrexate, omeprazole, folate, ciprofloxacin/dextrose-water, hydrocodone bitartrate/apap, tetracycline hydrochloride, fluocinonide, metronidazole, thimerosal/boric acid, cholestyramine/sucrose, ciprofloxacin hydrochloride, hyoscyamine sulfate, meperidine hydrochloride, midazolam hydrochloride, oxycodone hcl/acetaminophen, promethazine hydrochloride, sodium phosphate, sulfamethoxazole/trimethoprim, celecoxib, polycarbophil, propoxyphene napsylate, hydrocortisone, multivitamins, balsalazide disodium, codeine phosphate/apap, colesevelam hcl, cyanocobalamin, folic acid, levofloxacin, methylprednisolone, natalizumab and interferon-gamma
Non-limiting examples of therapeutic agents for multiple sclerosis with which an antibody, or antibody portion, of the invention can be combined include the following: corticosteroids; prednisolone; methylprednisolone; azathioprine; cyclophosphamide; cyclosporine; methotrexate; 4-ammopyridine; tizanidine; interferon-βla (AVONEX; Biogen); interferon-βlb (BETASERON; Chiron/Berlex); interferon α-n3) (Interferon Sciences/Fujimoto), interferon-α (Alfa Wassermann/J&J), interferon β IA-IF (Serono/Inhale Therapeutics), Peginterferon α 2b (Enzon/Schering-Plough), Copolymer 1 (Cop-1; COPAXONE; Teva Pharmaceutical Industries, Inc.); hyperbaric oxygen; intravenous immunoglobulin; clabribine; antibodies to or antagonists of other human cytokines or growth factors and their receptors, for example, TNF, LT, IL-I, DL-2, IL-6, IL-7, IL-8, 1L-23, IL-15, IL-16, IL-18, EMAP-II, GM-CSF, FGF, and PDGF. Antibodies of the invention, or antigen binding portions thereof, can be combined with antibodies to cell surface molecules such as CD2, CD3, CD4, CD8, CD19, CD20, CD25, CD28, CD30, CD40, CD45, CD69, CD80, CD86, CD90 or their ligands. The antibodies of the invention, or antigen binding portions thereof, may also be combined with agents, such as methotrexate, cyclosporine, FK506, rapamycin, mycophenolate mofetil, leflunomide, NSAIDs, for example, ibuprofen, corticosteroids such as prednisolone, phosphodiesterase inhibitors, adensosine agonists, antithrombotic agents, complement inhibitors, adrenergic agents, agents which interfere with signalling by proinflammatory cytokines such as TNFα or IL-I (e.g. IRAK, NIK, IKK, p38 or MAP kinase inhibitors), EL-lβ converting enzyme inhibitors, TACE inhibitors, T-cell signaling inhibitors such as kinase inhibitors, metalloproteinase inhibitors, sulfasalazine, azathioprine, 6-mercaptoρurines, angiotensin converting enzyme inhibitors, soluble cytokine receptors and derivatives thereof (e.g. soluble p55 or p75 TNF receptors, sIL-lRI, sIL-lRII, sIL- 6R) and antiinflammatory cytokines (e.g. IL-4, IL-10, IL-13 and TGFβ).
Preferred examples of therapeutic agents for multiple sclerosis in which the antibody or antigen binding portion thereof can be combined to include interferon-β, for example, IFNβla and IFNβlb; Copaxone, corticosteroids, caspase inhibitors, for example inhibitors of caspase-1, IL-I inhibitors, TNF inhibitors, and antibodies to CD40 ligand and CD80.
The antibodies of the invention, or antigen binding portions thereof, may also be combined with agents, such as alemtuzumab, dronabinol, Unimed, daclizumab, mitoxantrone, xaliproden hydrochloride, fampridine, glatiramer acetate, natalizumab, sinnabidol, a-immunokine NNSO3, ABR-215062, AnergiX.MS, chemokine receptor antagonists, BBR-2778, calagualine, CPI-1189, LEM (liposome encapsulated mitoxantrone), THCCBD (cannabinoid agonist) MBP- 8298, mesopram (PDE4 inhibitor), MNA-715, anti-IL-6 receptor antibody, neurovax, pirfenidone allotrap 1258 (RDP-1258), sTNF-Rl, talampanel, teriflunomide,TGF-beta2, tiplimotide, VLA-4 antagonists (for example, TR-14035, VLA4 Ultrahaler, Antegran-ELAN/Biogen), interferon gamma antagonists, IL-4 agonists.
Non-limiting examples of therapeutic agents for Angina with which an antibody, or antibody portion, of the invention can be combined include the following: aspirin, nitroglycerin, isosorbide mononitrate, metoprolol succinate, atenolol, metoprolol tartrate, amlodipine besylate, diltiazem hydrochloride, isosorbide dinitrate, clopidogrel bisulfate, nifedipine, atorvastatin calcium, potassium chloride, furosemide, simvastatin, verapamil hcl, digoxin, propranolol hydrochloride, carvedilol, lisinopril, spironolactone, hydrochlorothiazide, enalapril maleate, nadolol, ramipril, enoxaparin sodium, heparin sodium, valsartan, sotalol hydrochloride, fenofibrate, ezetimibe, bumetanide, losartan potassium, lisinopril/hydrochlorothiazide, felodipine, captopril, bisoprolol fumarate.
Non-limiting examples of therapeutic agents for Ankylosing Spondylitis with which an antibody, or antibody portion, of the invention can be combined include the following: ibuprofen, diclofenac and misoprostol, naproxen, meloxicam, indomethacin, diclofenac, celecoxib, rofecoxib, Sulfasalazine, Methotrexate, azathioprine, minocyclin, prednisone, etanercept, infliximab.
Non-limiting examples of therapeutic agents for Asthma with which an antibody, or antibody portion, of the invention can be combined include the following: albuterol, salmeterol/fluticasone, montelukast sodium, fluticasone propionate, budesonide, prednisone, salmeterol xinafoate, levalbuterol hcl, albuterol sulfate/ipratropium, prednisolone sodium phosphate, triamcinolone acetonide, beclomethasone dipropionate, ipratropium bromide, azithromycin, pirbuterol acetate, prednisolone, theophylline anhydrous, methylprednisolone sodium succinate, clarithromycin, zafirlukast, formoterol fumarate, influenza virus vaccine, methylprednisolone, amoxicillin trihydrate, flunisolide, allergy injection, cromolyn sodium, fexofenadine hydrochloride, flunisolide/menthol, amoxicillin/clavulanate, levofloxacin, inhaler assist device, guaifenesin, dexamethasone sodium phosphate, moxifloxacin hcl, doxycycline hyclate, guaifenesin/d-methorphan, p-ephedrine/cod/chlorphenir, gatifloxacin, cetirizine hydrochloride, mometasone furoate, salmeterol xinafoate, benzonatate, cephalexin, pe/hydrocodone/chlorphenir, cetirizine hcl/pseudoephed, phenylephrine/cod/promethazine, codeine/promethazine, cefprozil, dexamethasone, guaifenesin/pseudoephedrine, chlorpheniramine/hydrocodone, nedocromil sodium, terbutaline sulfate, epinephrine, methylprednisolone, metaproterenol sulfate.
Non-limiting examples of therapeutic agents for COPD with which an antibody, or antibody portion, of the invention can be combined include the following: albuterol sulfate/ipratropium, ipratropium bromide, salmeterol/fluticasone, albuterol, salmeterol xinafoate, fluticasone propionate, prednisone, theophylline anhydrous, methylprednisolone sodium succinate, montelukast sodium, budesonide, formoterol fumarate, triamcinolone acetonide, levofloxacin, guaifenesin, azithromycin, beclomethasone dipropionate, levalbuterol hcl, flunisolide, ceftriaxone sodium, amoxicillin trihydrate, gatifloxacin, zafirlukast, amoxicillin/clavulanate, flunisolide/menthol, chlorpheniramine/hydrocodone, metaproterenol sulfate, methylprednisolone, mometasone furoate, p-ephedrine/cod/chlorphenir, pirbuterol acetate, p-ephedrine/loratadine, terbutaline sulfate, tiotropium bromide, (R,R)-formoterol, TgAAT, Cilomilast, Roflumilast.
Non-limiting examples of therapeutic agents for HCV with which an antibody, or antibody portion, of the invention can be combined include the following: Interferon-alpha-2a, Interferon-alpha-2b, Lnterferon-alpha conl, Interferon-alpha-nl, Pegylated interferon-alpha-2a, Pegylated interferon-alpha-2b, ribavirin, Peginterferon alfa-2b + ribavirin, Ursodeoxycholic Acid, Glycyrrhizic Acid, Thymalfasin, Maxamine, VX-497 and any compounds that are used to treat HCV through intervention with the following targets:HCV polymerase, HCV protease, HCV helicase, HCV IRES (internal ribosome entry site).
Non-limiting examples of therapeutic agents for Idiopathic Pulmonary Fibrosis with which an antibody, or antibody portion, of the invention can be combined include the following: prednisone, azathioprine, albuterol, colchicine, albuterol sulfate, digoxin, gamma interferon, methylprednisolone sod succ, lorazepam, furosemide, lisinopril, nitroglycerin, spironolactone, cyclophosphamide, ipratropium bromide, actinomycin d, alteplase, fluticasone propionate, levofloxacin, metaproterenol sulfate, morphine sulfate, oxycodone hcl, potassium chloride, triamcinolone acetonide, tacrolimus anhydrous, calcium, interferon-alpha, methotrexate, mycophenolate mofetil, Interferon-gamma-lβ. Non-limiting examples of therapeutic agents for Myocardial Infarction with which an antibody, or antibody portion, of the invention can be combined include the following: aspirin, nitroglycerin, metoprolol tartrate, enoxaparin sodium, heparin sodium, clopidogrel bisulfate, carvedilol, atenolol, morphine sulfate, metoprolol succinate, warfarin sodium, lisinopril, isosorbide mononitrate, digoxin, furosemide, simvastatin, ramipril, tenecteplase, enalapril maleate, torsemide, retavase, losartan potassium, quinapril hcl/mag carb, bumetanide, alteplase, enalaprilat, amiodarone hydrochloride, tirofiban hcl m-hydrate, diltiazem hydrochloride, captopril, irbesartan, valsartan, propranolol hydrochloride, fosinopril sodium, lidocaine hydrochloride, eptifibatide, cefazolin sodium, atropine sulfate, aminocaproic acid, spironolactone, interferon, sotalol hydrochloride, potassium chloride, docusate sodium, dobutamine hcl, alprazolam, pravastatin sodium, atorvastatin calcium, midazolam hydrochloride, meperidine hydrochloride, isosorbide dinitrate, epinephrine, dopamine hydrochloride, bivalirudin, rosuvastatin, ezetimibe/simvastatin, avasimibe, cariporide.
Non-limiting examples of therapeutic agents for Psoriasis with which an antibody, or antibody portion, of the invention can be combined include the following: small molecule inhibitor of KDR (ABT-123), small molecule inhibitor of Tie-2, calcipotriene, clobetasol propionate, triamcinolone acetonide, halobetasol propionate, tazarotene, methotrexate, fluocinonide, betamethasone diprop augmented, fluocinolone acetonide, acitretin, tar shampoo, betamethasone valerate, mometasone furoate, ketoconazole, pramoxine/fluocinolone, hydrocortisone valerate, flurandrenolide, urea, betamethasone, clobetasol propionate/emoll, fluticasone propionate, azithromycin, hydrocortisone, moisturizing formula, folic acid, desonide, pimecrolimus, coal tar, diflorasone diacetate, etanercept folate, lactic acid, methoxsalen, hc/bismuth subgal/znox/resor, methylprednisolone acetate, prednisone, sunscreen, halcinonide, salicylic acid, anthralin, clocortolone pivalate, coal extract, coal tar/salicylic acid, coal tar/salicylic acid/sulfur, desoximetasone, diazepam, emollient, fluocinonide/emoUient, mineral oil/castor oil/na lact, mineral oil/peanut oil, petroleum/isopropyl myristate, psoralen, salicylic acid, soap/tribromsalan, thimerosal/boric acid, celecoxib, infliximab, cyclosporine, alefacept, efalizumab, tacrolimus, pimecrolimus, PUVA, UVB, sulfasalazine.
Non-limiting examples of therapeutic agents for Psoriatic Arthritis with which an antibody, or antibody portion, of the invention can be combined include the following: methotrexate, etanercept, rofecoxib, celecoxib, folic acid, sulfasalazine, naproxen, leflunomide, methylprednisolone acetate, indomethacin, hydroxychloroquine sulfate, prednisone, sulindac, betamethasone diprop augmented, infliximab, methotrexate, folate, triamcinolone acetonide, diclofenac, dimethylsulfoxide, piroxicam, diclofenac sodium, ketoprofen, meloxicam, methylprednisolone, nabumetone, tolmetin sodium, calcipotriene, cyclosporine, diclofenac sodium/misoprostol, fluocinonide, glucosamine sulfate, gold sodium thiomalate, hydrocodone bitartrate/apap, ibuprofen, risedronate sodium, sulfadiazine, thioguanine, valdecoxib, alefacept, efalizumab.
Non-limiting examples of therapeutic agents for Restenosis with which an antibody, or antibody portion, of the invention can be combined include the following: sirolimus, paclitaxel, everolimus, tacrolimus, ABT-578, acetaminophen.
Non-limiting examples of therapeutic agents for Sciatica with which an antibody, or antibody portion, of the invention can be combined include the following: hydrocodone bitartrate/apap, rofecoxib, cyclobenzaprine hcl, methylprednisolone, naproxen, ibuprofen, oxycodone hcl/acetaminophen, celecoxib, valdecoxib, methylprednisolone acetate, prednisone, codeine phosphate/apap, tramadol hcl/acetaminophen, metaxalone, meloxicam, methocarbamol, lidocaine hydrochloride, diclofenac sodium, gabapentin, dexamethasone, carisoprodol, ketorolac tromethamine, indomethacin, acetaminophen, diazepam, nabumetone, oxycodone hcl, tizanidine hcl, diclofenac sodium/misoprostol, propoxyphene napsylate/apap, asa/oxycod/oxycodone ter, ibuprofen/hydrocodone bit, tramadol hcl, etodolac, propoxyphene hcl, amitriptyline hcl, carisoprodol/codeine phos/asa, morphine sulfate, multivitamins, naproxen sodium, orphenadrine citrate, temazepam.
Preferred examples of therapeutic agents for SLE (Lupus) in which an antibody or an antigen binding portion can be combined include the following: NSAIDS, for example, diclofenac, naproxen, ibuprofen, piroxicam, indomethacin; COX2 inhibitors, for example, Celecoxib, rofecoxib, valdecoxib; anti-malarials, for example, hydroxychloroquine; Steroids, for example, prednisone, prednisolone, budenoside, dexamethasone; Cytotoxics, for example, azathioprine, cyclophosphamide, mycophenolate mofetil, methotrexate; inhibitors of PDE4 or purine synthesis inhibitor, for example Cellcept. Antibodies of the invention or antigen binding portions thereof, may also be combined with agents such as sulfasalazine, 5-aminosalicylic acid, olsalazine, Imuran and agents which interfere with synthesis, production or action of proinflammatory cytokines such as IL-I, for example, caspase inhibitors like IL-lβ converting enzyme inhibitors and IL- Ira. Antibodies of the invention or antigen binding portion thereof may also be used with T cell signaling inhibitors, for example, tyrosine kinase inhibitors; or molecules that target T cell activation molecules, for example, CTLA-4-IgG or anti-B7 family antibodies, anti-PD-1 family antibodies. Antibodies of the invention, or antigen binding portions thereof, can be combined with IL-Il or anti-cytokine antibodies, for example, fonotolizumab (anti-IFNg antibody), or anti-receptor receptor antibodies, for example, anti-IL-6 receptor antibody and antibodies to B-cell surface molecules. Antibodies of the invention or antigen binding portion thereof may also be used with LJP 394 (abetimus), agents that deplete or inactivate B-cells, for example, Rituximab (anti-CD20 antibody), lymphostat-B (anti-BlyS antibody), TNF antagonists, for example, anti-TNF antibodies, D2E7 (PCT Publication No. WO 97/29131; HUMIRA), CA2 (REMICADE), CDP 571, TNFR-Ig constructs, (p75TNFRIgG (ENBREI^ and p55TNFRIgG (LENERCEPT)).
The pharmaceutical compositions of the invention may include a "therapeutically effective amount" or a "prophylactically effective amount" of an antibody or antibody portion of the invention. A "therapeutically effective amount" refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic result. A therapeutically effective amount of the antibody or antibody portion may be determined by a person skilled in the art and may vary according to factors such as the disease state, age, sex, and weight of the individual, and the ability of the antibody or antibody portion to elicit a desired response in the individual. A therapeutically effective amount is also one in which any toxic or detrimental effects of the antibody, or antibody portion, are outweighed by the therapeutically beneficial effects. A "prophylactically effective amount" refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired prophylactic result. Typically, since a prophylactic dose is used in subjects prior to or at an earlier stage of disease, the prophylactically effective amount will be less than the therapeutically effective amount.
Dosage regimens may be adjusted to provide the optimum desired response (e.g., a therapeutic or prophylactic response). For example, a single bolus may be administered, several divided doses may be administered over time or the dose may be proportionally reduced or increased as indicated by the exigencies of the therapeutic situation. It is especially advantageous to formulate parenteral compositions in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the mammalian subjects to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. The specification for the dosage unit forms of the invention are dictated by and directly dependent on (a) the unique characteristics of the active compound and the particular therapeutic or prophylactic effect to be achieved, and (b) the limitations inherent in the art of compounding such an active compound for the treatment of . sensitivity in individuals.
An exemplary, non-limiting range for a therapeutically or prophylactically effective amount of an antibody or antibody portion of the invention is 0.1-20 mg/kg, more preferably 1-10 mg/kg. It is to be noted that dosage values may vary with the type and severity of the condition to be alleviated. It is to be further understood that for any particular subject, specific dosage regimens should be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the compositions, and that dosage ranges set forth herein are exemplary only and are not intended to limit the scope or practice of the claimed composition. It will be readily apparent to those skilled in the art that other suitable modifications and adaptations of the methods of the invention described herein are obvious and may be made using suitable equivalents without departing from the scope of the invention or the embodiments disclosed herein. Having now described the present invention in detail, the same will be more clearly understood by reference to the following examples, which are included for purposes of illustration only and are not intended to be limiting of the invention.
Examples
Example 1: Generation and isolation of anti human IL-12 monoclonal antibodies
Example 1.1: Assays to identify anti human IL-12 antibodies
Throughout Example 1 the following assays were used to identify and characterize anti human IL-12 antibodies unless otherwise stated.
Example 1.1 A: ELISA
Enzyme Linked Immunosorbent Assays to screen for antibodies that bind human BL-12 were performed as follows.
Example 1.1 A.I: ELISA to detect binding of anti human IL-12 antibodies to IL-12 p70
ELISA plates (Corning Costar, Acton, MA) were coated with 50μL/well of 5μg/ml goat anti-mouse IgG Fc specific (Pierce # 31170, Rockford, IL.) in Phosphate Buffered Saline (PBS) overnight at 4 degrees Celsius. Plates were washed once with PBS containing 0.05% Tween-20. Plates were blocked by addition of 200 μL/well blocking solution diluted to 2% in PBS (BioRad #170-6404, Hercules, CA.) for 1 hour at room temperature. Plates were washed once after blocking with PBS containing 0.05% Tween-20.
Fifty microliters per well of mouse sera or hybridoma supernatants diluted in PBS containing 0.1% Bovine Serum Albumin (BSA) (Sigma, St. Louis, MO.) was added to the ELISA plate prepared as described above and incubated for 1 hour at room temperature. Wells were washed three times with PBS containing 0.05% Tween-20. Fifty microliters of biotinylated recombinant purified human IL-12 p70 diluted to lOOng/mL in PBS containing 0.1% BSA was added to each well and incubated for 1 hour at room temperature. Plates were washed 3 times with PBS containing 0.05% Tween-20. Streptavidin HRP (Pierce # 21126, Rockland, IL.) was diluted 1:20000 in PBS containing 0.1% BSA; 50 μL/well was added and the plates incubated for 1 hour at room temperature. Plates were washed 3 times with PBS containing 0.05% Tween-20. Fifty microliters of TMB solution (Sigma # T0440, St. Louis, MO.) was added to each well and incubated for 10 minutes at room temperature. The reaction was stopped by addition of IN sulphuric acid. Plates were read spectrophotmetrically at a wavelength of 450 nm.
Example 1.1.A.2: ELISA to assess ability of IL-12 p70 or IL-12 p40 to compete with binding of anti human IL-12 antibodies to IL-12 p70
ELISA plates (Corning Costar, Acton, MA) were coated with 50μL/well of 5μg/rnl goat anti-mouse IgG Fc specific (Pierce # 31170, Rockford, IL.) in PBS overnight at 4 degrees Celsius. Plates were washed once with PBS+ 0.05% Tween-20. Plates were blocked by addition of PBS + 10% powdered milk for 1 hour at room temperature. Plates were washed three times after blocking with PBS+ 0.05% Tween-20.
Example 1.1.A 2(a): IL-12 p70 competition ELISA protocol
