WO2009021754A2 - Monospecific and multispecific antibodies and method of use - Google Patents

Monospecific and multispecific antibodies and method of use Download PDF

Info

Publication number
WO2009021754A2
WO2009021754A2 PCT/EP2008/006750 EP2008006750W WO2009021754A2 WO 2009021754 A2 WO2009021754 A2 WO 2009021754A2 EP 2008006750 W EP2008006750 W EP 2008006750W WO 2009021754 A2 WO2009021754 A2 WO 2009021754A2
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
cooh
linker
antibodies
protease
Prior art date
Application number
PCT/EP2008/006750
Other languages
French (fr)
Other versions
WO2009021754A3 (en
Inventor
Bing Liu
David Light
Zhuozhi Wang
Douglas Schneider
Original Assignee
Bayer Schering Pharma Aktiengesellschaft
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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=40110987&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO2009021754(A2) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Bayer Schering Pharma Aktiengesellschaft filed Critical Bayer Schering Pharma Aktiengesellschaft
Priority to JP2010520502A priority Critical patent/JP5485152B2/en
Priority to CA2696263A priority patent/CA2696263C/en
Priority to EP08785592A priority patent/EP2178914A2/en
Priority to US12/671,942 priority patent/US9624309B2/en
Publication of WO2009021754A2 publication Critical patent/WO2009021754A2/en
Publication of WO2009021754A3 publication Critical patent/WO2009021754A3/en
Priority to US15/466,587 priority patent/US20170210825A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/46Hybrid immunoglobulins
    • C07K16/468Immunoglobulins having two or more different antigen binding sites, e.g. multifunctional antibodies
    • 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
    • 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
    • A61K39/39558Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against tumor tissues, cells, antigens
    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6801Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
    • A61K47/6803Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6801Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
    • A61K47/6803Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates
    • A61K47/6807Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates the drug or compound being a sugar, nucleoside, nucleotide, nucleic acid, e.g. RNA antisense
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6801Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
    • A61K47/6803Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates
    • A61K47/6807Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates the drug or compound being a sugar, nucleoside, nucleotide, nucleic acid, e.g. RNA antisense
    • A61K47/6809Antibiotics, e.g. antitumor antibiotics anthracyclins, adriamycin, doxorubicin or daunomycin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6801Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
    • A61K47/6803Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates
    • A61K47/6811Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates the drug being a protein or peptide, e.g. transferrin or bleomycin
    • A61K47/6817Toxins
    • A61K47/6819Plant toxins
    • A61K47/6821Plant heterodimeric toxins, e.g. abrin or modeccin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6801Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
    • A61K47/6803Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates
    • A61K47/6811Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates the drug being a protein or peptide, e.g. transferrin or bleomycin
    • A61K47/6817Toxins
    • A61K47/6819Plant toxins
    • A61K47/6825Ribosomal inhibitory proteins, i.e. RIP-I or RIP-II, e.g. Pap, gelonin or dianthin
    • A61K47/6827Ricin A
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6801Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
    • A61K47/6803Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates
    • A61K47/6811Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates the drug being a protein or peptide, e.g. transferrin or bleomycin
    • A61K47/6817Toxins
    • A61K47/6829Bacterial toxins, e.g. diphteria toxins or Pseudomonas exotoxin A
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6851Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6851Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell
    • A61K47/6869Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell the tumour determinant being from a cell of the reproductive system: ovaria, uterus, testes, prostate
    • 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
    • A61P1/12Antidiarrhoeals
    • 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
    • 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
    • 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
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • 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
    • A61P31/04Antibacterial agents
    • A61P31/06Antibacterial agents for tuberculosis
    • 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
    • A61P33/02Antiprotozoals, e.g. for leishmaniasis, trichomoniasis, toxoplasmosis
    • A61P33/06Antimalarials
    • 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
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/14Drugs for disorders of the endocrine system of the thyroid hormones, e.g. T3, T4
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • C07K16/3069Reproductive system, e.g. ovaria, uterus, testes, prostate
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • C07K16/3076Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells against structure-related tumour-associated moieties
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/36Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against blood coagulation factors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/31Immunoglobulins specific features characterized by aspects of specificity or valency multispecific
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/64Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising a combination of variable region and constant region components
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • This invention relates to monospecific and multispecific antibodies that may be utilized for the diagnosis and treatment of various diseases.
  • these antibodies may be modified by protease cleavage.
  • Protease control or regulation may be provided by a protease site located in, for example, a linker.
  • protease-regulated antibodies may also be utilized for the diagnosis and treatment of various diseases.
  • An antibody may be directed against one or more different antigens or one or more different epitopes on the same antigen.
  • a bispecific antibody is directed against two different antigens or two different epitopes on the same antigen.
  • these antibodies have great potential for antibody- based diagnosis and for the treatment of various diseases and disorders such as cancer, infectious diseases, autoimmune diseases, and blood diseases.
  • bispecific antibodies can selectively stimulate and expand T lymphocytes (Wong, et al., J. Immunol. 139:1369-1374, 1987; Wong, et al., Clin. Immunol. Immunopathol.
  • a bispecific antibody may be used as a substitute for Factor VIII to enhance enzymatic reaction (US Patent Application No. 2007/0041978) or to direct stem cells to the site of injury in patients with myocardial infarction (Lum, et al., Blood Cells MoI. Dis. 32:82-87, 2004).
  • Bispecific antibodies targeting tumor-associated antigens and toxic agents may be used in cancer therapy.
  • one arm of the bispecific antibody may be directed to a tumor-associated antigen such as Her2, EGF receptor, CD20, CD22, CD30, CD33, CD52, and CA-125, and the other arm of the bispecific antibody may target a toxin, drug, or cytokine. That is, bispecific antibodies may selectively direct toxic agents to tumor cells enhancing the efficacy of therapeutic antibodies and decreasing systemic toxicity. Examples of a tumor-associated antigen such as Her2, EGF receptor, CD20, CD22, CD30, CD33, CD52, and CA-125, and the other arm of the bispecific antibody may target a toxin, drug, or cytokine. That is, bispecific antibodies may selectively direct toxic agents to tumor cells enhancing the efficacy of therapeutic antibodies and decreasing systemic toxicity. Examples of
  • SUBSTITUT SUBSTITUT toxin/drug include calicheamicin, doxorubicin, epirubicin, methotrexate, ricin A, saporin, gelonin, and vinca alkaloids, and cytokine examples include tumor necrosis factor alpha (TNF-alpha) and IL-2.
  • TNF-alpha tumor necrosis factor alpha
  • proteases of defined sites in biologically important effector proteins is a well known method for the natural control of cellular and extracellular physiological processes. Examples include protease activation and inhibition of the coagulation cascade (Butenas, et al., Biochemistry 67:3-12, 2002; Esmon, Chest, 124:26S-32S, 2003), protease activation of protease-activatable receptors (Coughlin, Arterioscler. Thromb. Vase. Biol. 18:514- 518, 1998), protease release of membrane associated cytokines (Amour, et al., FEBS Lett.
  • protease processing of prohormones in secretory vesicles (Moore, et al., Arch. Physiol. Biochem. 110:16-25, 2002), and protease processing of proproteins during secretion (Scamuffa, et al., FASEB J. 20:1954-1963, 2006).
  • Proteases are often expressed or located in a tissue-specific or tumor-specific manner and examples include the membrane serine protease conn in heart tissue (Yan, et al., Proc. Natl. Acad. Sci.
  • proteases expressed in the pancreas and released to the duodenum
  • specific cleavage of amino acid sequences by human proteases include thrombin (Chang, Eur. J. Biochem. 151 :217-224, 1985), factor Xa (Nagai, et al., Methods Enzymol. 153:461-481, 1987), furin (Brennan, et al., FEBS Lett. 347:80-84, 1994), subtilisin-like prohormone convertases (Lipkind, et al., J. Biol. Chem.
  • Protease cleavage is widely used in in vitro studies to specifically remove protein or peptide tags from recombinant proteins or to process hybrid recombinant proteins.
  • human rhinovirus 3C protease, thrombin, or factor Xa have been used to remove glutathione S- transferase (GST) tags (Dian, et al., Life Sciences News - Amersham Biosciences 10: 1-5, 2002) and factor Xa has been use to process hybrid proteins (Nagai, et al., 1987).
  • GST glutathione S- transferase
  • Proteases are often targets for drugs as a means to regulate biological processes; and examples include factor Xa (Phillips, et al., J. Med. Chem.
  • the present invention relates to a novel antibody format, for example, monospecifc and multispecific antibodies.
  • the antibodies of the present invention may be constructed by tandem linking of two different heavy chain (H) variable region domains (V H ) and two different light chain (L) variable region domains (V t ).
  • the heavy chain and light chain may form a Fab-like or IgG- like molecule through the disulfide bond between constant (C) regions.
  • Multispecific antibodies may be generated by linking more than two antibody variable domains.
  • the antibodies of the present invention may be modified by protease cleavage.
  • protease-regulated antibodies may be, for example, monospecific antibodies, bispecific antibodies, or antibodies with sequential binding-activity upon protease digestion in either, for example, Fab- like or IgG-like format.
  • Protease control or regulation may be provided by a protease site located in, for example, a linker.
  • protease-regulated antibodies may be utilized for the diagnosis and treatment of various diseases, and provide an additional level of control for biological drugs for therapeutic or diagnostic applications.
  • Figure 1 Schematic drawing of a monospecific protease-regulated antibody with a linker which contains a protease site between variable domain and Fc domain ("Type 1").
  • Figure 2 Schematic drawing of a bispecific protease-regulated antibody with a linker which contains a protease cleavage sequence that allows removal of one antigen-binding site ("Type 2").
  • Figure 3 Schematic drawing of another bispecific protease-regulated antibody with a linker which contains a protease cleavage sequence that allows removal of one antigen-binding site ("Type 2").
  • FIG. 1 Schematic drawing of the application of a bispecific protease-regulated antibody that simultaneously binds two different antigens.
  • FIG. 1 Schematic drawing of the application of a protease-regulated antibody that cannot bind to two different antigens simultaneously.
  • Figure 7 Schematic drawing of a monospecific protease-regulated antibody 'prodrug' that can only bind antigen following protease activation to remove inactive blocking antibody domains ("Type 4").
  • FIG. 8 Map of an expression vector for an IgG-like bispecific antibody.
  • SignalP signal peptide
  • VLani3E10 variable region of 3E10 lambda chain
  • Vkl9G9 variable region of 19G9 kappa chain
  • C kappa constant region of kappa chain
  • DHFR dihydrofolate reductase
  • V H variable region of heavy chain
  • Neo neomycin resistant gene
  • 3E10VH variable region of 3E10 heavy chain
  • 19G9VH variable region of 19G9 heavy chain
  • CH constant region of heavy chain
  • Amp ampicillin resistant gene.
  • FIG. 9 Map of expression vector of Fab-like bispecific antibody.
  • LacZ lac Z promoter; ompA and pho A, signal peptide;
  • VL-linkl-VK variable region of light chain of bispecific antibody against tissue factor and RGl;
  • CL-kappa constant region of kappa chain;
  • VHs with linker variable region of heavy cham of bispecific antibody against tissue factor and RGl;
  • CHl the first constant region of IgG heavy chain; cat, chloramphenicol resistant gene.
  • FIG. 10 TF-binding ELISA.
  • Four bispecific antibodies and parental antibodies were analyzed for binding to TF.
  • Antibodies were detected with HRP-conjugated anti-human IgG was used for detection.
  • Curve fitting of the data was performed using a 4-parameter equation with the Solver function in Microsoft Excel.
  • FIG. 11 RGl -binding ELISA.
  • Figure 12 Measurement of the antigen-binding activity of a bispecific protease-regulated antibody using a sandwich antigen-binding ELISA.
  • the linker of this antibody contained cleavage sites for enterokinase.
  • Figure 13 Measurement of the antigen-binding activity of protease-regulated antibodies 3E10-Typel-Fab and 19G9-Typel-Fab. The controls are designated 3El O-Reg-Fab, 19G9-Reg- Fab, and HuFab.
  • Figure 14 Measurement of the antigen-binding activity of Fab-like protease-regulated antibodies HlLl, H1L4, H1L7, H4L7, and H5L5 (Type 2) in the absence and presence of enterokinase. Parental antibodies 3E10 and 19G9, and polyclonal human Fab were used as control.
  • Figure 15 Measurement of the antigen-binding activity of Fab-like protease-regulated antibodies H2L1, H2L2, and H2L8 (Type 3) and H3L1, H3L4, and H5L4 (Type 4) in the absence and presence of enterokinase.
  • Parental antibodies 3E10 and 19G9, and polyclonal human Fab were used as control.
  • FIG. 16 Western blots of protease-regulated antibody 3E 10-Type 1 -Fab detected with anti-Myc antibody (A) or anti-kappa chain antibody (B). Lane 1 and 2: 3E10-Typel-Fab without or with enterokinase digestion, respectively. Lane 3 and 4: 3E10-Reg-Fab without or with enterokinase digestion, respectively.
  • FIG. Western blots of Fab-like protease-regulated antibodies HlLl, H 1L7, and H5L5 (Type 2) in the absence and presence of enterokinase. Antibodies were detected with anti- IgG(H+L) antibody. Lane 1 and 2: HlLl without or with enterokinase digestion, respectively. Lane 3 and 4: H1L7 without or with enterokinase digestion, respectively. Lane 5 and 6: H5L5 without or with enterokinase digestion, respectively. Lane 7: 3E10-Reg-Fab.
  • FIG. 1 Western blots of Fab-like protease-regulated antibodies H2L2 and H2L8 (Type 3) and H3L4 (Type 4) in the absence and presence of enterokinase. Antibodies were detected with anti-Myc antibody. Lane 1 and 2: H2L2 without or with enterokinase digestion, respectively. Lane 3 and 4: H2L8 without or with enterokinase digestion, respectively. Lane 5 and 6: H3L4 without or with enterokinase digestion, respectively.
  • Antibody as used herein includes intact immunoglobulin molecules (e.g., IgGl, IgG2a, IgG2b, IgG3, IgM, IgD, IgE, IgA), as well as fragments thereof, such as Fab, F(ab') 2 , scFv, Fv, and diabody which are capable of specific binding to an epitope of a protein.
  • immunoglobulin molecules e.g., IgGl, IgG2a, IgG2b, IgG3, IgM, IgD, IgE, IgA
  • fragments thereof such as Fab, F(ab') 2 , scFv, Fv, and diabody which are capable of specific binding to an epitope of a protein.
  • antibody also extends to other protein scaffolds that are able to orient antibody complementarity- determining region (CDR) inserts into the same active binding conformation as that found in natural antibodies such that the binding to the target antigen observed with these chimeric proteins is maintained relative to the binding activity of the natural antibody from which the CDRs were derived.
  • CDR complementarity- determining region
  • Antibody fragments comprise a portion of a full length antibody, generally the antigen binding or variable domain thereof.
  • Examples of antibody fragments include Fab, Fab', F(ab') 2> and Fv fragments; diabodies; linear antibodies; single-chain antibody molecules; monospecific antibodies; bispecific antibodies; and multispecif ⁇ c antibodies formed from antibody fragments.
  • autoimmune diseases includes, but is not limited to, multiple sclerosis, rheumatoid arthritis, lupus, type I diabetes mellitus, Crohn's disease, autoimmune hemolytic anemia, autoimmune hepatitis, glomerulonephritis, inflammatory bowel disease, myocarditis, psoriasis, thyroiditis, ulcerative colitis, and Graves'disease.
  • biological sample or "patient sample” as used herein, refers to a sample obtained from an organism or from components (e.g., cells) of an organism.
  • the sample may be of any biological tissue or fluid.
  • the sample may be a "clinical sample” which is a sample derived from a patient.
  • Such samples include, but are not limited to, sputum, blood, serum, plasma, blood cells (e.g., white cells), tissue samples, biopsy samples, urine, peritoneal fluid, and pleural fluid, saliva, semen, breast exudate, cerebrospinal fluid, tears, mucous, lymph, cytosols, ascites, amniotic fluid, bladder washes, and bronchioalveolar lavages or cells therefrom, among other body fluid samples.
  • the patient samples may be fresh or frozen, and may be treated with heparin, citrate, or EDTA.
  • Biological samples may also include sections of tissues such as frozen sections taken for histological purposes.
  • cancer includes, but is not limited to, solid tumors, such as cancers of the breast, respiratory tract, brain, reproductive organs, digestive tract, urinary tract, eye, liver, skin, head and neck, thyroid, parathyroid, and their distant metastases.
  • solid tumors such as cancers of the breast, respiratory tract, brain, reproductive organs, digestive tract, urinary tract, eye, liver, skin, head and neck, thyroid, parathyroid, and their distant metastases.
  • lymphomas, sarcomas, and leukemias include lymphomas, sarcomas, and leukemias.
  • conjugate refers to an antibody chemically linked to a chemical moiety, such as a therapeutic or cytotoxic agent.
  • infectious diseases includes, but is not limited to, HIV/ AIDS, lower respiratory infections, diarrheal diseases, tuberculosis, malaria, measles, pertussis, tetanus, meningitis, syphilis, hepatitis B and tropical diseases.
  • linker refers to a peptide (or polypeptide) comprising two or more amino acid residues joined by peptide bonds and used to link one or more antibody domains.
  • the linker may contain one or more protease cleavage sites.
  • protease cleavage sites refers to any enzyme, including the endopeptidases and exopeptidases, which catalyze the hydrolytic breakdown of proteins into peptides or amino acids.
  • the present invention is directed to the design and production of monospecific, bispecific antibodies, and multispecif ⁇ c antibodies.
  • the bispecific antibodies and multispecific antibodies may comprise tandem linked V H a-V H b-V H C...C H in one polypeptide and V L a-V L b- V L c...C L in another polypeptide.
  • the V H and V L domains may be exchanged from one polypeptide to another to create polypeptides such as V H a-V L b-V H C...C H and V L a-V H b-V L c...C L .
  • the two polypeptides may form Dimmers in the Fab format or the half IgG-like format, or two of each polypeptide may form a four polypeptide-containing homodimer of the IgG-like format.
  • These bispecific or multispecific antibodies or antibody fragments thereof may simultaneously bind different antigens or different epitopes of the same antigen.
  • a recombinant IgG-like bispecific antibody may be constructed by the tandem linking of two different V H domains of a heavy chain and two different V L domains of a light chain.
  • the present invention also relates to protease-regulated antibodies.
  • Protease-regulated antibodies may be, for example, monospecific antibodies, bispecific antibodies, multispecific antibodies, or antibodies with sequential binding-activity upon protease digestion in either, for example, Fab-like or IgG-like format.
  • Protease control or regulation may be provided by a selective protease site located in, for example, a linker.
  • These protease-regulated antibodies may be utilized for the diagnosis and treatment of various diseases including but not limited to cancer, infectious disease, and autoimmune diseases, and provide an additional level of control for biological drugs for therapeutic or diagnostic applications.
  • Protease-regulated antibodies may comprise a heavy chain (H) variable domain (V H )- linker-heavy chain constant domain (CH) in one polypeptide and a light chain (L) variable domain (V L )-linker-light chain constant domain (CL) in another polypeptide.
  • Bispecific protease- regulated antibodies may comprise, for example, Vm-linker-V ⁇ -CH in one polypeptide and V L r linker- V L2 -CL in another polypeptide, both regulated by proteolytic cleavage of the linker.
  • V H and V L domains in the bispecific protease-regulated antibodies may be exchanged from one polypeptide to another polypeptide to create polypeptides such as, for example, Vm-linker-V ⁇ -CH and V L i-linker-V H2 -CL.
  • the two polypeptides may form Dimmers, for example, in a Fab-like format, a half-IgG-like format, or an IgG-like format (e.g., two of each polypeptide forming a four polypeptide-containing homodimer).
  • the bispecific and sequential protease-regulated antibodies or antibody fragments may (1) simultaneously bind two different antigens or different epitopes of the same antigen, (2) sequentially bind two different antigens or different epitopes on the same antigen in a manner that may be dependent on the length, adjacent sequence, and design of the linker, or (3) a monospecific protease-activated binder which is in latent or prodrug form prior to protease digestion and which is switched on by protease cleavage.
  • protease-regulated antibodies are described herein whereby antibody formats are designed with selectivity due to specific protease-dependent binding or protease- specific functionality.
  • these protease-regulated antibodies may be described by deletions and/or additions of antibody framework, location of the linker, and its properties including length and solvent accessibility.
  • the linker may contain a cleavage site specific for a protease found in a target cell or tissue.
  • a protease-regulated antibody may contain a protease site in a linker located between the variable domain and the constant region domain and this antibody may bind only one antigen as illustrated in Figure 1 ("Type 1").
  • protease-regulated antibody may simultaneously bind two different antigens or two different epitopes as shown in Figures 2 and 3 ("Type 2") in the absence of a protease.
  • the first V H /V L domains of this antibody bind to an antigen without blocking the second V H /V L domains from binding to a second antigen.
  • This antibody can bind to antigens without steric blocking of the CDR regions of the second variable domains.
  • the simultaneous binding of this protease-regulated antibody to both antigens is prevented by proteolytic cleavage. That is, when the linker is cleaved by a protease, the antibody can only bind to the second antigen or separately bind two antigens. Simultaneous antigen binding is important for antibody function, for example, in cross-linking receptors, which may be prevented by proteolytic cleavage. Thus, an additional degree of specificity is added by including a protease site in the linker.
  • This bispecific protease-regulated antibody is more selective than a monospecific antibody because this antibody will specifically target cells or tissues expressing both antigens.
  • the additional degree of specificity is provided by the specific protease site in the linker.
  • Cell A and Cell B express both Antigen 1 and Antigen 2, however only Cell B expresses the selective protease.
  • the bispecif ⁇ c protease-regulated antibody with the uncleavable linker i.e., this linker does not contain the protease cleavage site
  • the bispecif ⁇ c protease-regulated antibody will bind to Cell B with lower avidity because the Antigen 1 binding domain is removed by proteolytic cleavage of the linker by the selective protease expressed by Cell B (alternately, the selective protease may be expressed by adjacent cells localized in the same tissue as Cell B).
  • Figure 5 illustrates a protease-regulated antibody that may sequentially bind to each antigen in a protease-dependent manner. That is, prior to protease cleavage of the linker, the protease-regulated antibody binds to a first antigen and following protease cleavage, the antibody binds to a second antigen ("Type 3").
  • the V H /V L domains of the N-terminal antibody bind to an antigen, but block the CDR regions of the downstream V H /V L domains from binding to a second antigen.
  • Protease cleavage of the linker allows removal of the N-terminal antibody, and removing the N-terminal antibody domains then permits binding to a second antigen. This allows for greater cell and/or tissue selectivity by requiring sequential binding.
  • Cell A and Cell B express both Antigen 1 and Antigen 2, but only Cell B expresses the selective protease.
  • Antigen 2 is a cell surface receptor that internalizes into the cell and allows internalization of antibodies that bind to it.
  • the protease-regulated antibody will bind to Antigen 1 expressed by Cell A and Cell B. However, only Cell B expresses the selective protease (or possibly cells adjacent to Cell B in the same tissue).
  • the protease- regulated antibody will be activated by proteolytic cleavage and internalized via Antigen 2 expressed on Cell B. Thus, this protease-regulated antibody will be specifically internalized by cells expressing Antigen 1 , Antigen 2, and the selective protease.
  • a protease-regulated antibody may not bind to an antigen before protease digestion, but may bind to antigen following protease digestion ("Type 4").
  • An example of this antibody is illustrated in Figure 7.
  • This monospecific protease-regulated antibody also contains a protease cleavage linker that allows removal of the N-terminal non-functional antibody which then leads to binding to an antigen by the functional antibody domains that are thus exposed.
  • Type 4 protease-regulated antibodies may be created by three approaches. In the first approach the protease cleavable linker sequence is modified so that it prevents the N-terminal V H and V L domains of a Type HI antibody (V H 1 and V L 1) from binding to the first antigen.
  • the linkers utilized in Type III antibodies shown in Tables 6 and 7 are combined with homodimeric N-terminal domains derived from the constant regions of antibodies.
  • the complete V H 1 and V L 1 domains of a Type III antibody are both replaced by the same constant domain that is capable of heterodimerization, for example, the CH3 domain of IgG or the CH4 domain of IgE.
  • protease-regulated antibodies may be modified by protease cleavage of the linker as described below.
  • the protease-regulated antibody illustrated in Figure 1 (Type 1) contains a protease site in the linker between the antigen binding domains and the Fc domain. This antibody will specifically target cells or tissues that present the antigen. When the linker is cleaved by the protease, the resultant protease-regulated antibody releases the functional Fc portion. In tissues where the protease is present, this antibody will release the Fc portion which is essential to antigen crosslinking, and induce an immune response such as antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC).
  • ADCC antibody-dependent cellular cytotoxicity
  • CDC complement-dependent cytotoxicity
  • hepsin a serine protease
  • the antibody In a cancer patient treated with a protease-regulated antibody against hepsin, the antibody would attack the tumor cells via the Fc portion-induced ADCC and CDC. However, in the liver, the protease- regulated antibody would initially bind to hepsin, but the Fc portion would be cleaved by a liver- specific protease prior to initiation of ADCC or CDC preventing liver toxicity.
  • the peptide (or polypeptide) linker of the protease-regulated antibody may comprise two or more amino acid residues and may contain one or more protease cleavage sites.
  • the linkers may alter antibody conformation, stability, and antigen-binding activities.
  • the length of linkers may range, for example, from 0 to about 100 amino acid residues. The following are examples of linkers:
  • Linker 1 SDDDDK (SEQ ID NO: 1)
  • Linker 2 GGGGSDDDDK (SEQ ID NO: 2)
  • Linker 3 GGGGSDDDDKGGGGS (SEQ ID NO: 3)
  • Linker 4 GGGGSGGGGSGGGGS (SEQ ID NO: 4)
  • Linker 5 fflPVLSGLSRTVNGEDAVPG (SEQ ID NO: 5)
  • Linker 6 VAAPFDDDDKTVGGYICEEN (SEQ ID NO: 6)
  • Linker 7 ELLESYIDGRIVEGSDAEIG (SEQ ID NO: 7)
  • Linker 8 STQSFNDFTRWGGEDAKPG (SEQ ID NO: 8)
  • Linker 9 PERGDNNLTRIVGGQECKDG (SEQ ID NO: 9)
  • Linker 10 EDQEDQVDPRLIDGKMTRRG (SEQ ID NO: 10)
  • Linker 11 KRNASKPQGRIVGGKVCPKG (SEQ ID NO: 11)
  • Linker 12
  • Linker 14 GSLVSGSCSQIINGEDCSPH (SEQ ED NO: 14)
  • Linker 15 SRIIN (SEQ ID NO: 15)
  • Linker 18 FNVLG (SEQ ID NO: 18)
  • Table 1 illustrates the excision site of several proteases.
  • ADAM metallopeptidase domain 9 (meltrin gamma)
  • ADAM metallopeptidase domain 17 (TNFalpha, converting enzyme)
  • Caspase 1 apoptosis-related cysteine peptidase (IL- l ⁇ convertase)
  • Caspase 2 apoptosis-related cysteine peptidase
  • Granzyme A (granzyme 1 , CTL-associated serine esterase 3)
  • the protease-regulated antibodies of the present invention may bind one or more antigens. These antigens may be selected from the group consisting of cytokines, cell surface receptors, enzymes, and receptors. These antigens include, but are not limited to, CD3, CD4, CD8, CD20, CD25, CD28, CD33, CD52, IL-2, IL-7, IL-8, TNF-alpha, TGF-beta INF-beta, INF-gamma, GMCSF, GCSF, VEGF, C5, EpCAM, EGF receptor, CD2 receptor, IL 2 receptor, IgE receptor, intergrin, and MHC class ⁇ .
  • the antibodies of the present invention may be utilized for the diagnosis and therapy of various diseases. For example, antibodies directed against human immunological cells and tumor- associated antigen may be used for cancer therapy. These antibodies may also be directed against tumor-associated antigen and toxic agents or enzymes for use as a cancer therapeutic. The antibodies of the present invention may also be utilized for the treatment of hemophilia and thrombosis as well as stem cell transplantation. These antibodies may be used for the selective stimulation and expansion of lymphocyte subset, hi addition, these antibodies may used for the detection of disease-related antigens.
  • bispecif ⁇ c antibodies may be used to recruit the immune system to attach tumor cells.
  • Targets on immunological cells include, but are not limited to, CD3, CD8, and Fc receptor.
  • Tumor-associated antigens include, but are not limited to, Her2, EGF receptor, CD20, CA-125, and carcinoembryonic antigen (CEA).
  • CEA carcinoembryonic antigen
  • a bispecif ⁇ c antibody against CD8 and Her2 can direct CD8-expressing cytotoxic lymphocytes to attack Her2 expressing breast cancer cells.
  • the antibodies or antibody fragments of the invention, or compositions including the antibodies or fragments can include a cytoxic agent that is conjugated to the antibody or fragment.
  • the cytotoxic agent is monomethylauristatin-E (MMAE), however, other cytoxic agents are also provided, which can include, for example, functional analogs of MMAE (e.g.
  • cytotoxic agents e.g., aplidin, azaribine, anastrozole, azacytidine, bleomycin, bortezomib, bryostatin-1, busulfan, calicheamycin, camptothecin, 10- hydroxycamptothecin, carmustine, celebrex, chlorambucil, cisplatin, irinotecan (CPT-I 1), SN-38, carboplatin, cladribine, cyclophosphamide, cytarabine, dacarbazine, docetaxel, dacti ⁇ omycin, daunomycin glucuronide, daunorubicin, dexamethasone, diethylstilbestrol, doxorubicin, doxorubicin glucuronide, epirubicin glucuronide, ethinyl estradiol, estramustine, etoposide