Mouse sera or hybridoma supernatants were diluted in PBS containing 0.1% BSA (Sigma, St. Louis, MO.) depending on anticipated antibody titer. Biotinylated recombinant purified human IL-12 p70 was prepared as a three times concentrated (3x) stock at O.lμg/ml in PBS containing 0.1% BSA. Recombinant purified human IL-12 p70 was prepared at various concentrations ranging from 0.1 to lOμg/ml in PBS containing 0.1% BSA. Equal volumes (75μL) of each of the following solutions were mixed: diluted mouse sera or hybridoma supernatant, biotinylated recombinant purified human IL-12 p70, and recombinant purified human IL-12 p70. Fifty microliters of this mixture was added to the coated ELISA plates described above and were incubated for 1 hour at room temperature. Wells were washed three times with PBS containing 0.05% Tween-20. Streptavidin HRP (Pierce # 21126, Rockland, IL.) was diluted 1:20000 in PBS containing 0.1% BSA; 50 μL/well was added and the plates incubated for 1 hour at room temperature. Plates were washed 3 times with PBS containing 0.05% Tween-20. Fifty microliters of TMB solution (Sigma # T0440, St. Louis, MO.) was added to each well and incubated for 10 minutes at room temperature. The reaction was stopped by addition of IN sulphuric acid. Plates were read spectrophotmetrically at a wavelength of 450 nm.
Example 1.1.A 2(b): IL-12 p40 competition ELISA protocol
Mouse sera or hybridoma supernatants were diluted in PBS containing 0.1% BSA (Sigma, St. Louis, MO.) depending on anticipated antibody titer. Biotinylated recombinant purified human IL-12 p70 was prepared as a three times concentrated (3x) stock at O.lμg/ml in PBS containing 0.1% BSA. Recombinant purified human IL-12 p40 was prepared at various concentrations ranging from 0.1 to lOμg/ml in PBS containing 0.1% BSA. Equal volumes (75μL) of each of the following solutions were mixed: diluted mouse sera or hybridoma supernatant, biotinylated recombinant purified human EL- 12 p70, and recombinant purified human IL- 12 p40. Fifty microliters of this mixture was added to the coated ELISA plates and incubated for 1 hour at room temperature. Wells were washed three times with PBS containing 0.05% Tween-20. Streptavidin HRP (Pierce # 21126, Rockland, IL.) was diluted 1:20000 in PBS containing 0.1% BSA; 50 μL/well was added and the plates incubated for 1 hour at room temperature. Plates were washed 3 times with PBS containing 0.05% Tween-20. Fifty microliters of TMB solution (Sigma # T0440, St. Louis, MO.) was added to each well and incubated for 10 minutes at room temperature. The reaction was stopped by addition of IN sulphuric acid. Plates were read spectrophotmetrically at a wavelength of 450 nm.
Example 1.1.B: Affinity Determinations using BIACORE technology
The BIACORE assay (Biacore, Inc, Piscataway, NJ) determines the affinity of antibodies with kinetic measurements of on-, off-rate constants. Binding of antibodies to recombinant purified human IL-12 p70 or recombinant purified human IL-12 p40 were determined by surface plasmon resonance-based measurements with a Biacore® 3000 instrument (Biacore® AB, Uppsala, Sweden) using running HBS-EP (10 mM HEPES [pH 7.4], 150 mM NaCl, 3 mM EDTA, and 0.005% surfactant P20) at 25° C. AU chemicals were obtained from Biacore® AB (Uppsala, Sweden) or otherwise from a different source as described in the text. Approximately 5000 RU of goat anti-mouse IgG, (Fcγ), fragment specific polyclonal antibody (Pierce Biotechnology Inc, Rockford, IL) diluted in 10 mM sodium acetate (pH 4.5) was directly immobilized across a CM5 research grade biosensor chip using a standard amine coupling kit according to manufacturer's instructions and procedures at 25 μg/ml. Unreacted moieties on the biosensor surface were blocked with ethanolamine. Modified carboxymethyl dextran surface in flowcell 2 and 4 was used as a reaction surface. Unmodified carboxymethyl dextran without goat anti-mouse IgG in flow cell 1 and 3 was used as the reference surface. For kinetic analysis, rate equations derived from the 1:1 Langmuir binding model were fitted simultaneously to association and dissociation phases of all eight injections (using global fit analysis) with the use of Biaevaluation 4.0.1 software. Purified antibodies were diluted in HEPES-buffered saline for capture across goat anti-mouse IgG specific reaction surfaces. Mouse antibodies to be captured as a ligand (25 μg/ml) were injected over reaction matrices at a flow rate of 5 μl/min. The association and dissociation rate constants, Ic0n (unit MT1S"1) and koff (unit s'1) were determined under a continuous flow rate of 25 μl/min. Rate constants were derived by making kinetic binding measurements at ten different antigen concentrations ranging from 10 - 200 nM. The equilibrium dissociation constant (unit M) of the reaction between mouse antibodies and recombinant purified human IL-12 ρ70 or recombinant purified human IL-12 p40 was then calculated from the kinetic rate constants by the following formula: KD = IWk0n. Binding is recorded as a function of time and kinetic rate constants are calculated. In this assay, on-rates as fast as 106M-1S'1 and off- rates as slow as 10'6 s'1 can be measured.
Example l.l.C: Functional Activity of anti human IL-12 antibodies
To examine the functional activity of the anti-human IL-12 antibodies of the invention, the antibodies were used in the following assays that measure the ability of an antibody to inhibit IL-12 activity.
Example l.l.C 1: Preparation of Human PHA-activated Lymphoblasts
Human peripheral blood mononuclear cells (PBMCs) were isolated from a leukopac collected from a healthy donor by Ficoll-Hypaque gradient centrifugation for 45 minutes at 1500 rpm as described in Current Protocols in Immunology, Unit 7.1. PBMC at the interface of the aqueous blood solution and the lymphocyte separation medium were collected and washed three times with phosphate-buffered saline (PBS) by centrifugation for 15 minutes at 1500 rpm to remove Ficoll-Paque particles.
The PBMC were then activated to form lymphoblasts as described in Current Protocols in Immunology, Unit 6.16. The washed PBMC were resuspended at 0.5-lxIO6 cells/mL in RPMI complete medium (RPMI 1640 medium, 10% fetal bovine serum (FBS), 100 U/ml penicillin, 100 tg/ml streptomycin), supplemented with O.Olmg/mL PHA-P (Sigma #L8754, St. Louis, MO) and cultured for 4 days at 370C in a 5% CO2 atmosphere. After four days, cell cultures were then re- seeded at IxIO6 cells/mL in culture media with 0.01mg/mL PHA-P and 50U/mL recombinant human IL-2 (R&D Systems #202-EL, Minneapolis, MN.). Cells were incubated at 37°C for 24 hours, washed with RPMI complete medium, then frozen in 95% FBS, 5% DMSO at IxIO7 cells/ml.
Example l.l.C 2: PHA blast IFN-γ induction assay: Inhibition of human IL-12 activity
The ability of anti-human IL-12 antibodies to inhibit the human IL-12 induced production of IFN-γ by PHA blasts was analyzed as follows. Various concentrations of immunized mouse serum, murine hybridoma supernatant or purified anti-human IL-12 antibodies were preincubated for one hour at 37 degrees C with 400 pg/ml recombinant purified human IL-12 p70 in lOOμL RPMI complete medium in a microtiter plate (U-bottom, 96-well, Costar). PHA blasts isolated as described above, were washed once and resuspended in RPMI complete medium to a cell density of IXlO7 cells/ml. PHA blasts (lOOμL of IXlO6 cells/ml) were added to the antibody plus recombinant purified human IL-12 p70 mixture (final IL-12 p70 concentration was 200 pg/ml) and incubated for 18 hours at 37 deg C. After incubation, 150 μL of cell-free supernatant was withdrawn from each well and the level of human IFN-γ produced was measured using a human IFN-γ ELISA (R&D Systems Cat#DIF50). Example l.l.C 3: PHA blast IFN-Y induction assay: Inhibition of cynomolgus monkey (cyno) IL-12 activity
The ability of anti-human IL-12 antibodies to inhibit the cynomolgus monkey IL-12 induced production of IFN-γ by PHA blasts was analyzed as follows. Various concentrations of immunized mouse serum, murine hybridoma supernatant or purified anti-human IL-12 antibodies were preincubated for one hour at 37 degrees C with 150 pg/mL recombinant purified cyno IL-12 p70 in lOOμL RPMI complete medium in a microliter plate (U-bottom, 96-well, Costar). PHA blasts isolated as described above, were washed once and resuspended in RPMI complete medium to a cell density of IXlO7 cells/ml. PHA blasts (lOOμL of IXlO7 cells/mL) were added to the antibody plus recombinant purified cyno IL-12 ρ70 mixture (final cyno IL-12 p70 concentration was 75μg/ml) and incubated for 18 hours at 37 deg C. After incubation, 150 μL of cell-free supernatant was withdrawn from each well and the level of human IFN-γ produced was measured using a human IFN-γ ELISA (R&D Systems Cat#D]F50).
Example 1.2: Generation of Anti human IL-12 monoclonal antibodies
Anti human IL-12 mouse monoclonal antibodies were obtained as follows:
Example 1.2.A: Immunization of mice with human IL-12 antigen
Twenty micrograms of recombinant purified human IL-12 ρ70 mixed with complete Freund's adjuvant (Rockland Immunochemicals, Gilbertsville, PA) was injected subcutaneously into five 6-8 week-old Balb/C and 5 AJ mice on Day 1. On days 24, 38, and 49, twenty micrograms of recombinant purified human IL-12 p70 mixed with Immunoeasy adjuvant (Qiagen, Valencia, CA) was injected subcutaneously into the same 5 Balb/C and 5 AJ mice. On day 84 or day 112 or day 144, mice were injected intravenously with 10 ug recombinant purified human IL-12 p70 or 2ug recombinant purified human IL-12 p40 (R & D Systems, Minneapolis, MN).
Example 1.2.B: Generation of Hybridoma
Splenocytes obtained from the immunized mice described in Example 1.2. A were fused with SP2/O-Ag-14 cells at a ratio of 5: 1 according to the established method described in Kohler, G. and Milstein 1975, Nature, 256:495 to generate hybridomas. Fusion products were plated in selection media containing azaserine and hypoxanthine in 96-well plates at a density of 2.5xlO6 spleen cells per well. Seven to ten days post fusion, macroscopic hybridoma colonies were observed. Supernatant from each well containing hybridoma colonies was tested by ELISA for the presence of antibody to IL-12 p70 (as described in Example l.l.A.l). Supernatants testing positive for binding to IL-12 p70 were then tested to determine whether they were p40-specific by the IL-12 p70 or IL-12 p40 competition ELISA (as described in Example 1.1.A.2). Supernatants displaying IL-12 p40-specific activity were then tested for the ability to neutralize IL-12 in the PHA blast assay for IFN-γ (as described in Example l.l.C).
Table 8: Fusion and screening data following immunizations of mice with human IL-12
Figure imgf000090_0001
Example 1.2.C: Identification and characterization of anti human IL-12p40 monoclonal antibodies
Hybridomas producing antibodies that bound IL-12, generated according to Examples 1.2.B and 1.2.C, and capable of binding IL-12 p40 specifically and particularly those with IC50 values in the PHA blast assay of 12nM or less than 12nM were scaled up and cloned by limiting dilution.
Hybridoma cells were expanded into media containing 10% low IgG fetal bovine serum (Hyclone #SH30151, Logan, UT.). On average, 250 mL of each hybridoma supernatant (derived from a clonal population) was harvested, concentrated and purified by protein A affinity chromatography, as described in Harlow, E. and Lane, D. 1988 "Antibodies: A Laboratory Manual". The ability of purified mAbs to inhibit IL-12 activity was determined using the PHA blast assay as described in Examples l.l.C 2 and 1.1.C3. Table 9 shows IC50 values from the PHA blast assays for ten monoclonal antibodies. Table 9: Neutralization of IL-12 by anti IL-12p40 Murine Monoclonal Antibodies
Figure imgf000091_0001
The binding affinities of the monoclonal antibodies to recombinant purified human IL-12 p70 were determined using surface plasmon resonance (Biacore®) measurement as described in Example l.l.B. Table 10 shows the affinity of the ten monoclonal antibodies described above for human IL-12 p70.
Table 10: Affinity of anti IL-12p40 Murine Monoclonal Antibodies for IL-12 p70
Figure imgf000091_0002
Example 1.2.C.1: Species Specificity of murine monoclonal anti-human IL-12p40 antibodies
To determine whether the ten monoclonal antibodies described above recognize murine IL-12, two ELISAs were set up. First, a direct ELISA was set up by directly coating ELISA plates with 5 ug/ml of recombinant purified mouse IL-12 (Peprotech). Murine-anti-human IL-12 p40 mAbs were prepared at various concentrations ranging from 3 to 200 ng/ml in PBS containing 0.1% BSA (Sigma, StLouis, MO). 50 μl of each antibody dilution was added to the coated ELISA plate and incubated for 1 hour at room temperature. Wells were washed 3 times with PBS containing 0.05% Tween-20. Anti-mouse IgG-HRP antibody (R&D #HAF007, Minneapolis, MN) was diluted 1:2000 in PBS containing 0.1% BSA; 50 ul/well was added and the plates incubated for 1 hour at room temperature. Fifty microliters of TMB solution (Sigma # T0440, St. Louis, MO.) was added to each well and incubated for 10 minutes at room temperature. The reaction was stopped by addition of 2N sulphuric acid. Plates were read spectrophotmetrically at a wavelength of 450 nm.
Second, an indirect ELISA was set up by coating ELISA plates with 5ug/ml of goat anti- mouse IgG, Fc fragment specific antibody (Pierce # 31170, Rockland, IL). Murine anti-human IL-12 p40 mAbs were prepared at various concentrations ranging from 0.1 to 100 ng/ml in PBS containing 0.1% BSA; 50 ul of each antibody dilution was added to the coated ELISA plate and incubated for 1 hour at room temperature. Wells were washed 3 times with PBS containing 0.05% Tween-20. Recombinant purified mouse IL-12 (Preprotech) was diluted at 0.2 ug/ml in PBS containing 0.1% BSA; 50 ul/well was added and the plates incubated for 1 hour at room temperature. Wells were washed 3 times with PBS containing 0.05% Tween-20. Biotinylated anti-mouse IL-12 antibody (R&D # BAF419) was diluted at 0.2 ug/ml in PBS containing 0.1% BSA; 50 ul/well was added and the plates incubated for 1 hour at room temperature. Wells were washed 3 times with PBS containing 0.05% Tween-20. Streptavidin HRP (Pierce # 21126, Rockland, IL.) was diluted 1:20000 in PBS containing 0.1% BSA; 50 μL/well was added and the plates incubated for 1 hour at room temperature. Plates were washed 3 times with PBScontaining 0.05% Tween-20. Fifty microliters of TMB solution was added to each well and incubated for 10 minutes at room temperature. The reaction was stopped by addition of 2N sulphuric acid. Plates were read spectrophotmetrically at a wavelength of 450 nm. Results from the direct and indirect ELISAs performed with the ten monoclonal antibodies are shown in Table 11. Table 11: Binding of anti IL-12 Murine Monoclonal Antibodies to mouse IL-12 p40
Figure imgf000093_0001
Example 1.2.D: Determination of the amino acid sequence of the variable region for each murine anti-human IL-12 p40 mAb
For each amino acid sequence determination, approximately 10xl06hybridoma cells were isolated by centrifugation and processed to isolate total RNA with Trizol (Gibco BRL/Invitrogen, Carlsbad, CA.) following manufacturer's instructions. Total RNA was subjected to first strand DNA synthesis using the Superscript First-Strand Synthesis System (hivitrogen, Carlsbad, CA) per the manufacturers instructions. Oligo(dT) was used to prime first-strand synthesis to select for poly(A)+ RNA. The first-strand cDNA product was then amplified by PCR with primers designed for amplification of murine immunoglobulin variable regions (Ig-Primer Sets, Novagen, Madison, WI). PCR products were resolved on an agarose gel, excised, purified, and then subcloned with the TOPO Cloning kit into pCR2.1-TOPO vector (hwitrogen, Carlsbad, CA) and transformed into TOPlO chemically competent E. coli (Invitrogen, Carlsbad, CA). Colony PCR was performed on the transformants to identify clones containing insert. Plasmid DNA was isolated from clones containing insert using a QIAprep Miniprep kit (Qiagen, Valencia, CA). Inserts in the plasmids were sequenced on both strands to determine the variable heavy or variable light chain DNA sequences using M13 forward and M13 reverse primers ( Fermentas Life Sciences, Hanover MD). Variable heavy and variable light chain sequences of the ten monoclonal antibodies described in Example 1.2.C are described in Table 1.
Example 2: Recombinant anti human IL-12p40 antibodies Example 2.1: Construction and expression of recombinant chimeric anti human IL-12p40 antibodies
The DNA encoding the heavy chain constant region of murine anti-human IL~12p40 monoclonal antibodies 3G7, 8El, 1A6, and 1D4 was replaced by a cDNA fragment encoding the human IgGl constant region containing 2 hinge-region amino acid mutations by homologous recombination in bacteria. These mutations are a leucine to alanine change at position 234 (EU numbering) and a leucine to alanine change at position 235 (Lund et al., 1991, J. Immunol., 147:2657). The light chain constant region of each of these antibodies was replaced by a human kappa constant region. Full-length chimeric antibodies were transiently expressed in COS cells by co-transfection of chimeric heavy and light chain cDNAs ligated into the pBOS expression plasmid (Mizushima and Nagata, Nucleic Acids Research 1990, VoI 18, pg 5322) comprising a heavy chain signal sequence MEFGLSWLFLVAILKGVQC (SEQ ID NO. 110), and a light chain signal sequence MDMRVPAQLLGLLLLWFPGSRC ((SEQ ID NO. 111).
Cell supernatants containing recombinant chimeric antibody were purified by Protein A Sepharose chromatography and bound antibody was eluted by addition of acid buffer. Antibodies were neutralized and dialyzed into PBS.
The heavy chain cDNA encoding chimeric 3G7 (described above) was co-transfected with the 1D4 chimeric light chain cDNA (both ligated in the pBOS vector) into COS cells. Cell supernatant containing recombinant chimeric antibody was purified by Protein A Sepharose chromatography and bound antibody was eluted by addition of acid buffer. Antibodies were neutralized and dialyzed into PBS.
The purified chimeric anti-human IL-12 monoclonal antibodies were then tested for their ability to inhibit the IL-12 induced production of IFN-γ by PHA blasts as described in Examples l.l.C 2 and 1.1.C3. Table 12 shows IC50 values from the PHA blast assays for five chimeric antibodies.
Table 12: Neutralization of IL-12 by anti IL-12 Chimeric Antibodies
Figure imgf000094_0001
Example 2.2: Construction and expression of CDR grafted anti human IL-12p40 antibodies
CDR-grafted anti-human IL- 12 antibodies were generated as follows.
Example 2.2.1: Selection of human antibody frameworks
Each murine variable heavy and variable light chain gene sequence (as described in Table 3) was separately aligned against 44 human immunoglobulin germline variable heavy chain or 46 germline variable light chain sequences (derived from NCBI Ig Blast website at http://www.ncbi.nlm.nih.gov/igblast/retrieveig.html.') using Vector NTI software. Human variable domain sequences having the highest overall homology to the original murine sequences (as well as the highest homology at positions known to be important for antigen binding) (Welschof, M. and Krauss, J. Methods In Molecular Biology, VoI 207) were selected for each heavy chain and light chain sequence to provide the framework (FW) 1, 2 and 3 sequences for CDR-grafting purposes. Identification of a suitable human variable heavy and light chain FW4 region (also known as the "joining" region) was accomplished by separately aligning each murine heavy chain and light chain FW4 region with 6 human immunoglobulin germline joining heavy chain and 5 germline joining light chain sequences in the NCBI database. In silico construction of complete CDR grafted antibodies was accomplished by substitution of human variable domain CDR sequences (derived from the NCBI website) with murine CDR sequences (derived from the hybridomas) with addition of a FW4 region (derived from the NCBI website) to each 3' end.
Example 2.2.2: Construction of CDR-grafted antibodies
In silico constructed CDR grafted antibodies described above were constructed de novo using oligonucleotides. For each variable region cDNA, 6 oligonucleotides of 60-80 nucleotides each were designed to overlap each other by 20 nucleotides at the 5' and/or 3' end of each oligonucleotide. In an annealing reaction, all 6 oligos were combined, boiled, and annealed in the presence of dNTPs. Then DNA polymerase I, Large (Klenow) fragment (New England Biolabs #M0210, Beverley, MA.) was added to fill-in the approximately 40bp gaps between the overlapping oligonucleotides. PCR was then performed to amplify the entire variable region gene using two outermost primers containing overhanging sequences complementary to the multiple cloning site in a modified pBOS vector (Mizushima, S. and Nagata, S., (1990) Nucleic acids Research VoI 18, No. 17)).
The PCR products derived from each cDNA assembly were separated on an agarose gel and the band corresponding to the predicted variable region cDNA size was excised and purified. The variable heavy region was inserted in-frame onto a cDNA fragment encoding the human IgGl constant region containing 2 hinge-region amino acid mutations (SEQ ID NO. 3) by homologous recombination in bacteria. These mutations are a leucine to alanine change at position 234 (EU numbering) and a leucine to alanine change at position 235 (Lund et al., 1991, J. Immunol., 147:2657). The variable light chain region was inserted in-frame with the human kappa constant region (SEQ ID NO. 4) by homologous recombination. Bacterial colonies were isolated and plasmid DNA extracted; cDNA inserts were sequenced in their entirety. Correct CDR-grafted heavy and light chains corresponding to each antibody were co-transfected into COS cells to transiently produce full-length CDR-grafted anti-human IL- 12 antibodies. For 1A6, pBOS vectors containing the 1A6 heavy chain grafted cDNA and the 1D4 light chain grafted cDNA were co-transfected into COS cells (the 1A6 light chain sequence, when grafted, was identical to the 1D4 light chain sequence). Cell supernatants containing recombinant chimeric antibody were purified by Protein A Sepharose chromatography and bound antibody was eluted by addition of acid buffer. Antibodies were neutralized and dialyzed into PBS. Nine CDR grafted antibodies are described in Table 5.
The ability of purified CDR grafted antibodies to inhibit IL-12 activity was determined using the PHA blast assay as described in Examples l.l.C 2 and 1.1.C3. The binding affinities of the purified CDR grafted antibodies to recombinant purified human IL-12p70 were determined using surface plasmon resonance (Biacore®) measurement as described in Example 1.1. B. Table 13 shows shows IC50 values from the PHA blast assays and the affinity of the nine CDR grafted antibodies described in Table 7 for human EL-12p70 and cynomolgous IL-12p70.
Table 13: Neutralization of IL-12 by anti IL-12p40 CDR grafted Antibodies and Affinity of anti IL-12p40 CDR grafted Antibodies for human and cynomolgous IL-12p70 and
Figure imgf000096_0001
Figure imgf000097_0001
Example 2.3: Construction of Humanized anti human IL-12 antibodies
Humanization of the anti human IL-12 antibodies was carried out as follows. Example 2.3.1: Homology modeling with CDR-grafted antibodies
Homology modeling was used was to identify residues unique to the murine antibody sequences that are predicted to be critical to the structure of the antibody combining site (the CDRs).
Homology modeling is a computational method whereby approximate three dimensional coordinates are generated for a protein. The source of initial coordinates and guidance for their further refinement is a second protein, the reference protein, for which the three dimensional coordinates are known and the sequence of which is related to the sequence of the first protein. The relationship among the sequences of the two proteins is used to generate a correspondence between the reference protein and the protein for which coordinates are desired, the target protein. The primary sequences of the reference and target proteins are aligned with coordinates of identical portions of the two proteins transferred directly from the reference protein to the target protein. Coordinates for mismatched portions of the two proteins, e.g. from residue mutations, insertions, or deletions, are constructed from generic structural templates and energy refined to insure consistency with the already transferred model coordinates. This computational protein structure may be further refined or employed directly in modeling studies. It should be clear from this description that the quality of the model structure is determined by the accuracy of the contention that the reference and target proteins are related and the precision with which the sequence alignment is constructed:
For the murine sequences 1A6, 8El and 1D4, a combination of BLAST searching and visual inspection was used to identify suitable reference structures. The reference structures chosen for 1A6 and 1D4 was the PDB entry IJRH. For 8El, the heavy chain reference structure was PDB entry 1FL3 and the light chain reference was IMEX. Sequence identity of 25% between the reference and target amino acid sequences is considered the minimum necessary to attempt a homology modeling exercise. Sequence alignments were constructed manually and model coordinates were generated with the program Jackal (see Petrey, D., Xiang, Z., Tang, C.L., Xie, L., Gimpelev, M., Mitros, T., Soto, C.S., Goldsmith-Fischman, S., Kernytsky, A., Schlessinger, A., et al. 2003. Using multiple structure alignments, fast model building, and energetic analysis in fold recognition and homology modeling. Proteins 53 (Suppl. 6): 430-435).
The primary sequences of the murine and human framework regions of the selected antibodies share significant identity. Residue positions that differ are candidates for inclusion of the murine residue in the humanized sequence in order to retain the observed binding potency of the murine antibody. A list of framework residues that differ between the human and murine sequences was constructed manually.
The likelihood that a given framework residue would impact the binding properties of the antibody depends on its proximity to the CDR residues. Therefore, using the model structures, the residues that differ between the murine and human sequences were ranked according to their distance from any atom in the CDRs. Those residues that fell within 4.5 A of any CDR atom were identified as most important and were recommended to be candidates for retention of the murine residue in the humanized antibody (i.e. back mutation).
Examination of the computer model suggested that in antibody 1A6.1, residues Ll, L2, L36, and L67 of the light chain (1D4.1 VKB3) are in significant contact with the CDRs and therefore suggests that murine residues should be retained at these positions. In the case of 1D4.1, residues Hl and H98 of the heavy chain (1D4.1 VH2-70), as well as L2 and L67 of the light chain (1D4.1 VKB3) were identified as positions for back mutation.
Example 2.3.2: Generation of Hybrid antibodies
To determine which CDR-grafted chain (VH, VL or both) may benefit from framework back-mutations, "hybrid" antibodies were constructed by pairing a CDR-grafted H or L chain with an appropriate chimeric murine H or L chain followed by co-transfection into COS cells. Table 14 shows the VH and VL amino acid sequences of the hybrid antibodies 1A6.3, 1A6.4, 1A6.7, 1A6.8, 1D4.4, and 1D4.5. Table 14: Amino acid sequences of hybrid antibodies
Figure imgf000099_0001
Hybrid antibodies were purified by protein A affinity chromatography (Example 1.2.C) and tested for potency in the PHA blast assay as in Examples 1.1.C2 and 1.1.C3. Kinetic measurements were determined using BIAcore as in Example 1.1. B. Table 15 shows the KD and IC50 values of the hybrid antibodies. The potency and affinity data derived with the hybrid mAbs was compared to data generated with the appropriate CDR-grafted mAbs (Example 2.3.1) to identify changes in potency and affinity attributed to a particular VH or VL chain. Whether or not, a humanized VH or VL chain was the optimum chain, was assessed using the methods described in Example 2.3.1. In some cases residues that did not fall within 4.5 A0 of any CDR atom were additionally targeted for back mutations.
Table 15: Neutralization of IL-12 by anti-IL-12p40 hybrid antibodies and affinity of anti- IL-12p40 hybrid antibodies
Figure imgf000100_0001
Example 2.3.3: Construction of framework back mutations in CDR-grafted antibodies
To generate humanized antibodies framework back mutations were introduced into the CDR-grafted antibodies using mutagenic primers and the QuikChange Multi Site-Directed Mutagenesis kit (Stratagene #200513, La Jolla, CA) following manufacturers instructions. Different combinations of back mutations were constructed for each of the CDR-grafts to identify the relative importance of each residue. 1A6.1 light chain VKB3 variant 1 (Ll-D-^ S, L2-I->V, L36-Y-3-F, L67-S^Y), 1D4.1 VKB3 variant 2 (Ll-D^ S, L36-Y->F, L67-S^Y), 1D4.1 VKB3 variant 3 (Ll-D^ S, L67-S->Y), 1D4.1 VKB3 variant 4 (L2-I^V, L67-S^Y), and 1D4.1 VKB3 variant 5 (L67-S^Y). 1D4.1 heavy chain VH2-70 variant 1 (Hl-E^Q, H93-A^T), and 1D4.1 VH2-70 variant 2 (H93-A->T). Mutated single stranded DNA was then transformed into XLlO- GoId ultracompetent cells. Colony sequencing was performed on the transformants to identify clones bearing the desired mutations. Plasmid DNA was isolated from positive clones using a Qiagen Maxiprep kit (Qiagen, Valencia, CA) and the variable and constant regions were sequenced in their entirety. As described above several additional combinations of back mutations were constructed for each of the CDR grafted antibodies. Characterization of the humanized antibodies generated, as disclosed above, was carried out as disclosed below in Example 2.3.4.
Example 2.3.4: Expression and characterization of humanized antibodies
PBOS expression vectors (see Example 2.1 and 2.2.2) harboring heavy and light chains containing framework back mutations were co-transfected into COS cells to transiently produce full-length humanized antibodies. The amino acid sequences of the VH and VL regions of the humanized antibodies are disclosed in Table 16.
Table 16: Sequences of Humanized Antibodies
Figure imgf000101_0001
Figure imgf000102_0001
The amino acid positions back mutated in the VH and VL of the CDR-grafted antibodies are listed in Table 17.
Table 17: Back mutated amino acids present in humanized antibodies
Figure imgf000102_0002
Cell supernatants containing humanized antibodies were purified as described in Example I.2.C. The ability of the purified humanized antibodies to neutralize IL-12 in vitro was determined using the PHA blast assay as described in Examples 1.1. C2 and 1.1. C3. The binding affinities of the purified antibodies to recombinant purified human IL-12ρ70 were determined using BIAcore as described in Example 1.1. B. Table 18 shows the IC50 values from the PHA blast assay and the KpS from BIAcore.
Table 18: Neutralization of IL-12 by anti-IL-12p40 humanized antibodies and affinity of anti-IL-12p40 humanized antibodies
Figure imgf000103_0001
Example 2.4; Generation of additional humanized anti human IL-12 antibodies
Humanization of the variable regions of the murine monoclonal antibodies 8El and 1A6 were carried out essentially according to the procedure of Queen, C, et al., Proc. Natl. Acad. Sci. USA 86: 10029-10033 (1989). First, human V segments with high homology to the murine monoclonal antibody VH or VL amino acid sequences were identified. Next, the complementarity-determining region (CDR) sequences together with framework amino acids important for maintaining the structures of the CDRs were grafted into the selected human framework sequences, hi addition, human framework amino acids that were found to be rare in the corresponding V region subgroup were substituted with consensus amino acids to reduce potential immunogenicity. The resulting humanized monoclonal antibodies were expressed in cells, purified and characterized as described below.
Humanized monoclonal antibodies 8E1.4, 8E1.5, 8E1.6, 1A6.10, 1A6.11, and 1A6.12 were generated as follows.
Example 2.4.1 : Generation of Humanized monoclonal antibodies 8E1.4, 8E1.5, 8E1.6 For humanization of the 8El variable regions, human V region frameworks used as acceptors for the CDRs of 8El were chosen based on sequence homology. First, a molecular model of the 8El variable regions was constructed with the aid of the computer programs ABMOD and ENCAD (Levitt, M., J. MoI. Biol. 168: 595-620 (1983)). Next, based on a homology search against human V and J segment sequences, the VH segment HA3D1 (Olee, T., et al., J. Exp. Med. 175: 831-842 (1992)) and the J segment JH4 (Ravetch, J.V., et al., Cell 27: 583-591 (1981)) were selected to provide the frameworks for the 8E1.4, 8E1.5, and 8E1.6 heavy chain variable regions. For the 8El.4, 8El.5 and 8El.6 light chain variable regions, the VL segment HK137 (Bentley, D.L., and Rabbitts, T.H., Cell 32: 181-189 (1983)) and the J segment JK2 (Hieter, P.A., et al., J. Biol. Chem. 257: 1516-1522 (1982)) were used. The identity of the framework amino acids between 8El VH and the acceptor human HA3D1 and JH4 segments was 84%, while the identity between 8El VL and the acceptor human HK137 and JK4 segments was 67%. Human antibody heavy chain and light chain acceptor sequences used to generate additional humanized anti-human IL-12 antibodies are listed in Table 19 and 20.
Table 19: Heavy Chain Acceptor Sequences used for Additional Humanized Antibodies
Figure imgf000104_0001
Table 20: Light Chain Acceptor used for Additional Humanized Antibodies
Figure imgf000104_0002
Example 2.4.2 :Construction of 8E1.4, 8E1.5 and 8E1.6 antibodies.
The heavy and light chain variable region genes were constructed and amplified using approximately 30 overlapping synthetic oligonucleotides ranging in length from approximately 20 to 40 bases following a published method (Rouillard, J.-M., et al, Nucleic Acids Res. 32: W176-W180 (2004)). The oligonucleotides were annealed and assembled with the Expand High Fidelity PCR System (Roche Diagnostics Corporation, Indianapolis, IN), yielding a full-length product. The resulting product was amplified by the polymerase chain reaction (PCR) using the Expand High Fidelity PCR System. The PCR-amplified fragments were gel-purified, digested with MIuI and Xbal, gel-purified, and subcloned, respectively, into a modified form of pVgl.D.Tt (Cole, M.S., et al., J. Immunol. 159: 3613-3621 (1997); and see below) and pVk (Co, M.S., et al., J. Immunol. 148: 1149-1154 (1992)).
At framework positions in which the computer model suggested significant contact with the CDRs, the amino acids from the mouse V regions were substituted for the original human framework amino acids. This was done at residue 49 for 8E1.4 and 8E1.5 heavy chains, and additionally at residue 74 for the heavy chain of 8E1.6. For the light chains of 8E1.5 and 8E1.6, replacements were made at residue 46, and additionally at residue 60 for 8E1.5. Framework residues that occurred only rarely at their respective positions in the corresponding human V region subgroups were replaced with human consensus amino acids at those positions. This was done at residue 78 for both 8E1.5 and 8E1.6 heavy chains, and at residue 36 for the 8El.5 and 8El.6 light chains.
Site-directed mutagenesis of the synthetic V-genes was done using the QuikChange II Site-Directed Mutagenesis Kit (Stratagene, La Jolla, CA), following the manufacturer's recommendations. Specific mutations in the 8El.6 VH gene were created using mutagenesis oligos and PCR methods well known in the art. The PCR step was done using PfuUltra HF DNA Polymerase (Stratagene), following the manufacturer's recommendations, by incubating at 95°C for 30 sec, followed by 18 cycles of 95°C for 30 sec, 55°C for 1 min and 68°C for 1 min, followed by incubating at 68°C for 7 min. Following digestion with Dpnl, E. coli strain TOPlO Chemically Competent Cells (Invitrogen Corporation, Carlsbad, CA) were transformed with a small portion of the PCR product. Sequence verified miniprep DNA was digested with MIuI and Xbal, and the resulting restriction fragment containing the mutated 8El.6 VH gene was subcloned into the modified pVgl.D.Tt expression vector described below.
Similarly, specific mutations in the 8El.5 VL gene were created using mutagenesis oligos and PCR methods well known in the art. The PCR step was done as described above, and following digestion with MIuI and Xbal, the resulting restriction fragment was subcloned into the pVk expression vector. Mutations were verified by nucleotide sequencing. Genes encoding humanized VH or VL were designed as mini-exons including signal peptides, splice donor signals, and appropriate restriction enzyme sites for subsequent cloning into a mammalian expression vector. The splice donor signals in the VH and VL mini-exons were derived from the corresponding human germline JH and JK sequences, respectively. The signal peptide sequences in the humanized VH mini exon was MEFGLSWLFLV AILKGVQC (SEQ ID NO. 110), and in the humanized VL mini-exons was
MDMRVPAQLLGLLLLWFPGSRC(SEQ ID NO. 111). The 8E1.4, 8E1.5 and 8E1.6 VH and VL genes were constructed by assembly of overlapping synthetic oligonucleotides and PCR methods well known in the art.
Example 2.4.3: Generation of Humanized monoclonal antibodies 1A6.10, 1A6.11 and 1A6.12
For humanization of the 1A6.10, 1A6.11 and 1A6.12 variable regions, the general approach provided in the present invention was followed. First, a molecular model of the 1 A6 variable regions was constructed with the aid of the computer programs ABMOD and ENCAD (Levitt, M., J. MoI. Biol. 168: 595-620 (1983)). Next, based on a homology search against human V and J segment sequences, the VH segment M60 (Schroeder, Jr., H.W. and Wang, J. Y., Proc. Natl. Acad. Sci. USA 87: 6146-6150 (1990)) and the J segment JH4 (Ravetch, J.V., et al., Cell 27: 583-591 (1981)) were selected to provide the frameworks for the 1A6.10 and 1A6.12 heavy chain variable regions. For the 1A6.10, 1A6.11 and 1A6.12 light chain variable regions, the VL segment IU-3R (Manheimer-Lory, A., et al., J. Exp. Med. 174: 1639-1652 (1991)) and the J segment JK4 (Hieter, P.A., et al., J. Biol. Chem. 257: 1516-1522 (1982)) were used. The identity of the framework amino acids between 1A6 VH and the acceptor human M60 and JH4 segments was 74%, while the identity between 1A6 VL and the acceptor human UI-3R and JK4 segments was 71%. The antibody sequences were generated as disclosed in Example 2.3.3.
At framework positions in which the computer model suggested significant contact with the CDRs, the amino acids from the mouse V regions were substituted for the original human framework amino acids. This was done at residue 68 for the 1A6.10, 1A6.11 and 1A6.12 heavy chains. For the light chains, replacements were made at residues 36 and 67 for 1A6.11 and 1A6.12, and additionally at residue 60 for 1A6.12. Framework residues that occurred only rarely at their respective positions in the corresponding human V region subgroups were replaced with human consensus amino acids at those positions. This was done at residues 10, 46, 83, 84, 86 and 87 of the heavy chain, and at residue 62 of the light chains of 1A6.10, 1A6.11 and 1A6.12. Site directed mutagenesis was performed using mutagenic oligos and PCR method as described above. The amino acid sequences of the resulting VH and VL regions of the additional humanized anti-IL-12 antibodies generated are listed in Table 21.
Table 21: Sequences of Additional Humanized Antibodies
Figure imgf000107_0001
The amino acid positions in the framework that were mutated are listed in Table 22. Table 22: Positions of Framework Mutations in Additional Humanized Antibodies
Figure imgf000108_0001
Example 2.4.4: Expression of additional humanized antibodies
The allotype of the human gamma-1 constant region gene in the expression plasmid pVgl.D.Tt was modified from GIm (z,a) to the z, non-a allotype. The overlap-extension PCR method (Higuchi, R., in "PCR Technology: Principles and Applications for DNA Amplification", Stockton Press, New York (1989), pp. 61-70) was used to generate the amino acid substitutions D356E and L358M (numbered according to the EU index of Kabat, E.A., et al., "Sequences of Proteins of Immunological Interest", 5th ed., National Institutes of Health, Bethesda, MD (1991)), using mutagenesis" primers. The PCR step was done using the QuikChange II Site-Directed Mutagenesis Kit (Stratagene). Following digestion or PCR generated product with Sfil and Eagl, the resulting restriction fragment was subcloned into a modified form of the pVgl.D.Tt expression vector containing an Nhel restriction site in the intron between the hinge and CH2 exons.
Mutations to the lower hinge region of the gamma-1 constant region gene were also generated by site-directed mutagenesis. Specifically amino acid substitutions L234A and L235A (numbered according to the EU index of Kabat, E.A., et al.) were generated using mutagenesis oligos. The PCR step was done using the QuikChange II Site-Directed Mutagenesis Kit (Stratagene) as described above. Following digestion of PCR generated product with Nhel and Eagl, the resulting restriction fragment was subcloned into the modified pVgl.D.Tt expression vector described above containing the D356E and L358M mutations and an Nhel site in the intron between the hinge and CH2 exons. Mutations were verified by nucleotide sequencing.
The amino acid sequences of the humanized VH and VL mini-exons are shown in Table 21. The resulting V gene fragments were cloned, respectively, into a modified form of pVgLD.Tt and pVk.