  • phage- antibody technology may be used to generate antibodies (Knappik, et al., J. MoI. Biol. 296:57-86, 2000).
  • Another approach for obtaining antibodies is to screen a DNA library from B cells as described by Dower, et al., (WO 91/17271) and McCafferty, et al., (WO 92/01047).
  • libraries of phage are produced in which members display different antibodies on their outer surfaces.
  • Antibodies are usually displayed as Fv or Fab fragments. Phage displaying antibodies are selected by affinity enrichment for binding to a selected protein.
  • Antibodies may also be produced using trioma methodology (Oestberg, et al., Hybridoma 2:361-367, 1983; U.S. Patent No. 4,634,664; U.S. Patent No. 4,634,666).
  • Antibodies may also be purified from any cell that expresses the antibodies, including host cells that have been transfected with antibody-encoding expression constructs.
  • the host cells may be cultured under conditions whereby the antibodies are expressed.
  • Purified antibody may be separated from other cellular components that may associate with the antibody in the cell, such as certain proteins, carbohydrates, or lipids using methods well known in the art. Such methods include, but are not limited to, size exclusion chromatography, ammonium sulfate fractionation, ion exchange chromatography, affinity chromatography, and preparative gel electrophoresis. Purity of the preparations may be assessed by any means known in the art, such as SDS- polyacrylamide gel electrophoresis.
  • a preparation of purified antibodies may contain more than one type of antibody.
  • antibodies may be produced using chemical methods to synthesize its amino acid sequence, such as by direct peptide synthesis using solid-phase techniques (see, e.g., Merrifield, J. Am. Chem. Soc. 85:2149-2154, 1963; Roberge, et al., Science 269:202-204, 1995). Protein synthesis may be performed using manual techniques or by automation. Automated synthesis may be achieved, for example, using Applied Biosystems 43 IA Peptide Synthesizer (Perkin Elmer).
  • fragments of antibodies may be separately synthesized and combined using chemical methods to produce a full-length molecule.
  • the antibodies of the present invention may be generated from parental antibodies.
  • Parent antibodies may be selected from various antibodies capable of binding specific targets and well known in the art, such as, but not limited to, but are not limited to anti-TNF antibody, anti-IL-12 antibody; anti-IL-18 antibody, anti-C5, anti-CD147, anti-gpl20, anti-CDl la, anti-CD18, anti- VEGF, anti-CD40L, anti-ICAM-1, anti-CD2, anti-EGFR, anti-TGF-beta 2, anti-E-selectin, anti- Her2/neu, anti-CD14, anti-ICAM-3, anti-CD80, anti-CD4, anti-CD3, anti-CD23, anti-beta2- integrin, , anti-CD52, , anti-CD22, anti-CD20, anti-CD25, anti-CD33, anti-HLA, anti-IL-1 alpha, anti-IL-1, anti-IL-1 receptor, anti-IL-2 receptor, anti-IL-4, anti-IL4 receptor, anti-IL5, anti-IL-5 receptor, anti-IL-6, anti-
  • Parent antibodies may also be selected from various therapeutic antibodies including, but are not limited to, rituximab, trastuzumab, pertuzumab, cetuximab, alemtuzumab, muromonab, ibritumomab, gemtuzumab ozogamicin, alefacept, abciximab, basiliximab, palivizumab, infliximab, adalimumab, etanercept, natalizumab, bevacizumab, omalizumab, efalizumab, clenoliximab, labetuzumab, epratuzumab, and visilizumab.
  • trastuzumab pertuzumab
  • cetuximab cetuximab
  • alemtuzumab muromonab
  • ibritumomab gemtuzumab ozogamicin
  • the newly synthesized molecules may be substantially purified by preparative high performance liquid chromatography ⁇ see, e.g., Creighton, Proteins: Structures and Molecular Principles. WH Freeman and Co., New York, N.Y., 1983).
  • the composition of a synthetic polypeptide may be confirmed by amino acid analysis or sequencing (e.g., using Edman degradation).
  • the present invention also relates to bispecific or bifunctional antibodies that have one binding site that specifically binds to a first antigen and a second binding site that specifically binds to a second antigen. This results in multi-functional valency, that is, an ability to bind at least two different epitopes simultaneously.
  • the present invention also relates to polynucleotides encoding antibodies. These polynucleotides may be used, for example, to produce quantities of the antibodies for therapeutic or diagnostic use.
  • Polynucleotides of the present invention may also be isolated from host cells, free of other cellular components such as membrane components, proteins, and lipids.
  • Polynucleotides may be isolated using standard nucleic acid purification techniques, or synthesized using an amplification technique such as the polymerase chain reaction (PCR), or by using an automatic synthesizer. Methods for isolating polynucleotides are routine and are known in the art. Any such technique for obtaining a polynucleotide may be used to obtain isolated polynucleotides encoding antibodies of the invention. For example, restriction enzymes and probes may be used to isolate polynucleotides which encode antibodies.
  • Antibody-encoding cDNA molecules may be made with standard molecular biology techniques, using mRNA as a template. Thereafter, cDNA molecules may be replicated using molecular biology techniques known in the art and disclosed in manuals such as Sambrook, et al., (Molecular Cloning: A Laboratory Manual, (Second Edition, Cold Spring Harbor Laboratory Press; Cold Spring Harbor, N. Y.; 1989) Vol. 1-3). An amplification technique, such as PCR, may be used to obtain additional copies of the polynucleotides. Alternatively, synthetic chemistry techniques may be used to synthesize polynucleotides encoding antibodies of the invention.
  • the polynucleotide may be inserted into an expression vector that contains the necessary elements for the transcription and translation of the inserted coding sequence.
  • Methods that are well known to those skilled in the art may be used to construct expression vectors containing sequences encoding antibodies and appropriate transcriptional and translational control elements. These methods include in vitro recombinant DNA techniques, synthetic techniques, and in vivo genetic recombination. Such techniques are described, for example, in Sambrook, et al. (1989) and in Ausubel, et al., (Current Protocols in Molecular Biology, John Wiley & Sons, New York, N. Y., 1995).
  • a variety of expression vector/host systems may be utilized to contain and express sequences encoding antibodies. These include, but are not limited to, microorganisms, such as bacteria transformed with recombinant bacteriophage, plasmid, or cosmid DNA expression vectors; yeast transformed with yeast expression vectors; insect cell systems infected with virus expression vectors (e.g., baculovirus); plant cell systems transformed with virus expression vectors (e.g., cauliflower mosaic virus, CaMV; tobacco mosaic virus, TMV); or bacterial expression vectors (e.g., Ti or pBR322 plasmids), or animal cell systems.
  • microorganisms such as bacteria transformed with recombinant bacteriophage, plasmid, or cosmid DNA expression vectors
  • yeast transformed with yeast expression vectors insect cell systems infected with virus expression vectors (e.g., baculovirus)
  • plant cell systems transformed with virus expression vectors e.g., cauliflower mosaic virus, CaMV; tobacco mosaic virus, T
  • control elements or regulatory sequences are those non-translated regions of the vector - enhancers, promoters, 5' and 3' untranslated regions - which interact with host cellular proteins to carry out transcription and translation. Such elements may vary in strength and specificity. Depending on the vector system and host utilized, any number of suitable transcription and translation elements, including constitutive and inducible promoters, may be used. For example, when cloning in bacterial systems, inducible promoters can be used. The baculovirus polyhedrin promoter may be used in insect cells.
  • Promoters or enhancers derived from the genomes of plant cells may be cloned into the vector.
  • plant viruses e.g., viral promoters or leader sequences
  • promoters from mammalian genes or from mammalian viruses may be used. If it is necessary to generate a cell line that contains multiple copies of a nucleotide sequence encoding an antibody, vectors based on SV40 or EBV may be used with an appropriate selectable marker.
  • the affinity (Kj) of antibody binding to an antigen may be assayed using any method known in the art including, for example, immunoassays such as enzyme-linked immunospecif ⁇ c assay (ELISA), Bimolecular Interaction Analysis (BIA) (see, e.g., Sjolander and Urbaniczky, Anal. Chem. 63:2338-2345, 1991; Szabo, et al., Curr. Opin. Struct. Biol. 5:699-705, 1995), and fluorescence-activated cell sorting (FACS) for quantification of antibody binding to cells that express an antigen.
  • immunoassays such as enzyme-linked immunospecif ⁇ c assay (ELISA), Bimolecular Interaction Analysis (BIA) (see, e.g., Sjolander and Urbaniczky, Anal. Chem. 63:2338-2345, 1991; Szabo, et al., Curr. Opin. Struct. Biol. 5:699
  • BIA is a technology for analyzing biospecific interactions in real time, without labeling any of the interactants (e.g., BIAcoreTM). Changes in the optical phenomenon surface plasmon resonance (SPR) may be used as an indication of real-time reactions between biological molecules.
  • SPR surface plasmon resonance
  • the present invention also relates to the use of quantitative immunoassays to measure levels of proteins in patient samples.
  • Many formats may be adapted for use with the methods of the present invention.
  • the detection and quantitation of a protein in patient samples may be performed, by enzyme-linked immunosorbent assays, radioimmunoassays, dual antibody sandwich assays, agglutination assays, fluorescent immunoassays, immunoelectron and scanning microscopy, among other assays commonly known in the art.
  • the quantitation of a protein in such assays may be adapted by conventional methods known in the art.
  • Serial changes in circulating a protein levels may be detected and quantified by a sandwich assay in which the capture antibody has been immobilized using conventional techniques on the surface of the support.
  • Suitable supports include, for example, synthetic polymer supports, such as polypropylene, polystyrene, substituted polystyrene, polyacrylamides (such as polyamides and polyvinylchloride), glass beads, agarose, and nitrocellulose.
  • the antibodies useful to identify proteins may be labeled in any conventional manner.
  • An example of a label is horseradish peroxidase, and an example of a method of labeling antibodies is by using biotin-strepavidin complexes.
  • antibodies used in the immunoassays of this invention that are used as tracers may be labeled in any manner, directly or indirectly, that results in a signal that is visible or can be rendered visible.
  • Detectable marker substances include radionuclides, such as 3 H, 125 I, and 131 I; fluorescers, such as, fluorescein isothiocyanate and other fluorochromes, phycobiliproteins, phycoerythin, rare earth chelates, Texas red, dansyl and rhodamine; colorimetric reagents (chromogens); electron-opaque materials, such as colloidal gold; bioluminescers; chemiluminescers; dyes; enzymes, such as, horseradish peroxidase, alkaline phosphatases, glucose oxidase, glucose-6-phosphate dehydrogenase, acetylcholinesterase, alpha -, beta-galactosidase, among others; co
  • Another detection and quantitation systems produce luminescent signals, bioluminescent (BL) or chemiluminescent (CL).
  • chemiluminescent (CL) or bioluminescent (BL) assays the intensity or the total light emission is measured and related to the concentration of the unknown analyte.
  • Light can be measured quantitatively using a luminometer (photomultiplier tube as the detector) or charge-coupled device, or qualitatively by means of photographic or X-ray film.
  • the main advantages of using such assays is their simplicity and analytical sensitivity, enabling the detection and/or quantitation of very small amounts of analyte.
  • Exemplary luminescent labels are acridinium esters, acridinium sulfonyl carboxamides, luminol, umbelliferone, isoluminol derivatives, photoproteins, such as aequorin, and luciferases from fireflies, marine bacteria, Vargulla and Renilla.
  • Luminol can be used optionally with an enhancer molecule such as 4-iodophenol or 4-hydroxy-cinnamic acid.
  • a CL signal is generated by treatment with an oxidant under basic conditions.
  • Additional luminescent detection systems are those wherein the signal (detectable marker) is produced by an enzymatic reaction upon a substrate.
  • CL and BL detection schemes have been developed for assaying alkaline phosphatases (AP), glucose oxidase, glucose 6-phosphate dehydrogenase, horseradish peroxidase (HRP), and xanthine-oxidase labels, among others.
  • AP and HRP are two enzyme labels which can be quantitated by a range of CL and BL reactions.
  • AP can be used with a substrate, such as an adamantyl 1,2-dioxetane aryl phosphate substrate (e.g.
  • AMPPD or CSPD Kricka, L.J., "Chemiluminescence and Bioluminescence, Analysis by,” Molecular Biology and Biotechnology: A Comprehensive Desk Reference (ed. R.A. Meyers) (VCH Publishers; N.Y., N.Y.; 1995)); for example, a disodium salt of 4-methoxy-4-(3-phosphatephenyl) spiro [l,2-dioxetane-3,2'-adamantane], with or without an enhancer molecule such as l-(trioctylphosphonium methyl)-4- (tributylphosphonium methyl) benzene diochloride.
  • HRP is may be used with substrates, such as, 2',3',6'-trifluorophenyl- methoxy- 10-methylacridan-9-carboxylate.
  • CL and BL reactions may be adapted for analysis not only of enzymes, but also of other substrates, cofactors, inhibitors, metal ions, and the like.
  • luminol, firefly luciferase, and marine bacterial luciferase reactions are indicator reactions for the production or consumption of peroxide, ATP, and NADPH, respectively. They may be coupled to other reactions involving oxidases, kinases, and dehydrogenases, and may be used to measure any component of the coupled reaction (enzyme, substrate, cofactor).
  • the detectable marker may be directly or indirectly linked to an antibody used in an assay of this invention.
  • exemplary of an indirect linkage of the detectable label is the use of a binding pair between an antibody and a marker or the use of a signal amplification system.
  • binding pairs that may be used to link antibodies to detectable markers are biotin/avidin, streptavidin, or anti-biotin; avidin/anti-avidin; thyroxine/thyroxine-binding globulin; antigen/antibody; antibody/ anti-antibody; carbohydrate/lectins; hapten/anti-hapten antibody; dyes and hydrophobic molecules/hydrophobic protein binding sites; enzyme inhibitor, coenzyme or cofactor/enzyme; polynucleic acid/homologous polynucleic acid sequence; fluorescein/anti- fluorescein; dinitrophenol/anti-dinitrophenol; vitamin Bl 2/intrinsic factor; cortisone, cortisol/cortisol binding protein; and ligands for specific receptor protein/membrane associated specific receptor proteins.
  • labels may be bound either covalently or non-covalently.
  • Exemplary antibody conjugation methods are described in Avarmeas, et al., Scan. J. Immunol. 8(Suppl. 7): 7, 1978); Bayer, et al., Meth. Enzymol. 62:308, 1979; Chandler, et al., J. Immunol. Meth. 53:187, 1982; Ekeke and Abuknesha, J. Steroid Biochem. 11:1579, 1979; Engvall and Perlmann, J. Immunol. 109:129, 1972; Geoghegan, et al., Immunol. Comm. 7:1, 1978; and Wilson and Nakane, Immunofluorescence and Related Techniques. Elsevier/North Holland Biomedical Press; Amsterdam (1978).
  • a fluorescent, chemiluminescent, or colored product may be determined or measured fluorometrically, luminometrically, spectrophotometrically, or visually.
  • chemiluminescent compounds having an acridinium, benzacridinium, or acridan type of heterocyclic ring systems are other examples of labels.
  • acridinium esters include those compounds having heterocyclic rings or ring systems that contain the heteroatom in a positive oxidation state including such ring systems as acridinium, benz[a]acridinium, benz[b]acridinium, benz[c]acridinium, a benzimidazole cation, quinolinium, isoquinolinium, quinolizinium, a cyclic substituted quinolinium, phenanthridinium, and quinoxalinium.
  • the tracer may be prepared by attaching to the selected antibody either directly or indirectly a reactive functional group present on the acridinium or benzacridinium ester, as is well known to those skilled in the art ⁇ see, e.g., Weeks, et al., Clin. Chem. 29(8): 1474-1479, 1983).
  • Examples of compounds are acridinium and benzacridinium esters with an aryl ring leaving group and the reactive functional group present in either the para or the meta position of the aryl ring. (see, e.g., U.S. Patent No. 4,745,181 and WO 94/21823).
  • treatment includes any process, action, application, therapy, or the like, wherein a subject (or patient), including a human being, is provided medical aid with the object of improving the subject's condition, directly or indirectly, or slowing the progression of a condition or disorder in the subject.
  • combination therapy means the administration of two or more therapeutic agents to treat a disease, condition, and/or disorder.
  • administration encompasses co-administration of two or more therapeutic agents in a substantially simultaneous manner, such as in a single capsule having a fixed ratio of active ingredients or in multiple, separate capsules for each inhibitor agent.
  • administration encompasses use of each type of therapeutic agent in a sequential manner.
  • the antibodies of the inventin may be administered in combination with the following agents: cytotoxic agent, angiogenesis inhibitors, antirheumatic agent, muscle relaxant, narcotic, non-steroid anti-inflammatory drug, analgesic, anesthetic, sedative, local anesthetic, neuromuscular blocker, antimicrobial agent, immunoglobulins, antidepressant, asthma medication, cytokine, and cytokine antagonist.
  • agents cytotoxic agent, angiogenesis inhibitors, antirheumatic agent, muscle relaxant, narcotic, non-steroid anti-inflammatory drug, analgesic, anesthetic, sedative, local anesthetic, neuromuscular blocker, antimicrobial agent, immunoglobulins, antidepressant, asthma medication, cytokine, and cytokine antagonist.
  • the antibodies of the inventin may be administered in combination with various anti-cancer agents including, but not limited to, bleomycin, docetaxel, doxorubicin, edatrexate, erlotinib, etoposide, finasteride, flutamide, gemcitabine, genitinib, goserelin acetate, granisetron, imatinib, irinotecan, ondansetron, paclitaxel, pegaspargase, piloca ⁇ ine hydrochloride, porfimer sodium, interleukin-2, rituximab, topotecan, trastuzumab, triapine, vincristine, and vinorelbine tartrate, or therapeutic antibodies or fragments thereof, or anti-angiogenic agent, such as, for example, angiostatin, bevacizumab, sorafenib, baculostatin, canstatin, maspin, anti-angiogenic agent
  • terapéuticaally effective means the amount of each agent administered that will achieve the goal of improvement in a disease, condition, and/or disorder severity, while avoiding or minimizing adverse side effects associated with the given therapeutic treatment.
  • pharmaceutically acceptable means that the subject item is appropriate for use in a pharmaceutical product.
  • an embodiment of this invention includes a method of treating the various conditions in a patient (including mammals) which comprises administering to said patient a composition containing an amount of an antibody of the invention that is effective in treating the target condition.
  • the antibodies of the present invention may be used in the treatment or prevention of various diseases including, but not limited to, cancer, infectious disease, and autoimmune diseases.
  • the antibodies of the present invention or compositions including the antibodies may include a cytotoxic agent (e.g., monomethylauristatin-E) that is conjugated to the antibody.
  • a cytotoxic agent e.g., monomethylauristatin-E
  • Antibodies of the present invention may be administered alone or in combination with one or more additional therapeutic agents.
  • Combination therapy includes administration of a single pharmaceutical dosage formulation which contains an antibody of the present invention and one or more additional therapeutic agents, as well as administration of the antibody of the present invention and each additional therapeutic agents in its own separate pharmaceutical dosage formulation.
  • an antibody of the present invention and a therapeutic agent may be administered to the patient together in a single oral dosage composition or each agent may be administered in separate oral dosage formulations
  • the antibody of the present invention and one or more additional therapeutic agents may be administered at essentially the same time (e g , concurrently) or at separately staggered times (e g , sequentially)
  • the antibody may be tested in vivo in a mouse xenograft tumor model
  • An example of a therapeutic model is detailed in Example 8
  • the antibodies desc ⁇ bed herein may be provided in a pharmaceutical composition comp ⁇ sing a pharmaceutically acceptable earner
  • the pharmaceutically acceptable earner may be non-pyrogenic
  • the compositions may be administered alone or in combination with at least one other agent, such as stabilizing compound, which may be administered in any stenle, biocompatible pharmaceutical earner including, but not limited to, salme, buffered saline, dextrose, and water
  • a vanety of aqueous earners may be employed including, but not limited to saline, glycine, or the like These solutions are stenle and generally free of particulate matter These solutions may be stenhzed by conventional, well known stenhzation techniques (e g , filtration)
  • the compositions may contain pharmaceutically acceptable auxiliary substances as required to approximate physiological conditions such as pH adjusting and buffering agents, and the like
  • concentration of the antibody of the invention in such pharmaceutical formulation may vary widely, and may be selected pnmanly based on fluid volumes, viscosities, etc
  • compositions may be administered to a patient alone, or in combination with other agents, drugs or hormones
  • these pharmaceutical compositions may contain suitable pharmaceutically acceptable earners compnsing excipients and auxilianes that facilitate processing of the active compounds into preparations which may be used pharmaceutically
  • Pharmaceutical compositions of the invention may be administered by any number of routes including, but not limited to, oral, mtravenous, intramuscular, mtra-artal, intramedullary, intrathecal, mtraventncular, transdermal, subcutaneous, intrapentoneal, intranasal, parenteral, topical, sublingual, or rectal means
  • Formulations suitable for subcutaneous, intravenous, intramuscular, and the like, suitable pharmaceutical earners, and techniques for formulation and administration may be prepared by any of the methods well known in the art ⁇ see, e.g., Remington's Pharmaceutical Sciences, Mack Publishing Co., Easton, Pa., 20 th edition, 2000).
  • the present invention also provides diagnostic methods with which a particular antigen may be detected in a patient sample or biological sample. Such diagnostic methods may be used, for example, to diagnose disorders in which a particular antigen is elevated or reduced. Such disorders include, but are not limited to, cancer, infectious disease, and autoimmune diseases. As an example, when used for diagnosis, detection of an amount of the antibody-antigen complex in a sample from a patient which is greater than an amount of the complex in a normal sample identifies the patient as likely to have the disorder
  • the patient sample may be contacted with an antibody of the invention, and the patient sample may then be assayed for the presence of an antibody-antigen complex.
  • the antibody may comprise a detectable label, such as a fluorescent, radioisotopic, chemiluminescent, or enzymatic label, such as horseradish peroxidase, alkaline phosphatase, or luciferase.
  • the antibody may be bound to a solid support, which may accommodate automation of the assay.
  • Suitable solid supports include, but are not limited to, glass or plastic slides, tissue culture plates, microtiter wells, tubes, silicon chips, or particles such as beads (including, but not limited to, latex, polystyrene, or glass beads). Any method known in the art may be used to attach the antibody to the solid support, including use of covalent and non-covalent linkages, passive absorption, or pairs of binding moieties attached to the antibody and the solid support. Binding of antigen and the antibody may be accomplished in any vessel suitable for containing the reactants. Examples of such vessels include microtiter plates, test tubes, and microcentrifuge tubes.
  • a therapeutically effective dose refers to the amount of an antibody that may be used to effectively treat a disease (e.g., cancer) compared with the efficacy that is evident in the absence of the therapeutically effective dose.
  • the therapeutically effective dose may be estimated initially in animal models (e.g., rats, mice, rabbits, dogs, or pigs). The animal model may also be used to determine the appropriate concentration range and route of administration. Such information may then be used to determine useful doses and routes for administration in humans.
  • Therapeutic efficacy and toxicity e.g., ED 50 - the dose therapeutically effective in 50% of the population and LD 5O - the dose lethal to 50% of the population
  • Therapeutic efficacy and toxicity e.g., ED 50 - the dose therapeutically effective in 50% of the population and LD 5O - the dose lethal to 50% of the population
  • the dose ratio of toxic to therapeutic effects is the therapeutic index, and it may be expressed as the ratio, LD 5O /ED 5 o.
  • the data obtained from animal studies may used in formulating a range of dosage for human use.
  • the dosage contained in such compositions may be within a range of circulating concentrations that include the ED 50 with little or no toxicity. The dosage varies
  • the exact dosage may be determined by the practitioner, in light of factors related to the patient who requires treatment. Dosage and administration may be adjusted to provide sufficient levels of the antibody or to maintain the desired effect. Factors that may be taken into account include the severity of the disease state, general health of the subject, age, weight, and gender of the subject, diet, time and frequency of administration, drug combination(s), reaction sensitivities, and tolerance/response to therapy. Effective in vivo dosages of an antibody are in the range of about 5 ⁇ g to about 500 ⁇ g/kg of patient body weight.
  • the mode of administration of antibody-containing pharmaceutical compositions of the present invention may be any suitable route which delivers the antibody to the host.
  • pharmaceutical compositions of the invention may be useful for parenteral administration (e.g., subcutaneous, intramuscular, intravenous, or intranasal administration).
  • Antibodies 3E10 and 19G9 recognize tissue factor (TF) and tumor-associated antigen RGl, respectively. These two antibodies were used to construct protease-regulated antibodies containing the protease site DDDDK (SEQ TD NO: 26) linker located between the antigen binding domains and constant region domain. Specifically, these antibodies contained V L -DDDDK-CL on the light chain and V H -DDDDK-CH1-Myc-His6 on the heavy chain, where the linker is cleavable by enterokinase, and Myc and His6 are tags for detection and purification.
  • DDDDK SEQ TD NO: 26
  • the DNA sequences for the two antibodies were cloned into bacterial expression vectors using standard molecular biology technologies, and the constructs were confirmed by DNA sequencing. Examples of plasmid are shown in Figures 8 and 9.
  • the plasmid containing either 3E10 or 19G9 was expressed and purified from bacterial strain TGl . Briefly, a single colony of bacteria strain TGl containing the antibody expression plasmid was selected and grown overnight in 8 ml of 2xYT medium in the presence of 34 ⁇ g/ml chloramphenicol and 1% glucose. A volume of culture (7 ml) was transferred to 250 ml fresh 2xYT medium containing 34 ⁇ g/ml chloramphenicol and 0.1% glucose.
  • protease-regulated antibodies were also constructed using tandem linked variable regions from 3E10 and 19G9. These antibodies contained, for example, V L 3E10- DDDDK-V L 19G9-CL on the light chain and V H 3E1O-DDDDK-V H 19G9-CH1-Myc-His6 on the heavy chain, where the linker is cleavable by enterokinase, and Myc and His6 are tags for detection and purification.
  • An antibody library was also constructed using the framework regions (FR), for example, FR4 of 3E10 and FRl of 19G9 either intact or truncated.
  • FR4 of 3E10 and FRl of 19G9 either intact or truncated.
  • the expression vector pIE_SRgamma_fa contains cDNAs encoding the constant regions of human IgGl (fa haplotype) and kappa chains, respectively.
  • An overlap PCR was performed to link the variable regions of anti-TF antibody 3E10 and anti-RGl antibody 19G9.
  • the native signal peptide of 19G9 was used for secretion of the protease-regulated antibodies.
  • Four examples of peptide linkers located between the variable regions of 3E10 and 19G9 are Linker 1: SDDDDK (SEQ ID NO: 2), Linker 2: GGGGSDDDDK (SEQ ID NO: 3), Linker 3:
  • the primers for amplification of the variable region of the light chain introduced Hind III and Bsiw I sites into the 5' and 3' ends of PCR fragment, respectively.
  • the resulting PCR-amplif ⁇ ed V L genes were cloned into the Hindlll/Bsiw site of pIE SRgammal fa to create PIE-3E10V L - linker-19G9V L .
  • NHKPSNTKVDKKVEPKCEF SEQ ID NO: 50
  • VDDDDKEVQLVQSGGGLVQPGGSLRLSCAG VDDDDKEVQLVQSGGGLVQPGGSLRLSCAG
  • Biotinylated TF (1 ⁇ g/ml) was added to streptavidin pre-coated 96-well plates (Pierce Chemical, Rockford, IL) and incubated for 1 hr. The plates were then washed (5x) with PBS containing 0.5% Tween-20. Samples and controls (serially diluted) were added to the wells and incubated for 1 hr, followed by washes (5x) with PBS containing 0.5% Tween-20. Horseradish peroxidase (HRP)-conjugated anti-human IgG or HRP -conjugated anti-human Fab were diluted in PBS (1 :5000) and added to each well.
  • HRP horseradish peroxidase
  • HRP horseradish peroxidase
  • streptavidin 1:10000 diluted
  • Amplex Red 10 ⁇ g/ml
  • Parental antibodies 3E10, 19G9, and polyclonal human Fab were used as controls.
  • the results are the average of duplicate wells ( Figure 12).
  • the untreated bispecific protease-regulated antibody simultaneously binds to both TF and RG-I ("Linkl” and "Link2,” respectively). However, following enterokinase treatment, the binding to both antigens is greatly reduced (“Linkl /EK" and “Link2/EK,” respectively).
  • protease-regulated antibodies were also measured using this assay.
  • the antigen binding activity of protease-regulated antibodies 3E10-Typel-Fab and 19G9-Typel-Fab is shown in Figure 13.
  • the controls are designated 3E10-Reg-Fab, 19G9-Reg-Fab, and HuFab.
  • FIG 14 The antigen binding activity of Fab-like protease-regulated antibodies is demonstrated in Figure 14.
  • the activity of antibodies HlLl, H1L4, H1L7, H4L7, and H5L5 (Type 2) was measured in the absence and presence of enterokinase.
  • Parental antibodies 3E10, 19G9, and polyclonal human Fab were used as controls.
  • Figure 15 shows the antigen binding activity of Fab-like protease-regulated antibodies H2L1, H2L2, and H2L8 (Type 3) and H3L1, H3L4, and H5L4 (Type 4).
  • Protease-regulated antibodies were digested with EnterokinaseMaxTM, the catalytic subunit of enterokinase (Invitrogen, Carlsbad, CA). The concentration of antibodies was adjusted to 1-5 ⁇ g/ml. A volume of antibody (100 ⁇ l) was mixed with 20 ⁇ l 10x EnterokinaseMaxTM buffer and 75 ⁇ l sterile water in a tube. EnterokinaseMaxTM (5 ⁇ l) was added to each sample and the samples were incubated at 37°C for 16 hr. For the control group, a volume of water (5 ⁇ l ) was used.
  • HRP horseradish peroxidase
  • Approximately 50 ng of antibody samples were mixed with loading buffer containing DTT (Invitrogen, Carlsbad, CA) and boiled for 5 min. The samples were then loaded onto a 12% Bis-Tris NuP AGE® gel (Invitrogen, Carlsbad, CA), separated, and transferred to nitrocellulose membranes. After blocking with 5% dry milk for 2 hr, the nitrocellulose membrane was incubated with a detection antibody for 1.5 hr.
  • Example 8 Subcutaneous xenograft cancer model
  • Each experimental group consists of 10 mice and the dosing volume was 0.1 mL/10 g body weight.
  • the length and width of each tumor is measured by using an electronic caliper 2-3 times per week and tumor weights (mg) are calculated based on the formula of [length (mm) x width (mm) 2 ]/2. All data, including daily observations, obtained throughout the course of the study are documented.