Human kidney cell line 293T/17 (American Type Culture Collection, Manassus, VA) was maintained in DMEM (BioWhittaker, Walkersville, MD) containing 10% Fetal Bovine Serum (FBS) (HyClone, Logan, UT), 0.1 mM MEM non-essential amino acids (Invitrogen Corporation) and 2 mM L-glutamine (Invitrogen Corporation), hereinafter referred to as 293 medium, at 37°C in a 7.5% CO2 incubator. For expression and purification of monoclonal antibodies after transient transfection, 293T/17 cells were incubated in DMEM containing 10% low-IgG FBS (HyClone), 0.1 mM MEM non-essential amino acids and 2 mM L-glutamine, hereinafter referred to as low-IgG 293 medium.
Transient transfection of 293T/17 cells was carried out using Lipofectamine 2000 (Invitrogen Corporation) following the manufacturer's recommendations. Approximately 2 x 107 cells per transfection were plated in a T-175 flask in 50 ml of 293 medium and grown overnight to confluence. The next day, 35 μg of light chain plasmid and 35 μg of heavy chain plasmid were combined with 3.75 ml of Hybridoma-SFM (HSFM) (Life Technologies, Rockville, MD). In a separate tube, 175 μl of Lipofectamine 2000 reagent and 3.75 ml of HSFM were combined and incubated for 5 min at room temperature. The 3.75 ml Lipofectamine 2000-HSFM mixture was mixed gently with the 3.75ml DNA-HSFM mixture and incubated at room temperature for 20 min. The medium covering the 293T/17 cells was aspirated and replaced with low-IgG 293 medium, then the lipofectamine-DNA complexes were added dropwise to the cells, mixed gently by swirling, and the cells were incubated for 7 days at 37°C in a 7.5% CO2 incubator before harvesting the supernatants. Transient transfectants producing 8E1.4, 8E1.5 and 8E1.6 were generated as described above. Expression of humanized 8El.4, 8El.5 and 8El.6 antibodies was measured by sandwich ELISA. MaxiSorp ELISA plates (Nunc Nalge International, Rochester, NY) were coated overnight at 40C with 100 μl/well of a 1: 1000 dilution of AffiniPure goat anti-human IgG Fcγ- chain specific polyclonal antibodies (Jackson ImmunoResearch Laboratories, Inc., West Grove, PA) in 0.2 M sodium carbonate-bicarbonate buffer, pH 9.4, washed with Wash Buffer (PBS containing 0.1% Tween 20), and blocked for 1 hr at room temperature with 300 μl/well of SuperBlock Blocking Buffer in TBS (Pierce Chemical Company, Rockford, IL). After washing with Wash Buffer, samples containing 8El.4, 8El.5 or 8El.6 were appropriately diluted in ELISA Buffer (PBS containing 1% BSA and 0.1% Tween 20) and 100 μl/well was applied to the ELISA plates. As a standard, humanized IgGl/κ antibody daclizumab (PDL BioPharma, Inc.) was used. After incubating the plates for 1 hr at room temperature, and washing with Wash Buffer, bound antibodies were detected using 100 μl/well of a 1:1000 dilution of HRP-conjugated goat anti-human kappa light chain specific polyclonal antibodies (Southern Biotechnology Associates, Inc., Birmingham, AL). After incubating for 1 hr at room temperature, and washing with Wash Buffer, color development was performed by adding 100 μl/well of ABTS Peroxidase Substrate/Peroxidase Solution B (KPL, Inc., Gaithersburg, MD). After incubating for 7 min at room temperature, color development was stopped by adding 50 μl/well of 2% oxalic acid. Absorbance was read at 415 nm using a VersaMax microplate reader (Molecular Devices Corporation, Sunnyvale, CA). Culture supernatants obtained from transiently transfected 293T/17 cells were analyzed by ELISA for production of 8El.4, 8El.5 and 8El.6. Expression levels of approximately 30-50 μg/ml were typically observed. Samples that were positive were subjected purification as described below.
Example 2.4.5: Purification of additional humanized antibodies
8El.4, 8El.5 and 8El.6 IgGl/κ monoclonal antibodies were purified from exhausted culture supernatant with a protein A Sepharose column as follows. Culture supernatants from transient transfections were harvested by centrifugation, and sterile filtered. The pH of the filtered supernatants was adjusted by addition of 1/50 volume of 1 M sodium citrate, pH 7.0. Supernatants were run over a 1 ml HiTrap Protein A HP column (GE Healthcare Bio-Sciences Corporation, Piscataway, NJ) that was pre-equilibrated with 20 mM sodium citrate, 150 mM NaCl, pH 7.0. The column was washed with the same buffer, and bound antibody was eluted with 20 mM sodium citrate, pH 3.5. After neutralization by addition of 1/50 volume of 1.5 M sodium citrate, pH 6.5, the pooled antibody fractions were concentrated to -0.5-1.0 mg/ml using a 15 ml Amicon Ultra-15 centrifugal filter device (30,000 dalton MWCO) (Millipore Corporation, Bedford, MA). Samples were then filter sterilized using a 0.2 μm Acrodisc syringe filter with HT Tuffryn membrane (Pall Corporation, East Hills, NY). The concentrations of the purified antibodies were determined by UV spectroscopy by measuring the absorbance at 280 nm (l mg/ml = 1.4 A280)-
SDS-PAGE analysis under non-reducing conditions indicated that the antibodies had a molecular weight of about 150-160 kD. Analysis under reducing conditions indicated that the antibodies were comprised of a heavy chain with a molecular weight of about 50 kD and a light chain with a molecular weight of about 25 kD. The purity of the antibodies appeared to be more than 95%.
Example 2.4.6: Characterization of additional humanized antibodies using Competition ELISA
MaxiSorp ELISA plates (Nalge Nunc International) were coated overnight at 40C with 100 μl/well of 1.0 μg/ml human IL-12 in 0.2 M sodium carbonate-bicarbonate buffer, pH 9.4, washed with Wash Buffer (PBS containing 0.1% Tween 20), and blocked for 1 hr at room temperature with 300 μl/well of SuperBlock Blocking Buffer in TBS (Pierce Chemical Company). After washing with Wash Buffer, a mixture of biotinylated 8El (0.8 μg/ml final concentration) and competitor antibody (8El or 8El.4 or 8El.5 or 8El.6) starting at 100 μg/ml final concentration and serially diluted 3-fold) in 100 μl/well of ELISA buffer was added in duplicate. As isotype controls, 100 μl/well of 100 μg/ml mouse IgGl/κ (MuFd79) or humanized IgGl/κ (HuFd79) monoclonal antibodies in ELISA buffer was used. As a no-competitor control, 100 μl/well of ELISA Buffer was used. After incubating the plates for 1 hr at room temperature, and washing with Wash Buffer, bound antibodies were detected using 100 μl/well of 1 μg/ml HRP-conjugated streptavidin (Pierce Chemical Company) in ELISA buffer. After incubating for 1 hr at room temperature, and washing with Wash Buffer, color development was performed by adding 100 μl/well of ABTS Peroxidase Substrate/Peroxidase Solution B (KPL, Inc.). After ' incubating for 5 min at room temperature, color development was stopped by adding 50 μl/well of 2% oxalic acid. Absorbance was read at 415 nm.
The affinity of 8E1.4, 8E1.5 and 8E1.6 to human IL-12 was analyzed by competition ELISA as described above. Both 8El and the three humanized versions competed with biotinylated 8El in a concentration-dependent manner. Table 23 shows the ICs0 values of 8El, 8E1.4, 8E1.5 and 8E1.6 obtained using the computer software GraphPad Prism (GraphPad Software Inc., San Diego, CA). Table 23: Binding properties of 8E1.4, 8E1.5 and 8E1.6 antibodies
Figure imgf000112_0001
The values represent IC50 (μg/ml) required to compete 0.8 (μg/ml) biotinylated 8El antibody.
Antibodies 1A6.10, 1A6.11, 1A6.12, 8E1.4 and 8El.5 were also generated using methods described in Example 2.3. Antibodies were expressed in COS cells and purified by Protein A affinity chromatography as described in examples 2.2.2 and 1.2.C, respectively. These purified mAbs were characterized for IC50 and KD according to example 1.1. B andl.l.C 2. Table 24 shows the binding properties of 1A6.10, 1A6.11, 1A6.12, 8E1.4 and 8E1.5.
Table 24: Kinetic and potency parameters of additional humanized antibodies
Figure imgf000112_0002
The present invention incorporates by reference in their entirety techniques well known in the field of molecular biology. These techniques include, but are not limited to, techniques described in the following publications:
Ausubel, F.M. et al. eds., Short Protocols In Molecular Biology (4th Ed. 1999) John Wiley & Sons, NY. (ISBN 0-471-32938-X).
Lu and Weiner eds., Cloning and Expression Vectors for Gene Function Analysis (2001) BioTechniques Press. Westborough, MA. 298 pp. (ISBN 1-881299-21-X).
Ill Kontermann and Dubel eds., Antibody Engineering (2001) Springer-Verlag. New York. 790 pp. (ISBN 3-540-41354-5).
Old, R. W. & S. B. Primrose, Principles of Gene Manipulation: An Introduction To Genetic
Engineering (3d Ed. 1985) Blackwell Scientific Publications, Boston. Studies in
Microbiology; V.2:409 pp. (ISBN 0-632-01318-4). Sambrook, J. et al. eds., Molecular Cloning: A Laboratory Manual (2d Ed. 1989) Cold Spring
Harbor Laboratory Press, NY. VoIs. 1-3. (ISBN 0-87969-309-6). Winnacker, EX. From Genes To Clones: Introduction To Gene Technology (1987) VCH
Publishers, NY (translated by Horst Ibelgaufts). 634 pp. (ISBN 0-89573-614-4).
References:
U.S. Patents
4,816,397,
4,816,567,
4,880,078;
5, 985,309,
5,223,409;
5,225,539;
5,258, 498;
5,290, 540,
5,290,540,
5,403,484;
5,427,908;
5,516,637;
5,530,101;
5,571,698;
5,580,717;
5,585,089,
5,658,727;
5,693,762,
5,714,352,
5,723,323,
5,733,743
5,750,753;
5,766886, ,780, 225; ,817,483, ,821,047; ,824,514, ,855,913, ,874,064, ,934,272, ,939,598, ,969,108; ,976,862, ,985, 320, ,985,309, ,985,615, ,998,209, ,624,821 ,723,323, ,763,192, ,814,476, ,075,181, ,091,001, ,114,598 ,130,364 ,180,370, ,204,023, ,350,861, ,585,089, ,914,128 ,526, 938, ,980,286, ,912,015; ,946,778, , 916,597; ,128,326; ,565,352). ,679,377; 5,989,463;
5,225,539;
5,565,332,
5,698,426;
5,714,350,
5,807,715,
6, 019,968,
6,019, 968,
5,627,052,
5,916,771,
5,648,260,
6,699,658,
U.S. Application
09/428,082
U. S patent publication
20020137134,
20040018590,
20030186374,
20050042664 Al
US2006104968
Foreign Patents
WO 97/15327, WO 00/56772 Al WO 91/17271, WO 92/09690, WO9937682 A2, WO 92/15679, WO 92/01047, WO2002097048A2, WO9524918 Al, PCT/GB91/01134, WO 99/25044, WO 97/29131, WO 98/31700 WO 99/15154; O 92/19244, O 92/19244, O 00/37504, O 00/56772. O 91/05548, O 91/09967, O 91/09967; WO 92/02551 WO92/22324 WO96/20698, WO 97/29131 WO01/83525. WO2003016466A2, WO2005100584 A2). WO 03/035835; WO 90/02809, PCT/US91/05939, PCT7US91/09630, PCT/US94/01234, PCT/US96/18978, PCT/US98/16280, WO 92/20791, WO 90/05144 Al WO 00 09560, WO 00/037504* WO 02072636, WO 91/10737, WO 91/10741 WO 92/01047, WO 92/18619, WO 93/11236, WO 94/02602, WO 95/15982, WO 95/20401, WO 96/33735 O 97/32572, O 97/32572, WO 97/44013, O 97/44013, WO 98/16654 WO 98/24893, WO 98/31346, WO 98/31346, WO 98/50433, WO 99/45031, WO 99/53049, WO 99/543428, WO 99/66903 WO 99/66903, WO90/14424, WO90/14430, WO90/14443 WO2004078140,
European Patent No:
EP 229246,
EP 239,400,
EP 239,400;
EP 519,596,
EP 519,596;
EP 592,106;
EP 592,106;
EP 1,176,195;
GB89/01334,
GB91/01134,
GB92/01755;
Other references:
Ames et al., J. Immunol. Methods 184:177-186 (1995), Babcock, J.S. et al. (1996) Proc. Natl. Acad. ScL USA 93:7843-7848,
Barbas et al. (1991) PNAS 88:7978-7982,
Bentley, D.L., and Rabbitts, T.H., Cell 32: 181-189 (1983),
Berrebi et al. (1998) Am. J Path 152:667-672,
Better et al., Science 240:1041-1043 (1988),
Bird et al. (1988) Science 242:423-426,
Breitling et al. PCT Publication No. WO 93/01288,
Brinkman et al., J. Immunol. Methods 182:41-50 (1995),
Broberg, et al. (2002) J. Interferon Cytokine Res. 22:641-651,
Brown et al. (1991) Proc. Natl. Acad. Sci. 88:2663-2667,
Bucht et al., (1996) Clin. Exp. Immunol. 103:347-367,
Buchwald et al., 1980, Surgery 88:507;
Burton et al., Advances in Immunology 57:191-280 (1994),
Carter et al., Proc. Natl. Acad. Sci. U.S.A. 89:4285 (1992);
Chothia &Lesk, J. MoI. Biol. 196:901-917 (1987),
Chothia et al., J. MoI. Biol. 227:799 (1992),
Chothia et al., Nature 342:877-883 (1989),
Clackson et al. (1991) Nature 352:624-628,
Cleek et al., 1997, "Biodegradable Polymeric Carriers for a bFGF Antibody for Cardiovascular
Application," Pro. Int'l. Symp. Control. ReI. Bioact. Mater. 24:853-854,
Co, M.S., et al., J. Immunol. 148: 1149-1154 (1992)
Co, M.S., et al., MoI. Immunol. (1993) 30:1361- 1367,
Cole, M.S., et al., J. Immunol. 159: 3613-3621 (1997),
Cooper, et al. (2002) J. Immunol. 168:1322-1327,
Cua et al. (2003) Nature 421:744-748,
Duchmarm et al., J Immunol. 26:934- 938(1996),
During et al., 1989, Ann. Neurol. 25:351;
Durocher et al., Nucleic Acids Research 2002, VoI 30, No.2,
Elkins, et al. (2002) Infection Immunity 70:1936-1948,
Fais et al. (1994) J Interferon Res. 14:235-238;
Foote and Winter (1992), J. MoI. Biol. 224:487-499,
Fuchs et al. (1991) Bio/Technology 9: 1370-1372,
Fuss, et al., (1996) J Immunol. 157:1261-1270,
Garrad et al. (1991) Bio/Technology 9: 1373-1377,
Giege, R. and Ducruix, A. Barrett, Crystallization of Nucleic Acids and Proteins, a Practical
Approach, 2nd ea., pp. 20 1-16, Oxford University Press, New York, New York, (1999), Gillies et al., (1989) J. Immunol. Methods 125:191-202,
Goldspiel et al., 1993, Clinical Pharmacy 12:488-505;
Goodson, in Medical Applications of Controlled Release, supra, vol. 2, pp. 115-138 (1984),
Gram et al. (1992) PNAS 89:3576-3580,
Green and Jakobovits J. Exp. Med. 188:483-495 (1998),
Green et al. Nature Genetics 7:13-21 (1994)
Griffiths et al. (1993) EMBO J 12:725-734,
Hammerling, et al.,: Monoclonal Antibodies and T-CeIl Hybridomas 563-681 (Elsevier, N. Y.,
1981),
Harlow et al., Antibodies: A Laboratory Manual, (Cold Spring Harbor Laboratory Press, 2nd ed.
1988),
Hawkins et al. (1992) JMoI Biol 226:889-896
Hay et al. (1992) Hum Antibod Hybridomas 3:81-85,
Hieter, P.A., et al., J. Biol. Chem. 257: 1516-1522 (1982),
Higuchi, R., in "PCR Technology: Principles and Applications for DNA Amplification",
Stockton Press, New York (1989), pp. 61-70),
Holliger, P., et al. (1993) Proc. Natl. Acad. Sci. USA 90:6444-6448,
Hoogenboom et al. (1991) Nuc Acid Res 19:4133-4137,
HoogenboomRR., (1997) 775 Tech. 15:62-70;
Azzazy H., and Highsmith W.E., (2002) Clin. Biochem. 35:425-445;
Gavilondo J.V., and Larrick J.W. (2002) BioTechniques 29: 128-145;
Hoogenboom H., and Chames P. (2000) Immunology Today 21:371-378,
Howard et al., 1989, J. Neurosurg. 7 1:105);
Huse et al. (1989) Science 246:1275-1281,
Huston et al. (1988) Proc. Natl. Acad. Sci. USA 85:5879-5883,
Huston et al., Methods in Enzymology 203:46-88 (1991);
Johnnson, B., et al. (1991) Anal. Biochem. 198:268-277.
Johnsson, B., et al. (1995) J. MoI. Recognit. 8:125-131,
Joliot et al., 1991, Proc. Natl. Acad. Sci. USA 88:1864-1868,
Jones et al., Nature 321:522 (1986);
Jδnsson, U., et al. (1993) Ann. Biol. Clin. 51:19-26; Jonsson,
Junghans, et al. (1990) Cancer Res. 50:1495-1502,
Kabat et al. (1971) Ann. NY Acad, Sci. 190:382-391.
Kabat, E.A., et al. (1991) Sequences of Proteins of Immunological Interest, Fifth Edition, U.S.
Department of Health and Human Services, NIH Publication No. 91-3242,
Kang et al. PCT Publication No. WO 92/18619, Kellermann S-A., and Green L.L. (2002) Current Opinion in Biotechnology 13:593-597,
Kettleborough et al. (1991) Prot. Engineer. 4:773-783,
Kettleborough et al., Eur. J. Immunol. 24:952-958 (1994),
Kipriyanov, S.M., et al (1994) MoI. Immunol. 31:1047-1058,
Kipriyanov, S.M., et al. (1995) Human Antibodies and Hybridomas 6:93-101,
Kobayashi, et al. (1989) J Exp Med 170:827-845;
Kohler, G. and Milstein 1975, Nature, 256:495,
Kontermann and Dubel eds., Antibody Engineering (2001) Springer-Verlag. New York. 790 pp.
(ISBN 3-540-41354-5),
Ladner et al. U.S. Patent No. 5,223,409,
Lam et al., 1997, "Microencapsulation of Recombinant Humanized Monoclonal Antibody for
Local Delivery," Proc. Int'l. Symp. Control ReI. Bioact. Mater. 24:759- 760,
Langer (1990), Science 249:1527-1533,
Langrish et.al. (2004) Immunological Reviews202: 96-105,
Levitt, M., J. MoI. Biol. 168: 595-620 (1983),
Levy et al., 1985, Science 228:190;
Ling, et al. (1995) J Exp Med 154:116-127;
Little M. et al (2000) Immunology Today 21:364-370,
Lund et al., 1991, J. Immunol., 147:2657,
MacCallum (J MoI Biol 262(5):732-45 (1996),
Manheimer-Lory, A., et al., J. Exp. Med. 174: 1639-1652 (1991),
Marchalonis et al., Adv Exp Med Biol. 484:13-30 (2001),
McCafferty et al, Nature (1990) 348:552-554,
Medical Applications of Controlled Release, Langer and Wise (eds.), CRC Pres., Boca Raton,
FIa. (1974); Controlled Drug Bioavailability, Drug Product Design and Performance,
Mendez et al., Nature Genetics 15:146-156 (1997),
Mizushima and Nagata, Nucleic Acids Research 1990, VoI 18, pg 5322,
Mizushima, S. and Nagata, S., (1990) Nucleic acids Research VoI 18, No. 17,
Monteleone et al., (1997) Gastroent. 112:1169-1178,
Morgan and Anderson, 1993, Ann. Rev. Biochem. 62:191-217; May, 1993, TIBTECH 11(5): 155-
215,
Morita et al., (1998) Arth. and Rheumat. 41:306-314,
Morrison et al., 1984, Proc. Natl. Acad. Sci. 81:851-855,
Morrison, Science 229:1202 (1985),
Mulligan, Science 260:926- 932 (1993);
Mullinax et al., BioTechniques 12(6):864-869 (1992), Murphy et al. J Exp Med 198:1951-1957;
Neuberger et al., 1984, Nature 312:604-608,
Neurath et al., J Exp. Med 182:1281-1290 (1995),
Ning et al., 1996, "Intratumoral Radioimmunotheraphy of a Human Colon Cancer Xenograft
Using a Sustained-Release Gel," Radiotherapy &Oncology 39:179-189,
Oi et al., BioTechniques 4:214 (1986);
Olee, T., et al., J. Exp. Med. 175: 831-842 (1992),
Oppmann et al. (2000) Immunity 13:715-725,
Padlan (PASEB J. 9:133-139 (1995),
Padlan, Molecular Immunology 28(4/5):489-498 (1991),
Parham, et al. (2002) J. Immunol. 168:5699-5708,
Parronchi et al (1997) Am. J. Path. 150:823-832,
Persic et al., Gene 187 9-18 (1997),
Petrey, D., Xiang, Z., Tang, C.L., Xie, L., Gimpelev, M., Mitros, T., Soto, C.S., Goldsmith-
Fischman, S., Kernytsky, A., Schlessinger, A., et al. 2003. Using multiple structure alignments, fast model building, and energetic analysis in fold recognition and homology modeling. Proteins
53 (Suppl. 6): 430-435),
Pirhonen, et al. (2002) J. Immunol. 169:5673-5678,
Podlaski, et al. (1992) Arch. Biochem. Biophys. 294:230-237,
Poljak, RJ., et al. (1994) Structure 2:1121-1123,
Presta et al., J. Immunol. 151:2623 (1993),
Queen, C, et al., Proc. Natl. Acad. Sci. USA 86: 10029-10033 (1989),
R. Jefferis, Biotechnol. Prog. 21 (2005), pp. 11-16,
RJ. Kaufman and P.A. Sharp (1982) MoI. Biol. 159:601-6211
Ranger and Peppas, 1983, J., Macromol. Sci. Rev. Macromol. Chem. 23:61;
Ravetch, J. V., et al., Cell 27: 583-591 (1981),
Remington's Pharmaceutical Sciences and Introduction to Pharmaceutical Dosage Forms, 19th ed., Mack Pub. Co., Easton, Pa. (1995),
Riechmann et al., Nature 332:323 (1988),
Roberts, R.W. and Szostak, J. W. (1997) Proc. Natl. Acad. Sci. USA 94:12297-12302,
Roguska. et al. , PNAS 91:969-973 (1994);
Rouillard, J.-M., et al., Nucleic Acids Res. 32: W176-W180 (2004),
Saudek et al., 1989, N. Engl. J. Med. 321:574),
Sawai et al., AJRI 34:26-34 (1995),
Schroeder, Jr., H.W. and Wang, J. Y., Proc. Natl. Acad. Sci. USA 87: 6146-6150 (1990), Seder, et al. (1993) Proc. Natl. Acad. Sci. 90:10188-10192,
Sefton, 1987, CRC Crit. Ref. Biomed. Eng. 14:20;
Shapiro et al., Crit. Rev. Immunol. 22(3): 183-200 (2002),
Shields, R. L. et al. (2002) J. Biol. Chem. 277:26733-26740,
Shu et al., PNAS 90:7995-7999 (1993);
Sims et al., J. Immunol. 151: 2296 (1993);
Skerra et al., Science 240:1038-1040 (1988).
Smolen and Ball (eds.), Wiley, New York (1984);
Song et al., 1995, "Antibody Mediated Lung Targeting of Long- Circulating Emulsions," PDA
Journal of Pharmaceutical Science &Technology 50:372-397,
Studnicka et al., Protein Engineering 7(6):805-814 (1994);
Sustained and Controlled Release Drug Delivery Systems, J.R. Robinson, ed., Marcel Dekker,
Inc., New York, 1978.
Takeda et al., 1985, Nature 314:452-454
Taylor, L. D., et al. (1992) Nucl. Acids Res. 20:6287-6295,
Tolstoshev, 1993, Ann. Rev. Pharmacol. Toxicol. 32:573-596;
Trinchieri et al., (2003) Immunity 19:641-644,
Trinchieri, G. (2003) Nat. Rev. Irnmun. 3:133-146,
V., et al. (1991) Biotechniques 11:620-627,
Umana et al. (1999) Nat. Biotech. 17:176-1,
Urlaub and Chasin, (1980) Proc. Natl. Acad. Sci. USA 77:4216-4220,
Verhoeyen et al., Science 239:1534 (1988)),
Wallick, S.C., et al., Exp. Med. (1988) 168:1099-1109,
Ward et al, (1989) Nature 341:544-546,
Welschof, M. and Krauss, J. Methods In Molecular Biology, VoI 207,
Windhagen et al., (1995) J Exp. Med. 182:1985-1996,
Winnaker, From Genes to Clones (Verlagsgesellschaft, Weinheim, Germany 1987),
Wright, A., et al., EMBO J. (1991) 10:2717 2723),
Wu and Wu, 1991, Biotherapy 3:87-95;
Wu and Wu, J. Biol. Chem. 262:4429-4432 (1987),
Although a number of embodiments and features have been described above, it will be understood by those skilled in the art that modifications and variations of the described embodiments and features may be made without departing from the present disclosure or the invention as defined in the appended claims. Each of the publications mentioned herein is incorporated by reference.

Claims

We claim:
1. A binding protein comprising an antigen binding domain, said binding protein capable of binding a p40 subunit of IL-12, said antigen binding domain comprising at least one CDR comprising an amino acid sequence selected from the group consisting of:
CDR-Hl. X1-X2-X3-X4-X5-X6-XT (SEQ ID NO: 55) , wherein; X1 is D, K, T, or S; X2 is Y, S, or T; X3 is Y, V, G, W, S, or F; X4 is I, or M; X5 is H, G, E, or V; X5 is V, or is not present; and X7 is S, or is not present;
CDR-H2. X1- X2~ X3—X4— X5— X6- X7— X8~ X9— XlO- XlI- Xl2~ Xl3~ Xl4~ Xl5-Xl6- Xl 7
X18-X19-X20 (SEQ ID NO: 56) , wherein;
XI is H, D, G, W, S, Y or R; X2 is I, or F;
X3 is Y, W, L, S, N, D or G;
X4 is W, P, H, T, or S;
X5 is D, G, E, A, or I;
X6 is D, G, S, T, or N;
X7 is D, G, S, or P;
X8 is K, N, S, E, T, or H;
X9 is Y, T, P, I, or N;
X10 is Y, N, T, H, K, S, or G;
XII is N, or Y;
X12 is P, N, A, D, or S; X13 is S, E, D, or P; X14 is L, K, D, T, or Y; X15 is K, F, V, M, R, or A; X16 is S, K, Q, P, or is not present; X17 is D, G, R, or is not present; X18 is F, or is not present; X19 is Q, or is not present; and X20 is D, or is not present;
CDR-H3. X1-X2-X3-X4-X5-Xe-X7-X8-Xg-XiO-XiI-X^-XiS (SEQ ID NO: 57) , wherein; X1 is R, N, or W; X2 is G, T, R, P, or H; X3 is I, R, F, Y, or Q; X4 is R, V, Y, F, or A; X5 is S, N, G, A, or R; X6 is A, Y, L, F, or M; X7 is M, A, D, L, or F; X8 is D, M, Y, or W; X9 is Y, D, or N; X10 is Y, A, or is not present; X11 is M, or is not present; X12 is D, or is not present; and X13 is Y, or is not present;
CDR-Ll. XI-X2-X3-X4-XS-X6-X7-X8-X9-X1O-XII-XI2-XI3-XI4-XIS (SEQ ID NO: 58) , wherein;
XI is K, or R; X2 is A;
X3 is S; X4 is Q, or E; X5 is S, or N; X6 is V, or I; X7 is S, G, or D; X8 is N, T, or K; X9 is D, N, or Y; X10 is V, G, or L;
XII is A, I, or H;
X12 is S, or is not present;
X13 is F, or is not present;
Xj.4 is M, or is not present; and
X15 is N, or is not present; CDR-L2. X1-X2-X3-X4-X5-X6-X7-X8 (SEQ ID WO: 59), wherein;
X1 is Y, or S;
X2 is A, or T;
X3 is S, or A;
X4 is N, H, S, or Q;
X5 is R, W, or S;
X6 is Y, Q, or I;
X7 is T, S, or G; and
X8 is S, or is not present; and CDR-L3. X1-X2-X3-X4-X5-Xe-X7-Xs-Xg (SEQ ID NO: 60), wherein; X1 is Q; X2 is Q;
X3 is D, Y, or S;
X4 is Y, N, K, or I;
X5 is N, T, S, or E;
X6 is S, Y, V, or W; X7 is P;
X8 is W, F, Y, L, or P; and
X9 is T, or S.
2. The binding protein according to claim 1, wherein said at least one CDR comprises an amino acid sequence selected from the group consisting of:
residues 31-37 of SEQ ID NO.:35; residues 50-66 of SEQ ID NO. :45; residues 52-67 of SEQ ID NO.:35; residues 99-101 of SEQ ID NO. :45; residues 100-108 of SEQ ID NO.-.35; residues 24-34 of SEQ ID NO. :46; residues 24-34 of SEQ ID NO.:36; residues 50-56 of SEQ ID NO. :46; residues 50-56 of SEQ ID NO. :36; residues 89-97 of SEQ ID NO. :46; residues 89-97 of SEQ ID NO.:36; residues 31-35 of SEQ ID NO. :47; residues 31-37 of SEQ ID NO. :37; residues 50-66 of SEQ ID NO. :47; residues 52-67 of SEQ ID NO.:37; residues 99-106 of SEQ ID NO. :47; residues.100-109 of SEQ ID NO.:37; residues 24-34 of SEQ ID NO. :48; residues 24-34 of SEQ ID NO.:38; residues 50-56 of SEQ ID NO. :48; residues 50-56 of SEQ ID NO.:38; residues 89-97 of SEQ ID NO. :48; residues 89-97 of SEQ ID NO.:38; residues 31-35 of SEQ ID NO. :49; residues 31-35 of SEQ ID NO.:39; residues 50-66 of SEQ ID NO. :49; residues 50-66 of SEQ ID NO.:39; residues 99-111 of SEQ ID NO. :49; residues 99-106 of SEQ ID NO.:39; residues 24-38 of SEQ ID NO. :50; residues 24-34 of SEQ ID Nθ.:40; residues 5.3-60 Of SEQ ID NO. :50; residues 50-56 of SEQ ID Nθ.:40; residues 93-101 of SEQ ID NO. :50; residues 89-97 of SEQ ID NO.: 40; residues 31-37 of SEQ ID NO. :51; residues 31-35 of SEQ ID NO. -.41; residues 52-67 Of SEQ ID NO. :51; residues 50-66 of SEQ ID NO. :41 residues 100-109 of SEQ ID NO. :51; residues 99-106 of SEQ ID Nθ.:41; residues 24-34 of SEQ ID NO. :52; residues 24-34 of SEQ ID NO.:42; residues 50-56 of SEQ ID NO. :52; residues 50-56 of SEQ ID NO.-.42; residues 89-97 of SEQ ID NO. :52; residues 89-97 of SEQ ID NO.:42; residues 31-35 Of SEQ ID NO. -.53; residues 31-35 of SEQ ID NO.:43; residues 47-66 of SEQ ID NO. :53; residues 50-66 of SEQ ID NO.:43; residues 99-107 Of SEQ ID NO. :53; residues 99-106 of SEQ ID NO.:43; residues 24-34 of SEQ ID NO. :54; residues 24-34 of SEQ ID NO.:44; residues 50-56 Of SEQ ID NO. :54; residues 50-56 of SEQ ID NO. :44; and residues 89-97 of SEQ ID NO.:44; residues 89-97 of SEQ ID NO. :54. residues 31-35 of SEQ ID NO.:45;
3. The binding protein according to claim 1, wherein said binding protein comprises at least 3 CDRs.
4. The binding protein according to claim 3, wherein said at least 3 CDRs are selected from a variable domain CDR set consisting of:
VH 1D4 CDR Set
VH 1D4 CDR-Hl Residues 31-37 of SEQ ID NO. :35
VH 1D4 CDR-H2 Residues 52-67 of SEQ ID NO. : 35
VH 1D4 CDR-H3 Residues 100-108 of SEQ ID NO. : 35
VL 1D4 CDR Set VL 1D4 CDR-Ll Residues24-34 of SEQ ID NO. :36
VL 1D4 CDR-L2 Residues50-56 of SEQ ID NO. : 36
VL 1D4 CDR-L3 Residues89-97 of SEQ ID NO. : 36
VH 1A6 CDR Set
VH 1A6 CDR-Hl Residues31-37 of SEQ ID NO. : 37
VH 1A6 CDR-H2 Residues52-67 of SEQ ID NO. : 37
VH 1A6 CDR-H3 ResidueslOO-109 of SEQ ID NO. : 37
VL 1A6 CDR Sθt
VL 1A6 CDR-Ll Residues24-34 of SEQ ID NO. : 38
VL 1A6 CDR-L2 Residues50-56 of SEQ ID NO. : 38
VL 1A6 CDR-L3 Residues89-97 of SEQ ID NO. 38
VH 1D8 CDR Sθt
VH 1D8 CDR-Hl Residues 31-35 of SEQ ID NO. 39
VH 1D8 CDR-H2 • Residues 50-66 of SEQ ID NO. 39
VH 1D8 CDR-H3 Residues 99-106 of SEQ ID NO. 39
VL 1D8 CDR Set
VL 1D8 CDR-Ll Residues 24-34 of SEQ ID NO :40
VL 1D8 CDR-L2 Residues 50-56 of SEQ ID NO. 40
VL 1D8 CDR-L3 Residues 89-97 of SEQ ID NO. 40
VH 3G7 CDR Set
VH 3G7 CDR-Hl Residues 31-35 of SEQ ID NO. 41
VH 3G7 CDR-H2 Residues 50-66 of SEQ ID NO. 41
VH 3G7 CDR-H3 Residues 99-106 of SEQ ID NO. 41
VL 3G7 CDR Set
VL 3G7 CDR-Ll Residues 24-34 of SEQ ID NO. 42
VL 3G7 CDR-L2 Residues 50-56 of SEQ ID NO. 42
VL 3G7 CDR-L3 Residues 89-97 of SEQ ID NO. 42
VH 5E8 CDR Set
VH 5E8 CDR-Hl Residues 31-35 of SEQ ID NO. 43
VH 5E8 CDR-H2 Residues 50-66 of SEQ ID NO. 43
VH 5E8 CDR-H3 Residues 99-106 of SEQ ID NO. . 43
VL 5E8 CDR Set
VL 5E8 CDR-Ll Residues 24-34 of SEQ ID NO. 44
VL 5E8 CDR-L2 Residues 50-56 of SEQ ID NO. 44
VL 5E8 CDR-L3 Residues 89-97 of SEQ ID NO. 44
VH 8El CDR Set
VH 8El CDR-Hl Residues 31-35 of SEQ ID NO. 45
VH 8El CDR-H2 Residues 50-66 of SEQ ID NO . :45
VH 8El CDR-H3 Residues 99-101 of SEQ ID NO. . 45
VL 8El CDR Set
VL 8El CDR-Ll Residues 24-34 of SEQ ID NO. : 46
VL 8El CDR-L2 Residues 50-56 of SEQ ID NO. : 46
VL 8El CDR-L3 Residues 89-97 of SEQ ID NO. : 46
VH 1H6 CDR Sθt
VH 1H6 CDR-Hl Residues 31-35 of SEQ ID NO. : 47
VH 1H6 CDR-H2 Residues 50-66 of SEQ ID NO. : 47
VH 1H6 CDR-H3 Residues 99-106 of SEQ ID NO. : 47
VL 1H6 CDR Set
VL 1H6 CDR-Ll Residues 24-34 of SEQ ID NO. : 48.
VL 1H6 CDR-L2 Residues 50-56 of SEQ ID NO. : 48
VL 1H6 CDR-L3 Residues 89-97 of SEQ ID NO. : 48
VH 3A11 CDB . Set
VH 3A11 CDR-Hl Residues 31-35 of SEQ ID NO. : 49
VH 3All CDR-H2 Residues 50-66 of SEQ ID NO. : 49
VH 3A11 CDR-H3 Residues 99-111 of SEQ ID NO. : 49 VL 3Al1 CDB. Sθt
VL 3A11 CDR-Ll Residues 24-38 of SEQ ID NO. 50
VL 3A11 CDR-L2 Residues 53-60 of SEQ ID NO. 50
VL 3A11 CDR-L3 Residues 93-101 of SEQ ID NO. 50
VH 4B4 CDR Set
VH 4B4 CDR-Hl Residues 31-37 of SEQ ID NO. 51
VH 4B4 CDR-H2 Residues 52-67 of SEQ ID NO. 51
VH 4B4 CDR-H3 Residues 100-109 of SEQ ID NO. 51
VL 4B4 CDR Sθt
VL 4B4 CDR-Ll Residues 24-34 of SEQ ID NO. 52
VL 4B4 CDR-L2 Residues 50-56 of SEQ ID NO. 52
VL 4B4 CDR-L3 Residues 89-97 of SEQ ID NO. : 52
VH 7G3 CDR Set
VH 7G3 CDR-Hl Residues 31-35 of SEQ ID NO. : 53
VH 7G3 CDR-H2 Residues 47-66 of SEQ ID NO. : 53
VH 7G3 CDR-H3 Residues 99-107 of SEQ ID NO. . 53
And
VL 7G3 CDR J Set
VL 7G3 CDR-Ll Residues 24-34 of SEQ ID NO. : 54
VL 7G3 CDR-L2 Residues 50-56 of SEQ ID NO. : 54
VL 7G3 CDR-L3 Residues 89-97 of SEQ ID NO. : 54
The binding protein according to claim 4, comprising at least two variable domain CDR sets.
The binding protein according to claim 5, , wherein said at least two variable domain CDR sets are selected from a group consisting of:
VH 1D4 CDR Set & VL 1D4 CDR Set;
VH 1A6 CDR Set & VL 1A6 CDR Set;
VH 1D8 CDR Set & VL 1D8 CDR Set;
VH 3G7 CDR Set & VL 3G7 CDR Set;
VH 5E8 CDR Set & VL 5E8 CDR Set;
VH 8El CDR Set & VL 8El CDR Set;
VH 1H6 CDR Set & VL 1H6 CDR Set;
VH 3Al 1 CDR Set & VL 3Al 1 CDR Set;
VH 4B4 CDR Set & VL 4B4 CDR Set; and
VH 7G3 CDR Set & VL 7G3 CDR Set.
The binding protein according to claim 3, further comprising a human acceptor framework.
The binding protein according to claim 4, further comprising a human acceptor framework.
The binding protein according to claim 5, further comprising a human acceptor framework.
10. The binding protein according to claim 6, further comprising a human acceptor framework.
11. The binding protein according to claim 7, wherein said human acceptor framework comprises a amino acid sequence selected from the group consisting of:
SEQ ID NO.: 6 SEQ ID NO.: 19 SEQ ID NO.: 32
SEQ ID NO.: 7 SEQ ID NO.: 20 SEQ ID NO.: 33
SEQ ID NO.: 8 SEQ ID NO.: 21 SEQ ID NO.: 34
SEQ ID NO.: 9 SEQ ID NO.: 22 SEQ ID NO.: 92
SEQ ID NO.: 10 SEQ ID NO.: 23 SEQ ID NO.: 93
SEQ ID NO.: 11 SEQ ID NO.: 24 SEQ ID NO.: 94
SEQ ID NO.: 12 SEQ ID NO.: 25 SEQ ID NO.: 95
SEQ ID NO..-13 SEQ ID NO.: 26 SEQ ID NO.: 96
SEQ ID NO.: 14 SEQ ID NO.: 27 AND
SEQ ID NO.: 15 SEQ ID NO.: 28 SEQ ID NO.: 97.
SEQ ID NO.:16 SEQ ID NO.:29
SEQ ID NO..-17 SEQ ID NO.:30
SEQ ID NO.: 18 SEQ ID NO..-31
12. The binding protein according to claim 8, wherein said human acceptor framework comprises a amino aicd sequence selected from the group consisting of:
SEQ ID NO.: 6 SEQ ID NO.: 19 SEQ ID NO.: 32
SEQ ID NO.:7 SEQ ID NO.:20 SEQ ID NO.:33
SEQ ID NO.:8 SEQ ID NO.: 21 SEQ ID NO..-34
SEQ ID NO.: 9 SEQ ID NO-: 22 SEQ ID NO.: 92
SEQ ID NO.: 10 SEQ ID NO.: 23 SEQ ID NO.: 93
SEQ ID NO..-11 SEQ ID NO.:24 SEQ ID NO.:94
SEQ ID NO.: 12 SEQ ID NO.: 25 SEQ ID NO.: 95
SEQ ID NO.:13 SEQ ID NO.:26 SEQ ID NO.:96
SEQ ID NO.: 14 SEQ ID NO.: 27 AND
SEQ ID NO..-15 SEQ ID NO.:28 SEQ ID NO.:97
SEQ ID NO.: 16 SEQ ID NO.: 29
SEQ ID NO.: 17 SEQ ID NO.: 30
SEQ ID NO.: 18 SEQ ID NO.:31
13. The binding protein according to claim 9, wherein said human acceptor framework comprises a amino aicd sequence selected from the group consisting of:
SEQ ID NO.: 6 SEQ ID NO.: 19 SEQ ID NO.: 32
SEQ ID NO.:7 SEQ ID NO.:20 SEQ ID NO.:33
SEQ ID NO.:8 SEQ ID NO.:21 SEQ ID NO.:34
SEQ ID NO.: 9 SEQ ID NO.: 22 SEQ ID NO.: 92
SEQ ID NO.: 10 SEQ ID NO.: 23 SEQ ID NO-: 93
SEQ ID NO.: 11 SEQ ID NO.: 24 SEQ ID NO.: 94
SEQ ID NO.: 12 SEQ ID NO.: 25 SEQ ID NO.: 95
SEQ ID NO.:13 SEQ ID NO.:26 SEQ ID NO.:96
SEQ ID NO.: 14 SEQ ID NO.: 27 AND
SEQ ID NO.: 15 SEQ ID NO.: 28 SEQ ID NO.: 97.
SEQ ID NO.: 16 SEQ ID NO.: 29
SEQ ID NO.:17 SEQ ID NO.:30
SEQ ID NO.:18 SEQ ID NO.:31
14. The binding protein according to claim 10, wherein said human acceptor framework comprises a amino aicd sequence selected from the group consisting of:
SEQ ID NO.: 6 SEQ ID NO.: 19 SEQ ID NO.: 32
SEQ ID NO.:7 SEQ ID NO.:20 SEQ ID NO.:33
SEQ ID NO.: 8 SEQ ID NO.: 21 SEQ ID NO.: 34
SEQ ID NO.: 9 SEQ ID NO.: 22 SEQ ID NO.: 92
SEQ ID NO.: 10 SEQ ID NO.: 23 SEQ ID NO.: 93
SEQ ID NO.:11 SEQ ID NO.:24 SEQ ID NO.:94
SEQ ID NO.: 12 SEQ ID NO.: 25 SEQ ID NO.: 95
SEQ ID NO.: 13 SEQ ID NO.: 26 SEQ ID NO.: 96
SEQ ID NO.: 14 SEQ ID NO.: 27 AND
SEQ ID NO.: 15 SEQ ID NO.: 28 SEQ ID NO.: 97.
SEQ ID NO.: 16 SEQ ID NO.: 29
SEQ ID NO.: 17 SEQ ID NO.: 30
SEQ ID NO.:18 SEQ ID NO.:31
15. The binding protein according to claim 1, wherein said binding protein comprises at least one variable domain having an amino acid sequence selected from the group consisting of;
SEQ ID NO . : 61 SEQ ID NO . : 62 SEQ ID NO.: 63 SEQ ID NO.: 69 SEQ ID NO.: 75
SEQ ID NO.: 64 SEQ ID NO.: 70 SEQ ID NO.: 76
SEQ ID NO.:65 SEQ ID NO.: 71 SEQ ID NO.:77
SEQ ID NO.: 66 SEQ ID NO.: 72 and
SEQ ID NO.: 67 SEQ ID NO.: 73 SEQ ID NO.: 78.
SEQ ID NO.:68 SEQ ID NO.:74
16. The binding protein according to claim 15 wherein said binding protein comprises two variable domains, wherein said two variable domains have amino acid sequences selected from the group consisting of;
SEQ ID NO.: 61 & SEQ ID NO.: 62, SEQ ID NO.: 63 & SEQ ID NO.: 64, SEQ ID NO.: 65 & SEQ ID NO.: 66, SEQ ID NO.: 67 & SEQ ID NO.: 68, SEQ ID NO.: 69 & SEQ ID NO.: 70, SEQ ID NO.: 71 & SEQ ID NO.: 72, SEQ ID NO.: 73 & SEQ ID NO.: 74, SEQ ID NO.:75 & SEQ ID NO.: 76, SEQ ID NO. -.77 & SEQ ID NO.: 78 SEQ ID NO.: 67 & SEQ ID NO.: 70, AND SEQ ID NO.: 69 & SEQ ID NO.: 68.
17. The binding protein according to claim 7, wherein said human acceptor framework comprises at least one Framework Region amino acid substitution at a key residue, said key residue selected from the group consisting of: a residue adjacent to a CDR; a glycosylation site residue; a rare residue; a residue capable of interacting with a p40 subunit of human IL-12; a residue capable of interacting with a CDR; a canonical residue; a contact residue between heavy chain variable region and light chain variable region; a residue within a Vernier zone;and a residue in a region that overlaps between a Chothia-defined variable heavy chain CDRl and a Kabat-defined first heavy chain framework.
18. The binding protein according to claim 10, wherein said human acceptor framework comprises at least one Framework Region amino acid substitution at a key residue, said key residue selected from the group consisting of: a residue adjacent to a CDR; a glycosylation site residue; a rare residue; a residue capable of interacting with a p40 subunit of human IL-12; a residue capable of interacting with a CDR; a canonical residue; a contact residue between heavy chain variable region and light chain variable region; a residue within a Vernier zone;and a residue in a region that overlaps between a Chothia-defined variable heavy chain CDRl and a Kabat-defined first heavy chain framework.
19. The binding protein according to claim 16, wherein said human acceptor framework comprises at least one Framework Region amino acid substitution at a key residue, said key residue selected from the group consisting of: a residue adjacent to a CDR; a glycosylation site residue; a rare residue; a residue capable of interacting with a p40 subunit of human IL-12; a residue capable of interacting with a CDR; a canonical residue; a contact residue between heavy chain variable region and light chain variable region; a residue within a Vernier zone;and a residue in a region that overlaps between a Chothia-defined variable heavy chain CDRl and a Kabat-defined first heavy chain framework.
20. The binding protein according to claim 17, wherein key residue selected from the group consisting of 3H, 5H, 1OH, HH, 12H, 13H, 15H, 16H, 18H, 19H, 23H, 24H, 25H, 30H1 41H, 44H, 46H, 49H, 66H, 68H, 7 IH, 73H, 74H, 75H, 76H, 77H, 78H, 79H, 8 IH, 82H, 82AH, 82BH, 82CH, 83H, 84H, 85H, 86H, 87H, 89H, 93H, 98H, 108H, 109H, IL, 2L, 3L, 7L, 8L, 9L, 1OL, HL, 12L, 13L, 15L, 17L, 19L, 20L, 21L, 22L, 36L, 41L, 42L, 43L, 45L, 46L, 58L, 6OL, 62L, 63L, 67L, 7OL, 73L, 74L, 77L, 78L, 79L, 80L, 83L, 85L, 87L, 104L, and 106L.
21. The binding protein according to claim 18, wherein key residue selected from the group consisting of 3H, 5H, 1OH, HH, 12H, 13H, 15H, 16H, 18H, 19H, 23H, 24H, 25H, 3OH, 41H, 44H, 46H, 49H, 66H, 68H, 71H, 73H, 74H, 75H, 76H, 77H, 78H, 79H, 81H, 82H, 82AH, 82BH, 82CH, 83H, 84H, 85H, 86H, 87H, 89H, 93H, 98H, 108H, 109H, IL, 2L, 3L, 7L, 8L, 9L, 1OL, HL, 12L, 13L, 15L, 17L, 19L, 2OL, 21L, 22L, 36L, 41L, 42L, 43L, 45L, 46L, 58L, 6OL, 62L, 63L, 67L, 7OL, 73L, 74L, 77L, 78L, 79L, 80L, 83L, 85L, 87L, 104L, and 106L.
22. The binding protein according to claim 19, wherein key residue selected from the group consisting of 3H, 5H, 1OH, HH, 12H, 13H, 15H, 16H, 18H, 19H, 23H, 24H, 25H, 30H, 41H, 44H, 46H, 49H, 66H, 68H, 71H, 73H, 74H, 75H, 76H, 77H, 78H, 79H, 81H, 82H, 82AH, 82BH, 82CH, 83H, 84H, 85H, 86H, 87H, 89H, 93H, 98H, 108H, 109H, IL, 2L, 3L, 7L, 8L, 9L, 1OL, HL, 12L, 13L, 15L, 17L, 19L, 2OL, 21L, 22L, 36L, 41L, 42L, 43L, 45L, 46L, 58L, 6OL, 62L, 63L, 67L, 7OL, 73L, 74L, 77L, 78L, 79L, 80L, 83L, 85L, 87L, 104L, and 106L.
23. The binding protein according to claim 17, wherein the binding protein is a consensus human ' variable domain.
24. The binding protein according to claim 18, wherein the binding protein is a consensus human variable domain.
25. The binding protein according to claim 19, wherein the binding protein is a consensus human variable domain.
26. The binding protein according to claim 7, wherein said human acceptor framework comprises at least one Framework Region amino acid substitution, wherein the amino acid sequence of the framework is at least 65% identical to the sequence of said human acceptor framework and comprises at least 70 amino acid residues identical to said human acceptor framework.
27. The binding protein according to claim 10, wherein said human acceptor framework comprises at least one Framework Region amino acid substitution, wherein the amino acid sequence of the framework is at least 65% identical to the sequence of said human acceptor framework and comprises at least 70 amino acid residues identical to said human acceptor framework.
28. The binding protein according to claim 16, wherein said human acceptor framework comprises at least one Framework Region amino acid substitution, wherein the amino acid sequence of the framework is at least 65% identical to the sequence of said human acceptor framework and comprises at least 70 amino acid residues identical to said human acceptor framework.
29. The binding protein according to claim 1, wherein said binding protein comprises at least one variable domain having an amino acid sequence selected from the group consisting of;
SEQ ID NO.: 79 SEQ ID NO.: 88 SEQ ID NO.: 103
SEQ ID NO.:80 SEQ ID NO.:89 SEQ ID NO.:104
SEQ ID NO.:81 SEQ ID NO..-90 SEQ ID NO.:105
SEQ ID NO.:82 SEQ ID NO.: 91 SEQ ID NO..-106
SEQ ID NO.: 83 SEQ ID NO.: 98 SEQ ID NO.: 107
SEQ ID NO.:84 SEQ ID NO.:99 SEQ ID NO.:108
SEQ ID NO.: 85 SEQ ID NO..-100 and
SEQ ID NO.:86 SEQ ID NO.: 101 SEQ ID NO..-109.
SEQ ID NO.: 87 SEQ ID KfO.: 102
30. The binding protein according to claim 29 wherein said binding protein comprises two variable domains, wherein said two variable domains have amino acid sequences selected from the group consisting of;
SEQ ID NO.: 67 & SEQ ID NO.:79,
SEQ ID NO.: 80 & SEQ ID NO.: 81,
SEQ ID NO.: 82 & SEQ ID NO.: 83,
SEQ ID NO-: 84 & SEQ ID NO.: 85,
SEQ ID NO.: 86 & SEQ ID NO.: 87,
SEQ ID NO.: 88 & SEQ ID NO.: 89,
SEQ ID NO.:90 & SEQ ID NO.:91,
SEQ ID NO.:98 & SEQ ID NO. :99,
SEQ ID NO.: 100 & SEQ ID NO.: 101,
SEQ ID NO.: 102 & SEQ ID NO.:103,
SEQ ID NO.:104 & SEQ ID NO..-105,
SEQ ID NO-: 106 & SEQ ID NO.: 107, AND
SEQ ID NO.: 108 & SEQ ID NO.: 109.
1. The binding protein according to claim 20, wherein said binding protein comprises at least one variable domain having an amino acid sequence selected from the group consisting of;
SEQ ID NO. :79 SEQ ID NO. :88 SEQ ID NO. :103 SEQ ID NO. :80 SEQ ID NO. :89 SEQ ID NO. :104 SEQ ID NO. :81 SEQ ID NO. :90 SEQ ID NO. :105 SEQ ID NO. :82 SEQ ID NO. :91 SEQ ID NO. :106 SEQ ID NO. :83 SEQ ID NO. :98 SEQ ID NO. :107 SEQ ID NO. :84 SEQ ID NO. :99 SEQ ID NO. :108, SEQ ID NO. :85 SEQ ID NO. :100 AND SEQ ID NO. :86 SEQ ID NO. :101 SEQ ID NO. :109. SEQ ID NO. :87 SEQ ID NO. :102
2 The binding protein according to claim 21, wherein said binding protein comprises at least one variable domain having an amino acid sequence selected from the group consisting of;
SEQ ID NO. :79 SEQ ID NO. 88 SEQ ID NO. :103 SEQ ID NO. :80 SEQ ID NO. 89 SEQ ID NO. :104 SEQ ID NO. :81 SEQ ID NO. 90 SEQ ID NO. :105 SEQ ID NO. :82 SEQ ID NO. 91 SEQ ID NO. :106 SEQ ID NO. :83 SEQ ID NO. :98 SEQ ID NO. :107 SEQ ID NO. :84 SEQ ID NO. 99 SEQ ID NO. :108, SEQ ID NO. :85 SEQ ID NO. :100 AND SEQ ID NO. :86 SEQ ID NO. 101 SEQ ID NO. :109. SEQ ID NO. :87 SEQ ID NO. :102
3 The binding protein according to claim 22, wherein said binding protein comprises at least one variable domain having an amino acid sequence selected from the group consisting of;
SEQ ID NO. :79 SEQ ID NO. :87 SEQ ID NO. :101 SEQ ID NO. :80 SEQ ID NO. :88 SEQ ID NO. :102 SEQ ID NO. :81 SEQ ID NO. :89 SEQ ID NO. :103 SEQ ID NO. :82 SEQ ID NO. :90 SEQ ID NO. :104 SEQ ID NO. :83 SEQ ID NO. :91 SEQ ID NO. :105 SEQ ID NO. :84 SEQ ID NO. :98 SEQ ID NO. :106 SEQ ID NO. :85 SEQ ID NO. :99 SEQ ID NO. :107 SEQ ID NO. :86 SEQ ID NO. :100 SEQ ID NO . : 108 , AND SEQ ID KTO . : 109 .
34. The binding protein according to claim 1, wherein the binding protein is capable of binding a target selected from the group consisting of IL- 12 and IL-23.
35. The binding protein according to claim 4, wherein the binding protein is capable of binding a target selected from the group consisting of IL-12 and IL-23.
36. The binding protein according to claim 6, wherein the binding protein is capable of binding a target selected from the group consisting of DL- 12 and IL-23.
37. The binding protein according to claim 7, wherein the binding protein is capable of binding a target selected from the group consisting of IL-12 and 1L-23.
38. The binding protein according to claim 11, wherein the binding protein is capable of binding a target selected from the group consisting of IL-12 and IL-23.
39. The binding protein according to claim 15, wherein the binding protein is capable of binding a target selected from the group consisting of IL-12 and IL-23.
40. The binding protein according to claim 17, wherein the binding protein is capable of binding a target selected from the group consisting of IL-12 and IL-23.
41. The binding protein according to claim 20, wherein the binding protein is capable of binding a target selected from the group consisting of IL-12 and EL-23.
42. The binding protein according to claim 26, wherein the binding protein is capable of binding a target selected from the group consisting of IL-12 and IL-23.
43. The binding protein according to claim 29, wherein the binding protein is capable of binding a target selected from the group consisting of IL-12 and IL-23.
44. The binding protein according to claim 34, wherein the binding protein is capable of modulating a biological function of a target selected from the group consisting of IL-12 and IL-23.
45. The binding protein according to claim 39, wherein the binding protein is capable of modulating a biological function of a target selected from the group consisting of IL-12 and IL-23.
46. The binding protein according to claim 43, wherein the binding protein is capable of modulating a biological function of a target selected from the group consisting of IL-12 and IL-23.
47. The binding protein according to claim 34, wherein the binding protein is capable of neutralizing a target selected from the group consisting of IL-12 and IL-23.
48. The binding protein according to claim 39, wherein the binding protein is capable of neutralizing a target selected from the group consisting of IL-12 and IL-23.
49. The binding protein according to claim 43, wherein the binding protein is capable of neutralizing a target selected from the group consisting of IL-12 and IL-23.
50. The binding protein according to claim 34, wherein said binding protein has an on rate constant(Kon)to said target selected from the group consisting of: at least about 102M4S"1; at least about 103M-1S"1; at least about 104M-1S"1; at least about 105M-1S"1; and at least about
10 M" s"1, as measured by surface plasmon resonance.
51. The binding protein according to claim 39, wherein said binding protein has an on rate constant(Kon)to said target selected from the group consisting of: at least about 102M-1S"1; at least about 103M-1S"1; at least about 104M-1S-1; at least about 105M-1S"1; and at least about 106M-1S"1, as measured by surface plasmon resonance.
52. The binding protein according to claim 42, wherein said binding protein has an on rate constant(Kon)to said target selected from the group consisting of: at least about 102M-1S"1; at least about 103M-1S"1; at least about 104M-1S-1; at least about 105M-1S-1; and at least about 106M-1S"1, as measured by surface plasmon resonance.
53. The binding protein according to claim 43, wherein said binding protein has an on rate constant(Kon)to said target selected from the group consisting of: at least about 102MT1S"1; at least about 103M-1S"1; at least about 104M-1S'1; at least about 105M-1S"1; and at least about 106M-1S"1, as measured by surface plasmon resonance.
54. The binding protein according to claim 34, wherein said binding protein has an off rate constant(Koff) to said target selected from the group consisting of: at most about 10'3S"1; at most about 10"4S"1; at most about 10"5S"1; and at most about 10"V\ as measured by surface plasmon resonance.
55. The binding protein according to claim 39, wherein said binding protein has an off rate constant(Koff) to said target selected from the group consisting of: at most about 10"3S"1; at most about 10"4S"1; at most about 10"5S"1; and at most about 10-6s-1, as measured by surface plasmon resonance.