Abstract

This invention relates to monospecific and multispecific antibodies that may be utilized for the diagnosis and treatment of various diseases. In addition, these antibodies may be modified by protease cleavage. Protease control or regulation may be provided by a protease site located in, for example, a linker. These protease-regulated antibodies may also be utilized for the diagnosis and treatment of various diseases.

Description

MONOSPECIFIC AND MULTISPECD7IC ANTIBODIES AND METHOD OF USE
[001] This application claims benefit to U.S. Provisional Application Serial No. 60/955,912, filed August 15, 2007, and U.S. Provisional Application Serial No. 60/955,913, filed August 15, 2007, the contents of which are incorporated herein by reference in their entirety.
FIELD OF THE INVENTION
[002] This invention relates to monospecific and multispecific antibodies that may be utilized for the diagnosis and treatment of various diseases. In addition, these antibodies may be modified by protease cleavage. Protease control or regulation may be provided by a protease site located in, for example, a linker. These protease-regulated antibodies may also be utilized for the diagnosis and treatment of various diseases.
BACKGROUND OF THE INVENTION
[003] An antibody may be directed against one or more different antigens or one or more different epitopes on the same antigen. For example, a bispecific antibody is directed against two different antigens or two different epitopes on the same antigen. As bispecific antibodies can simultaneously bind to two distinct targets, these antibodies have great potential for antibody- based diagnosis and for the treatment of various diseases and disorders such as cancer, infectious diseases, autoimmune diseases, and blood diseases. For example, bispecific antibodies can selectively stimulate and expand T lymphocytes (Wong, et al., J. Immunol. 139:1369-1374, 1987; Wong, et al., Clin. Immunol. Immunopathol. 58:236-250, 1991), direct immune cells or toxic agents to kill tumor cells (Lum, et al., Exp. Hematol. 34:1-6,2006; Wolf, et al., Drug Discov. Today 10: 1237-1244, 2005; Cao, et al., Adv. Drug Deliv. Rev. 55:171-197, 2003; Talac, et al., J. Biol. Regul. Homeost. Agents 14:175-181, 2000), and simultaneously block two receptors (Lu, et al., J. Biol. Chem. 279:2856-2565, 2004). In addition, a bispecific antibody may be used as a substitute for Factor VIII to enhance enzymatic reaction (US Patent Application No. 2007/0041978) or to direct stem cells to the site of injury in patients with myocardial infarction (Lum, et al., Blood Cells MoI. Dis. 32:82-87, 2004).
[004] Bispecific antibodies targeting tumor-associated antigens and toxic agents may be used in cancer therapy. For example, using this technology, one arm of the bispecific antibody may be directed to a tumor-associated antigen such as Her2, EGF receptor, CD20, CD22, CD30, CD33, CD52, and CA-125, and the other arm of the bispecific antibody may target a toxin, drug, or cytokine. That is, bispecific antibodies may selectively direct toxic agents to tumor cells enhancing the efficacy of therapeutic antibodies and decreasing systemic toxicity. Examples of
SUBSTITUT" SHEET (RULE 26) toxin/drug include calicheamicin, doxorubicin, epirubicin, methotrexate, ricin A, saporin, gelonin, and vinca alkaloids, and cytokine examples include tumor necrosis factor alpha (TNF-alpha) and IL-2.
[005] Specific cleavage by proteases of defined sites in biologically important effector proteins is a well known method for the natural control of cellular and extracellular physiological processes. Examples include protease activation and inhibition of the coagulation cascade (Butenas, et al., Biochemistry 67:3-12, 2002; Esmon, Chest, 124:26S-32S, 2003), protease activation of protease-activatable receptors (Coughlin, Arterioscler. Thromb. Vase. Biol. 18:514- 518, 1998), protease release of membrane associated cytokines (Amour, et al., FEBS Lett. 435:39- 44, 1998), protease processing of prohormones in secretory vesicles (Moore, et al., Arch. Physiol. Biochem. 110:16-25, 2002), and protease processing of proproteins during secretion (Scamuffa, et al., FASEB J. 20:1954-1963, 2006). Proteases are often expressed or located in a tissue-specific or tumor-specific manner and examples include the membrane serine protease conn in heart tissue (Yan, et al., Proc. Natl. Acad. Sci. USA 97:8525-8529, 2000), the kallikrein serine protease prostate-specific antigen (PSA) in prostate tissue, prostate cancer, and seminal fluid (Veveris- Lowe, et al., Semin. Thromb. Hemost. 33:87-99, 2007), the membrane serine protease hepsin in liver tissue and tumors (Xuan, et al., Cancer Res. 66:3611-3619, 2006), coagulation protease factor X expressed in the liver and secreted into blood (Miao, et al., J. Biol. Chem. 267:7395-7401, 1992), and digestive proteases expressed in the pancreas and released to the duodenum (Belorgey, et al., Biochem. J. 313:555-560, 1996). Specific cleavage of amino acid sequences by human proteases include thrombin (Chang, Eur. J. Biochem. 151 :217-224, 1985), factor Xa (Nagai, et al., Methods Enzymol. 153:461-481, 1987), furin (Brennan, et al., FEBS Lett. 347:80-84, 1994), subtilisin-like prohormone convertases (Lipkind, et al., J. Biol. Chem. 270:13277-13284, 1995), and the matrix metalloproteinases (Minod, et al., J. Biol. Chem. 281 :38302-38313, 2006). Genes encoding specific proteases may be up-regulated in tumor tissue and Table 2 indicates proteases that are associated with cancer tissue.
[006] Protease cleavage is widely used in in vitro studies to specifically remove protein or peptide tags from recombinant proteins or to process hybrid recombinant proteins. For example, human rhinovirus 3C protease, thrombin, or factor Xa have been used to remove glutathione S- transferase (GST) tags (Dian, et al., Life Sciences News - Amersham Biosciences 10: 1-5, 2002) and factor Xa has been use to process hybrid proteins (Nagai, et al., 1987). Proteases are often targets for drugs as a means to regulate biological processes; and examples include factor Xa (Phillips, et al., J. Med. Chem. 41 :3557-3562,1998), thrombin (Riester, et al., Proc. Natl. Acad. Sci. USA 102:8597-8602, 2005), urokinase (Killeen, et al., Br. J. Cancer 96:262-268, 2007), and factor Vila (Kohrt, et al., Bioorg. Med. Chem. Lett. 15:4752-4756, 2005). Finally, proteins developed as biological drugs may be modified to prevent cleavage by proteases and to improve their stability in vitro or in vivo (Light, et al., Eur. J. Biochem. 262:522-533, 1999; Saenko, et al., Haemophilia 12:42-51, 2006).
[007] Specific protease cleavage sites have been incorporated into linkers that link a toxin molecule to a targeting antibody in order to allow protease specific release of the toxin by intracellular proteases (Trail, et al., Cancer Immunol. Immunother. 52:328-337, 2003). Furthermore, targeting antibodies have been created in many formats. For example, bispecific antibodies have been developed to allow binding to two different antigens or two different epitopes of an antigen by a single antibody molecule (Segal, et al., Curr. Opin. Immunol. 11 :558-562, 1999; Tomlinson, et al., Methods Enzymol. 326:461-479, 2000; Wu, et al., Nat Biotechnol. 25:1290- 1297, 2007). Other bispecific molecules have been generated with the ability to block two receptors (Lu, et al., J. Biol. Chem. 279:2856-2865, 2004) and to recruit immune cells to attack cancer cells and tumor tissue (Loffler, et al., Leukemia 17:900-909, 2003; Lum, et al., Exp. Hematol. 34:1-6, 2006).
[008] The present invention relates to a novel antibody format, for example, monospecifc and multispecific antibodies. The antibodies of the present invention may be constructed by tandem linking of two different heavy chain (H) variable region domains (VH) and two different light chain (L) variable region domains (Vt). The heavy chain and light chain may form a Fab-like or IgG- like molecule through the disulfide bond between constant (C) regions. Multispecific antibodies may be generated by linking more than two antibody variable domains.
[009] The antibodies of the present invention may be modified by protease cleavage. These protease-regulated antibodies may be, for example, monospecific antibodies, bispecific antibodies, or antibodies with sequential binding-activity upon protease digestion in either, for example, Fab- like or IgG-like format. Protease control or regulation may be provided by a protease site located in, for example, a linker. These protease-regulated antibodies may be utilized for the diagnosis and treatment of various diseases, and provide an additional level of control for biological drugs for therapeutic or diagnostic applications.
DESCRIPTION OF THE FIGURES
[100] Figure 1. Schematic drawing of a monospecific protease-regulated antibody with a linker which contains a protease site between variable domain and Fc domain ("Type 1").
[101] Figure 2. Schematic drawing of a bispecific protease-regulated antibody with a linker which contains a protease cleavage sequence that allows removal of one antigen-binding site ("Type 2"). [102] Figure 3. Schematic drawing of another bispecific protease-regulated antibody with a linker which contains a protease cleavage sequence that allows removal of one antigen-binding site ("Type 2").
[103] Figure 4. Schematic drawing of the application of a bispecific protease-regulated antibody that simultaneously binds two different antigens.
[104] Figure 5. Schematic drawing of a protease-regulated antibody that cannot bind to two different antigens simultaneously ("Type 3")-
[105] Figure 6. Schematic drawing of the application of a protease-regulated antibody that cannot bind to two different antigens simultaneously.
[106] Figure 7. Schematic drawing of a monospecific protease-regulated antibody 'prodrug' that can only bind antigen following protease activation to remove inactive blocking antibody domains ("Type 4").
[107] Figure 8. Map of an expression vector for an IgG-like bispecific antibody. SignalP: signal peptide; VLani3E10: variable region of 3E10 lambda chain; Vkl9G9: variable region of 19G9 kappa chain; C kappa: constant region of kappa chain; DHFR: dihydrofolate reductase; VH: variable region of heavy chain; Neo: neomycin resistant gene; 3E10VH: variable region of 3E10 heavy chain; 19G9VH: variable region of 19G9 heavy chain; CH: constant region of heavy chain; Amp: ampicillin resistant gene.
[108] Figure 9. Map of expression vector of Fab-like bispecific antibody. LacZ, lac Z promoter; ompA and pho A, signal peptide; VL-linkl-VK, variable region of light chain of bispecific antibody against tissue factor and RGl; CL-kappa, constant region of kappa chain; VHs with linker, variable region of heavy cham of bispecific antibody against tissue factor and RGl; CHl, the first constant region of IgG heavy chain; cat, chloramphenicol resistant gene.
[109] Figure 10. TF-binding ELISA. Four bispecific antibodies and parental antibodies were analyzed for binding to TF. Antibodies were detected with HRP-conjugated anti-human IgG was used for detection. Curve fitting of the data was performed using a 4-parameter equation with the Solver function in Microsoft Excel. Positive control anti-TF IgG 3ElOx: IC5O = 2.0 nM (filled diamond, solid line); Linker 1 (SEQ ID NO: 1) IgG-like bispecific antibody: IC50 = 0.78 nM (filled triangle, large dashed line); Linker 2 (SEQ ID NO: 2) IgG-like bispecific antibody: IC50 = 0.93 nM (open triangle, small dashed line); Linker 3 (SEQ ID NO: 3) IgG-like bispecific antibody: IC5O = 1.06 nM (filled square, alternating small and large dashed line); Linker 4 (SEQ ID NO: 4) IgG-like bispecific antibody: IC50 = 1.01 nM (open square, two large and one small dashed line); and negative control anti-RGl IgG 19G9: no binding (open diamond, solid line).
- A - [110] Figure 11. RGl -binding ELISA. A bispecific antibody containing Linker 1, anti-RGl antibody 19G9, and polyclonal nonimmune control human IgG kappa were analyzed for binding to RG-I . Curve fitting of the data was performed using a 4-parameter equation with the Solver function in Microsoft Excel. Positive control anti-RGl IgG 19G9: IC50 = 1.4 nM (filled diamond, solid line); Linker 1 (SEQ ID NO: 1) IgG-like bispecific antibody: IC50 = 1.1 nM (filled triangle, large dashed line); and negative control nonimmune polyclonal human IgG kappa: no binding (open triangle, small dashed line).
[111] Figure 12. Measurement of the antigen-binding activity of a bispecific protease-regulated antibody using a sandwich antigen-binding ELISA. The linker of this antibody contained cleavage sites for enterokinase.
[112] Figure 13. Measurement of the antigen-binding activity of protease-regulated antibodies 3E10-Typel-Fab and 19G9-Typel-Fab. The controls are designated 3El O-Reg-Fab, 19G9-Reg- Fab, and HuFab.
[113] Figure 14. Measurement of the antigen-binding activity of Fab-like protease-regulated antibodies HlLl, H1L4, H1L7, H4L7, and H5L5 (Type 2) in the absence and presence of enterokinase. Parental antibodies 3E10 and 19G9, and polyclonal human Fab were used as control.
[114] Figure 15. Measurement of the antigen-binding activity of Fab-like protease-regulated antibodies H2L1, H2L2, and H2L8 (Type 3) and H3L1, H3L4, and H5L4 (Type 4) in the absence and presence of enterokinase. Parental antibodies 3E10 and 19G9, and polyclonal human Fab were used as control.
[115] Figure 16. Western blots of protease-regulated antibody 3E 10-Type 1 -Fab detected with anti-Myc antibody (A) or anti-kappa chain antibody (B). Lane 1 and 2: 3E10-Typel-Fab without or with enterokinase digestion, respectively. Lane 3 and 4: 3E10-Reg-Fab without or with enterokinase digestion, respectively.
[116] Figure 17. Western blots of Fab-like protease-regulated antibodies HlLl, H 1L7, and H5L5 (Type 2) in the absence and presence of enterokinase. Antibodies were detected with anti- IgG(H+L) antibody. Lane 1 and 2: HlLl without or with enterokinase digestion, respectively. Lane 3 and 4: H1L7 without or with enterokinase digestion, respectively. Lane 5 and 6: H5L5 without or with enterokinase digestion, respectively. Lane 7: 3E10-Reg-Fab.
[117] Figure 18. Western blots of Fab-like protease-regulated antibodies H2L2 and H2L8 (Type 3) and H3L4 (Type 4) in the absence and presence of enterokinase. Antibodies were detected with anti-Myc antibody. Lane 1 and 2: H2L2 without or with enterokinase digestion, respectively. Lane 3 and 4: H2L8 without or with enterokinase digestion, respectively. Lane 5 and 6: H3L4 without or with enterokinase digestion, respectively.
DESCRIPTION OF THE INVENTION
[118] It is to be understood that this invention is not limited to the particular methodology, protocols, cell lines, animal species or genera, constructs, and reagents described and as such may vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to limit the scope of the present invention which will be limited only by the appended claims.
[119] It must be noted that as used herein and in the appended claims, the singular forms "a," "and," and "the" include plural reference unless the context clearly dictates otherwise. Thus, for example, reference to "an antibody" is a reference to one or more antibodies and includes equivalents thereof known to those skilled in the art, and so forth.
[120] Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood to one of ordinary skill in the art to which this invention belongs. Although any methods, devices, and materials similar or equivalent to those described herein can be used in the practice or testing of the invention, the preferred methods, devices and materials are now described.
[121] All publications and patents mentioned herein are hereby incorporated herein by reference for the purpose of describing and disclosing, for example, the constructs and methodologies that are described in the publications which might be used in connection with the presently described invention. The publications discussed above and throughout the text are provided solely for their disclosure prior to the filing date of the present application. Nothing herein is to be construed as an admission that the inventors are not entitled to antedate such disclosure by virtue of prior invention.
[122] For convenience, the meaning of certain terms and phrases employed in the specification, examples, and appended claims are provided below.
[123] "Antibody" as used herein includes intact immunoglobulin molecules (e.g., IgGl, IgG2a, IgG2b, IgG3, IgM, IgD, IgE, IgA), as well as fragments thereof, such as Fab, F(ab')2, scFv, Fv, and diabody which are capable of specific binding to an epitope of a protein. The term antibody also extends to other protein scaffolds that are able to orient antibody complementarity- determining region (CDR) inserts into the same active binding conformation as that found in natural antibodies such that the binding to the target antigen observed with these chimeric proteins is maintained relative to the binding activity of the natural antibody from which the CDRs were derived.
[124] "Antibody fragments" comprise a portion of a full length antibody, generally the antigen binding or variable domain thereof. Examples of antibody fragments include Fab, Fab', F(ab')2> and Fv fragments; diabodies; linear antibodies; single-chain antibody molecules; monospecific antibodies; bispecific antibodies; and multispecifϊc antibodies formed from antibody fragments.
[125] The term "autoimmune diseases" includes, but is not limited to, multiple sclerosis, rheumatoid arthritis, lupus, type I diabetes mellitus, Crohn's disease, autoimmune hemolytic anemia, autoimmune hepatitis, glomerulonephritis, inflammatory bowel disease, myocarditis, psoriasis, thyroiditis, ulcerative colitis, and Graves'disease.
[126] The terms "biological sample" or "patient sample" as used herein, refers to a sample obtained from an organism or from components (e.g., cells) of an organism. The sample may be of any biological tissue or fluid. The sample may be a "clinical sample" which is a sample derived from a patient. Such samples include, but are not limited to, sputum, blood, serum, plasma, blood cells (e.g., white cells), tissue samples, biopsy samples, urine, peritoneal fluid, and pleural fluid, saliva, semen, breast exudate, cerebrospinal fluid, tears, mucous, lymph, cytosols, ascites, amniotic fluid, bladder washes, and bronchioalveolar lavages or cells therefrom, among other body fluid samples. The patient samples may be fresh or frozen, and may be treated with heparin, citrate, or EDTA. Biological samples may also include sections of tissues such as frozen sections taken for histological purposes.
[127] The term "cancer" includes, but is not limited to, solid tumors, such as cancers of the breast, respiratory tract, brain, reproductive organs, digestive tract, urinary tract, eye, liver, skin, head and neck, thyroid, parathyroid, and their distant metastases. The term also includes lymphomas, sarcomas, and leukemias.
[128] The term " conjugate" refers to an antibody chemically linked to a chemical moiety, such as a therapeutic or cytotoxic agent.
[129] The term "infectious diseases" includes, but is not limited to, HIV/ AIDS, lower respiratory infections, diarrheal diseases, tuberculosis, malaria, measles, pertussis, tetanus, meningitis, syphilis, hepatitis B and tropical diseases.
[130] The term "linker" refers to a peptide (or polypeptide) comprising two or more amino acid residues joined by peptide bonds and used to link one or more antibody domains. The linker may contain one or more protease cleavage sites. [131] The term "protease" refers to any enzyme, including the endopeptidases and exopeptidases, which catalyze the hydrolytic breakdown of proteins into peptides or amino acids.
[132] The present invention is directed to the design and production of monospecific, bispecific antibodies, and multispecifϊc antibodies. For example, the bispecific antibodies and multispecific antibodies may comprise tandem linked VHa-VHb-VHC...CH in one polypeptide and VLa-VLb- VLc...CL in another polypeptide. Alternately, the VH and VL domains may be exchanged from one polypeptide to another to create polypeptides such as VHa-VLb-VHC...CH and VLa-VHb-VLc...CL. The two polypeptides may form Dimmers in the Fab format or the half IgG-like format, or two of each polypeptide may form a four polypeptide-containing homodimer of the IgG-like format. These bispecific or multispecific antibodies or antibody fragments thereof may simultaneously bind different antigens or different epitopes of the same antigen.
[133] As an example, a recombinant IgG-like bispecific antibody may be constructed by the tandem linking of two different VH domains of a heavy chain and two different VL domains of a light chain. The construct is exemplified as follows: heavy chain = NH2-Vn 1 -VH2-CH1-CH2-CH3 -COOH light chain = NH2-VL1-VL2-CL.
[134] Another bispecific antibody may comprise the following: heavy chain = NH2-VL1-VH2-CH1 -CH2-CH3-COOH light chain = NH2-VH1 -V^-CL-COOH.