56. The binding protein according to claim 42, wherein said binding protein has an off rate constant(Koff) to said target selected from the group consisting of: at most about 10"3S"1; at most about 10"4S"1; at most about 10"5S"1; and at most about 10"6S"1, as measured by surface plasmon resonance.
57. The binding protein according to claim 43, wherein said binding protein has an off rate constant(Koff) to said target selected from the group consisting of: at most about 10"3S"1; at most about 10"4S"1; at most about 10"5S"1; and at most about 10"6S"1, as measured by surface plasmon resonance.
58. The binding protein according to claim 34, wherein said binding protein has a dissociation constant (KD) to said target selected from the group consisting of: at most about 10"7 M; at most about lO"8 M; at most about 10"9 M; at most about 10"10 M; at most about 10"11 M; at most about 10"12 M; and at most 10"13 M.
59. The binding protein according to claim 39, wherein said binding protein has a dissociation constant (KD) to said target selected from the group consisting of: at most about 10"7 M; at most about 10"8 M; at most about 10"9 M; at most about 10"10 M; at most about 10"11 M; at most about 10"12 M; and at most 10"13M.
60. The binding protein according to claim 42, wherein said binding protein has a dissociation constant (KD) to said target selected from the group consisting of: at most about 10"7 M; at most about 10'8 M; at most about 10'9 M; at most about 10"10 M; at most about 10 M; at most about 10"12 M; and at most 10'13M.
61. The binding protein according to claim 43, wherein said binding protein has a dissociation constant (KD) to said target selected from the group consisting of: at most about 10"7 M; at most about 10'8 M; at most about 10'9 M; at most about 10'10 M; at most about 10"u M; at most about 10'12 M; and at most 10"13 M.
62. An antibody construct comprising a binding protein described in any one of claims 1-61, said antibody construct further comprising a linker polypeptide or an immunoglobulin constant domain.
63. The antibody construct according to claim 62, wherein said binding protein is selected from the group consisting of;
an immunoglobulin molecule, a disulfide linked Fv, a monoclonal antibody, a scFv, a chimeric antibody, a single domain antibody, a CDR-grafted antibody, a diabody, a humanized antibody, a multispecific antibody, a Fab, a dual specific antibody, and a Fab' , a bispecific antibody. aF(ab')2, a Fv,
64. The antibody construct according to claim 62, wherein said binding protein comprises a heavy chain immunoglobulin constant domain selected from the group consisting of;
a human IgM constant domain, a human IgG4 constant domain, a human IgGl constant domain, a human IgE constant domain, a human IgG2 constant domain, and a human IgG3 constant domain, a human IgA constant domain.
65. The antibody construct according to claim 62, comprising an immunnoglobulin constant domain having an amino acid sequence selected from the group consisting of: SEQ ID NO.:2 SEQ ID NO.:4 SEQ ID NO.:5.
SEQ ID NO.:3 and
66. An antibody conjugate comprising an antibody construct described in any one of claims 62-65, said antibody conjugate further comprising an agent selected from the group consisting of; an immunoadhension molecule, an imaging agent, a therapeutic agent, and a cytotoxic agent.
67. The antibody conjugate according to claim 66, wherein said agent is an imaging agent selected from the group consisting of a radiolabel, an enzyme, a fluorescent label, a luminescent label, a bioluminescent label, a magnetic label, and biotin.
68. The antibody conjugate according to claim 66, wherein said imaging agent is a radiolabel selected from the group consisting of: 3H, 14C, 35S, 90Y, 99Tc, 111In, 1231, 1311, 177Lu, 166Ho, and 153Sm.
69. The antibody conjugate according to claim 66, wherein said agent is a therapeutic or cytotoxic agent selected from the group consisting of; an anti-metabolite, an alkylating agent, an antibiotic, a growth factor, a cytokine, an anti-angiogenic agent, an anti-mitotic agent, an anthracycline, toxin, and an apoptotic agent.
70. The antibody construct according to claim 64, wherein said binding protein possesses a human glycosylation pattern.
71. The antibody conjugate according to claim 66, wherein said binding protein possesses a human glycosylation pattern.
72. The binding protein according to claim 3, wherein said binding protein exists as a crystal.
73. The antibody construct according to claim 62, wherein said antibody construct exists as a crystal.
74. The antibody conjugate according to claim 66, wherein said antibody construct exists as a crystal.
75. The binding protein according to claim 72, wherein said crystal is a carrier-free pharmaceutical controlled release crystal.
76. The antibody construct according to claim 73, wherein said crystal is a carrier-free pharmaceutical controlled release crystal.
77. The antibody conjugate according to claim 74, wherein said crystal is a carrier-free pharmaceutical controlled release crystal.
78. The binding protein according to claim 72, wherein said binding protein has a greater half life in vivo than the soluble counterpart of said binding protein.
79. The antibody construct according to claim 73, wherein said antibody construct has a greater half life in vivo than the soluble counterpart of said antibody construct.
80. The antibody conjugate according to claim 74, wherein said antibody conjugate has a greater half life in vivo than the soluble counterpart of said antibody conjugate.
81. The binding protein according to claim 72, wherein said binding protein retains biological activity.
82. The antibody construct according to claim 73, wherein said antibody construct retains biological activity.
83. The antibody conjugate according to claim 74, wherein said antibody conjugate retains biological activity.
84. An isolated nucleic acid encoding a binding protein amino acid sequence of any one of claims 1- 61.
85. An isolated nucleic acid encoding an antibody construct amino acid sequence of any one of claims 62-65.
86. An isolated nucleic acid encoding an antibody conjugate amino acid sequence of any one of claims 66-69.
87. A vector comprising an isolated nucleic acid according to any one of claims 84-86.
88. The vector of claim 87 wherein said vector is selected from the group consisting of pcDNA, pTT, pTT3, pEFBOS, pBV, pJV, and pBJ.
89. A host cell comprising a vector according to any one of claims 87-88.
90. The host cell according to claim 89, wherein said host cell is a prokaryotic cell.
91. The host cell according to claim 90, wherein said host cell is E.Coli.
92. The host cell according to claim 89, wherein said host cell is a eukaryotic cell.
93. The host cell according to claim 92, wherein said eukaryotic cell is selected from the group consisting of protist cell, animal cell, plant cell and fungal cell.
94. The host cell according to claim 92, wherein said eukaryotic cell is an animal cell selected from the group consisting of; a mammalian cell, an avian cell, and an insect cell.
95. The host cell according to claim 92, wherein said host cell is a CHO cell.
96. The host cell according to claim 92, wherein said host cell is COS.
97. The host cell according to claim 92, wherein said host cell is a yeast cell.
98. The host cell according to claim 97, wherein said yeast cell is Saccharomγces cerevisiae.
99. The host cell according to claim 92, wherein said host cell is an insect Sf9 cell.
100. A method of producing a protein capable of binding the p40 subunit of IL-12, comprising culturing a host cell of any one of claims 89-99 in culture medium under conditions sufficient to produce a binding protein capable of binding the p40 subunit of IL-12.
101. A protein produced according to the method of claim 100.
102. A composition for the release of a binding protein said composition comprising:
(a) a formulation, wherein said formulation comprises a crystallized binding protein, according to any one of claims 72-83, and an ingredient; and
(b) at least one polymeric carrier.
103. The composition according to claim 102, wherein said polymeric carrier is a polymer selected from one or more of the group consisting of: poly (acrylic acid), poly (cyanoacrylates), poly (amino acids), poly (anhydrides), poly (depsipeptide), poly (esters), poly (lactic acid), poly (lactic-co-glycolic acid) or PLGA, poly (b-hydroxybutryate), poly (caprolactone), poly (dioxanone); poly (ethylene glycol), poly ((hydroxypropyl) methacrylamide, poly [(organo) phosphazene], poly (ortho esters), poly (vinyl alcohol), poly (vinylpyrrolidone), maleic anhydride- alkyl vinyl ether copolymers, pluronic polyols, albumin, alginate, cellulose and cellulose derivatives, collagen, fibrin, gelatin, hyaluronic acid, oligosaccharides, glycaniinoglycans, sulfated polyeaccharides, blends and copolymers thereof.
104. The composition according to claim 102, wherein said ingredient is selected from the group consisting of albumin, sucrose, trehalose, lactitol, gelatin, hydroxypropyl-β- cyclodextrin, methoxypolyethylene glycol and polyethylene glycol.
105. A method for treating a mammal comprising the step of administering to the mammal an effective amount of the composition according to claim 96.
106. A pharmaceutical composition comprising the binding protein of any one of claims 1-61, and a pharmaceutically acceptable carrier.
107. The pharmaceutical composition of claim 106 wherein said pharmaceutically acceptable carrier functions as adjuvant useful to increase the absorption, or dispersion of said binding protein.
108. The pharmaceutical composition of claim 107 wherein said adjuvant is hyaluronidase.
109. The pharmaceutical composition of claim 106 further comprising at least one additional therapeutic agent for treating a disorder in which IL-12 activity is detrimental.
110. The pharmaceutical composition of claim 109, wherein said additional agent is selected from the group consisting of: Therapeutic agent, imaging agent, cytotoxic agent, angiogenesis inhibitors; kinase inhibitors; co-stimulation molecule blockers; adhesion molecule blockers; anti-cytokine antibody or functional fragment thereof; methotrexate; cyclosporin; rapamycin; FK506; detectable label or reporter; a TNF antagonist; an antirheumatic; a muscle relaxant, a narcotic, a non-steroid anti-inflammatory drug (NSADD), an analgesic, an anesthetic, a sedative, a local anesthetic, a neuromuscular blocker, an antimicrobial, an antipsoriatic, a corticosteriod, an anabolic steroid, an erythropoietin, an immunization, an immunoglobulin, an immunosuppressive, a growth hormone, a hormone replacement drug, a radiopharmaceutical, an antidepressant, an antipsychotic, a stimulant, an asthma medication, a beta agonist, an inhaled steroid, an epinephrine or analog, a cytokine, and a cytokine antagonist.
111. A method for reducing human IL-I? activity comprising contacting human 1L-12 with the binding protein of any one of claims 1-61 such that human IL- 12 activity is reduced.
112. A method for reducing human IL-12 activity in a human subject suffering from a disorder in which IL-12 activity is detrimental, comprising administering to the human subject the binding protein of any one of claims 1-61 such that human IL-12 activity in the human subject is reduced.
113. A method for treating a subject for a disease or a disorder in which IL-12 activity is detrimental by administering to the subject the binding protein of any one of claims 1-61 such that treatment is achieved.
114. The method of claim 113, wherein said disorder is selected from the group comprising rheumatoid arthritis, osteoarthritis, juvenile chronic arthritis, septic arthritis, Lyme arthritis, psoriatic arthritis, reactive arthritis, spondyloarthropathy, systemic lupus erythematosus, Crohn's disease, ulcerative colitis, inflammatory bowel disease, insulin dependent diabetes mellitus, thyroiditis, asthma, allergic diseases, psoriasis, dermatitis scleroderma, graft versus host disease, organ transplant rejection, acute or chronic immune disease associated with organ transplantation, sarcoidosis, atherosclerosis, disseminated intravascular coagulation, Kawasaki's disease, Grave's disease, nephrotic syndrome, chronic fatigue syndrome, Wegener's granulomatosis, Henoch-Schoenlein purpurea, microscopic vasculitis of the kidneys, chronic active hepatitis, uveitis, septic shock, toxic shock syndrome, sepsis syndrome, cachexia, infectious diseases, parasitic diseases, acquired immunodeficiency syndrome, acute transverse myelitis, Huntington's chorea, Parkinson's disease, Alzheimer's disease, stroke, primary biliary cirrhosis, hemolytic anemia, malignancies, heart failure, myocardial infarction, Addison's disease, sporadic, polyglandular deficiency type I and polyglandular deficiency type π, Schmidt's syndrome, adult (acute) respiratory distress syndrome, alopecia, alopecia areata, seronegative arthopathy, arthropathy, Reiter's disease, psoriatic arthropathy, ulcerative colitic arthropathy, enteropathic synovitis, chlamydia, yersinia and salmonella associated arthropathy, spondyloarthopathy, atheromatous disease/arteriosclerosis, atopic allergy, autoimmune bullous disease, pemphigus vulgaris, pemphigus foliaceus, pemphigoid, linear IgA disease, autoimmune haemolytic anaemia, Coombs positive haemolytic anaemia, acquired pernicious anaemia, juvenile pernicious anaemia, myalgic encephalitis/Royal Free Disease, chronic mucocutaneous candidiasis, giant cell arteritis, primary sclerosing hepatitis, cryptogenic autoimmune hepatitis, Acquired Immunodeficiency Disease Syndrome, Acquired Immunodeficiency Related Diseases, Hepatitis B, Hepatitis C, common varied immunodeficiency (common variable hypogammaglobulinaemia), dilated cardiomyopathy, female infertility, ovarian failure, premature ovarian failure, fibrotic lung disease, cryptogenic fibrosing alveolitis, post-inflammatory interstitial lung disease, interstitial pneumonitis, connective tissue disease associated interstitial lung disease, mixed connective tissue disease associated lung disease, systemic sclerosis associated interstitial lung disease, rheumatoid arthritis associated interstitial lung disease, systemic lupus erythematosus associated lung disease, dermatomyositis/polymyositis associated lung disease, Sjogren's disease associated lung disease, ankylosing spondylitis associated lung disease, vasculitic diffuse lung disease, haemosiderosis associated lung disease, drug-induced interstitial lung disease, fibrosis, radiation fibrosis, bronchiolitis obliterans, chronic eosinophilic pneumonia, lymphocytic infiltrative lung disease, postinfectious interstitial lung disease, gouty arthritis, autoimmune hepatitis, type-1 autoimmune hepatitis (classical autoimmune or lupoid hepatitis), type-2 autoimmune hepatitis (anti-LKM antibody hepatitis), autoimmune mediated hypoglycaemia, type B insulin resistance with acanthosis nigricans, hypoparathyroidism, acute immune disease associated with organ transplantation, chronic immune disease associated with organ transplantation, osteoarthrosis, primary sclerosing cholangitis, psoriasis type 1, psoriasis type 2, idiopathic leucopaenia, autoimmune neutropaenia, renal disease NOS, glomerulonephritides, microscopic vasulitis of the kidneys, lyme disease, discoid lupus erythematosus, male infertility idiopathic or NOS, sperm autoimmunity, multiple sclerosis (all subtypes), sympathetic ophthalmia, pulmonary hypertension secondary to connective tissue disease, Goodpasture's syndrome, pulmonary manifestation of polyarteritis nodosa, acute rheumatic fever, rheumatoid spondylitis, Still's disease, systemic sclerosis, Sjδrgren's syndrome, Takayasu's disease/arteritis, autoimmune thrombocytopaenia, idiopathic thrombocytopaenia, autoimmune thyroid disease, hyperthyroidism, goitrous autoimmune hypothyroidism (Hashimoto's disease), atrophic autoimmune hypothyroidism, primary myxoedema, phacogenic uveitis, primary vasculitis, vitiligo acute liver disease, chronic liver diseases, alcoholic cirrhosis, alcohol-induced liver injury, choleosatatis, idiosyncratic liver disease, Drug-Induced hepatitis, Non-alcoholic Steatohepatitis, allergy and asthma, group B streptococci (GBS) infection, mental disorders (e.g., depression and schizophrenia), Th2 Type and ThI Type mediated diseases, acute and chronic pain (different forms of pain), and cancers such as lung, breast, stomach, bladder, colon, pancreas, ovarian, prostate and rectal cancer and hematopoietic malignancies (leukemia and lymphoma), Abetalipoprotemia, Acrocyanosis, acute and chronic parasitic or infectious processes, acute leukemia, acute lymphoblastic leukemia (ALL), acute myeloid leukemia (AML), acute or chronic bacterial infection, acute pancreatitis, acute renal failure, adenocarcinomas, aerial ectopic beats, AIDS dementia complex, alcohol-induced hepatitis, allergic conjunctivitis, allergic contact dermatitis, allergic rhinitis, allograft rejection, alpha-1- antitrypsin deficiency, amyotrophic lateral sclerosis, anemia, angina pectoris, anterior horn cell degeneration, anti cd3 therapy, antiphospholipid syndrome, anti-receptor hypersensitivity reactions, aordic and peripheral aneuryisms, aortic dissection, arterial hypertension, arteriosclerosis, arteriovenous fistula, ataxia, atrial fibrillation (sustained or paroxysmal), atrial flutter, atrioventricular block, B cell lymphoma, bone graft rejection, bone marrow transplant (BMT) rejection, bundle branch block, Burkitt's lymphoma, Burns, cardiac arrhythmias, cardiac stun syndrome, cardiac tumors, cardiomyopathy, cardiopulmonary bypass inflammation response, cartilage transplant rejection, cerebellar cortical degenerations, cerebellar disorders, chaotic or multifocal atrial tachycardia, chemotherapy associated disorders, chromic myelocytic leukemia (CML), chronic alcoholism, chronic inflammatory pathologies, chronic lymphocytic leukemia (CLL), chronic obstructive pulmonary disease (COPD), chronic salicylate intoxication, colorectal carcinoma, congestive heart failure, conjunctivitis, contact dermatitis, cor pulmonale, coronary artery disease, Creutzfeldt-Jakob disease, culture negative sepsis, cystic fibrosis, cytokine therapy associated disorders, Dementia pugilistica, demyelinating diseases, dengue hemorrhagic fever, dermatitis, dermatologic conditions, diabetes, diabetes mellitus, diabetic aterosclerotic disease, Diffuse Lewy body disease, dilated congestive cardiomyopathy, disorders of the basal ganglia, Down's Syndrome in middle age, drug- induced movement disorders induced by drugs which block CNS dopamine receptors, drug sensitivity, eczema, encephalomyelitis, endocarditis, endocrinopathy, epiglottitis, epstein-barr virus infection, erythromelalgia, extrapyramidal and cerebellar disorders, familial hematophagocytic lymphohistiocytosis, fetal thymus implant rejection, Friedreich's ataxia, functional peripheral arterial disorders, fungal sepsis, gas gangrene, gastric ulcer, glomerular nephritis, graft rejection of any organ or tissue, gram negative sepsis, gram positive sepsis, granulomas due to intracellular organisms, hairy cell leukemia, Hallerrorden- Spatz disease, hashimoto's thyroiditis, hay fever, heart transplant rejection, hemachromatosis, hemodialysis, hemolytic uremic syndrome/thrombolytic thrombocytopenic purpura, hemorrhage, hepatitis (A), His bundle arrythmias, HTV infection/HIV neuropathy, Hodgkin's disease, hyperkinetic movement disorders, hypersensitity reactions, hypersensitivity pneumonitis, hypertension, hypokinetic movement disorders, hypothalamic-pituitary-adrenal axis evaluation, idiopathic Addison's disease, idiopathic pulmonary fibrosis, antibody mediated cytotoxicity, Asthenia, infantile spinal muscular atrophy, inflammation of the aorta, influenza a, ionizing radiation exposure, iridocyclitis/uveitis/optic neuritis, ischemia- reperfusion injury, ischemic stroke, juvenile rheumatoid arthritis, juvenile spinal muscular atrophy, Kaposi's sarcoma, kidney transplant rejection, legionella, leishmaniasis, leprosy, lesions of the corticospinal system, lipedema, liver transplant rejection, lymphedema, malaria, malignamt Lymphoma, malignant histiocytosis, malignant melanoma, meningitis, meningococcemia, metabolic/idiopathic, migraine headache, mitochondrial multi.system disorder, mixed connective tissue disease, monoclonal gammopathy, multiple myeloma, multiple systems degenerations (Mencel Dejerine- Thomas Shi-Drager and Machado- Joseph), myasthenia gravis, mycobacterium avium intracellulare, mycobacterium tuberculosis, myelodyplastic syndrome, myocardial infarction, myocardial ischemic disorders, nasopharyngeal carcinoma, neonatal chronic lung disease, nephritis, nephrosis, neurodegenerative diseases, neurogenic I muscular atrophies , neutropenic fever, non- hodgkins lymphoma, occlusion of the abdominal aorta and its branches, occulsive arterial disorders, okt3 therapy, orchitis/epidydimitis, orchitis/vasectomy reversal procedures, organomegaly, osteoporosis, pancreas transplant rejection, pancreatic carcinoma, paraneoplastic syndrome/hypercalcemia of malignancy, parathyroid transplant rejection, pelvic inflammatory disease, perennial rhinitis, pericardial disease, peripheral atherlosclerotic disease, peripheral vascular disorders, peritonitis, pernicious anemia, Pneumocystis carinii pneumonia, pneumonia, POEMS syndrome (polyneuropathy, organomegaly, endocrinopathy, monoclonal gammopathy, and skin changes syndrome), post perfusion syndrome, post pump syndrome, post-MI cardiotomy syndrome, preeclampsia, Progressive supranucleo Palsy, primary pulmonary hypertension, radiation therapy, Raynaud's phenomenon and disease, Raynoud's disease, Refsum's disease, regular narrow QRS tachycardia, renovascular hypertension, reperfusion injury, restrictive cardiomyopathy, sarcomas, scleroderma, senile chorea, Senile Dementia of Lewy body type, seronegative arthropathies, shock, sickle cell anemia, skin allograft rejection, skin changes syndrome, small bowel transplant rejection, solid tumors, specific arrythmias, spinal ataxia, spinocerebellar degenerations, streptococcal myositis, structural lesions of the cerebellum, Subacute sclerosing panencephalitis, Syncope, syphilis of the cardiovascular system, systemic anaphalaxis, systemic inflammatory response syndrome, systemic onset juvenile rheumatoid arthritis, T-cell or FAB ALL, Telangiectasia, thromboangitis obliterans, thrombocytopenia, toxicity, transplants, trauma/hemorrhage, type IH hypersensitivity reactions, type IV hypersensitivity, unstable angina, uremia, urosepsis, urticaria, valvular heart diseases, varicose veins, vasculitis, venous diseases, venous thrombosis, ventricular fibrillation, viral and fungal infections, vital encephalitis/aseptic meningitis, vital-associated hemaphagocytic syndrome, Wernicke- Korsakoff syndrome, Wilson's disease, xenograft rejection of any organ or tissue.
A method of treating a patient suffering from a disorder in which IL- 12 is detrimental comprising the step of administering the binding protein of any one of claims 1-61 before, concurrent, or after the administration of a second agent, wherein the second agent is selected from the group consisting of budenoside, epidermal growth factor, corticosteroids, cyclosporin, sulfasalazine, aminosalicylates, 6-mercaptopurine, azathioprine, metronidazole, lipoxygenase inhibitors, mesalamine, olsalazine, balsalazide, antioxidants, thromboxane inhibitors, IL-I receptor antagonists, anti-IL-lβ monoclonal antibodies, anti-IL-6 monoclonal antibodies, growth factors, elastase inhibitors, pyridinyl-imidazole compounds, antibodies or agonists of TNF, LT, IL-I, IL- 2, IL-6, IL-7, IL-8, IL-15, IL-16, IL-18, EMAP-H, GM-CSF, FGF, and PDGF, antibodies of CD2, CD3, CD4, CD8, CD25, CD28, CD30, CD40, CD45, CD69, CD90 or their ligands, methotrexate, cyclosporin, FK506, rapamycin, mycophenolate mofetil, leflunomide, NSAIDs, ibuprofen, corticosteroids, prednisolone, phosphodiesterase inhibitors, adensosine agonists, antithrombotic agents, complement inhibitors, adrenergic agents, IRAK, NIK, IKK, p38, MAP kinase inhibitors, IL-lβ converting enzyme inhibitors, TNFαconverting enzyme inhibitors, T-cell signalling inhibitors, metalloproteinase inhibitors, sulfasalazine, azathioprine, 6- mercaptopurines, angiotensin converting enzyme inhibitors, soluble cytokine receptors, soluble p55 TNF receptor, soluble p75 TNF receptor, sIL-lRI, sEL-lRII, sIL-6R, antiinflammatory cytokines, IL-4, IL-10, EL-Il, IL-13 and TGFβ.
The method according to claim 113, wherein said administering to the subject is by at least one mode selected from parenteral, subcutaneous, intramuscular, intravenous, intrarticular, intrabronchial, intraabdominal, intracapsular, intracartilaginous, intracavitary, intracelial, intracerebellar, intracerebroventricular, intracolic, intracervical, intragastric, intrahepatic, intramyocardial, intraosteal, intrapelvic, intrapericardiac, intraperitoneal, intrapleural, intraprostatic, intrapulmonary, intrarectal, intrarenal, intraretinal, intraspinal, intrasynovial, intrathoracic, intrauterine, intravesical, bolus, vaginal, rectal, buccal, sublingual, intranasal, and transdermal.
PCT/US2006/025584 2005-06-30 2006-06-29 Il-12/p40 binding proteins WO2007005608A2 (en)

Priority Applications (12)

Application Number Priority Date Filing Date Title
BRPI0611714-7A BRPI0611714A2 (en) 2005-06-30 2006-06-29 il-12 / p40 binding proteins
JP2008519612A JP5396080B2 (en) 2005-06-30 2006-06-29 IL-12 / p40 binding protein
CA002612239A CA2612239A1 (en) 2005-06-30 2006-06-29 Il-12/p40 binding proteins
EP20060785967 EP1907421A4 (en) 2005-06-30 2006-06-29 Il-12/p40 binding proteins
CN2006800240973A CN101379085B (en) 2005-06-30 2006-06-29 IL-12/P40 binding proteins
MX2007016401A MX2007016401A (en) 2005-06-30 2006-06-29 Il-12/p40 binding proteins.
AU2006265932A AU2006265932B2 (en) 2005-06-30 2006-06-29 IL-12/p40 binding proteins
KR1020137014441A KR20130080058A (en) 2005-06-30 2006-06-29 Il-12/p40 binding proteins
IL187691A IL187691A (en) 2005-06-30 2007-11-27 Binding protein capable of binding a p40 subunit of il-12 and il-23, method for preparing the same and a pharmaceutical composition for reducing or inhibiting il-12 activity
NO20080557A NO20080557L (en) 2005-06-30 2008-01-30 IL-12 / P40 binding proteins
IL216803A IL216803A0 (en) 2005-06-30 2011-12-06 A binding protein capable of binding a p40 subunit of il-12 and il-23, a method for preparing the same and a pharmaceutical composition for reducing or inhibiting il-12 activity
IL228793A IL228793A0 (en) 2005-06-30 2013-10-08 Il-12/p40 binding proteins

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US69567905P 2005-06-30 2005-06-30
US60/695,679 2005-06-30

Publications (2)

Publication Number Publication Date
WO2007005608A2 true WO2007005608A2 (en) 2007-01-11
WO2007005608A3 WO2007005608A3 (en) 2008-10-09

Family

ID=37605037

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2006/025584 WO2007005608A2 (en) 2005-06-30 2006-06-29 Il-12/p40 binding proteins

Country Status (18)

Country Link
US (3) US7700739B2 (en)
EP (1) EP1907421A4 (en)
JP (2) JP5396080B2 (en)
KR (2) KR20130080058A (en)
CN (11) CN103145837A (en)
AU (1) AU2006265932B2 (en)
BR (1) BRPI0611714A2 (en)
CA (2) CA2612239A1 (en)
CR (10) CR9599A (en)
IL (3) IL187691A (en)
MX (1) MX2007016401A (en)
NO (1) NO20080557L (en)
NZ (1) NZ596992A (en)
RU (1) RU2461571C2 (en)
TW (2) TW201444869A (en)
UA (1) UA96922C2 (en)
WO (1) WO2007005608A2 (en)
ZA (1) ZA201201216B (en)

Cited By (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2148888A1 (en) * 2007-05-30 2010-02-03 Abbott Laboratories Humanized antibodies which bind to a (1-42) globulomer and uses thereof
EP2220120A2 (en) * 2007-11-27 2010-08-25 Ablynx N.V. Amino acid sequences directed against heterodimeric cytokines and/or their receptors and polypeptides comprising the same
WO2010102251A3 (en) * 2009-03-05 2010-11-25 Abbott Laboratories Il-17 binding proteins
CN102177178A (en) * 2008-08-14 2011-09-07 赛福伦澳大利亚私人有限公司 Anti-IL-12/IL-23 antibodies
WO2011029892A3 (en) * 2009-09-11 2011-09-15 F. Hoffmann-La Roche Ag Highly concentrated pharmaceutical formulations comprising anti - cd20 antibody
US8168760B2 (en) 2007-03-29 2012-05-01 Abbott Laboratories Crystalline anti-human IL-12 antibodies
CN101935687B (en) * 2009-06-29 2012-11-07 中国科学院上海生命科学研究院 Application of IL-12p40 siRNA to promoting liver regeneration
US8524230B2 (en) 2005-09-01 2013-09-03 Merck Sharp & Dohme Corp. Use of IL-23 and IL-17 antagonists to treat autoimmune ocular inflammatory disease
US8580954B2 (en) 2006-03-28 2013-11-12 Hospira, Inc. Formulations of low dose diclofenac and beta-cyclodextrin
US9303085B2 (en) 2010-05-14 2016-04-05 Abbvie Inc. IL-1 binding proteins
US9610301B2 (en) 2008-01-15 2017-04-04 Abbvie Deutschland Gmbh & Co Kg Powdered protein compositions and methods of making same
US9695232B2 (en) 2012-10-03 2017-07-04 Philogen S.P.A. Anti-ED-A immunoconjugates for inflammatory bowel disease
WO2018081074A1 (en) * 2016-10-26 2018-05-03 Cedars-Sinai Medical Center Neutralizing anti-tl1a monoclonal antibodies
US10316083B2 (en) 2013-07-19 2019-06-11 Cedars-Sinai Medical Center Signature of TL1A (TNFSF15) signaling pathway
US10633449B2 (en) 2013-03-27 2020-04-28 Cedars-Sinai Medical Center Treatment and reversal of fibrosis and inflammation by inhibition of the TL1A-DR3 signaling pathway
US10689439B2 (en) 2018-04-25 2020-06-23 Prometheus Biosciences, Inc. Optimized anti-TL1A antibodies
US11001623B2 (en) 2014-01-30 2021-05-11 Coherus Biosciences, Inc. Method of manufacturing a protein by perfusion in media with a low amino acid concentration
US11186872B2 (en) 2016-03-17 2021-11-30 Cedars-Sinai Medical Center Methods of diagnosing inflammatory bowel disease through RNASET2
US11236393B2 (en) 2008-11-26 2022-02-01 Cedars-Sinai Medical Center Methods of determining responsiveness to anti-TNFα therapy in inflammatory bowel disease
EP3827845B1 (en) 2015-11-03 2022-03-30 Janssen Biotech, Inc. Subcutaneous formulations of anti-cd38 antibodies and their uses
US11292848B2 (en) 2019-10-24 2022-04-05 Prometheus Biosciences, Inc. Humanized antibodies to TNF-like ligand 1A (TL1A) and uses thereof
US11459404B2 (en) 2013-02-26 2022-10-04 Roche Glycart Ag Bispecific T cell activating antigen binding molecules

Families Citing this family (35)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2612239A1 (en) * 2005-06-30 2007-01-11 Abbott Laboratories Il-12/p40 binding proteins
US20110165063A1 (en) * 2009-01-29 2011-07-07 Abbott Laboratories Il-1 binding proteins
JO3244B1 (en) 2009-10-26 2018-03-08 Amgen Inc Human il-23 antigen binding proteins
BR112012013148A2 (en) * 2009-12-29 2017-03-21 F Hoffmann - La Roche Ag pharmaceutical formulation and use
MX2012010116A (en) * 2010-03-01 2013-02-26 Alexion Pharma Inc Methods and compositions for treating degos' disease.
MX2012011629A (en) * 2010-04-07 2013-03-05 Abbvie Inc Tnf-î± binding proteins.
ES2395801B1 (en) * 2011-06-23 2014-06-06 María Carmen PARDINA PALLEJÀ "PENTOXIFILINA BY TRANSVAGINAL ROUTE FOR THE TREATMENT OF INFERTILITY"   
SG11201401360XA (en) 2011-10-25 2014-05-29 Onclave Therapeutics Ltd Antibody formulations and methods
US20140010820A1 (en) * 2012-05-21 2014-01-09 Abbvie, Inc. Novel purification of non-human antibodies using protein a affinity chromatography
US10247730B2 (en) * 2013-02-14 2019-04-02 Faron Pharmaceuticals Oy Method for determining acute respiratory distress syndrome (ARDS) related biomarkers, a method to monitor the development and treatment of ARDS in a patient
CA2920835A1 (en) 2013-08-20 2015-02-26 Anutra Medical, Inc. Syringe fill system and method
USD763433S1 (en) 2014-06-06 2016-08-09 Anutra Medical, Inc. Delivery system cassette
USD750768S1 (en) 2014-06-06 2016-03-01 Anutra Medical, Inc. Fluid administration syringe
USD774182S1 (en) 2014-06-06 2016-12-13 Anutra Medical, Inc. Anesthetic delivery device
US9616114B1 (en) 2014-09-18 2017-04-11 David Gordon Bermudes Modified bacteria having improved pharmacokinetics and tumor colonization enhancing antitumor activity
CN105353135A (en) * 2015-11-23 2016-02-24 中国人民解放军第三军医大学第一附属医院 Use of Alzheimer's disease marker
CN109641960A (en) 2016-06-29 2019-04-16 检查点治疗公司 PD-L1 specific antibody and the method for using it
US11129906B1 (en) 2016-12-07 2021-09-28 David Gordon Bermudes Chimeric protein toxins for expression by therapeutic bacteria
US11180535B1 (en) 2016-12-07 2021-11-23 David Gordon Bermudes Saccharide binding, tumor penetration, and cytotoxic antitumor chimeric peptides from therapeutic bacteria
US11166985B2 (en) 2017-05-12 2021-11-09 Crispr Therapeutics Ag Materials and methods for engineering cells and uses thereof in immuno-oncology
JP7356354B2 (en) 2017-05-12 2023-10-04 クリスパー セラピューティクス アクチェンゲゼルシャフト Materials and methods for the manipulation of cells and their use in immuno-oncology
EP4233989A3 (en) 2017-06-07 2023-10-11 Shifamed Holdings, LLC Intravascular fluid movement devices, systems, and methods of use
JP7319266B2 (en) 2017-11-13 2023-08-01 シファメド・ホールディングス・エルエルシー Intravascular fluid transfer devices, systems and methods of use
US10722631B2 (en) 2018-02-01 2020-07-28 Shifamed Holdings, Llc Intravascular blood pumps and methods of use and manufacture
CN112105420A (en) 2018-05-11 2020-12-18 克里斯珀医疗股份公司 Methods and compositions for treating cancer
JP2021530246A (en) * 2018-07-03 2021-11-11 マレンゴ・セラピューティクス,インコーポレーテッド Anti-TCR antibody molecule and its use
KR20230148273A (en) 2018-09-24 2023-10-24 얀센 바이오테크 인코포레이티드 Safe and effective method of treating ulcerative colitis with anti-IL12/IL23 antibody
EP3941934A4 (en) 2019-03-18 2022-12-07 Janssen Biotech, Inc. Method of treating psoriasis in pediatric subjects with anti-il12/il23 antibody
MA55797A (en) 2019-04-30 2022-03-09 Crispr Therapeutics Ag ALLOGENIC CELL THERAPY OF B LYMPHOCYTE MALIGNITIES USING GENETICALLY MODIFIED T LYMPHOCYTES TARGETING CD19
WO2021016372A1 (en) 2019-07-22 2021-01-28 Shifamed Holdings, Llc Intravascular blood pumps with struts and methods of use and manufacture
KR20220038775A (en) * 2019-07-30 2022-03-29 아케소 바이오파마, 인크. Anti-human P40 protein domain antibodies and uses thereof
EP4034192A4 (en) 2019-09-25 2023-11-29 Shifamed Holdings, LLC Intravascular blood pump systems and methods of use and control thereof
CN111812014B (en) * 2020-08-05 2023-04-18 上海市血液中心 Platelet antibody detection method and platelet preservation method based on nano material
CN113092202A (en) * 2021-03-25 2021-07-09 上海福君基因生物科技有限公司 Gastric cancer prognosis prediction device
WO2023070038A2 (en) * 2021-10-20 2023-04-27 Synthekine, Inc. Human il-12p40 variants and uses thereof