[135] The present invention also relates to protease-regulated antibodies. Protease-regulated antibodies may be, for example, monospecific antibodies, bispecific antibodies, multispecific antibodies, or antibodies with sequential binding-activity upon protease digestion in either, for example, Fab-like or IgG-like format. Protease control or regulation may be provided by a selective protease site located in, for example, a linker. These protease-regulated antibodies may be utilized for the diagnosis and treatment of various diseases including but not limited to cancer, infectious disease, and autoimmune diseases, and provide an additional level of control for biological drugs for therapeutic or diagnostic applications.
[136] Protease-regulated antibodies may comprise a heavy chain (H) variable domain (VH)- linker-heavy chain constant domain (CH) in one polypeptide and a light chain (L) variable domain (VL)-linker-light chain constant domain (CL) in another polypeptide. Bispecific protease- regulated antibodies may comprise, for example, Vm-linker-V^-CH in one polypeptide and VLr linker- VL2-CL in another polypeptide, both regulated by proteolytic cleavage of the linker. Alternately, the VH and VL domains in the bispecific protease-regulated antibodies may be exchanged from one polypeptide to another polypeptide to create polypeptides such as, for example, Vm-linker-V^-CH and VLi-linker-VH2-CL. The two polypeptides may form Dimmers, for example, in a Fab-like format, a half-IgG-like format, or an IgG-like format (e.g., two of each polypeptide forming a four polypeptide-containing homodimer). The bispecific and sequential protease-regulated antibodies or antibody fragments may (1) simultaneously bind two different antigens or different epitopes of the same antigen, (2) sequentially bind two different antigens or different epitopes on the same antigen in a manner that may be dependent on the length, adjacent sequence, and design of the linker, or (3) a monospecific protease-activated binder which is in latent or prodrug form prior to protease digestion and which is switched on by protease cleavage. Libraries of bispecific protease-regulated antibodies in the Fab-like format can be readily created, expressed in bacteria, and screened for specific functionalities, including susceptibility of the linker to cleavage by a specific protease and optimization of this cleavage step.
[137] Several types of protease-regulated antibodies are described herein whereby antibody formats are designed with selectivity due to specific protease-dependent binding or protease- specific functionality. In particular, these protease-regulated antibodies may be described by deletions and/or additions of antibody framework, location of the linker, and its properties including length and solvent accessibility. Furthermore, the linker may contain a cleavage site specific for a protease found in a target cell or tissue. One example of a protease-regulated antibody may contain a protease site in a linker located between the variable domain and the constant region domain and this antibody may bind only one antigen as illustrated in Figure 1 ("Type 1").
[138] Another example of a protease-regulated antibody may simultaneously bind two different antigens or two different epitopes as shown in Figures 2 and 3 ("Type 2") in the absence of a protease. The first VH/VL domains of this antibody bind to an antigen without blocking the second VH/VL domains from binding to a second antigen. This antibody can bind to antigens without steric blocking of the CDR regions of the second variable domains. The simultaneous binding of this protease-regulated antibody to both antigens is prevented by proteolytic cleavage. That is, when the linker is cleaved by a protease, the antibody can only bind to the second antigen or separately bind two antigens. Simultaneous antigen binding is important for antibody function, for example, in cross-linking receptors, which may be prevented by proteolytic cleavage. Thus, an additional degree of specificity is added by including a protease site in the linker.
[139] This bispecific protease-regulated antibody is more selective than a monospecific antibody because this antibody will specifically target cells or tissues expressing both antigens. The additional degree of specificity is provided by the specific protease site in the linker. In Figure 4, Cell A and Cell B express both Antigen 1 and Antigen 2, however only Cell B expresses the selective protease. The bispecifϊc protease-regulated antibody with the uncleavable linker (i.e., this linker does not contain the protease cleavage site) will bind to Cell A with greater avidity because the bispecific antibody is able to bind to both antigens. In contrast, the bispecifϊc protease-regulated antibody will bind to Cell B with lower avidity because the Antigen 1 binding domain is removed by proteolytic cleavage of the linker by the selective protease expressed by Cell B (alternately, the selective protease may be expressed by adjacent cells localized in the same tissue as Cell B).
[140] In contrast, Figure 5 illustrates a protease-regulated antibody that may sequentially bind to each antigen in a protease-dependent manner. That is, prior to protease cleavage of the linker, the protease-regulated antibody binds to a first antigen and following protease cleavage, the antibody binds to a second antigen ("Type 3"). The VH/VL domains of the N-terminal antibody bind to an antigen, but block the CDR regions of the downstream VH/VL domains from binding to a second antigen. Protease cleavage of the linker allows removal of the N-terminal antibody, and removing the N-terminal antibody domains then permits binding to a second antigen. This allows for greater cell and/or tissue selectivity by requiring sequential binding.
[141] In Figure 6, Cell A and Cell B express both Antigen 1 and Antigen 2, but only Cell B expresses the selective protease. In addition, Antigen 2 is a cell surface receptor that internalizes into the cell and allows internalization of antibodies that bind to it. The protease-regulated antibody will bind to Antigen 1 expressed by Cell A and Cell B. However, only Cell B expresses the selective protease (or possibly cells adjacent to Cell B in the same tissue). The protease- regulated antibody will be activated by proteolytic cleavage and internalized via Antigen 2 expressed on Cell B. Thus, this protease-regulated antibody will be specifically internalized by cells expressing Antigen 1 , Antigen 2, and the selective protease.
[142] In an additional example, a protease-regulated antibody may not bind to an antigen before protease digestion, but may bind to antigen following protease digestion ("Type 4"). An example of this antibody is illustrated in Figure 7. This monospecific protease-regulated antibody also contains a protease cleavage linker that allows removal of the N-terminal non-functional antibody which then leads to binding to an antigen by the functional antibody domains that are thus exposed. Type 4 protease-regulated antibodies may be created by three approaches. In the first approach the protease cleavable linker sequence is modified so that it prevents the N-terminal VH and VL domains of a Type HI antibody (VH1 and VL1) from binding to the first antigen. Examples of these linkers are shown in the sequences in Table 8. In the second approach, the linkers utilized in Type III antibodies shown in Tables 6 and 7 are now combined with heterodimeric N-terminal VH and VL domains that have been mutated to destroy their antigen binding function. Examples of this approach are shown in the sequences in Table 9 in which the CDR3 of VH1 and CDR3 of VL1
- IO - are replaced by a poly-alanine sequence of a similar length as the respective CDR. In the third approach, the linkers utilized in Type III antibodies shown in Tables 6 and 7 are combined with homodimeric N-terminal domains derived from the constant regions of antibodies. For example, the complete VH1 and VL1 domains of a Type III antibody are both replaced by the same constant domain that is capable of heterodimerization, for example, the CH3 domain of IgG or the CH4 domain of IgE.
[143] These protease-regulated antibodies may be modified by protease cleavage of the linker as described below. For example, the protease-regulated antibody illustrated in Figure 1 (Type 1) contains a protease site in the linker between the antigen binding domains and the Fc domain. This antibody will specifically target cells or tissues that present the antigen. When the linker is cleaved by the protease, the resultant protease-regulated antibody releases the functional Fc portion. In tissues where the protease is present, this antibody will release the Fc portion which is essential to antigen crosslinking, and induce an immune response such as antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC). To illustrate, hepsin, a serine protease, is expressed in both tumor tissue and normal liver tissue. In a cancer patient treated with a protease-regulated antibody against hepsin, the antibody would attack the tumor cells via the Fc portion-induced ADCC and CDC. However, in the liver, the protease- regulated antibody would initially bind to hepsin, but the Fc portion would be cleaved by a liver- specific protease prior to initiation of ADCC or CDC preventing liver toxicity.
[144] The peptide (or polypeptide) linker of the protease-regulated antibody may comprise two or more amino acid residues and may contain one or more protease cleavage sites. The linkers may alter antibody conformation, stability, and antigen-binding activities. The length of linkers may range, for example, from 0 to about 100 amino acid residues. The following are examples of linkers:
Linker 1: SDDDDK (SEQ ID NO: 1) Linker 2: GGGGSDDDDK (SEQ ID NO: 2) Linker 3: GGGGSDDDDKGGGGS (SEQ ID NO: 3) Linker 4: GGGGSGGGGSGGGGS (SEQ ID NO: 4) Linker 5: fflPVLSGLSRTVNGEDAVPG (SEQ ID NO: 5) Linker 6: VAAPFDDDDKTVGGYICEEN (SEQ ID NO: 6) Linker 7: ELLESYIDGRIVEGSDAEIG (SEQ ID NO: 7) Linker 8: STQSFNDFTRWGGEDAKPG (SEQ ID NO: 8) Linker 9: PERGDNNLTRIVGGQECKDG (SEQ ID NO: 9) Linker 10: EDQEDQVDPRLIDGKMTRRG (SEQ ID NO: 10) Linker 11 : KRNASKPQGRIVGGKVCPKG (SEQ ID NO: 11) Linker 12: SVCTTKTSTRIVGGTNSSWG (SEQ ID NO: 12)
Linker 13: SRIVG (SEQ ID NO: 13)
Linker 14: GSLVSGSCSQIINGEDCSPH (SEQ ED NO: 14)
Linker 15: SRIIN (SEQ ID NO: 15)
Linker 16: NKLVH (SEQ ID NO: 16)
Linker 17: DKIID (SEQ ID NO: 17)
Linker 18: FNVLG (SEQ ID NO: 18)
Linker 19: TRAIG (SEQ ID NO: 19)
Linker 20: TRLDP (SEQ ID NO: 20)
Linker 21 : TRIDC (SEQ ID NO: 21)
Linker 22: SGSNQ (SEQ ID NO: 22)
Linker 23: SKVLN (SEQ ID NO: 23)
Linker 24: NKJIG (SEQ ID NO: 24)
Linker 25: DKLLE (SEQ ID NO: 25)
Table 1 illustrates the excision site of several proteases.
TABLE l
Figure imgf000013_0001
[146] The cleavage sites of additional proteases that may be incorporated in a linker are described in Table 2.
TABLE 2
TUMOR ASSOCIATED PROTEASES (Extracellular Or Intracellular)
ADAM metallopeptidase domain 9 (meltrin gamma)
ADAM metallopeptidase domain 10
ADAM metallopeptidase domain 17 (TNFalpha, converting enzyme)
ADAM metallopeptidase domain 28
ADAM-like, decysin 1
ADAM metallopeptidase, thrombospondin type 1 motif 1
ADAM metallopeptidase, thrombospondin type 1 motif 5, aggrecanase-2
ADAMTS-like 3
ADAMTS-like 4
Beta-site APP-cleaving enzyme 1
Bleomycin hydrolase
Bone morphogenetic protein 1
Complement component 1, r subcomponent
Complement component 1, s subcomponent
Calpain 2, (m/II) large subunit
Caspase 1, apoptosis-related cysteine peptidase (IL- lβ convertase) Caspase 2, apoptosis-related cysteine peptidase
Caspase 3, apoptosis-related cysteine peptidase
Caspase 4, apoptosis-related cysteine peptidase
Caspase 6, apoptosis-related cysteine peptidase
Caspase 7, apoptosis-related cysteine peptidase
Caspase 9, apoptosis-related cysteine peptidase
Complement factor D (adipsin)
CASP8 and FADD-like apoptosis regulator
Cathepsin B
Cathepsin F
Cathepsin H
Cathepsin K
Cathepsin L
Cathepsin L2
Cathepsin O
Cathepsin S
Cylindromatosis (turban tumor syndrome)
Extra spindle pole bodies homolog 1 (S. Cerevisiae)
Granzyme A (granzyme 1 , CTL-associated serine esterase 3)
Histocompatibility (minor) 13
Hepsin (transmembrane protease, serine 1)
Figure imgf000015_0001
[147] The protease-regulated antibodies of the present invention may bind one or more antigens. These antigens may be selected from the group consisting of cytokines, cell surface receptors, enzymes, and receptors. These antigens include, but are not limited to, CD3, CD4, CD8, CD20, CD25, CD28, CD33, CD52, IL-2, IL-7, IL-8, TNF-alpha, TGF-beta INF-beta, INF-gamma, GMCSF, GCSF, VEGF, C5, EpCAM, EGF receptor, CD2 receptor, IL 2 receptor, IgE receptor, intergrin, and MHC class π.
[148] The antibodies of the present invention may be utilized for the diagnosis and therapy of various diseases. For example, antibodies directed against human immunological cells and tumor- associated antigen may be used for cancer therapy. These antibodies may also be directed against tumor-associated antigen and toxic agents or enzymes for use as a cancer therapeutic. The antibodies of the present invention may also be utilized for the treatment of hemophilia and thrombosis as well as stem cell transplantation. These antibodies may be used for the selective stimulation and expansion of lymphocyte subset, hi addition, these antibodies may used for the detection of disease-related antigens.
[149] For cancer immunotherapy, bispecifϊc antibodies may be used to recruit the immune system to attach tumor cells. Targets on immunological cells include, but are not limited to, CD3, CD8, and Fc receptor. Tumor-associated antigens include, but are not limited to, Her2, EGF receptor, CD20, CA-125, and carcinoembryonic antigen (CEA). For example, a bispecifϊc antibody against CD8 and Her2 can direct CD8-expressing cytotoxic lymphocytes to attack Her2 expressing breast cancer cells.
[150] The antibodies or antibody fragments of the invention, or compositions including the antibodies or fragments, can include a cytoxic agent that is conjugated to the antibody or fragment. In one aspect, the cytotoxic agent is monomethylauristatin-E (MMAE), however, other cytoxic agents are also provided, which can include, for example, functional analogs of MMAE (e.g. monomethylauristatin-F), and other cytotoxic agents, e.g., aplidin, azaribine, anastrozole, azacytidine, bleomycin, bortezomib, bryostatin-1, busulfan, calicheamycin, camptothecin, 10- hydroxycamptothecin, carmustine, celebrex, chlorambucil, cisplatin, irinotecan (CPT-I 1), SN-38, carboplatin, cladribine, cyclophosphamide, cytarabine, dacarbazine, docetaxel, dactiπomycin, daunomycin glucuronide, daunorubicin, dexamethasone, diethylstilbestrol, doxorubicin, doxorubicin glucuronide, epirubicin glucuronide, ethinyl estradiol, estramustine, etoposide, etoposide glucuronide, etoposide phosphate, floxuridine (FUdR), 3',5'-0-dioleoyl-FudR (FUdR- dθ), fludarabine, flutamide, fluorouracil, fluoxymesterone, gemcitabine, hydroxyprogesterone caproate, hydroxyurea, idarubicin, ifosfamide, L-asparaginase, leucovorin, lomustine, mechlorethamine, medroprogesterone acetate, megestrol acetate, melphalan, mercaptopurine, 6- mercaptopurine, methotrexate, mitoxantrone, mithramycin, mitomycin, mitotane, phenyl butyrate, prednisone, procarbazine, paclitaxel, pentostatin, PSI-341, semustine streptozocin, tamoxifen, taxanes, taxol, testosterone propionate, thalidomide, thioguanine, thiotepa, teniposide, topotecan, uracil mustard, velcade, vinblastine, vinorelbine, vincristine, ricin, abrin, ribomiclease, onconase, rapLRl, DNase I, Staphylococcal enterotoxin-A, pokeweed antiviral protein, gelonin, diphtheria toxin, Pseudomonas exotoxin, and Pseudomonas endotoxin, or combinations thereof. Any of the cytoxic agents can also include functional analogs thereof.
Antibody Technology
[151] A number of technologies are available to produce antibodies. For example, phage- antibody technology may be used to generate antibodies (Knappik, et al., J. MoI. Biol. 296:57-86, 2000). Another approach for obtaining antibodies is to screen a DNA library from B cells as described by Dower, et al., (WO 91/17271) and McCafferty, et al., (WO 92/01047). In these methods, libraries of phage are produced in which members display different antibodies on their outer surfaces. Antibodies are usually displayed as Fv or Fab fragments. Phage displaying antibodies are selected by affinity enrichment for binding to a selected protein. Antibodies may also be produced using trioma methodology (Oestberg, et al., Hybridoma 2:361-367, 1983; U.S. Patent No. 4,634,664; U.S. Patent No. 4,634,666).
[152] Antibodies may also be purified from any cell that expresses the antibodies, including host cells that have been transfected with antibody-encoding expression constructs. The host cells may be cultured under conditions whereby the antibodies are expressed. Purified antibody may be separated from other cellular components that may associate with the antibody in the cell, such as certain proteins, carbohydrates, or lipids using methods well known in the art. Such methods include, but are not limited to, size exclusion chromatography, ammonium sulfate fractionation, ion exchange chromatography, affinity chromatography, and preparative gel electrophoresis. Purity of the preparations may be assessed by any means known in the art, such as SDS- polyacrylamide gel electrophoresis. A preparation of purified antibodies may contain more than one type of antibody.
[153] Alternatively, antibodies may be produced using chemical methods to synthesize its amino acid sequence, such as by direct peptide synthesis using solid-phase techniques (see, e.g., Merrifield, J. Am. Chem. Soc. 85:2149-2154, 1963; Roberge, et al., Science 269:202-204, 1995). Protein synthesis may be performed using manual techniques or by automation. Automated synthesis may be achieved, for example, using Applied Biosystems 43 IA Peptide Synthesizer (Perkin Elmer). Optionally, fragments of antibodies may be separately synthesized and combined using chemical methods to produce a full-length molecule. [154] The antibodies of the present invention may be generated from parental antibodies. Parent antibodies may be selected from various antibodies capable of binding specific targets and well known in the art, such as, but not limited to, but are not limited to anti-TNF antibody, anti-IL-12 antibody; anti-IL-18 antibody, anti-C5, anti-CD147, anti-gpl20, anti-CDl la, anti-CD18, anti- VEGF, anti-CD40L, anti-ICAM-1, anti-CD2, anti-EGFR, anti-TGF-beta 2, anti-E-selectin, anti- Her2/neu, anti-CD14, anti-ICAM-3, anti-CD80, anti-CD4, anti-CD3, anti-CD23, anti-beta2- integrin, , anti-CD52, , anti-CD22, anti-CD20, anti-CD25, anti-CD33, anti-HLA, anti-IL-1 alpha, anti-IL-1, anti-IL-1 receptor, anti-IL-2 receptor, anti-IL-4, anti-IL4 receptor, anti-IL5, anti-IL-5 receptor, anti-IL-6, anti-IL-8, anti-IL-9, anti-IL-13, anti-IL-13 receptor, anti-IL-17, and anti-IL-23. Parent antibodies may also be selected from various therapeutic antibodies including, but are not limited to, rituximab, trastuzumab, pertuzumab, cetuximab, alemtuzumab, muromonab, ibritumomab, gemtuzumab ozogamicin, alefacept, abciximab, basiliximab, palivizumab, infliximab, adalimumab, etanercept, natalizumab, bevacizumab, omalizumab, efalizumab, clenoliximab, labetuzumab, epratuzumab, and visilizumab.
[155] The newly synthesized molecules may be substantially purified by preparative high performance liquid chromatography {see, e.g., Creighton, Proteins: Structures and Molecular Principles. WH Freeman and Co., New York, N.Y., 1983). The composition of a synthetic polypeptide may be confirmed by amino acid analysis or sequencing (e.g., using Edman degradation).
[156] The present invention also relates to bispecific or bifunctional antibodies that have one binding site that specifically binds to a first antigen and a second binding site that specifically binds to a second antigen. This results in multi-functional valency, that is, an ability to bind at least two different epitopes simultaneously.
Polynucleotides encoding antibodies
[157] The present invention also relates to polynucleotides encoding antibodies. These polynucleotides may be used, for example, to produce quantities of the antibodies for therapeutic or diagnostic use.
[158] Polynucleotides of the present invention may also be isolated from host cells, free of other cellular components such as membrane components, proteins, and lipids. Polynucleotides may be isolated using standard nucleic acid purification techniques, or synthesized using an amplification technique such as the polymerase chain reaction (PCR), or by using an automatic synthesizer. Methods for isolating polynucleotides are routine and are known in the art. Any such technique for obtaining a polynucleotide may be used to obtain isolated polynucleotides encoding antibodies of the invention. For example, restriction enzymes and probes may be used to isolate polynucleotides which encode antibodies.
[159] Antibody-encoding cDNA molecules may be made with standard molecular biology techniques, using mRNA as a template. Thereafter, cDNA molecules may be replicated using molecular biology techniques known in the art and disclosed in manuals such as Sambrook, et al., (Molecular Cloning: A Laboratory Manual, (Second Edition, Cold Spring Harbor Laboratory Press; Cold Spring Harbor, N. Y.; 1989) Vol. 1-3). An amplification technique, such as PCR, may be used to obtain additional copies of the polynucleotides. Alternatively, synthetic chemistry techniques may be used to synthesize polynucleotides encoding antibodies of the invention.