Family Cites Families (100)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0092918B1 (en) 1982-04-22 1988-10-19 Imperial Chemical Industries Plc Continuous release formulations
GB8308235D0 (en) 1983-03-25 1983-05-05 Celltech Ltd Polypeptides
US4816567A (en) * 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US5807715A (en) 1984-08-27 1998-09-15 The Board Of Trustees Of The Leland Stanford Junior University Methods and transformed mammalian lymphocyte cells for producing functional antigen-binding protein including chimeric immunoglobulin
US5128326A (en) 1984-12-06 1992-07-07 Biomatrix, Inc. Drug delivery systems based on hyaluronans derivatives thereof and their salts and methods of producing same
EP0590689B2 (en) 1985-03-30 2006-08-16 KAUFFMAN, Stuart A. Method for obtaining DNA, RNA, peptides, polypeptides or proteins by means of a DNA-recombinant technique
US6492107B1 (en) 1986-11-20 2002-12-10 Stuart Kauffman Process for obtaining DNA, RNA, peptides, polypeptides, or protein, by recombinant DNA technique
US4980286A (en) 1985-07-05 1990-12-25 Whitehead Institute For Biomedical Research In vivo introduction and expression of foreign genetic material in epithelial cells
US5618920A (en) 1985-11-01 1997-04-08 Xoma Corporation Modular assembly of antibody genes, antibodies prepared thereby and use
US5225539A (en) 1986-03-27 1993-07-06 Medical Research Council Recombinant altered antibodies and methods of making altered antibodies
GB8607679D0 (en) 1986-03-27 1986-04-30 Winter G P Recombinant dna product
US4946778A (en) 1987-09-21 1990-08-07 Genex Corporation Single polypeptide chain binding molecules
DE3883899T3 (en) 1987-03-18 1999-04-22 Sb2 Inc CHANGED ANTIBODIES.
US5258498A (en) 1987-05-21 1993-11-02 Creative Biomolecules, Inc. Polypeptide linkers for production of biosynthetic proteins
US4880078A (en) 1987-06-29 1989-11-14 Honda Giken Kogyo Kabushiki Kaisha Exhaust muffler
EP0768377A1 (en) 1988-09-02 1997-04-16 Protein Engineering Corporation Generation and selection of recombinant varied binding proteins
US5223409A (en) 1988-09-02 1993-06-29 Protein Engineering Corp. Directed evolution of novel binding proteins
ES2052027T5 (en) 1988-11-11 2005-04-16 Medical Research Council IMMUNOGLOBULINE VARIABLE DOMAIN SEQUENCE CLONING.
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
WO1990014443A1 (en) 1989-05-16 1990-11-29 Huse William D Co-expression of heteromeric receptors
CA2016841C (en) 1989-05-16 1999-09-21 William D. Huse A method for producing polymers having a preselected activity
CA2016842A1 (en) 1989-05-16 1990-11-16 Richard A. Lerner Method for tapping the immunological repertoire
AU6430190A (en) 1989-10-10 1991-05-16 Pitman-Moore, Inc. Sustained release composition for macromolecular proteins
WO1991006287A1 (en) 1989-11-06 1991-05-16 Enzytech, Inc. Protein microspheres and methods of using them
GB8928874D0 (en) 1989-12-21 1990-02-28 Celltech Ltd Humanised antibodies
AU7247191A (en) 1990-01-11 1991-08-05 Molecular Affinities Corporation Production of antibodies using gene libraries
US5780225A (en) 1990-01-12 1998-07-14 Stratagene Method for generating libaries of antibody genes comprising amplification of diverse antibody DNAs and methods for using these libraries for the production of diverse antigen combining molecules
JP3068180B2 (en) 1990-01-12 2000-07-24 アブジェニックス インコーポレイテッド Generation of heterologous antibodies
US6075181A (en) 1990-01-12 2000-06-13 Abgenix, Inc. Human antibodies derived from immunized xenomice
US5427908A (en) 1990-05-01 1995-06-27 Affymax Technologies N.V. Recombinant library screening methods
JPH06508511A (en) 1990-07-10 1994-09-29 ケンブリッジ アンティボディー テクノロジー リミティド Method for producing specific binding pair members
GB9015198D0 (en) 1990-07-10 1990-08-29 Brien Caroline J O Binding substance
WO1992002551A1 (en) 1990-08-02 1992-02-20 B.R. Centre Limited Methods for the production of proteins with a desired function
ATE174598T1 (en) 1990-08-24 1999-01-15 Ixsys Inc METHOD FOR PRODUCING OLIGONUCLEOTIDES WITH RANDOM CODONS
US5698426A (en) 1990-09-28 1997-12-16 Ixsys, Incorporated Surface expression libraries of heteromeric receptors
CA2095633C (en) 1990-12-03 2003-02-04 Lisa J. Garrard Enrichment method for variant proteins with altered binding properties
IL100460A (en) 1990-12-20 1997-06-10 Ixsys Method for optimization of binding proteins and nucleic acids encoding a binding protein produced thereby
AU1545692A (en) 1991-03-01 1992-10-06 Protein Engineering Corporation Process for the development of binding mini-proteins
WO1992018619A1 (en) 1991-04-10 1992-10-29 The Scripps Research Institute Heterodimeric receptor libraries using phagemids
ATE221379T1 (en) 1991-05-01 2002-08-15 Jackson H M Found Military Med METHOD FOR TREATING INFECTIOUS RESPIRATORY DISEASES
DE69233482T2 (en) 1991-05-17 2006-01-12 Merck & Co., Inc. Method for reducing the immunogenicity of antibody variable domains
WO1992022324A1 (en) 1991-06-14 1992-12-23 Xoma Corporation Microbially-produced antibody fragments and their conjugates
DE4122599C2 (en) 1991-07-08 1993-11-11 Deutsches Krebsforsch Phagemid for screening antibodies
ES2136092T3 (en) 1991-09-23 1999-11-16 Medical Res Council PROCEDURES FOR THE PRODUCTION OF HUMANIZED ANTIBODIES.
DE69229477T2 (en) 1991-09-23 1999-12-09 Cambridge Antibody Tech Methods for the production of humanized antibodies
ES2313867T3 (en) 1991-12-02 2009-03-16 Medical Research Council ANTI-AUTO ANTIBODY PRODUCTION OF ANTIBODY SEGMENT REPERTORIES EXPRESSED ON THE PAYMENT SURFACE.
CA2507749C (en) 1991-12-13 2010-08-24 Xoma Corporation Methods and materials for preparation of modified antibody variable domains and therapeutic uses thereof
US5714350A (en) 1992-03-09 1998-02-03 Protein Design Labs, Inc. Increasing antibody affinity by altering glycosylation in the immunoglobulin variable region
US5912015A (en) 1992-03-12 1999-06-15 Alkermes Controlled Therapeutics, Inc. Modulated release from biocompatible polymers
US5733743A (en) 1992-03-24 1998-03-31 Cambridge Antibody Technology Limited Methods for producing members of specific binding pairs
ES2301158T3 (en) 1992-07-24 2008-06-16 Amgen Fremont Inc. XENOGENIC ANTIBODY PRODUCTION.
US5639641A (en) 1992-09-09 1997-06-17 Immunogen Inc. Resurfacing of rodent antibodies
US5934272A (en) 1993-01-29 1999-08-10 Aradigm Corporation Device and method of creating aerosolized mist of respiratory drug
WO1994018219A1 (en) 1993-02-02 1994-08-18 The Scripps Research Institute Methods for producing antibody libraries using universal or randomized immunoglobulin light chains
US5565352A (en) 1993-11-24 1996-10-15 Arch Development Corporation Deubiquitinating enzyme: compositions and methods
WO1995015982A2 (en) 1993-12-08 1995-06-15 Genzyme Corporation Process for generating specific antibodies
US5516637A (en) 1994-06-10 1996-05-14 Dade International Inc. Method involving display of protein binding pairs on the surface of bacterial pili and bacteriophage
US6130364A (en) 1995-03-29 2000-10-10 Abgenix, Inc. Production of antibodies using Cre-mediated site-specific recombination
US6091001A (en) 1995-03-29 2000-07-18 Abgenix, Inc. Production of antibodies using Cre-mediated site-specific recombination
US6019968A (en) 1995-04-14 2000-02-01 Inhale Therapeutic Systems, Inc. Dispersible antibody compositions and methods for their preparation and use
JPH11503914A (en) 1995-04-21 1999-04-06 セル ジェネシス,インコーポレイテッド Generation of large genomic DNA deletions
US5916597A (en) 1995-08-31 1999-06-29 Alkermes Controlled Therapeutics, Inc. Composition and method using solid-phase particles for sustained in vivo release of a biologically active agent
JP2978435B2 (en) 1996-01-24 1999-11-15 チッソ株式会社 Method for producing acryloxypropyl silane
WO1997032572A2 (en) 1996-03-04 1997-09-12 The Penn State Research Foundation Materials and methods for enhancing cellular internalization
US5714352A (en) 1996-03-20 1998-02-03 Xenotech Incorporated Directed switch-mediated DNA recombination
US5855913A (en) 1997-01-16 1999-01-05 Massachusetts Instite Of Technology Particles incorporating surfactants for pulmonary drug delivery
US5985309A (en) 1996-05-24 1999-11-16 Massachusetts Institute Of Technology Preparation of particles for inhalation
US5874064A (en) 1996-05-24 1999-02-23 Massachusetts Institute Of Technology Aerodynamically light particles for pulmonary drug delivery
US6699658B1 (en) 1996-05-31 2004-03-02 Board Of Trustees Of The University Of Illinois Yeast cell surface display of proteins and uses thereof
US5916771A (en) 1996-10-11 1999-06-29 Abgenix, Inc. Production of a multimeric protein by cell fusion method
US5989463A (en) 1997-09-24 1999-11-23 Alkermes Controlled Therapeutics, Inc. Methods for fabricating polymer-based controlled release devices
KR100764256B1 (en) * 1998-01-23 2007-10-05 에프. 호프만-라 로슈 아게 Antibodies against human il-12
US5989263A (en) * 1998-03-11 1999-11-23 Arteria Medical Science L.L.C. Hydraulically actuated dilatation mechanism for vessel dilatation and vascular prosthesis delivery and methods of use
US6660843B1 (en) * 1998-10-23 2003-12-09 Amgen Inc. Modified peptides as therapeutic agents
US6914128B1 (en) 1999-03-25 2005-07-05 Abbott Gmbh & Co. Kg Human antibodies that bind human IL-12 and methods for producing
DK2168984T3 (en) * 1999-03-25 2012-12-10 Abbott Gmbh & Co Kg Human antibodies that bind human IL-12 and methods for its production
AU3672800A (en) 1999-04-09 2000-11-14 Kyowa Hakko Kogyo Co. Ltd. Method for controlling the activity of immunologically functional molecule
DK1292680T3 (en) * 2000-06-22 2010-03-08 Genentech Inc Agonist-anti-TrkC monoclonal antibodies
US7449308B2 (en) 2000-06-28 2008-11-11 Glycofi, Inc. Combinatorial DNA library for producing modified N-glycans in lower eukaryotes
AU2001276842B2 (en) 2000-06-28 2007-04-26 Glycofi, Inc. Methods for producing modified glycoproteins
US6902734B2 (en) * 2000-08-07 2005-06-07 Centocor, Inc. Anti-IL-12 antibodies and compositions thereof
EP2325205A3 (en) 2000-12-28 2011-10-12 Altus Pharmaceuticals Inc. Crystals of whole antibodies and fragments thereof and methods for making and using them
US7667004B2 (en) 2001-04-17 2010-02-23 Abmaxis, Inc. Humanized antibodies against vascular endothelial growth factor
ES2312569T3 (en) 2001-04-30 2009-03-01 Eli Lilly And Company HUMANIZED ANTIBODIES.
AU2002314825A1 (en) * 2001-05-30 2002-12-09 Centocor, Inc. Anti-p40 immunoglobulin derived proteins, compositions, methods and uses
EP1432444A4 (en) 2001-08-17 2005-11-02 Lilly Co Eli Anti-a-beta antibodies
ATE346866T1 (en) 2001-09-14 2006-12-15 Affimed Therapeutics Ag MULTIMERIC, SINGLE CHAIN, TANDEM FV ANTIBODIES
JP4578098B2 (en) 2001-10-01 2010-11-10 ダイアックス、コープ Multi-chain eukaryotic display vectors and their use
CA2462203A1 (en) * 2001-10-12 2003-11-20 University Of Iowa Research Foundation Methods and products for enhancing immune responses using imidazoquinoline compounds
WO2003035835A2 (en) 2001-10-25 2003-05-01 Genentech, Inc. Glycoprotein compositions
CN101824088B (en) * 2002-10-30 2012-05-30 健泰科生物技术公司 Inhibition of il-17 production
US20060104968A1 (en) 2003-03-05 2006-05-18 Halozyme, Inc. Soluble glycosaminoglycanases and methods of preparing and using soluble glycosaminogly ycanases
CN102943067B (en) 2003-03-05 2016-06-22 海洋酶公司 Soluble hyaluronidase glycoprotein (sHASEGP), prepare their method, their purposes and the pharmaceutical composition comprising them
WO2004101511A2 (en) 2003-05-09 2004-11-25 Protein Design Labs, Inc Anti-ip-10 antibodies and methods of using thereof for the treatment of inflamatory bowel diseases
AU2004239288B2 (en) * 2003-05-09 2010-01-28 Centocor, Inc. IL-23p40 specific immunoglobulin derived proteins, compositions, methods and uses
EP1629012B1 (en) 2003-05-31 2018-11-28 Amgen Research (Munich) GmbH Pharmaceutical compositions comprising bispecific anti-cd3, anti-cd19 antibody constructs for the treatment of b-cell related disorders
CA2537055A1 (en) 2003-08-22 2005-04-21 Medimmune, Inc. Humanization of antibodies
JP2007535317A (en) 2004-04-15 2007-12-06 グライコフィ, インコーポレイテッド Production of galactosylated glycoproteins in lower eukaryotes
BRPI0515858A (en) * 2004-12-21 2008-08-12 Centocor Inc anti-il-12 antibodies, epitopes, compositions, methods and uses
CA2612239A1 (en) * 2005-06-30 2007-01-11 Abbott Laboratories Il-12/p40 binding proteins

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of EP1907421A4 *

Cited By (55)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8524230B2 (en) 2005-09-01 2013-09-03 Merck Sharp & Dohme Corp. Use of IL-23 and IL-17 antagonists to treat autoimmune ocular inflammatory disease
US8946292B2 (en) 2006-03-28 2015-02-03 Javelin Pharmaceuticals, Inc. Formulations of low dose diclofenac and beta-cyclodextrin
US8580954B2 (en) 2006-03-28 2013-11-12 Hospira, Inc. Formulations of low dose diclofenac and beta-cyclodextrin
RU2476442C2 (en) * 2007-03-29 2013-02-27 Эббот Лэборетриз Crystalline human il-12 antibodies
US8940873B2 (en) 2007-03-29 2015-01-27 Abbvie Inc. Crystalline anti-human IL-12 antibodies
US8168760B2 (en) 2007-03-29 2012-05-01 Abbott Laboratories Crystalline anti-human IL-12 antibodies
US8404819B2 (en) 2007-03-29 2013-03-26 Abbvie Inc. Crystalline anti-human IL-12 antibodies
EP2148888A1 (en) * 2007-05-30 2010-02-03 Abbott Laboratories Humanized antibodies which bind to a (1-42) globulomer and uses thereof
US8975382B2 (en) 2007-11-27 2015-03-10 Ablynx N.V. Amino acid sequences directed against HER2 and polypeptides comprising the same for the treatment of cancers and/or tumors
US9969805B2 (en) 2007-11-27 2018-05-15 Ablynx N.V. Amino acid sequences directed against HER2 and polypeptides comprising the same for the treatment of cancers and/or tumors
EP2650311A3 (en) * 2007-11-27 2014-06-04 Ablynx N.V. Amino acid sequences directed against heterodimeric cytokines and/or their receptors and polypeptides comprising the same
EP2220120A2 (en) * 2007-11-27 2010-08-25 Ablynx N.V. Amino acid sequences directed against heterodimeric cytokines and/or their receptors and polypeptides comprising the same
US9610301B2 (en) 2008-01-15 2017-04-04 Abbvie Deutschland Gmbh & Co Kg Powdered protein compositions and methods of making same
KR101781965B1 (en) 2008-08-14 2017-09-27 테바 파마슈티컬즈 오스트레일리아 피티와이 엘티디 Anti-il-12/il-23 antibodies
KR20160116056A (en) 2008-08-14 2016-10-06 테바 파마슈티컬즈 오스트레일리아 피티와이 엘티디 Anti-il-12/il-23 antibodies
CN102177178A (en) * 2008-08-14 2011-09-07 赛福伦澳大利亚私人有限公司 Anti-IL-12/IL-23 antibodies
US11236393B2 (en) 2008-11-26 2022-02-01 Cedars-Sinai Medical Center Methods of determining responsiveness to anti-TNFα therapy in inflammatory bowel disease
US8835610B2 (en) 2009-03-05 2014-09-16 Abbvie Inc. IL-17 binding proteins
US8779101B2 (en) 2009-03-05 2014-07-15 Abbvie, Inc. IL-17 binding proteins
KR101579771B1 (en) 2009-03-05 2015-12-28 애브비 인코포레이티드 IL-17 binding proteins
KR20110128909A (en) * 2009-03-05 2011-11-30 아보트 러보러터리즈 Il-17 binding proteins
US9663587B2 (en) 2009-03-05 2017-05-30 Abbvie Inc. IL-17 binding proteins
US9481736B2 (en) 2009-03-05 2016-11-01 Abbvie, Inc. IL-17 binding proteins
US9481735B2 (en) 2009-03-05 2016-11-01 Abbvie Inc. IL-17 binding proteins
WO2010102251A3 (en) * 2009-03-05 2010-11-25 Abbott Laboratories Il-17 binding proteins
CN101935687B (en) * 2009-06-29 2012-11-07 中国科学院上海生命科学研究院 Application of IL-12p40 siRNA to promoting liver regeneration
US10752696B2 (en) 2009-09-11 2020-08-25 Genentech, Inc. Highly concentrated pharmaceutical formulations
EA031972B1 (en) * 2009-09-11 2019-03-29 Ф. Хоффманн-Ля Рош Аг Highly concentrated pharmaceutical formulations comprising an anti-cd20 antibody
EP3064196A1 (en) * 2009-09-11 2016-09-07 F. Hoffmann-La Roche AG Highly concentrated pharmaceutical formulations
EP3061445A1 (en) * 2009-09-11 2016-08-31 F. Hoffmann-La Roche AG Highly concentrated pharmaceutical formulations
WO2011029892A3 (en) * 2009-09-11 2011-09-15 F. Hoffmann-La Roche Ag Highly concentrated pharmaceutical formulations comprising anti - cd20 antibody
EP4218715A1 (en) * 2009-09-11 2023-08-02 F. Hoffmann-La Roche AG Highly concentrated pharmaceutical formulations
US10377831B2 (en) 2009-09-11 2019-08-13 Genentech, Inc. Highly concentrated pharmaceutical formulations
AU2010294186B2 (en) * 2009-09-11 2013-09-19 F. Hoffmann-La Roche Ag Highly concentrated pharmaceutical formulations comprising anti - CD20 antibody
US10280227B2 (en) 2009-09-11 2019-05-07 Genentech, Inc. Highly concentrated pharmaceutical formulations
EP4218716A1 (en) * 2009-09-11 2023-08-02 F. Hoffmann-La Roche AG Highly concentrated pharmaceutical formulations
US9447183B2 (en) 2010-05-14 2016-09-20 Abbvie Inc. IL-1 binding proteins
US9441038B2 (en) 2010-05-14 2016-09-13 Abbvie Inc. IL-1 binding proteins
US9303085B2 (en) 2010-05-14 2016-04-05 Abbvie Inc. IL-1 binding proteins
US9409986B2 (en) 2010-05-14 2016-08-09 Abbvie Inc. IL-1 binding proteins
US9447184B2 (en) 2010-05-14 2016-09-20 Abbvie Inc. IL-1 binding proteins
US9695232B2 (en) 2012-10-03 2017-07-04 Philogen S.P.A. Anti-ED-A immunoconjugates for inflammatory bowel disease
US10239939B2 (en) 2012-10-03 2019-03-26 Philogen S.P.A. Anti-ED-A immunoconjugates for inflammatory bowel disease
US11459404B2 (en) 2013-02-26 2022-10-04 Roche Glycart Ag Bispecific T cell activating antigen binding molecules
US10633449B2 (en) 2013-03-27 2020-04-28 Cedars-Sinai Medical Center Treatment and reversal of fibrosis and inflammation by inhibition of the TL1A-DR3 signaling pathway
US10316083B2 (en) 2013-07-19 2019-06-11 Cedars-Sinai Medical Center Signature of TL1A (TNFSF15) signaling pathway
US11312768B2 (en) 2013-07-19 2022-04-26 Cedars-Sinai Medical Center Signature of TL1A (TNFSF15) signaling pathway
US11001623B2 (en) 2014-01-30 2021-05-11 Coherus Biosciences, Inc. Method of manufacturing a protein by perfusion in media with a low amino acid concentration
EP3827845B1 (en) 2015-11-03 2022-03-30 Janssen Biotech, Inc. Subcutaneous formulations of anti-cd38 antibodies and their uses
US11186872B2 (en) 2016-03-17 2021-11-30 Cedars-Sinai Medical Center Methods of diagnosing inflammatory bowel disease through RNASET2
US10322174B2 (en) 2016-10-26 2019-06-18 Cedars-Sinai Medical Center Neutralizing anti-TL1A monoclonal antibodies
WO2018081074A1 (en) * 2016-10-26 2018-05-03 Cedars-Sinai Medical Center Neutralizing anti-tl1a monoclonal antibodies
US11440954B2 (en) 2018-04-25 2022-09-13 Prometheus Biosciences, Inc. Optimized anti-TL1A antibodies
US10689439B2 (en) 2018-04-25 2020-06-23 Prometheus Biosciences, Inc. Optimized anti-TL1A antibodies
US11292848B2 (en) 2019-10-24 2022-04-05 Prometheus Biosciences, Inc. Humanized antibodies to TNF-like ligand 1A (TL1A) and uses thereof

Also Published As

Publication number Publication date
CR20130153A (en) 2013-05-23
CR20130155A (en) 2013-05-23
CN103172737A (en) 2013-06-26
KR20080028895A (en) 2008-04-02
MX2007016401A (en) 2008-02-20
TW201444869A (en) 2014-12-01
IL187691A (en) 2013-10-31
CR20130156A (en) 2013-05-23
CN103145840A (en) 2013-06-12
CN103145838A (en) 2013-06-12
CR20130151A (en) 2013-05-23
TW200745160A (en) 2007-12-16
CA2848662A1 (en) 2007-01-11
CN103145837A (en) 2013-06-12
IL187691A0 (en) 2008-08-07
CR20130154A (en) 2013-05-23
CN103145839A (en) 2013-06-12
CN103145841A (en) 2013-06-12
IL228793A0 (en) 2013-12-31
CN101379085A (en) 2009-03-04
US20100196315A1 (en) 2010-08-05
UA96922C2 (en) 2011-12-26
CN103146708A (en) 2013-06-12
WO2007005608A3 (en) 2008-10-09
CN103172739A (en) 2013-06-26
CA2612239A1 (en) 2007-01-11
US20100047245A1 (en) 2010-02-25
EP1907421A4 (en) 2012-03-28
IL216803A0 (en) 2012-01-31
JP2009500018A (en) 2009-01-08
CR20130152A (en) 2013-05-23
RU2461571C2 (en) 2012-09-20
CN101379085B (en) 2013-03-27
NO20080557L (en) 2008-01-30
CR9599A (en) 2009-06-05
JP5396080B2 (en) 2014-01-22
ZA201201216B (en) 2012-11-28
AU2006265932B2 (en) 2012-10-25
US20140178332A1 (en) 2014-06-26
EP1907421A2 (en) 2008-04-09
RU2008103312A (en) 2009-08-10
US7700739B2 (en) 2010-04-20
CR20130157A (en) 2013-05-23
CR20130149A (en) 2013-05-23
CN103145842A (en) 2013-06-12
BRPI0611714A2 (en) 2009-01-13
KR20130080058A (en) 2013-07-11
CN103172738A (en) 2013-06-26
JP2013177405A (en) 2013-09-09
NZ596992A (en) 2013-07-26
AU2006265932A1 (en) 2007-01-11
CR20130150A (en) 2013-05-23
US8629257B2 (en) 2014-01-14

Similar Documents

Publication Publication Date Title
US7700739B2 (en) IL-12/p40 binding proteins
US8383778B2 (en) IL-1 binding proteins
US8398966B2 (en) IL-1 binding proteins
US20120275996A1 (en) IL-1 Binding Proteins
US20110165063A1 (en) Il-1 binding proteins
AU2012238334A1 (en) IL-12/p40 binding proteins

Legal Events

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

Ref document number: 200680024097.3

Country of ref document: CN

121 Ep: the epo has been informed by wipo that ep was designated in this application
DPE2 Request for preliminary examination filed before expiration of 19th month from priority date (pct application filed from 20040101)
WWE Wipo information: entry into national phase

Ref document number: 563241

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 2006265932

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 187691

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 12007502756

Country of ref document: PH

ENP Entry into the national phase

Ref document number: 2612239

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 9729/DELNP/2007

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: MX/a/2007/016401

Country of ref document: MX

Ref document number: CR2007-009599

Country of ref document: CR

WWE Wipo information: entry into national phase

Ref document number: 07135361

Country of ref document: CO

ENP Entry into the national phase

Ref document number: 2008519612

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 1020077030917

Country of ref document: KR

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2006785967

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2008103312

Country of ref document: RU

Ref document number: A20080099

Country of ref document: BY

ENP Entry into the national phase

Ref document number: PI0611714

Country of ref document: BR

DPE2 Request for preliminary examination filed before expiration of 19th month from priority date (pct application filed from 20040101)
WWE Wipo information: entry into national phase

Ref document number: CR2013-000157

Country of ref document: CR

Ref document number: CR2013-000151

Country of ref document: CR

Ref document number: CR2013-000150

Country of ref document: CR

Ref document number: CR2013-000155

Country of ref document: CR

Ref document number: CR2013-000149

Country of ref document: CR

Ref document number: CR2013-000148

Country of ref document: CR

Ref document number: CR2013-000153

Country of ref document: CR

Ref document number: CR2013-000156

Country of ref document: CR

Ref document number: CR2013-000152

Country of ref document: CR

Ref document number: CR2013-000154

Country of ref document: CR

WWE Wipo information: entry into national phase

Ref document number: 1020137014441

Country of ref document: KR