[160] To express a polynucleotide encoding an antibody, the polynucleotide may be inserted into an expression vector that contains the necessary elements for the transcription and translation of the inserted coding sequence. Methods that are well known to those skilled in the art may be used to construct expression vectors containing sequences encoding antibodies and appropriate transcriptional and translational control elements. These methods include in vitro recombinant DNA techniques, synthetic techniques, and in vivo genetic recombination. Such techniques are described, for example, in Sambrook, et al. (1989) and in Ausubel, et al., (Current Protocols in Molecular Biology, John Wiley & Sons, New York, N. Y., 1995).
[161] A variety of expression vector/host systems may be utilized to contain and express sequences encoding antibodies. These include, but are not limited to, microorganisms, such as bacteria transformed with recombinant bacteriophage, plasmid, or cosmid DNA expression vectors; yeast transformed with yeast expression vectors; insect cell systems infected with virus expression vectors (e.g., baculovirus); plant cell systems transformed with virus expression vectors (e.g., cauliflower mosaic virus, CaMV; tobacco mosaic virus, TMV); or bacterial expression vectors (e.g., Ti or pBR322 plasmids), or animal cell systems.
[162] The control elements or regulatory sequences are those non-translated regions of the vector - enhancers, promoters, 5' and 3' untranslated regions - which interact with host cellular proteins to carry out transcription and translation. Such elements may vary in strength and specificity. Depending on the vector system and host utilized, any number of suitable transcription and translation elements, including constitutive and inducible promoters, may be used. For example, when cloning in bacterial systems, inducible promoters can be used. The baculovirus polyhedrin promoter may be used in insect cells. Promoters or enhancers derived from the genomes of plant cells (e.g., heat shock, RUBISCO, and storage protein genes) or from plant viruses (e.g., viral promoters or leader sequences) may be cloned into the vector. In mammalian cell systems, promoters from mammalian genes or from mammalian viruses may be used. If it is necessary to generate a cell line that contains multiple copies of a nucleotide sequence encoding an antibody, vectors based on SV40 or EBV may be used with an appropriate selectable marker.
[163] General texts describing additional molecular biological techniques useful herein, including the preparation of antibodies include Berger and Kimmel (Guide to Molecular Cloning Techniques. Methods in Enzvmology, Vol. 152, Academic Press, Inc.); Sambrook, et al., (Molecular Cloning: A Laboratory Manual. (Second Edition, Cold Spring Harbor Laboratory Press; Cold Spring Harbor, N.Y.; 1989) Vol. 1-3); Current Protocols in Molecular Biology. (F. M. Ausabel et al. [Eds.], Current Protocols, a joint venture between Green Publishing Associates, Inc. and John Wiley & Sons, Inc. (supplemented through 2000)); Harlow et al., (Monoclonal Antibodies: A Laboratory Manual. Cold Spring Harbor Laboratory Press (1988), Paul [Ed.]); Fundamental Immunology. (Lippincott Williams & Wilkins (1998)); and Harlow, et al., (Using Antibodies: A Laboratory Manual. Cold Spring Harbor Laboratory Press (1998)).
Assays
[164] The affinity (Kj) of antibody binding to an antigen may be assayed using any method known in the art including, for example, immunoassays such as enzyme-linked immunospecifϊc assay (ELISA), Bimolecular Interaction Analysis (BIA) (see, e.g., Sjolander and Urbaniczky, Anal. Chem. 63:2338-2345, 1991; Szabo, et al., Curr. Opin. Struct. Biol. 5:699-705, 1995), and fluorescence-activated cell sorting (FACS) for quantification of antibody binding to cells that express an antigen. BIA is a technology for analyzing biospecific interactions in real time, without labeling any of the interactants (e.g., BIAcore™). Changes in the optical phenomenon surface plasmon resonance (SPR) may be used as an indication of real-time reactions between biological molecules.
[165] The present invention also relates to the use of quantitative immunoassays to measure levels of proteins in patient samples. Many formats may be adapted for use with the methods of the present invention. For example, the detection and quantitation of a protein in patient samples may be performed, by enzyme-linked immunosorbent assays, radioimmunoassays, dual antibody sandwich assays, agglutination assays, fluorescent immunoassays, immunoelectron and scanning microscopy, among other assays commonly known in the art. The quantitation of a protein in such assays may be adapted by conventional methods known in the art. Serial changes in circulating a protein levels may be detected and quantified by a sandwich assay in which the capture antibody has been immobilized using conventional techniques on the surface of the support.
[166] Suitable supports include, for example, synthetic polymer supports, such as polypropylene, polystyrene, substituted polystyrene, polyacrylamides (such as polyamides and polyvinylchloride), glass beads, agarose, and nitrocellulose. [167] The antibodies useful to identify proteins may be labeled in any conventional manner. An example of a label is horseradish peroxidase, and an example of a method of labeling antibodies is by using biotin-strepavidin complexes.
[168] As appropriate, antibodies used in the immunoassays of this invention that are used as tracers may be labeled in any manner, directly or indirectly, that results in a signal that is visible or can be rendered visible. Detectable marker substances include radionuclides, such as 3H, 125I, and 131I; fluorescers, such as, fluorescein isothiocyanate and other fluorochromes, phycobiliproteins, phycoerythin, rare earth chelates, Texas red, dansyl and rhodamine; colorimetric reagents (chromogens); electron-opaque materials, such as colloidal gold; bioluminescers; chemiluminescers; dyes; enzymes, such as, horseradish peroxidase, alkaline phosphatases, glucose oxidase, glucose-6-phosphate dehydrogenase, acetylcholinesterase, alpha -, beta-galactosidase, among others; coenzymes; enzyme substrates; enzyme cofactors; enzyme inhibitors; enzyme subunits; metal ions; free radicals; or any other immunologically active or inert substance which provides a means of detecting or measuring the presence or amount of immunocomplex formed. Exemplary of enzyme substrate combinations are horseradish peroxidase and tetramethyl benzidine (TMB), and alkaline phosphatases and paranitrophenyl phosphate (pNPP).
[169] Another detection and quantitation systems according to this invention produce luminescent signals, bioluminescent (BL) or chemiluminescent (CL). In chemiluminescent (CL) or bioluminescent (BL) assays, the intensity or the total light emission is measured and related to the concentration of the unknown analyte. Light can be measured quantitatively using a luminometer (photomultiplier tube as the detector) or charge-coupled device, or qualitatively by means of photographic or X-ray film. The main advantages of using such assays is their simplicity and analytical sensitivity, enabling the detection and/or quantitation of very small amounts of analyte.
[170] Exemplary luminescent labels are acridinium esters, acridinium sulfonyl carboxamides, luminol, umbelliferone, isoluminol derivatives, photoproteins, such as aequorin, and luciferases from fireflies, marine bacteria, Vargulla and Renilla. Luminol can be used optionally with an enhancer molecule such as 4-iodophenol or 4-hydroxy-cinnamic acid. Typically, a CL signal is generated by treatment with an oxidant under basic conditions.
[171] Additional luminescent detection systems are those wherein the signal (detectable marker) is produced by an enzymatic reaction upon a substrate. CL and BL detection schemes have been developed for assaying alkaline phosphatases (AP), glucose oxidase, glucose 6-phosphate dehydrogenase, horseradish peroxidase (HRP), and xanthine-oxidase labels, among others. AP and HRP are two enzyme labels which can be quantitated by a range of CL and BL reactions. For example, AP can be used with a substrate, such as an adamantyl 1,2-dioxetane aryl phosphate substrate (e.g. AMPPD or CSPD; Kricka, L.J., "Chemiluminescence and Bioluminescence, Analysis by," Molecular Biology and Biotechnology: A Comprehensive Desk Reference (ed. R.A. Meyers) (VCH Publishers; N.Y., N.Y.; 1995)); for example, a disodium salt of 4-methoxy-4-(3-phosphatephenyl) spiro [l,2-dioxetane-3,2'-adamantane], with or without an enhancer molecule such as l-(trioctylphosphonium methyl)-4- (tributylphosphonium methyl) benzene diochloride. HRP is may be used with substrates, such as, 2',3',6'-trifluorophenyl- methoxy- 10-methylacridan-9-carboxylate.
[172] CL and BL reactions may be adapted for analysis not only of enzymes, but also of other substrates, cofactors, inhibitors, metal ions, and the like. For example, luminol, firefly luciferase, and marine bacterial luciferase reactions are indicator reactions for the production or consumption of peroxide, ATP, and NADPH, respectively. They may be coupled to other reactions involving oxidases, kinases, and dehydrogenases, and may be used to measure any component of the coupled reaction (enzyme, substrate, cofactor).
[173] The detectable marker may be directly or indirectly linked to an antibody used in an assay of this invention. Exemplary of an indirect linkage of the detectable label is the use of a binding pair between an antibody and a marker or the use of a signal amplification system.
[174] Examples of binding pairs that may be used to link antibodies to detectable markers are biotin/avidin, streptavidin, or anti-biotin; avidin/anti-avidin; thyroxine/thyroxine-binding globulin; antigen/antibody; antibody/ anti-antibody; carbohydrate/lectins; hapten/anti-hapten antibody; dyes and hydrophobic molecules/hydrophobic protein binding sites; enzyme inhibitor, coenzyme or cofactor/enzyme; polynucleic acid/homologous polynucleic acid sequence; fluorescein/anti- fluorescein; dinitrophenol/anti-dinitrophenol; vitamin Bl 2/intrinsic factor; cortisone, cortisol/cortisol binding protein; and ligands for specific receptor protein/membrane associated specific receptor proteins.
[175] Various means for linking labels directly or indirectly to antibodies are known in the art. For example, labels may be bound either covalently or non-covalently. Exemplary antibody conjugation methods are described in Avarmeas, et al., Scan. J. Immunol. 8(Suppl. 7): 7, 1978); Bayer, et al., Meth. Enzymol. 62:308, 1979; Chandler, et al., J. Immunol. Meth. 53:187, 1982; Ekeke and Abuknesha, J. Steroid Biochem. 11:1579, 1979; Engvall and Perlmann, J. Immunol. 109:129, 1972; Geoghegan, et al., Immunol. Comm. 7:1, 1978; and Wilson and Nakane, Immunofluorescence and Related Techniques. Elsevier/North Holland Biomedical Press; Amsterdam (1978).
[176] Depending upon the nature of the label, various techniques may be employed for detecting and quantitating the label. For fluorescers, a large number of fluorometers are available. For chemiluminescers, luminometers or films are available. With enzymes, a fluorescent, chemiluminescent, or colored product may be determined or measured fluorometrically, luminometrically, spectrophotometrically, or visually.
[177] Various types of chemiluminescent compounds having an acridinium, benzacridinium, or acridan type of heterocyclic ring systems are other examples of labels. Examples of acridinium esters include those compounds having heterocyclic rings or ring systems that contain the heteroatom in a positive oxidation state including such ring systems as acridinium, benz[a]acridinium, benz[b]acridinium, benz[c]acridinium, a benzimidazole cation, quinolinium, isoquinolinium, quinolizinium, a cyclic substituted quinolinium, phenanthridinium, and quinoxalinium.
[178] The tracer may be prepared by attaching to the selected antibody either directly or indirectly a reactive functional group present on the acridinium or benzacridinium ester, as is well known to those skilled in the art {see, e.g., Weeks, et al., Clin. Chem. 29(8): 1474-1479, 1983). Examples of compounds are acridinium and benzacridinium esters with an aryl ring leaving group and the reactive functional group present in either the para or the meta position of the aryl ring. (see, e.g., U.S. Patent No. 4,745,181 and WO 94/21823).
Methods of Use
[179] As used herein, various terms are defined below.
[180] The term "treatment" includes any process, action, application, therapy, or the like, wherein a subject (or patient), including a human being, is provided medical aid with the object of improving the subject's condition, directly or indirectly, or slowing the progression of a condition or disorder in the subject.
[181] The term "combination therapy" or "co-therapy" means the administration of two or more therapeutic agents to treat a disease, condition, and/or disorder. Such administration encompasses co-administration of two or more therapeutic agents in a substantially simultaneous manner, such as in a single capsule having a fixed ratio of active ingredients or in multiple, separate capsules for each inhibitor agent. In addition, such administration encompasses use of each type of therapeutic agent in a sequential manner.
[182] The antibodies of the inventin may be administered in combination with the following agents: cytotoxic agent, angiogenesis inhibitors, antirheumatic agent, muscle relaxant, narcotic, non-steroid anti-inflammatory drug, analgesic, anesthetic, sedative, local anesthetic, neuromuscular blocker, antimicrobial agent, immunoglobulins, antidepressant, asthma medication, cytokine, and cytokine antagonist. [183] For example, the antibodies of the inventin may be administered in combination with various anti-cancer agents including, but not limited to, bleomycin, docetaxel, doxorubicin, edatrexate, erlotinib, etoposide, finasteride, flutamide, gemcitabine, genitinib, goserelin acetate, granisetron, imatinib, irinotecan, ondansetron, paclitaxel, pegaspargase, pilocaφine hydrochloride, porfimer sodium, interleukin-2, rituximab, topotecan, trastuzumab, triapine, vincristine, and vinorelbine tartrate, or therapeutic antibodies or fragments thereof, or anti-angiogenic agent, such as, for example, angiostatin, bevacizumab, sorafenib, baculostatin, canstatin, maspin, anti-VEGF antibodies or peptides, anti-placental growth factor antibodies or peptides, anti-Flk-1 antibodies, anti-Fit- 1 antibodies or peptides, laminin peptides, fibronectin peptides, plasminogen activator inhibitors, tissue metalloproteinase inhibitors, interferons, interleukin 12, IP-IO, Gro-β, thrombospondin, 2- methoxyoestradiol, proliferin-related protein, carboxiamidotriazole, CMlOl, Marimastat, pentosan polysulphate, angiopoietin 2, interferon-alpha, herbimycin A, PNU145156E, 16K prolactin fragment, Linomide, thalidomide, pentoxifylline, genistein, TNP-470, endostatin, paclitaxel, accutin, cidofovir, vincristine, bleomycin, AGM- 1470, platelet factor 4 or minocycline.
[184] The phrase "therapeutically effective" means the amount of each agent administered that will achieve the goal of improvement in a disease, condition, and/or disorder severity, while avoiding or minimizing adverse side effects associated with the given therapeutic treatment.
[185] The term "pharmaceutically acceptable" means that the subject item is appropriate for use in a pharmaceutical product.
[186] The antibodies of this invention are expected to be valuable as therapeutic agents. Accordingly, an embodiment of this invention includes a method of treating the various conditions in a patient (including mammals) which comprises administering to said patient a composition containing an amount of an antibody of the invention that is effective in treating the target condition.
[187] The antibodies of the present invention may be used in the treatment or prevention of various diseases including, but not limited to, cancer, infectious disease, and autoimmune diseases.
[188] The antibodies of the present invention or compositions including the antibodies may include a cytotoxic agent (e.g., monomethylauristatin-E) that is conjugated to the antibody.
[189] Antibodies of the present invention may be administered alone or in combination with one or more additional therapeutic agents. Combination therapy includes administration of a single pharmaceutical dosage formulation which contains an antibody of the present invention and one or more additional therapeutic agents, as well as administration of the antibody of the present invention and each additional therapeutic agents in its own separate pharmaceutical dosage formulation. For example, an antibody of the present invention and a therapeutic agent may be administered to the patient together in a single oral dosage composition or each agent may be administered in separate oral dosage formulations
[100] Where separate dosage formulations are used, the antibody of the present invention and one or more additional therapeutic agents may be administered at essentially the same time (e g , concurrently) or at separately staggered times (e g , sequentially)
[101] To assess the ability of a particular antibody to be therapeutically useful to treat cancer, as an example, the antibody may be tested in vivo in a mouse xenograft tumor model An example of a therapeutic model is detailed in Example 8
Pharmaceutical compositions
[102] The antibodies descπbed herein may be provided in a pharmaceutical composition compπsing a pharmaceutically acceptable earner The pharmaceutically acceptable earner may be non-pyrogenic The compositions may be administered alone or in combination with at least one other agent, such as stabilizing compound, which may be administered in any stenle, biocompatible pharmaceutical earner including, but not limited to, salme, buffered saline, dextrose, and water A vanety of aqueous earners may be employed including, but not limited to saline, glycine, or the like These solutions are stenle and generally free of particulate matter These solutions may be stenhzed by conventional, well known stenhzation techniques (e g , filtration) The compositions may contain pharmaceutically acceptable auxiliary substances as required to approximate physiological conditions such as pH adjusting and buffering agents, and the like The concentration of the antibody of the invention in such pharmaceutical formulation may vary widely, and may be selected pnmanly based on fluid volumes, viscosities, etc , according to the particular mode of administration selected If desired, more than one type of antibody may be included in a pharmaceutical composition
[103] The compositions may be administered to a patient alone, or in combination with other agents, drugs or hormones In addition to the active ingredients, these pharmaceutical compositions may contain suitable pharmaceutically acceptable earners compnsing excipients and auxilianes that facilitate processing of the active compounds into preparations which may be used pharmaceutically Pharmaceutical compositions of the invention may be administered by any number of routes including, but not limited to, oral, mtravenous, intramuscular, mtra-artenal, intramedullary, intrathecal, mtraventncular, transdermal, subcutaneous, intrapentoneal, intranasal, parenteral, topical, sublingual, or rectal means
[104] Formulations suitable for subcutaneous, intravenous, intramuscular, and the like, suitable pharmaceutical earners, and techniques for formulation and administration may be prepared by any of the methods well known in the art {see, e.g., Remington's Pharmaceutical Sciences, Mack Publishing Co., Easton, Pa., 20th edition, 2000).
Diagnostic methods
[105] The present invention also provides diagnostic methods with which a particular antigen may be detected in a patient sample or biological sample. Such diagnostic methods may be used, for example, to diagnose disorders in which a particular antigen is elevated or reduced. Such disorders include, but are not limited to, cancer, infectious disease, and autoimmune diseases. As an example, when used for diagnosis, detection of an amount of the antibody-antigen complex in a sample from a patient which is greater than an amount of the complex in a normal sample identifies the patient as likely to have the disorder
[106] The patient sample may be contacted with an antibody of the invention, and the patient sample may then be assayed for the presence of an antibody-antigen complex. As described above, the antibody may comprise a detectable label, such as a fluorescent, radioisotopic, chemiluminescent, or enzymatic label, such as horseradish peroxidase, alkaline phosphatase, or luciferase.
[107] Optionally, the antibody may be bound to a solid support, which may accommodate automation of the assay. Suitable solid supports include, but are not limited to, glass or plastic slides, tissue culture plates, microtiter wells, tubes, silicon chips, or particles such as beads (including, but not limited to, latex, polystyrene, or glass beads). Any method known in the art may be used to attach the antibody to the solid support, including use of covalent and non-covalent linkages, passive absorption, or pairs of binding moieties attached to the antibody and the solid support. Binding of antigen and the antibody may be accomplished in any vessel suitable for containing the reactants. Examples of such vessels include microtiter plates, test tubes, and microcentrifuge tubes.
Determination of a Therapeutically Effective Dose
[108] The determination of a therapeutically effective dose is well within the capability of those skilled in the art. A therapeutically effective dose refers to the amount of an antibody that may be used to effectively treat a disease (e.g., cancer) compared with the efficacy that is evident in the absence of the therapeutically effective dose.
[109] The therapeutically effective dose may be estimated initially in animal models (e.g., rats, mice, rabbits, dogs, or pigs). The animal model may also be used to determine the appropriate concentration range and route of administration. Such information may then be used to determine useful doses and routes for administration in humans. [110] Therapeutic efficacy and toxicity (e.g., ED50 - the dose therapeutically effective in 50% of the population and LD5O - the dose lethal to 50% of the population) of an antibody may be determined by standard pharmaceutical procedures in cell cultures or experimental animals. The dose ratio of toxic to therapeutic effects is the therapeutic index, and it may be expressed as the ratio, LD5O/ED5o. The data obtained from animal studies may used in formulating a range of dosage for human use. The dosage contained in such compositions may be within a range of circulating concentrations that include the ED50 with little or no toxicity. The dosage varies within this range depending upon the dosage form employed, sensitivity of the patient, and the route of administration.
[111] The exact dosage may be determined by the practitioner, in light of factors related to the patient who requires treatment. Dosage and administration may be adjusted to provide sufficient levels of the antibody or to maintain the desired effect. Factors that may be taken into account include the severity of the disease state, general health of the subject, age, weight, and gender of the subject, diet, time and frequency of administration, drug combination(s), reaction sensitivities, and tolerance/response to therapy. Effective in vivo dosages of an antibody are in the range of about 5 μg to about 500 μg/kg of patient body weight.
[112] The mode of administration of antibody-containing pharmaceutical compositions of the present invention may be any suitable route which delivers the antibody to the host. As an example, pharmaceutical compositions of the invention may be useful for parenteral administration (e.g., subcutaneous, intramuscular, intravenous, or intranasal administration).
[113] All patents and patent applications cited in this disclosure are expressly incorporated herein by reference. The above disclosure generally describes the present invention. A more complete understanding can be obtained by reference to the following specific examples, which are provided for purposes of illustration only and are not intended to limit the scope of the invention.
EXAMPLES
[114] In order that this invention may be better understood, the following examples are set forth. These examples are for the purpose of illustration only, and are not to be construed as limiting the scope of the invention in any manner. All publications mentioned herein are incorporated by reference in their entirety.
Example 1: Construction, expression, and purification ofFab-like antibody
[115] Antibodies 3E10 and 19G9 recognize tissue factor (TF) and tumor-associated antigen RGl, respectively. These two antibodies were used to construct protease-regulated antibodies containing the protease site DDDDK (SEQ TD NO: 26) linker located between the antigen binding domains and constant region domain. Specifically, these antibodies contained VL-DDDDK-CL on the light chain and VH-DDDDK-CH1-Myc-His6 on the heavy chain, where the linker is cleavable by enterokinase, and Myc and His6 are tags for detection and purification. The DNA sequences for the two antibodies were cloned into bacterial expression vectors using standard molecular biology technologies, and the constructs were confirmed by DNA sequencing. Examples of plasmid are shown in Figures 8 and 9. The plasmid containing either 3E10 or 19G9 was expressed and purified from bacterial strain TGl . Briefly, a single colony of bacteria strain TGl containing the antibody expression plasmid was selected and grown overnight in 8 ml of 2xYT medium in the presence of 34 μg/ml chloramphenicol and 1% glucose. A volume of culture (7 ml) was transferred to 250 ml fresh 2xYT medium containing 34 μg/ml chloramphenicol and 0.1% glucose. After 3 hours of incubation, 0.5 mM IPTG was added to induce Fab expression. The culture was incubated overnight at 25 °C. Following incubation, the culture was centrifuged to pellet the bacterial cells, and the pellet was resuspended in a Bug Buster® lysis buffer (Novagen, Madison, WI). After centrifugation, the bacterial lysis supernatant was filtered, and the Fab fragments were affinity-purified through a Ni-NTA column (Qiagen, Valencia, CA) according to the manufacturer's instruction.
[116] Other examples of protease-regulated antibodies were also constructed using tandem linked variable regions from 3E10 and 19G9. These antibodies contained, for example, VL3E10- DDDDK-VL19G9-CL on the light chain and VH3E1O-DDDDK-VH19G9-CH1-Myc-His6 on the heavy chain, where the linker is cleavable by enterokinase, and Myc and His6 are tags for detection and purification. An antibody library was also constructed using the framework regions (FR), for example, FR4 of 3E10 and FRl of 19G9 either intact or truncated. Several types of protease-regulated antibodies were screened from this library. The cloning, expression, and purification were performed as described above. Example 2: Cloning and expression oflgG-like antibodies
[117] The expression vector pIE_SRgamma_fa contains cDNAs encoding the constant regions of human IgGl (fa haplotype) and kappa chains, respectively. An overlap PCR was performed to link the variable regions of anti-TF antibody 3E10 and anti-RGl antibody 19G9. The native signal peptide of 19G9 was used for secretion of the protease-regulated antibodies. Four examples of peptide linkers located between the variable regions of 3E10 and 19G9 are Linker 1: SDDDDK (SEQ ID NO: 2), Linker 2: GGGGSDDDDK (SEQ ID NO: 3), Linker 3:
GGGGSDDDDKGGGGS (SEQ ID NO: 4), and Linker 4: GGGGSGGGGSGGGGS (SEQ ID NO: 5). The primers for amplification of the variable region of the light chain introduced Hind III and Bsiw I sites into the 5' and 3' ends of PCR fragment, respectively. The resulting PCR-amplifϊed VL genes were cloned into the Hindlll/Bsiw site of pIE SRgammal fa to create PIE-3E10VL- linker-19G9VL. The same strategy was used to clone in frame VH fusions of 3E10 and 19G9 (including linkers 1-4) into pEE-3E10VL-linker-19G9VL. Briefly, the primer pairs of the variable regions of 3E10 and 19G9 contained Notl/Apal sites. The PCR products were digested with Notl/Apal and inserted upstream of the CH region of pffi-3E10VH-linker-19G9VH ensuring that the VH regions were in frame with the CH region in the respective pIE derivatives. The final constructs were verified by DNA sequencing analysis.
[118] Transfection and transient expression of the protease-regulated antibodies were conducted using mammalian cells. Approximately 4 x 108 CHO-S cells supplemented with CHO-SF medium were prepared for transfection. Transfection was carried out using Lipofectamine™ 2000 (Invitrogen, Carlsbad, CA) and 1 mg plasmid DNA following the manufacturer's instruction. The cells were grown for three days after transfection, and the culture media was harvested and filtered for antibody isolation and purification.
[119] Examples of the protease-regulated antibodies are described in Tables 3-9
TABLE 3: PROTE ASE-REGUL ATED ANTIBODIES (Type 1)
Fab-like protease-regulated antibodies against TF (3E10)
Light chain Heavy chain
DIVLTQPHSVSASPGKTVTISCTRSSGSVA DLVESGGTLVQPGGSLRLSCAASGFSFTDAW SYYVQWYQQRPGSSPTTVIYEDNHRPSGVP MSWVRQAPGKELEWVSSISGSGGSTYYAGSV DRFSGSIDTSSNSASLTISGLKTEDEADYY KGRFTI SRDNSKNTLYLQMNSLRAEDTAVYY CQSYDSNNLWFGGGTKLTVLGAGGGGSDD CARVLSLTDYYWYGMDVWGQGTLVTVSASDD DDKRTVAAPSVFIFPPSDEQLKSGTASWC DDKSSASTKGPSVFPLAPSSKSTSGGTAALG LLNNFYPREAKVQWKVDNALQSGNSQESVT CLVKDYFPEPVTVSWNSGALTSGVHTFPAVL EQDSKDSTYSLSSTLTLSKADYEKHKVYAC QSSGLYSLSSWTVPSSSLGTQTYICNVNHK EVTHQGLSSPVTKSFNRGEC (SEQ ID NO:33 ) PSNTKVDKKVEPKCEF (SEQ ID NO: 34 )
Fab-like protease-regulated antibodies against RGl (19G9)
Light chain Heavy chain
DIVLTQSPGTLSLSPGERATLSCRASQSVSS QLVQSGGGLVQPGGSLRLSCAGSGFTFSSYV SYLAWYQQKPGQAPRLLIYGASSRATGIPDR MHWLRQAPGKGLEWVSVIGTGGVTHYADSVK FSGSGSGTDFTLTISRLEPEDFAVYYCQQYS GRFTI SRDNAKNSLYLQMNSLRAEDTAVYYC SSLTFGGGTKVEIKDDDDKRTVAAPSVFIFP ARWGYYGSGSYENDAFDIWGQGTMVTVDDDD
PSDEQLKSGTASWCLLNNFYPREAKVQWKV KSSASTKGPSVFPLAPSSKSTSGGTAALGCL DNALQSGNSQESVTEQDSKDSTYSLSSTLTL VKDYFPEPVTVSWNSGALTSGVHTFPAVLQS SKADYEKHKVYACEVTHQGLSSPVTKSFNRG SGLYSLSSWTVPSSSLGTQTYICNVNHKPS EC (SEQ ID NO: 35 ) NTKVDKKVEPKCEF (SEQ ID NO: 36)
IgG-like protease-regulated antibodies against TF (3E10)
Light chain Heavy chain
NFMLTQPHSVSASPGKTVTISCTRSSGSVAS QVNLRESGGTLVQPGGSLRLSCAASGFSFTD YYVQWYQQRPGSSPTTVIYEDNHRPSGVPDR AWMSWVRQAPGKELEWVSSISGSGGSTYYAG FSGSIDTSSNSASLTISGLKTEDEADYYCQS SVKGRFTISRDNSKNTLYLQMNSLRAEDTAV YDSNNLWFGGGTKLTVLGQSDDDDKPKAAP YYCARVLSLTDYYWYGMDVWGQGTLVTVSAS SVTLFPPSSEELQANKATLVCLISDFYPGAV DDDDKTKGPSVFPLAPSSKSTSGGTAALGCL TVAWKADSSPVKAGVETTTPSKQSNNKYAAS VKDYFPEPVTVSWNSGALTSGVHTFPAVLQS SYLSLTPEQWKSHRSYSCQVTHEGSTVEKTV SGLYSLSSWTVPSSSLGTQTYICNVNHKPS APTECS (SEQ ID NO: 37 ) NTKVDKRVEPKSCDKTHTCPPCPAPELLGGP
SVFLFPPKPKDTLMISRTPEVTCVWDVSHE
DPEVKFNWYVDGVEVHNAKTKPREEQYNSTY
RWSVLTVLHQDWLNGKEYKCKVSNKALPAP
IEKTISKAKGQPREPQVYTLPPSREEMTKNQ
VSLTCLVKGFYPSDIAVEWESNGQPENNYKT
TPPVLDSDGSFFLYSKLTVDKSRWQQGNVFS
CSVMHEALHNHYTQKSLSLSPGK
(SEQ ID NO: 38 )
IgG-like protease-regulated antibodies against RGl (19G9)
Light chain Heavy chain
EIVLTQSPGTLSLSPGERATLSCRASQSVS EVQLVQSGGGLVQPGGSLRLSCAGSGFTFS
SSYLAWYQQKPGQAPRLLIYGASSRATGIP SYVMHWLRQAPGKGLEWVSVIGTGGVTHYA
DRFSGSGSGTDFTLTISRLEPEDFAVYYCQ DSVKGRFTISRDNAKNSLYLQMNSLRAEDT
QYSSSLTFGGGTKVEIKRTSDDDDKVAAPS AVYYCARWGYYGSGSYENDAFDIWGQGTMV
VFIFPPSDEQLKSGTASWCLLNNFYPREA TVSSASDDDDDKTKGPSVFPLAPSSKSTSG
KVQWKVDNALQSGNSQESVTEQDSKDSTYS GTAALGCLVKDYFPEPVTVSWNSGALTSGV
LSSTLTLSKADYEKHKVYACEVTHQGLSSP HTFPAVLQSSGLYSLSSWTVPSSSLGTQT
VTKSFNRGEC YICNVNHKPSNTKVDKRVEPKSCDKTHTCP
(SEQ ID NO: 39) PCPAPELLGGPSVFLFPPKPKDTLMISRTP EVTCVWDVSHEDPEVKFNWYVDGVEVHNA KTKPREEQYNSTYRWSVLTVLHQDWLNGK EYKCKVSNKALPAPIEKTISKAKGQPREPQ VYTLPPSREEMTKNQVSLTCLVKGFYPSDI AVEWESNGQPENNYKTTPPVLDSDGSFFLY SKLTVDKSRWQQGNVFSCSVMHEALHNHYT QKSLSLSPGK (SEQ ID NO: 40) TABLE 4: PROTEASE-REGULATED ANTIBODIES (Type 2)
Fab-like protease-regulated antibodies against TF and RGl
HlLl
Light chain Heavy chain
DIVLTQPHSVSASPGKTVTISCTRSSGSVA QVQLVESGGTLVQPGGSLRLSCAASGFSFT SYYVQWYQQRPGSSPTTVIYEDNHRPSGVP DAWMSWVRQAPGKELEWVSSISGSGGSTYY DRFSGSIDTSSNSASLTISGLKTEDEADYY AGSVKGRFTISRDNSKNTLYLQMNSLRAED CQSYDSNNLWFGGGTKLTVLGASDDDDKE TAVYYCARVLSLTDYYWYGMDVWGQGTLVT IVLTQSPGTLSLSPGERATLSCRASQSVSS VSASDDDDKEVQLVQSGGGLVQPGGSLRLS SYLAWYQQKPGQAPRLLIYGASSRATGIPD CAGSGFTFSSYVMHWLRQAPGKGLEWVSVI RFSGSGSGTDFTLTISRLEPEDFAVYYCQQ GTGGVTHYADSVKGRFTISRDNAKNSLYLQ YSSSLTFGGGTKVEIKRTVAAPSVFIFPPS MNSLRAEDTAVYYCARWGYYGSGSYENDAF DEQLKSGTASWCLLNNFYPREAKVQWKVD DIWGQGTMVTVSSASTKGPSVFPLAPSSKS NALQSGNSQESVTEQDSKDSTYSLSSTLTL TSGGTAALGCLVKDYFPEPVTVSWNSGALT SKADYEKHKVYACEVTHQGLSSPVTKSFNR SGVHTFPAVLQSSGLYSLSSWTVPSSSLG GEC (SEQ ID NO: 41) TQTYICNVNHKPSNTKVDKKVEPKCEF
(SEQ ID NO: 42)
H1L4
Light chain Heavy chain
DIVLTQPHSVSASPGKTVTISCTRSSGSVA QVQLVESGGTLVQPGGSLRLSCAASGFSFT SYYVQWYQQRPGSSPTTVIYEDNHRPSGVP DAWMSWVRQAPGKELEWVSSISGSGGSTYY DRFSGSIDTSSNSASLTISGLKTEDEADYY AGSVKGRFTISRDNSKNTLYLQMNSLRAED CQSYDSNNLWFGGGTKLTVLGASDDDDKL TAVYYCARVLSLTDYYWYGMDVWGQGTLVT TQSPGTLSLSPGERATLSCRASQSVSSSYL VSASDDDDKEVQLVQSGGGLVQPGGSLRLS AWYQQKPGQAPRLLIYGASSRATGIPDRFS CAGSGFTFSSYVMHWLRQAPGKGLEWVSVI GSGSGTDFTLTISRLEPEDFAVYYCQQYSS GTGGVTHYADSVKGRFTISRDNAKNSLYLQ SLTFGGGTKVEIKRTVAAPSVFIFPPSDEQ MNSLRAEDTAVYYCARWGYYGSGSYENDAF LKSGTASWCLLNNFYPREAKVQWKVDNAL DIWGQGTMVTVSSASTKGPSVFPLAPSSKS QSGNSQESVTEQDSKDSTYSLSSTLTLSKA TSGGTAALGCLVKDYFPEPVTVSWNSGALT
DYEKHKVYACEVTHQGLSSPVTKSFNRGEC SGVHTFPAVLQSSGLYSLSSWTVPSSSLG (SEQ ID NO: 43) TQTYICNVNHKPSNTKVDKKVEPKCEF
(SEQ ID NO: 44)
H1L7
Light chain Heavy chain
DIVLTQPHSVSASPGKTVTISCTRSSGSVA QVQLVESGGTLVQPGGSLRLSCAASGFSFT
SYYVQWYQQRPGSSPTTVIYEDNHRPSGVP DAWMSWVRQAPGKELEWVSSISGSGGSTYY
DRFSGSIDTSSNSASLTISGLKTEDEADYY AGSVKGRFTISRDNSKNTLYLQMNSLRAED
CQSYDSNNLWFGGGTKLTVLGASDDDDKS TAVYYCARVLSLTDYYWYGMDVWGQGTLVT
PGTLSLSPGERATLSCRASQSVSSSYLAWY VSASDDDDKEVQLVQSGGGLVQPGGSLRLS
QQKPGQAPRLLIYGASSRATGIPDRFSGSG CAGSGFTFSSYVMHWLRQAPGKGLEWVSVI
SGTDFTLTISRLEPEDFAVYYCQQYSSSLT GTGGVTHYADSVKGRFTISRDNAKNSLYLQ
FGGGTKVEIKRTVAAPSVFIFPPSDEQLKS MNSLRAEDTAVYYCARWGYYGSGSYENDAF
GTASWCLLNNFYPREAKVQWKVDNALQSG DIWGQGTMVTVSSASTKGPSVFPLAPSSKS
NSQESVTEQDSKDSTYSLSSTLTLSKADYE TSGGTAALGCLVKDYFPEPVTVSWNSGALT
KHKVYACEVTHQGLSSPVTKSFNRGEC SGVHTFPAVLQSSGLYSLSSWTVPSSSLG
(SEQ ID NO: 45 ) TQTYICNVNHKPSNTKVDKKVEPKCEF (SEQ ID NO: 46 ) TABLE 4 (cont'd)
H4L2
Light chain Heavy chain
DIVLTQPHSVSASPGKTVTISCTRSSGSVA QVQLVESGGTLVQPGGSLRLSCAASGFSFT SYYVQWYQQRPGSSPTTVIYEDNHRPSGVP DAWMSWVRQAPGKELEWVSSISGSGGSTYY DRFSGSIDTSSNSASLTISGLKTEDEADYY AGSVKGRFTISRDNSKNTLYLQMNSLRAED CQSYDSNNLWFGGGTKLTVLGDDDDKEIV TAVYYCARVLSLTDYYWYGMDVWGQGTLVT LTQSPGTLSLSPGERATLSCRASQSVSSSY VDDDDKQSGGGLVQPGGSLRLSCAGSGFTF LAWYQQKPGQAPRLLIYGASSRATGIPDRF SSYVMHWLRQAPGKGLEWVSVIGTGGVTHY SGSGSGTDFTLTISRLEPEDFAVYYCQQYS ADSVKGRFTISRDNAKNSLYLQMNSLRAED SSLTFGGGTKVEIKRTVAAPSVFIFPPSDE TAVYYCARWGYYGSGSYENDAFDIWGQGTM QLKSGTASWCLLNNFYPREAKVQWKVDNA VTVSSASTKGPSVFPLAPSSKSTSGGTAAL LQSGNSQESVTEQDSKDSTYSLSSTLTLSK GCLVKDYFPEPVTVSWNSGALTSGVHTFPA ADYEKHKVYACEVTHQGLSSPVTKSFNRGE VLQSSGLYSLSSWTVPSSSLGTQTYICNV C (SEQ ID NO: 47) NHKPSNTKVDKKVEPKCEF (SEQ ID NO: 48)
H4L5
Light chain Heavy chain
DIVLTQPHSVSASPGKTVTISCTRSSGSVA QVQLVESGGTLVQPGGSLRLSCAASGFSFT
SYYVQWYQQRPGSSPTTVIYEDNHRPSGVP DAWMSWVRQAPGKELEWVSSISGSGGSTYY
DRFSGSIDTSSNSASLTISGLKTEDEADYY AGSVKGRFTISRDNSKNTLYLQMNSLRAED
CQSYDSNNLWFGGGTKLTVLGDDDDKLTQ TAVYYCARVLSLTDYYWYGMDVWGQGTLVT
SPGTLSLSPGERATLSCRASQSVSSSYLAW VDDDDKQSGGGLVQPGGSLRLSCAGSGFTF
YQQKPGQAPRLLIYGASSRATGIPDRFSGS SSYVMHWLRQAPGKGLEWVSVIGTGGVTHY
GSGTDFTLTISRLEPEDFAVYYCQQYSSSL ADSVKGRFTISRDNAKNSLYLQMNSLRAED
TFGGGTKVEIKRTVAAPSVFIFPPSDEQLK TAVYYCARWGYYGSGSYENDAFDIWGQGTM
SGTASWCLLNNFYPREAKVQWKVDNALQS VTVSSASTKGPSVFPLAPSSKSTSGGTAAL
GNSQESVTEQDSKDSTYSLSSTLTLSKADY GCLVKDYFPEPVTVSWNSGALTSGVHTFPA
EKHKVYACEVTHQGLSSPVTKSFNRGEC VLQSSGLYSLSSWTVPSSSLGTQTYICNV
(SEQ IDNO: 49) NHKPSNTKVDKKVEPKCEF (SEQ ID NO: 50)
H4L7
Light chain Heavy chain
DIVLTQPHSVSASPGKTVTISCTRSSGSVA QVQLVESGGTLVQPGGSLRLSCAASGFSFT
SYYVQWYQQRPGSSPTTVIYEDNHRPSGVP DAWMSWVRQAPGKELEWVSSISGSGGSTYY
DRFSGSIDTSSNSASLTISGLKTEDEADYY AGSVKGRFTISRDNSKNTLYLQMNSLRAED
CQSYDSNNLWFGGGTKLTVLGASDDDDKS TAVYYCARVLSLTDYYWYGMDVWGQGTLVT
PGTLSLSPGERATLSCRASQSVSSSYLAWY VDDDDKQSGGGLVQPGGSLRLSCAGSGFTF
QQKPGQAPRLLIYGASSRATGIPDRFSGSG SSYVMHWLRQAPGKGLEWVSVIGTGGVTHY
SGTDFTLTISRLEPEDFAVYYCQQYSSSLT ADSVKGRFTISRDNAKNSLYLQMNSLRAED
FGGGTKVEIKRTVAAPSVFIFPPSDEQLKS TAVYYCARWGYYGSGSYENDAFDIWGQGTM
GTASWCLLNNFYPREAKVQWKVDNALQSG VTVSSASTKGPSVFPLAPSSKSTSGGTAAL
NSQESVTEQDSKDSTYSLSSTLTLSKADYE GCLVKDYFPEPVTVSWNSGALTSGVHTFPA
KHKVYACEVTHQGLSSPVTKSFNRGEC VLQSSGLYSLSSWTVPSSSLGTQTYICNV
(SEQIDNO: 51) NHKPSNTKVDKKVEPKCEF (SEQ ID NO: 52) TABLE 4 (cont'd)
H5L5
Light chain Heavy chain
DIVLTQPHSVSASPGKTVTISCTRSSGSVA QVQLVESGGTLVQPGGSLRLSCAASGFSFT
SYYVQWYQQRPGSSPTTVIYEDNHRPSGVP DAWMSWVRQAPGKELEWVSSISGSGGSTYY
DRFSGSIDTSSNSASLTISGLKTEDEADYY AGSVKGRFTISRDNSKNTLYLQMNSLRAED
CQSYDSNNLWFGGGTKLTVLGDDDDKLTQ TAVYYCARVLSLTDYYWYGMDVWGQGTLVT
SPGTLSLSPGERATLSCRASQSVSSSYLAW VSASDDDDKLVQPGGSLRLSCAGSGFTFSS
YQQKPGQAPRLLIYGASSRATGIPDRFSGS YVMHWLRQAPGKGLEWVSVIGTGGVTHYAD
GSGTDFTLTISRLEPEDFAVYYCQQYSSSL SVKGRFTISRDNAKNSLYLQMNSLRAEDTA
TFGGGTKVEIKRTVAAPSVFIFPPSDEQLK VYYCARWGYYGSGSYENDAFDIWGQGTMVT
SGTASWCLLNNFYPREAKVQWKVDNALQS VSSASTKGPSVFPLAPSSKSTSGGTAALGC
GNSQESVTEQDSKDSTYSLSSTLTLSKADY LVKDYFPEPVTVSWNSGALTSGVHTFPAVL
EKHKVYACEVTHQGLSSPVTKSFNRGEC QSSGLYSLSSWTVPSSSLGTQTYICNVNH
(SEQ ID NO: 53) KPSNTKVDKKVEPKCEF (SEQ ID NO: 54)
TABLE 5: PROTEASE-REGULATED ANTIBODIES (Type 2)
IgG-like protease-regulated antibodies against TF and RGl
3E10-Linkerl-19G9
Light chain Heavy chain
NFMLTQPHSVSASPGKTVTISCTRSSGSVA QVNLRESGGTLVQPGGSLRLSCAASGFSFT SYYVQWYQQRPGSSPTTVIYEDNHRPSGVP DAWMSWVRQAPGKELEWVSSISGSGGSTYY DRFSGSIDTSSNSASLTISGLKTEDEADYY AGSVKGRFTISRDNSKNTLYLQMNSLRAED CQSYDSNNLWFGGGTKLTVLGASDDDDKE TAVYYCARVLSLTDYYWYGMDVWGQGTLVT IVLTQSPGTLSLSPGERATLSCRASQSVSS VSASDDDDKEVQLVQSGGGLVQPGGSLRLS SYLAWYQQKPGQAPRLLIYGASSRATGIPD CAGSGFTFSSYVMHWLRQAPGKGLEWVSVI RFSGSGSGTDFTLTISRLEPEDFAVYYCQQ GTGGVTHYADSVKGRFTISRDNAKNSLYLQ YSSSLTFGGGTKVEIKRTVAAPSVFIFPPS MNSLRAEDTAVYYCARWGYYGSGSYENDAF DEQLKSGTASWCLLNNFYPREAKVQWKVD DIWGQGTMVTVSSASTKGPSVFPLAPSSKS NALQSGNSQESVTEQDSKDSTYSLSSTLTL TSGGTAALGCLVKDYFPEPVTVSWNSGALT SKADYEKHKVYACEVTHQGLSSPVTKSFNR SGVHTFPAVLQSSGLYSLSSWTVPSSSLG GEC (SEQ ID NO: 55) TQTYICNVNHKPSNTKVDKRVEPKSCDKTH TCPPCPAPELLGGPSVFLFPPKPKDTLMIS RTPEVTCVWDVSHEDPEVKFNWYVDGVEV HNAKTKPREEQYNSTYRWSVLTVLHQDWL NGKEYKCKVSNKALPAPIEKTISKAKGQPR EPQVYTLPPSREEMTKNQVSLTCLVKGFYP SDIAVEWESNGQPENNYKTTPPVLDSDGSF FLYSKLTVDKSRWQQGNVFSCSVMHEALHN HYTQKSLSLSPGK (SEQ ID NO: 56)
3E10-Linker2-19G9
Light chain Heavy chain
NFMLTQPHSVSASPGKTVTISCTRSSGSVA QVNLRESGGTLVQPGGSLRLSCAASGFSFT SYYVQWYQQRPGSSPTTVIYEDNHRPSGVP DAWMSWVRQAPGKELEWVSSISGSGGSTYY DRFSGSIDTSSNSASLTISGLKTEDEADYY AGSVKGRFTISRDNSKNTLYLQMNSLRAED CQSYDSNNLWFGGGTKLTVLGAGGGGSDD TAVYYCARVLSLTDYYWYGMDVWGQGTLVT DDKEIVLTQSPGTLSLSPGERATLSCRASQ VSAGGGGSDDDDKEVQLVQSGGGLVQPGGS SVSSSYLAWYQQKPGQAPRLLIYGASSRAT LRLSCAGSGFTFSSYVMHWLRQAPGKGLEW GIPDRFSGSGSGTDFTLTISRLEPEDFAVY VSVIGTGGVTHYADSVKGRFTISRDNAKNS YCQQYSSSLTFGGGTKVEIKRTVAAPSVFI LYLQMNSLRAEDTAVYYCARWGYYGSGSYE FPPSDEQLKSGTASWCLLNNFYPREAKVQ NDAFDIWGQGTMVTVSSASTKGPSVFPLAP WKVDNALQSGNSQESVTEQDSKDSTYSLSS SSKSTSGGTAALGCLVKDYFPEPVTVSWNS TLTLSKADYEKHKVYACEVTHQGLSSPVTK GALTSGVHTFPAVLQSSGLYSLSSWTVPS SFNRGEC (SEQ ID NO: 57) SSLGTQTYICNVNHKPSNTKVDKRVEPKSC DKTHTCPPCPAPELLGGPSVFLFPPKPKDT LMISRTPEVTCVWDVSHEDPEVKFNWYVD GVEVHNAKTKPREEQYNSTYRWSVLTVLH QDWLNGKEYKCKVSNKALPAPIEKTISKAK GQPREPQVYTLPPSREEMTKNQVSLTCLVK GFYPSDIAVEWESNGQPENNYKTTPPVLDS DGSFFLYSKLTVDKSRWQQGNVFSCSVMHE ALHNHYTQKSLSLSPGK (SEQ ID NO: 58) TABLE 5 (contd)
3E10-Linker3-19G9
Light chain Heavy chain
NFMLTQPHSVSASPGKTVTISCTRSSGSVA QVNLRESGGTLVQPGGSLRLSCAASGFSFT SYYVQWYQQRPGSSPTTVIYEDNHRPSGVP DAWMSWVRQAPGKELEWVSSISGSGGSTYY DRFSGSIDTSSNSASLTISGLKTEDEADYY AGSVKGRFTISRDNSKNTLYLQMNSLRAED CQSYDSNNLWFGGGTKLTVLGAGGGGSDD TAVYYCARVLSLTDYYWYGMDVWGQGTLVT
DDKGGGGSEIVLTQSPGTLSLSPGERATLS VSAGGGGSDDDDKGGGGSEVQLVQSGGGLV CRASQSVSSSYLAWYQQKPGQAPRLLIYGA QPGGSLRLSCAGSGFTFSSYVMHWLRQAPG SSRATGIPDRFSGSGSGTDFTLTISRLEPE KGLEWVSVIGTGGVTHYADSVKGRFTISRD DFAVYYCQQYSSSLTFGGGTKVEIKRTVAA NAKNSLYLQMNSLRAEDTAVYYCARWGYYG PSVFIFPPSDEQLKSGTASWCLLNNFYPR SGSYENDAFDIWGQGTMVTVSSASTKGPSV EAKVQWKVDNALQSGNSQESVTEQDSKDST FPLAPSSKSTSGGTAALGCLVKDYFPEPVT YSLSSTLTLSKADYEKHKVYACEVTHQGLS VSWNSGALTSGVHTFPAVLQSSGLYSLSSV SPVTKSFNRGEC (SEQ ID NO: 59) VTVPSSSLGTQTYICNVNHKPSNTKVDKRV EPKSCDKTHTCPPCPAPELLGGPSVFLFPP KPKDTLMISRTPEVTCVWDVSHEDPEVKF NWYVDGVEVHNAKTKPREEQYNSTYRWSV LTVLHQDWLNGKEYKCKVSNKALPAPIEKT ISKAKGQPREPQVYTLPPSREEMTKNQVSL TCLVKGFYPSDIAVEWESNGQPENNYKTTP PVLDSDGSFFLYSKLTVDKSRWQQGNVFSC SVMHEALHNHYTQKSLSLSPGK (SEQ ID NO: 60)
3E10-Linker4-19G9
Light chain Heavy chain
NFMLTQPHSVSASPGKTVTISCTRSSGSVASYYVQ QVNLRESGGTLVQPGGSLRLSCAASGFSFTDAWMS
WYQQRPGSSPTTVIYEDNHRPSGVPDRFSGSIDTS WVRQAPGKELEWVSSISGSGGSTYYAGSVKGRFTI
SNSASLTISGLKTEDEADYYCQSYDSNNLWFGGG SRDNSKNTLYLQMNSLRAEDTAVYYCARVLSLTDY
TKLTVLGAGGGGSGGGGSGGGGSEIVLTQSPGTLS YWYGMDVWGQGTLVTVSAGGGGSGGGGSGGGGSEV
LSPGERATLSCRASQSVSSSYLAWYQQKPGQAPRL QLVQSGGGLVQPGGSLRLSCAGSGFTFSSYVMHWL
LIYGASSRATGIPDRFSGSGSGTDFTLTISRLEPE RQAPGKGLEWVSVIGTGGVTHYADSVKGRFTISRD
DFAVYYCQQYSSSLTFGGGTKVEIKRTVAAPSVFI NAKNSLYLQMNSLRAEDTAVYYCARWGYYGSGSYE
FPPSDEQLKSGTASWCLLNNFYPREAKVQWKVDN NDAFDIWGQGTMVTVSSASTKGPSVFPLAPSSKST
ALQSGNSQESVTEQDSKDSTYSLSSTLTLSKADYE SGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTF
KHKVYACEVTHQGLSSPVTKSFNRGEC PAVLQSSGLYSLSSWTVPSSSLGTQTYICNVNHK
(SEQ ID NO: 61) PSNTKVDKRVEPKSCDKTHTCPPCPAPELLGGPSV
FLFPPKPKDTLMISRTPEVTCVWDVSHEDPEVKF
NWYVDGVEVHNAKTKPREEQYNSTYRWSVLTVLH
QDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPRE
PQVYTLPPSREEMTKNQVSLTCLVKGFYPSDIAVE
WESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKS
RWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK
(SEQ ID NO: 62)
3E10-Linkl-19G9 Fab
Light chain Heavy chain
DIVLTQPHSVSASPGKTVTISCTRSSGSVASYYVQ QVQLVESGGTLVQPGGSLRLSCAASGFSFTDAWMS
WYQQRPGSSPTTVIYEDNHRPSGVPDRFSGSIDTS WVRQAPGKELEWVSSISGSGGSTYYAGSVKGRFTI
SNSASLTISGLKTEDEADYYCQSYDSNNLWFGGG SRDNSKNTLYLQMNSLRAEDTAVYYCARVLSLTDY
TKLTVLGASDDDDKEIVLTQSPGTLSLSPGERATL YWYGMDVWGQGTLVTVSASDDDDKEVQLVQSGGGL
SCRASQSVSSSYLAWYQQKPGQAPRLLIYGASSRA VQPGGSLRLSCAGSGFTFSSYVMHWLRQAPGKGLE
TGIPDRFSGSGSGTDFTLTISRLEPEDFAVYYCQQ WVSVIGTGGVTHYADSVKGRFTISRDNAKNSLYLQ
YSSSLTFGGGTKVEIKRTVAAPSVFIFPPSDEQLK MNSLRAEDTAVYYCARWGYYGSGSYENDAFDIWGQ
SGTASWCLLNNFYPREAKVQWKVDNALQSGNSQE GTMVTVSSASTKGPSVFPLAPSSKSTSGGTAALGC
SVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEV LVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGL
THQGLSSPVTKSFNRGEA YSLSSWTVPSSSLGTQTYICNVNHKPSNTKVDKK
(SEQ ID NO: 63) VEPKSEF (SEQ ID NO: 64) TABLE 6: PROTEASE-REGULATED ANTIBODIES (Type 3)
Fab-like protease-regulated antibodies
H1L5
Light chain Heavy chain
DIVLTQPHSVSASPGKTVTISCTRSSGSVA QVQLVESGGTLVQPGGSLRLSCAASGFSFT
SYYVQWYQQRPGSSPTTVIYEDNHRPSGVP DAWMSWVRQAPGKELEWVSSISGSGGSTYY
DRFSGSIDTSSNSASLTISGLKTEDEADYY AGSVKGRFTISRDNSKNTLYLQMNSLRAED
CQSYDSNWLWFGGGTKLTVLGDDDDKLTQ TAVYYCARVLSLTDYYWYGMDVWGQGTLVT
SPGTLSLSPGERATLSCRASQSVSSSYLAW VSASDDDDKEVQLVQSGGGLVQPGGSLRLS
YQQKPGQAPRLLIYGASSRATGIPDRFSGS CAGSGFTFSSYVMHWLRQAPGKGLEWVSVI
GSGTDFTLTISRLEPEDFAVYYCQQYSSSL GTGGVTHYADSVKGRFTISRDNAKNSLYLQ
TFGGGTKVEIKRTVAAPSVFIFPPSDEQLK MNSLRAEDTAVYYCARWGYYGSGSYENDAF
SGTASWCLLNNFYPREAKVQWKVDNALQS DIWGQGTMVTVSSASTKGPSVFPLAPSSKS
GNSQESVTEQDSKDSTYSLSSTLTLSKADY TSGGTAALGCLVKDYFPEPVTVSWNSGALT
EKHKVYACEVTHQGLSSPVTKSFNRGEC SGVHTFPAVLQSSGLYSLSSWTVPSSSLG
(SEQ ID NO: 65) TQTYICNVNHKPSNTKVDKKVEPKCEF
(SEQ ID NO: 66)
H2L1
Light chain Heavy chain
DIVLTQPHSVSASPGKTVTISCTRSSGSVA QVQLVESGGTLVQPGGSLRLSCAASGFSFT SYYVQWYQQRPGSSPTTVIYEDNHRPSGVP DAWMSWVRQAPGKELEWVSSISGSGGSTYY DRFSGSIDTSSNSASLTISGLKTEDEADYY AGSVKGRFTISRDNSKNTLYLQMNSLRAED CQSYDSNNLWFGGGTKLTVLGASDDDDKE TAVYYCARVLSLTDYYWYGMDVWGQGTLVT IVLTQSPGTLSLSPGERATLSCRASQSVSS VDDDDKEVQLVQSGGGLVQPGGSLRLSCAG SYLAWYQQKPGQAPRLLIYGASSRATGIPD SGFTFSSYVMHWLRQAPGKGLEWVSVIGTG RFSGSGSGTDFTLTISRLEPEDFAVYYCQQ GVTHYADSVKGRFTISRDNAKNSLYLQMNS YSSSLTFGGGTKVEIKRTVAAPSVFIFPPS LRAEDTAVYYCARWGYYGSGSYENDAFDIW DEQLKSGTASWCLLNNFYPREAKVQWKVD GQGTMVTVSSASTKGPSVFPLAPSSKSTSG NALQSGNSQESVTEQDSKDSTYSLSSTLTL GTAALGCLVKDYFPEPVTVSWNSGALTSGV SKADYEKHKVYACEVTHQGLSSPVTKSFNR HTFPAVLQSSGLYSLSSWTVPSSSLGTQT GEC (SEQ ID NO: 67) YICNVNHKPSNTKVDKKVEPKCEF
(SEQ ID NO: 68)
H2L2
Light chain Heavy chain
DIVLTQPHSVSASPGKTVTISCTRSSGSVA QVQLVESGGTLVQPGGSLRLSCAASGFSFT SYYVQWYQQRPGSSPTTVIYEDNHRPSGVP DAWMSWVRQAPGKELEWVSSISGSGGSTYY DRFSGSIDTSSNSASLTISGLKTEDEADYY AGSVKGRFTISRDNSKNTLYLQMNSLRAED CQSYDSNNLWFGGGTKLTVLGDDDDKEIV TAVYYCARVLSLTDYYWYGMDVWGQGTLVT LTQSPGTLSLSPGERATLSCRASQSVSSSY VDDDDKEVQLVQSGGGLVQPGGSLRLSCAG LAWYQQKPGQAPRLLIYGASSRATGIPDRF SGFTFSSYVMHWLRQAPGKGLEWVSVIGTG SGSGSGTDFTLTISRLEPEDFAVYYCQQYS GVTHYADSVKGRFTISRDNAKNSLYLQMNS SSLTFGGGTKVEIKRTVAAPSVFIFPPSDE LRAEDTAVYYCARWGYYGSGSYENDAFDIW QLKSGTASWCLLNNFYPREAKVQWKVDNA GQGTMVTVSSASTKGPSVFPLAPSSKSTSG LQSGNSQESVTEQDSKDSTYSLSSTLTLSK GTAALGCLVKDYFPEPVTVSWNSGALTSGV ADYEKHKVYACEVTHQGLSSPVTKSFNRGE HTFPAVLQSSGLYSLSSWTVPSSSLGTQT C (SEQ ID NO: 69) YICNVNHKPSNTKVDKKVEPKCEF
(SEQ ID NO: 70) TABLE 6 (contd)
H2L4
Light chain Heavy chain
DIVLTQPHSVSASPGKTVTISCTRSSGSVA QVQLVESGGTLVQPGGSLRLSCAASGFSFT SYYVQWYQQRPGSSPTTVIYEDNHRPSGVP DAWMSWVRQAPGKELEWVSSISGSGGSTYY DRFSGSIDTSSNSASLTISGLKTEDEADYY AGSVKGRFTISRDNSKNTLYLQMNSLRAED CQSYDSNNLWFGGGTKLTVLGASASDDDD TAVYYCARVLSLTDYYWYGMDVWGQGTLVT KLTQSPGTLSLSPGERATLSCRASQSVSSS VDDDDKEVQLVQSGGGLVQPGGSLRLSCAG YLAWYQQKPGQAPRLLIYGASSRATGIPDR SGFTFSSYVMHWLRQAPGKGLEWVSVIGTG FSGSGSGTDFTLTISRLEPEDFAVYYCQQY GVTHYADSVKGRFTISRDNAKNSLYLQMNS SSSLTFGGGTKVEIKRTVAAPSVFIFPPSD LRAEDTAVYYCARWGYYGSGSYENDAFDIW EQLKSGTASWCLLNNFYPREAKVQWKVDN GQGTMVTVSSASTKGPSVFPLAPSSKSTSG ALQSGNSQESVTEQDSKDSTYSLSSTLTLS GTAALGCLVKDYFPEPVTVSWNSGALTSGV KADYEKHKVYACEVTHQGLSSPVTKSFNRG HTFPAVLQSSGLYSLSSWTVPSSSLGTQT EC (SEQ ID NO: 71) YICNVNHKPSNTKVDKKVEPKCEF
(SEQ ID NO: 72)
H2L5
Light chain Heavy chain
DIVLTQPHSVSASPGKTVTISCTRSSGSVA QVQLVESGGTLVQPGGSLRLSCAASGFSFT
SYYVQWYQQRPGSSPTTVIYEDNHRPSGVP DAWMSWVRQAPGKELEWVSSISGSGGSTYY
DRFSGSIDTSSNSASLTISGLKTEDEADYY AGSVKGRFTISRDNSKNTLYLQMNSLRAED
CQSYDSNNLWFGGGTKLTVLGDDDDKLTQ TAVYYCARVLSLTDYYWYGMDVWGQGTLVT
SPGTLSLSPGERATLSCRASQSVSSSYLAW VDDDDKEVQLVQSGGGLVQPGGSLRLSCAG
YQQKPGQAPRLLIYGASSRATGIPDRFSGS SGFTFSSYVMHWLRQAPGKGLEWVSVIGTG
GSGTDFTLTISRLEPEDFAVYYCQQYSSSL GVTHYADSVKGRFTISRDNAKNSLYLQMNS
TFGGGTKVEIKRTVAAPSVFIFPPSDEQLK LRAEDTAVYYCARWGYYGSGSYENDAFDIW
SGTASWCLLNNFYPREAKVQWKVDNALQS GQGTMVTVSSASTKGPSVFPLAPSSKSTSG
GNSQESVTEQDSKDSTYSLSSTLTLSKADY GTAALGCLVKDYFPEPVTVSWNSGALTSGV
EKHKVYACEVTHQGLSSPVTKSFNRGEC HTFPAVLQSSGLYSLSSWTVPSSSLGTQT
(SEQ ID NO: 73) YICNVNHKPSNTKVDKKVEPKCEF
(SEQ ID NO: 74)
H2L7
Light chain Heavy chain
DIVLTQPHSVSASPGKTVTISCTRSSGSVA QVQLVESGGTLVQPGGSLRLSCAASGFSFT
SYYVQWYQQRPGSSPTTVIYEDNHRPSGVP DAWMSWVRQAPGKELEWVSSISGSGGSTYY
DRFSGSIDTSSNSASLTISGLKTEDEADYY AGSVKGRFTISRDNSKNTLYLQMNSLRAED
CQSYDSNNLWFGGGTKLTVLGASDDDDKS TAVYYCARVLSLTDYYWYGMDVWGQGTLVT
PGTLSLSPGERATLSCRASQSVSSSYLAWY VDDDDKEVQLVQSGGGLVQPGGSLRLSCAG
QQKPGQAPRLLIYGASSRATGIPDRFSGSG SGFTFSSYVMHWLRQAPGKGLEWVSVIGTG
SGTDFTLTISRLEPEDFAVΎYCQQYSSSLT GVTHYADSVKGRFTISRDNAKNSLYLQMNS
FGGGTKVEIKRTVAAPSVFIFPPSDEQLKS LRAEDTAVYYCARWGYYGSGSYENDAFDIW
GTASWCLLNNFYPREAKVQWKVDNALQSG GQGTMVTVSSASTKGPSVFPLAPSSKSTSG
NSQESVTEQDSKDSTYSLSSTLTLSKADYE GTAALGCLVKDYFPEPVTVSWNSGALTSGV
KHKVYACEVTHQGLSSPVTKSFNRGEC HTFPAVLQSSGLYSLSSWTVPSSSLGTQT
(SEQ ID NO: 75) YICNVNHKPSNTKVDKKVEPKCEF
(SEQ ID NO: 76) TABLE 6 (contd)
H2L8
Light chain Heavy chain
DIVLTQPHSVSASPGKTVTISCTRSSGSVA QVQLVESGGTLVQPGGSLRLSCAASGFSFT
SYYVQWYQQRPGSSPTTVIYEDNHRPSGVP DAWMSWVRQAPGKELEWVSSISGSGGSTYY
DRFSGSIDTSSNSASLTISGLKTEDEADYY AGSVKGRFTISRDNSKNTLYLQMNSLRAED
CQSYDSNNLWFGGGTKLTVLGDDDDKSPG TAVYYCARVLSLTDYYWYGMDVWGQGTLVT
TLSLSPGERATLSCRASQSVSSSYLAWYQQ VDDDDKEVQLVQSGGGLVQPGGSLRLSCAG
KPGQAPRLLIYGASSRATGIPDRFSGSGSG SGFTFSSYVMHWLRQAPGKGLEWVSVIGTG
TDFTLTISRLEPEDFAVYYCQQYSSSLTFG GVTHYADSVKGRFTISRDNAKNSLYLQMNS
GGTKVEIKRTVAAPSVFIFPPSDEQLKSGT LRAEDTAVYYCARWGYYGSGSYENDAFDIW
ASWCLLNNFYPREAKVQWKVDNALQSGNS GQGTMVTVSSASTKGPSVFPLAPSSKSTSG
QESVTEQDSKDSTYSLSSTLTLSKADYEKH GTAALGCLVKDYFPEPVTVSWNSGALTSGV
KVYACEVTHQGLSSPVTKSFNRGEC HTFPAVLQSSGLYSLSSWTVPSSSLGTQT
(SEQ ID NO: 77) YICNVNHKPSNTKVDKKVEPKCEF
(SEQ ID NO: 78)
TABLE 7: PROTEASE-REGULATED ANTIBODIES (Type 3)
IgG-like protease-regulated antibodies
3E10-Linkerla-19G9
Light chain Heavy chain
NFMLTQPHSVSASPGKTVTISCTRSSGSVA QVNLRESGGTLVQPGGSLRLSCAASGFSFT SYYVQWYQQRPGSSPTTVIYEDNHRPSGVP DAWMSWVRQAPGKELEWVSSISGSGGSTYY
DRFSGSIDTSSNSASLTISGLKTEDEADYY AGSVKGRFTISRDNSKNTLYLQMNSLRAED CQSYDSNNLWFGGGTKLTVSDDDDKEIVL TAVYYCARVLSLTDYYWYGMDVWGQGTLVT TQSPGTLSLSPGERATLSCRASQSVSSSYL VDDDDKEVQLVQSGGGLVQPGGSLRLSCAG AWYQQKPGQAPRLLIYGASSRATGIPDRFS SGFTFSSYVMHWLRQAPGKGLEWVSVIGTG GSGSGTDFTLTISRLEPEDFAVYYCQQYSS GVTHYADSVKGRFTISRDNAKNSLYLQMNS SLTFGGGTKVEIKRTVAAPSVFIFPPSDEQ LRAEDTAVYYCARWGYYGSGSYENDAFDIW LKSGTASWCLLNNFYPREAKVQWKVDNAL GQGTMVTVSSASTKGPSVFPLAPSSKSTSG QSGNSQESVTEQDSKDSTYSLSSTLTLSKA GTAALGCLVKDYFPEPVTVSWNSGALTSGV DYEKHKVYACEVTHQGLSSPVTKSFNRGEC HTFPAVLQSSGLYSLSSWTVPSSSLGTQT (SEQ ID NO: 79) YICNVNHKPSNTKVDKRVEPKSCDKTHTCP PCPAPELLGGPSVFLFPPKPKDTLMISRTP EVTCVWDVSHEDPEVKFNWYVDGVEVHNA KTKPREEQYNSTYRWSVLTVLHQDWLNGK EYKCKVSNKALPAPIEKTISKAKGQPREPQ VYTLPPSREEMTKNQVSLTCLVKGFYPSDI AVEWESNGQPENNYKTTPPVLDSDGSFFLY SKLTVDKSRWQQGNVFSCSVMHEALHNHYT QKSLSLSPGK (SEQ ID NO: 80)
3E10-Linkerlb-19G9
Light chain Heavy chain
NFMLTQPHSVSASPGKTVTISCTRSSGSVA QVNLRESGGTLVQPGGSLRLSCAASGFSFT
SYYVQWYQQRPGSSPTTVIYEDNHRPSGVP DAWMSWVRQAPGKELEWVSSISGSGGSTYY
DRFSGSIDTSSNSASLTISGLKTEDEADYY AGSVKGRFTISRDNSKNTLYLQMNSLRAED
CQSYDSNNLWFGGGTKLTVSDDDDKLTQS TAVYYCARVLSLTDYYWYGMDVWGQGTLVT
PGTLSLSPGERATLSCRASQSVSSSYLAWY VDDDDKQSGGGLVQPGGSLRLSCAGSGFTF
QQKPGQAPRLLIYGASSRATGIPDRFSGSG SSYVMHWLRQAPGKGLEWVSVIGTGGVTHY
SGTDFTLTISRLEPEDFAVYYCQQYSSSLT ADSVKGRFTISRDNAKNSLYLQMNSLRAED
FGGGTKVEIKRTVAAPSVFIFPPSDEQLKS TAVYYCARWGYYGSGSYENDAFDIWGQGTM
GTASWCLLNNFYPREAKVQWKVDNALQSG VTVSSASTKGPSVFPLAPSSKSTSGGTAAL
NSQESVTEQDSKDSTYSLSSTLTLSKADYE GCLVKDYFPEPVTVSWNSGALTSGVHTFPA
KHKVYACEVTHQGLSSPVTKSFNRGEC VLQSSGLYSLSSWTVPSSSLGTQTYICNV
(SEQ ID NO: 81) NHKPSNTKVDKRVEPKSCDKTHTCPPCPAP ELLGGPSVFLFPPKPKDTLMISRTPEVTCV WDVSHEDPEVKFNWYVDGVEVHNAKTKPR EEQYNSTYRWSVLTVLHQDWLNGKEYKCK VSNKALPAPIEKTISKAKGQPREPQVYTLP PSREEMTKNQVSLTCLVKGFYPSDIAVEWE SNGQPENNYKTTPPVLDSDGSFFLYSKLTV DKSRWQQGNVFSCSVMHEALHNHYTQKSLS LSPGK (SEQ ID NO: 82) TABLE 7 (contd)
3E10-Linkerlc-19G9
Light chain Heavy chain
NFMLTQPHSVSASPGKTVTISCTRSSGSVA QVNLRESGGTLVQPGGSLRLSCAASGFSFT SYYVQWYQQRPGSSPTTVIYEDNHRPSGVP DAWMSWVRQAPGKELEWVSSISGSGGSTYY DRFSGSIDTSSNSASLTISGLKTEDEADYY AGSVKGRFTISRDNSKNTLYLQMNSLRAED CQSYDSNNLWFGGGTKLTVLGASDDDDKL TAVΎYCARVLSLTDYYWYGMDVWGQGTLVT TQSPGTLSLSPGERATLSCRASQSVSSSYL VSASDDDDKQSGGGLVQPGGSLRLSCAGSG AWYQQKPGQAPRLLIYGASSRATGIPDRFS FTFSSYVMHWLRQAPGKGLEWVSVIGTGGV GSGSGTDFTLTISRLEPEDFAVYYCQQYSS THYADSVKGRFTISRDNAKNSLYLQMNSLR SLTFGGGTKVEIKRTVAAPSVFIFPPSDEQ AEDTAVYYCARWGYYGSGSYENDAFDIWGQ LKSGTASWCLLNNFYPREAKVQWKVDNAL GTMVTVSSASTKGPSVFPLAPSSKSTSGGT QSGNSQESVTEQDSKDSTYSLSSTLTLSKA AALGCLVKDYFPEPVTVSWNSGALTSGVHT DYEKHKVYACEVTHQGLSSPVTKSFNRGEC FPAVLQSSGLYSLSSWTVPSSSLGTQTYI
(SEQ ID NO: 83) CNVNHKPSNTKVDKRVEPKSCDKTHTCPPC PAPELLGGPSVFLFPPKPKDTLMISRTPEV TCVWDVSHEDPEVKFNWYVDGVEVHNAKT KPREEQYNSTYRWSVLTVLHQDWLNGKEY KCKVSNKALPAPIEKTISKAKGQPREPQVY TLPPSREEMTKNQVSLTCLVKGFYPSDIAV EWESNGQPENNYKTTPPVLDSDGSFFLYSK LTVDKSRWQQGNVFSCSVMHEALHNHYTQK SLSLSPGK (SEQ ID NO: 84)
Figure imgf000041_0001
TABLE 8 (contd)
H3L7
Light chain Heavy chain
DIVLTQPHSVSASPGKTVTISCTRSSGSVA QVQLVESGGTLVQPGGSLRLSCAASGFSFT
SYYVQWYQQRPGSSPTTVIYEDNHRPSGVP DAWMSWVRQAPGKELEWVSSISGSGGSTYY
DRFSGSIDTSSNSASLTISGLKTEDEADYY AGSVKGRFTISRDNSKNTLYLQMNSLRAED
CQSYDSNNLWFGGGTKLTVLGASDDDDKS TAVYYCARVLSLTDYYWYGMDVWGQGTLVT
PGTLSLSPGERATLSCRASQSVSSSYLAWY VSASDDDDKQSGGGLVQPGGSLRLSCAGSG
QQKPGQAPRLLIYGASSRATGIPDRFSGSG FTFSSYVMHWLRQAPGKGLEWVSVIGTGGV
SGTDFTLTISRLEPEDFAVYYCQQYSSSLT THYADSVKGRFTISRDNAKNSLYLQMNSLR
FGGGTKVEIKRTVAAPSVFIFPPSDEQLKS AEDTAVYYCARWGYYGSGSYENDAFDIWGQ
GTASWCLLNNFYPREAKVQWKVDNALQSG GTMVTVSSASTKGPSVFPLAPSSKSTSGGT
NSQESVTEQDSKDSTYSLSSTLTLSKADYE AALGCLVKDYFPEPVTVSWNSGALTSGVHT
KHKVYACEVTHQGLSSPVTKSFNRGEC FPAVLQSSGLYSLSSWTVPSSSLGTQTYI
(SEQ ID NO: 93) CNVNHKPSNTKVDKKVEPKCEF (SEQ ID NO:94 )
H1L2
Light chain Heavy chain
DIVLTQPHSVSASPGKTVTISCTRSSGSVA QVQLVESGGTLVQPGGSLRLSCAASGFSFT SYYVQWYQQRPGSSPTTVIYEDNHRPSGVP DAWMSWVRQAPGKELEWVSSISGSGGSTYY DRFSGSIDTSSNSASLTISGLKTEDEADYY AGSVKGRFTISRDNSKNTLYLQMNSLRAED CQSYDSNNLWFGGGTKLTVLGDDDDKEIV TAVYYCARVLSLTDYYWYGMDVWGQGTLVT LTQSPGTLSLSPGERATLSCRASQSVSSSY VSASDDDDKEVQLVQSGGGLVQPGGSLRLS LAWYQQKPGQAPRLLIYGASSRATGIPDRF CAGSGFTFSSYVMHWLRQAPGKGLEWVSVI SGSGSGTDFTLTISRLEPEDFAVYYCQQYS GTGGVTHYADSVKGRFTISRDNAKNSLYLQ SSLTFGGGTKVEIKRTVAAPSVFIFPPSDE MNSLRAEDTAVYYCARWGYYGSGSYENDAF QLKSGTASWCLLNNFYPREAKVQWKVDNA DIWGQGTMVTVSSASTKGPSVFPLAPSSKS LQSGNSQESVTEQDSKDSTYSLSSTLTLSK TSGGTAALGCLVKDYFPEPVTVSWNSGALT ADYEKHKVYACEVTHQGLSSPVTKSFNRGE SGVHTFPAVLQSSGLYSLSSWTVPSSSLG C (SEQ ID NO: 95) TQTYICNVNHKPSNTKVDKKVEPKCEF (SEQ ID NO: 96)
H5L1
Light chain Heavy chain
DIVLTQPHSVSASPGKTVTISCTRSSGSVA QVQLVESGGTLVQPGGSLRLSCAASGFSFT SYYVQWYQQRPGSSPTTVIYEDNHRPSGVP DAWMSWVRQAPGKELEWVSSISGSGGSTYY DRFSGSIDTSSNSASLTISGLKTEDEADYY AGSVKGRFTISRDNSKNTLYLQMNSLRAED CQSYDSNNLWFGGGTKLTVLGASDDDDKE TAVYYCARVLSLTDYYWYGMDVWGQGTLVT IVLTQSPGTLSLSPGERATLSCRASQSVSS VSASDDDDKLVQPGGSLRLSCAGSGFTFSS SYLAWYQQKPGQAPRLLIYGASSRATGIPD YVMHWLRQAPGKGLEWVSVIGTGGVTHYAD RFSGSGSGTDFTLTISRLEPEDFAVYYCQQ SVKGRFTISRDNAKNSLYLQMNSLRAEDTA YSSSLTFGGGTKVEIKRTVAAPSVFIFPPS VYYCARWGYYGSGSYENDAFDIWGQGTMVT DEQLKSGTASWCLLNNFYPREAKVQWKVD VSSASTKGPSVFPLAPSSKSTSGGTAALGC NALQSGNSQESVTEQDSKDSTYSLSSTLTL LVKDYFPEPVTVSWNSGALTSGVHTFPAVL SKADYEKHKVYACEVTHQGLSSPVTKSFNR QSSGLYSLSSWTVPSSSLGTQTYICNVNH GEC (SEQ ID NO: 97) KPSNTKVDKKVEPKCEF (SEQ ID NO: 98)
Figure imgf000043_0001
TABLE 9: PROTEASE-REGULATED ANTIBODIES (Type 4)
HlL5a
Heavy chain Heavy chain
QVQLVESGGTLVQPGGSLRLSCAASGFSFT QVQLVESGGTLVQPGGSLRLSCAASGFSFT DAWMSWVRQAPGKELEWVSSISGSGGSTYY DAWMSWVRQAPGKELEWVSSISGSGGSTYY AGSVKGRFTISRDNSKNTLYLQMNSLRAED AGSVKGRFTISRDNSKNTLYLQMNSLRAED TAVYYCARAAAAAAAAAAAAAAWGQGTLVT TAVYYCARAAAAAAAAAAAAAAWGQGTLVT
VSASDDDDKEVQLVQSGGGLVQPGGSLRLS VSASDDDDKEVQLVQSGGGLVQPGGSLRLS
CAGSGFTFSSYVMHWLRQAPGKGLEWVSVI CAGSGFTFSSYVMHWLRQAPGKGLEWVSVI GTGGVTHYADSVKGRFTISRDNAKNSLYLQ GTGGVTHYADSVKGRFTISRDNAKNSLYLQ MNSLRAEDTAVYYCARWGYYGSGSYENDAF MNSLRAEDTAVYYCARWGYYGSGSYENDAF DIWGQGTMVTVSSASTKGPSVFPLAPSSKS DIWGQGTMVTVSSASTKGPSVFPLAPSSKS TSGGTAALGCLVKDYFPEPVTVSWNSGALT TSGGTAALGCLVKDYFPEPVTVSWNSGALT SGVHTFPAVLQSSGLYSLSSWTVPSSSLG SGVHTFPAVLQSSGLYSLSSWTVPSSSLG TQTYICNVNHKPSNTKVDKKVEPKCEF TQTYICNVNHKPSNTKVDKKVEPKCEF (SEQIDNO: 107) (SEQ ID NO: 108)
H2Lla
Heavy chain Heavy chain
QVQLVESGGTLVQPGGSLRLSCAASGFSFT QVQLVESGGTLVQPGGSLRLSCAASGFSFT DAWMSWVRQAPGKELEWVSSISGSGGSTYY DAWMSWVRQAPGKELEWVSSISGSGGSTYY AGSVKGRFTISRDNSKNTLYLQMNSLRAED AGSVKGRFTISRDNSKNTLYLQMNSLRAED TAVYYCARAAAAAAAAAAAAAAWGQGTLVT TAVYYCARAAAAAAAAAAAAAAWGQGTLVT VDDDDKEVQLVQSGGGLVQPGGSLRLSCAG VDDDDKEVQLVQSGGGLVQPGGSLRLSCAG SGFTFSSYVMHWLRQAPGKGLEWVSVIGTG SGFTFSSYVMHWLRQAPGKGLEWVSVIGTG GVTHYADSVKGRFTISRDNAKNSLYLQMNS GVTHYADSVKGRFTISRDNAKNSLYLQMNS
LRAEDTAVYYCARWGYYGSGSYENDAFDIW LRAEDTAVYYCARWGYYGSGSYENDAFDIW
GQGTMVTVSSASTKGPSVFPLAPSSKSTSG GQGTMVTVSSASTKGPSVFPLAPSSKSTSG
GTAALGCLVKDYFPEPVTVSWNSGALTSGV GTAALGCLVKDYFPEPVTVSWNSGALTSGV
HTFPAVLQSSGLYSLSSWTVPSSSLGTQT HTFPAVLQSSGLYSLSSWTVPSSSLGTQT
YICNVNHKPSNTKVDKKVEPKCEF YICNVNHKPSNTKVDKKVEPKCEF
(SEQ ID NO: 109) (SEQ ID NO: 110)
H2L2a
Heavy chain Heavy chain
QVQLVESGGTLVQPGGSLRLSCAASGFSFT QVQLVESGGTLVQPGGSLRLSCAASGFSFT
DAWMSWVRQAPGKELEWVSSISGSGGSTYY DAWMSWVRQAPGKELEWVSSISGSGGSTYY
AGSVKGRFTISRDNSKNTLYLQMNSLRAED AGSVKGRFTISRDNSKNTLYLQMNSLRAED
TAVYYCARAAAAAAAAAAAAAAWGQGTLVT TAVYYCARAAAAAAAAAAAAAAWGQGTLVT
VDDDDKEVQLVQSGGGLVQPGGSLRLSCAG VDDDDKEVQLVQSGGGLVQPGGSLRLSCAG
SGFTFSSYVMHWLRQAPGKGLEWVSVIGTG SGFTFSSYVMHWLRQAPGKGLEWVSVIGTG
GVTHYADSVKGRFTISRDNAKNSLYLQMNS GVTHYADSVKGRFTISRDNAKNSLYLQMNS
LRAEDTAVYYCARWGYYGSGSYENDAFDIW LRAEDTAVYYCARWGYYGSGSYENDAFDIW
GQGTMVTVSSASTKGPSVFPLAPSSKSTSG GQGTMVTVSSASTKGPSVFPLAPSSKSTSG
GTAALGCLVKDYFPEPVTVSWNSGALTSGV GTAALGCLVKDYFPEPVTVSWNSGALTSGV
HTFPAVLQSSGLYSLSSWTVPSSSLGTQT HTFPAVLQSSGLYSLSSWTVPSSSLGTQT
YICNVNHKPSNTKVDKKVEPKCEF YICNVNHKPSNTKVDKKVEPKCEF
(SEQIDNO: 111) (SEQ IDNO:112)
Figure imgf000045_0001
TABLE 9 (contd)
H2L8a
Heavy chain Heavy chain
QVQLVESGGTLVQPGGSLRLSCAASGFSFT QVQLVESGGTLVQPGGSLRLSCAASGFSFT DAWMSWVRQAPGKELEWVSSISGSGGSTYY DAWMSWVRQAPGKELEWVSSISGSGGSTYY AGSVKGRFTISRDNSKNTLYLQMNSLRAED AGSVKGRFTISRDNSKNTLYLQMNSLRAED TAVYYCARAAAAAAAAAAAAAAWGQGTLVT TAVYYCARAAAAAAAAAAAAAAWGQGTLVT VDDDDKEVQLVQSGGGLVQPGGSLRLSCAG VDDDDKEVQLVQSGGGLVQPGGSLRLSCAG SGFTFSSYVMHWLRQAPGKGLEWVSVIGTG SGFTFSSYVMHWLRQAPGKGLEWVSVIGTG GVTHYADSVKGRFTISRDNAKNSLYLQMNS GVTHYADSVKGRFTISRDNAKNSLYLQMNS LRAEDTAVYYCARWGYYGSGSYENDAFDIW LRAEDTAVYYCARWGYYGSGSYENDAFDIW GQGTMVTVSSASTKGPSVFPLAPSSKSTSG GQGTMVTVSSASTKGPSVFPLAPSSKSTSG GTAALGCLVKDYFPEPVTVSWNSGALTSGV GTAALGCLVKDYFPEPVTVSWNSGALTSGV
HTFPAVLQSSGLYSLSSWTVPSSSLGTQT HTFPAVLQSSGLYSLSSWTVPSSSLGTQT YICNVNHKPSNTKVDKKVEPKCEF YICNVNHKPSNTKVDKKVEPKCEF (SEQ ID NO: 119) (SEQ ID NO.120)
Example 3: TF-binding ELISA
[120] Biotinylated TF (1 μg/ml) was added to streptavidin pre-coated 96-well plates (Pierce Chemical, Rockford, IL) and incubated for 1 hr. The plates were then washed (5x) with PBS containing 0.5% Tween-20. Samples and controls (serially diluted) were added to the wells and incubated for 1 hr, followed by washes (5x) with PBS containing 0.5% Tween-20. Horseradish peroxidase (HRP)-conjugated anti-human IgG or HRP -conjugated anti-human Fab were diluted in PBS (1 :5000) and added to each well. Following a 1 hr incubation, the plates were washed again. Amplex Red (10 μg/ml) was added to each well, and the signal was read using a plate reader. The data was analyzed using Softmax (Molecular Devices, Sunnyvale, CA). Results are shown in Figure 10.
Example 4: RGl-binding ELISA
[121] Ninety-six well plates were coated with RGl (1 μg/ml) by overnight incubation, and the plates were then washed (5x) with PBS containing 0.5% Tween-20. Samples and controls (serially diluted) were added to the wells and incubated for 1 hr, followed by washes (5x) with PBS containing 0.5% Tween-20. Horserandish peroxidase (HRP)-conjugated anti-human IgG or HRP- conjugated anti-human Fab were diluted in PBS (1 :5000) and added to each well. Following a 1 hr incubation, the plates were washed again. Amplex Red (10 μg/ml) was added to each well, and the signal was read using a plate reader. The data was analyzed using Softmax (Molecular Devices, Sunnyvale, CA). Results are shown in Figure 11.
Example 5: Sandwich antigen-binding ELISA
[122] The antigen binding activity of a bispecifϊc protease-regulated antibody (illustrated in Figure 2) was measured using a sandwich antigen-binding ELISA. This antibody binds two antigens, RG-I and TF, and the linker contains cleavage sites for enterokinase ("EK").
[123] Ninety-six well plates were coated with RGl (1 μg/ml) by overnight incubation. The plates were then washed five times with PBS containing 0.5% Tween-20. Antibody samples and controls were digested with 30 units of enterokinase for 16 hr at 37°C (see Example 4). The antibody samples, with or without enterokinase digestion, were serially diluted and added to the wells of the ELISA plates. The samples were incubated for one hour, followed by washes (5x) with PBS containing 0.5% Tween-20. Biotinylated TF (0.1 μg/ml) was added to each well and incubated for one hour. Horseradish peroxidase (HRP)-conjugated streptavidin (1:10000 diluted) was then added to each well. Following a one-hour incubation, the plates were washed again. Amplex Red (10 μg/ml) was added to each well, and the signal was read using a plate reader. The data was analyzed using Soflmax® (Molecular Devices, Sunnyvale, CA). Parental antibodies 3E10, 19G9, and polyclonal human Fab were used as controls. The results are the average of duplicate wells (Figure 12). The untreated bispecific protease-regulated antibody simultaneously binds to both TF and RG-I ("Linkl" and "Link2," respectively). However, following enterokinase treatment, the binding to both antigens is greatly reduced ("Linkl /EK" and "Link2/EK," respectively).
[124] The antigen binding activity of several examples of protease-regulated antibodies was also measured using this assay. For example, the antigen binding activity of protease-regulated antibodies 3E10-Typel-Fab and 19G9-Typel-Fab is shown in Figure 13. The controls are designated 3E10-Reg-Fab, 19G9-Reg-Fab, and HuFab.
[125] The antigen binding activity of Fab-like protease-regulated antibodies is demonstrated in Figure 14. The activity of antibodies HlLl, H1L4, H1L7, H4L7, and H5L5 (Type 2) was measured in the absence and presence of enterokinase. Parental antibodies 3E10, 19G9, and polyclonal human Fab were used as controls. Similarly, Figure 15 shows the antigen binding activity of Fab-like protease-regulated antibodies H2L1, H2L2, and H2L8 (Type 3) and H3L1, H3L4, and H5L4 (Type 4).
Example 6. Enterokinase digestion of protease-regulated antibodies
[126] Protease-regulated antibodies were digested with EnterokinaseMax™, the catalytic subunit of enterokinase (Invitrogen, Carlsbad, CA). The concentration of antibodies was adjusted to 1-5 μg/ml. A volume of antibody (100 μl) was mixed with 20 μl 10x EnterokinaseMax™ buffer and 75 μl sterile water in a tube. EnterokinaseMax™ (5 μl) was added to each sample and the samples were incubated at 37°C for 16 hr. For the control group, a volume of water (5 μl ) was used.
Example 7. Western blots of antibodies
[127] Three detection antibodies were used to probe protease-regulated antibodies: anti-human kappa antibody, anti-human IgG(H+L), and anti-Myc tag antibodies. These detection antibodies were conjugated with horseradish peroxidase (HRP). Approximately 50 ng of antibody samples were mixed with loading buffer containing DTT (Invitrogen, Carlsbad, CA) and boiled for 5 min. The samples were then loaded onto a 12% Bis-Tris NuP AGE® gel (Invitrogen, Carlsbad, CA), separated, and transferred to nitrocellulose membranes. After blocking with 5% dry milk for 2 hr, the nitrocellulose membrane was incubated with a detection antibody for 1.5 hr. The membrane was then washed in PBS containing 0.5% Tween-20, and incubated with SuperSignal West Femto (Pierce Chemical, Rockford, IL), and expose to X-ray film for development. Results are shown in Figures 16-18.
Example 8: Subcutaneous xenograft cancer model
[128] Human mammary xenograft, MaTu cells are maintained as adherent cultures in RPMI supplemented with 10% FBS. Ncr nude mice (8-12 weeks of age) are inoculated subcutaneously in the right flank with 5 x 106 cells in 0.1 mL of 80% matrigel/20% HBSS. When tumors reach an average size of -180 mg (6 days), treatment is initiated. Antibodies are administered i.v. once every four days (Q4Dx3) at a dose of 10 mg/kg. Control mice are treated with PBS or an unconjugated monoclonal antibody. Daily examinations into the health status of each animal are conducted. Each experimental group consists of 10 mice and the dosing volume was 0.1 mL/10 g body weight. The length and width of each tumor is measured by using an electronic caliper 2-3 times per week and tumor weights (mg) are calculated based on the formula of [length (mm) x width (mm)2]/2. All data, including daily observations, obtained throughout the course of the study are documented. Tumor growth inhibition (TGI) is calculated as 1-T/CxlOO, where T = final tumor weights from a treated group, and C = final tumor weights from the control group. The data demonstrates the therapeutic utility of antibodies for the treatment of tumors.
[129] Other embodiments of the invention will be apparent to the skilled in the art from a consideration of this specification or practice of the invention disclosed herein. It is intended that the specification and examples be considered as exemplary only, with the true scope and spirit of the invention being indicated by the following claims.

Claims

Claims
1. An antibody comprising one or more variable regions, wherein said antibody binds one or more antigens or epitopes.
2. The antibody of claim 1, wherein said antibody comprises two variable regions, and wherein a first variable domain Vl is specific for a first antigen, a second variable domain V2 is specific for a second antigen
3. The antibody of claim 2, wherein the first variable domain Vl comprises VL1 and VH1 domains which are specific for a first antigen, and wherein the second variable domain V2 comprises VL2 and VH2 domains which are specific for a second antigen.
4. The antibody of claim 1, 2, or 3, wherein the antibody is of the Fab-like or IgG-like type.
5. The antibody of claim 1 , 2, 3, or 4, wherein heavy and light chains are selected from the group consisting of:
a) heavy chain: NH2-VL1-VH2-CH1-CH2-CH3-COOH light chain: NH2-VH 1 -V1^-CL-COOH, b) heavy chain: NH2-VH1-VL2-CH1-CH2-CH3-COOH light chain: NH2-VL1-VH2-CL-COOH, c) heavy chain: NH2-VL1-VL2-CH1-CH2-CH3-COOH light chain: NH2-VH 1 -VH2-CL-COOH, d) heavy chain: NH2-VL1-VH2-CH1-COOH light chain: NH2-VH 1 -VL2-CL-COOH, e) heavy chain: NH2-VH1-VL2-CH1-COOH light chain: NH2-VL1-VH2-CL-COOH, f) heavy chain: NH2-VL1-VL2-CH1-COOH light chain: NH2-VH 1 -VH2-CL-COOH, g) heavy chain: NH2-VH1-VH2-CH1-COOH light chain: NH2-VL1-VL2-CL-COOH, h) heavy chain: NH2-VH1-VH2-CH1-CH2-CH3-COOH, light chain: NH2-VL1-VL2-CL-COOH, i) heavy chain: NH2-VH1-CH1-CH2-CH3-COOH, light chain: NH2-VL1 -CL-COOH, and j) heavy chain: NH2-VL1-CH1-CH2-CH3-COOH, light chain: NH2-VH1 -CL-COOH.
6. The antibody of claims 1 to 5, further comprising a linker, wherein said linker comprises two or more amino acids.
7. The linker of claim 6, wherein said linker comprises one or more protease cleavage sites.
8. The linker of claim 7, wherein said protease cleavage sites are selected from the group consisting of SEQ ID NO: 1-32.
9. The antibody of claim 6, wherein heavy and light chains are selected from the group consisting of: a) heavy chain: NH2-VL1 -linker- VH2-CH 1 -CH2-CH3 -COOH light chain: NH2-VH 1 -linker-VL2-CL-COOH, b) heavy chain: NH2-VHl-linker-VL2-CHl-CH2-CH3-COOH light chain: NH2-VL1 -linker- VH2-CL-COOH, c) heavy chain: NH2-VLl-linker-VL2-CHl-CH2-CH3-COOH light chain: NH2-VH1 -linker- VH2-CL-COOH, d) heavy chain: NH2-VL1 -linker- VH2-CH1 -COOH light chain: NH2-VHl-lmker-VL2-CL-COOH, e) heavy chain: NH2-VH1 -linker- VL2-CH1 -COOH light chain: NH2-V, 1 -linker-VH2-CL-C00H, f) heavy chain: NH2-VL1 -linker- VL2-CH1 -COOH light chain: NH2-VH1 -linker- VH2-CL-COOH, g) heavy chain: NH2-VHl-linker-VH2-CHl-COOH light chain: NH2-VL1 -linker-V^-CL-COOH, and h) heavy chain: NH2-VIIl-linker-VH2-CHl-CH2-CH3-COOH, light chain: NH2-VLl-linker-VL2-CL-COOH.
10. The antibody of claims 6 to 9, wherein the antibody simultaneously binds to two different antigens or two different epitopes.
11. The antibody of claims 6 to 9, wherein the antibody sequentially binds to two antigens in a protease-dependent manner.
12. The antibody of claims 6 to 9, wherein the antibody binds to antigen following protease digestion.
13. The antibody of claims 1-12 selected from the group consisting of SEQ ID NO: 33-120.
14. The antibody of claims 1-13, wherein said antibody is conjugated to a therapeutic or cytotoxic agent.
15. The antibody of claim 14, wherein said antibody is conjugated to an agent selected from the group consisting of monomethylauristatin-E (MMAE), aplidin, azaribine, anastrozole, azacytidine, bleomycin, bortezomib, bryostatin-1, busulfan, calicheamycin, camptothecin, 10-hydroxycamptothecin, carmustine, celebrex, chlorambucil, cisplatin, irinotecan (CPT-Il), SN-38, carboplatin, cladribine, cyclophosphamide, cytarabine, dacarbazine, docetaxel, dactiπomycin, daunomycin glucuronide, daunorubicin, dexamethasone, diethylstilbestrol, doxorubicin, doxorubicin glucuronide, epirubicin glucuronide, ethinyl estradiol, estramustine, etoposide, etoposide glucuronide, etoposide phosphate, floxuridine (FUdR), 3',5'-O-dioleoyl-FudR (FUdR-dO), fludarabine, flutamide, fluorouracil, fluoxymesterone, gemcitabine, hydroxyprogesterone caproate, hydroxyurea, idarubicin, ifosfamide, L- asparaginase, leucovorin, lomustine, mechlorethamine, medroprogesterone acetate, megestrol acetate, melphalan, mercaptopurine, 6-mercaptopurine, methotrexate, mitoxantrone, mithramycin, mitomycin, mitotane, phenyl butyrate, prednisone, procarbazine, paclitaxel, pentostatin, PSI-341, semustine streptozocin, tamoxifen, taxanes, taxol, testosterone propionate, thalidomide, thioguanine, thiotepa, teniposide, topotecan, uracil mustard, velcade, vinblastine, vinorelbine, vincristine, ricin, abrin, ribomiclease, onconase, rapLRl, DNase I, Staphylococcal enterotoxin-A, pokeweed antiviral protein, gelonin, diphtheria toxin, Pseudomonas exotoxin, and Pseudomonas endotoxin, and functional analogs thereof
16. A pharmaceutical composition comprising a therapeutically effective amount of an antibody of any of claims 1 to 14, in combination with a pharmaceutically acceptable carrier.
17. A pharmaceutical composition comprising a therapeutically effective amount of an antibody of any of claims 1 to 14, in combination with a pharmaceutically acceptable carrier and one or more pharmaceutical agents.
18. A method for the treatment of cancer, infectious diseases, and autoimmune diseases comprising administering to a subject in need an effective amount of of an antibody of any of claims 1 to 14 or a pharmaceutical composition of claims 16 or 17.
19. The method of claim 18, wherein the cancer is selected from cancers of the breast, respiratory tract, brain, reproductive organs, digestive tract, urinary tract, eye, liver, skin, head and neck, thyroid, parathyroid, lymphomas, sarcomas, and leukemias.
20. The method of claim 18, wherein the autoimmune disease is selected from multiple sclerosis, rheumatoid arthritis, lupus, type I diabetes mellitus, Crohn's disease, autoimmune hemolytic anemia, autoimmune hepatitis, glomerulonephritis, inflammatory bowel disease, myocarditis, psoriasis, thyroiditis, ulcerative colitis, and Graves'disease.
21. The method of claim 18 , wherein the infectious disease is selected from HIV/AH) S , lower respiratory infections, diarrheal diseases, tuberculosis, malaria, measles, pertussis, tetanus, meningitis, syphilis, hepatitis B and tropical diseases.
22. Use of an antibody according to claims 1 to 14 in the manufacture of a medicament for the treatment of a disease selected from the group of diseases consisting of cancer, infectious diseases, and autoimmune diseases.
23. Use of an antibody according to claims 1 to 14 in a method of diagnosing a disease.
24. Use according to claim 23, wherein the disease is selected from the group of diseases consisting of cancer, infectious diseases and autoimmune diseases.
25. A diagnostic method to detect a disease in a patient comprising:
(a) immunologically detecting and quantifying the level of a protein associated with a disease in control samples taken from individuals of a control population;
(b) immunologically detecting and quantifying changes in the protein in samples of a patient sample taken from a patient over time; and
(c) comparing the levels of the protein in the patient's samples to the level of the protein in the control samples; wherein a level of the protein in the patient's samples that is above the level of the protein in the control samples is indicative the presence of disease in the patient.
26. A method for monitoring the status of a disease in a patient, and/or monitoring how a patient with said disease is responding to a therapy comprising immunologically detecting and quantifying changes in the levels of a protein associated with a disease in patient samples taken over time, wherein increasing levels of the protein over time indicate disease progression or a negative response to said therapy, and wherein decreasing levels of the protein over time indicate disease remission or a positive response to said therapy.
27. A kit comprising an antibody of any one of claims 1-14.
8. The kit of claim 27, further comprising solutions for suspending or fixing the cells, detectable labels, solutions for rendering a polypeptide susceptible to the binding of an antibody, solutions for lysing cells, and/or solutions for the purification of polypeptides.
PCT/EP2008/006750 2007-08-15 2008-08-16 Monospecific and multispecific antibodies and method of use WO2009021754A2 (en)

Priority Applications (5)

Application Number Priority Date Filing Date Title
JP2010520502A JP5485152B2 (en) 2007-08-15 2008-08-16 Monospecific and multispecific antibodies and methods of use
CA2696263A CA2696263C (en) 2007-08-15 2008-08-16 Monospecific and multispecific antibodies and method of use
EP08785592A EP2178914A2 (en) 2007-08-15 2008-08-16 Monospecific and multispecific antibodies and method of use
US12/671,942 US9624309B2 (en) 2007-08-15 2008-08-16 Monospecific and multispecific antibodies and method of use
US15/466,587 US20170210825A1 (en) 2007-08-15 2017-03-22 Monospecific and multispecific antibodies and method of use

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US95591307P 2007-08-15 2007-08-15
US60/955,913 2007-08-15

Related Child Applications (3)

Application Number Title Priority Date Filing Date
US12/671,942 A-371-Of-International US9624309B2 (en) 2007-08-15 2008-08-16 Monospecific and multispecific antibodies and method of use
EP12172879.4A Previously-Filed-Application EP2532676B1 (en) 2007-08-15 2008-08-16 Protease-regulated antibody
US15/466,587 Continuation US20170210825A1 (en) 2007-08-15 2017-03-22 Monospecific and multispecific antibodies and method of use

Publications (2)

Publication Number Publication Date
WO2009021754A2 true WO2009021754A2 (en) 2009-02-19
WO2009021754A3 WO2009021754A3 (en) 2009-06-18

Family

ID=40110987

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2008/006750 WO2009021754A2 (en) 2007-08-15 2008-08-16 Monospecific and multispecific antibodies and method of use

Country Status (6)

Country Link
US (2) US9624309B2 (en)
EP (2) EP2178914A2 (en)
JP (2) JP5485152B2 (en)
CA (1) CA2696263C (en)
ES (1) ES2628395T3 (en)
WO (1) WO2009021754A2 (en)

Cited By (135)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011030107A1 (en) * 2009-09-10 2011-03-17 Ucb Pharma S.A. Multivalent antibodies
WO2011086091A1 (en) * 2010-01-12 2011-07-21 Ucb Pharma S.A. Multivalent antibodies
WO2012025525A1 (en) 2010-08-24 2012-03-01 Roche Glycart Ag Activatable bispecific antibodies
US8217147B2 (en) 2005-08-10 2012-07-10 Macrogenics, Inc. Identification and engineering of antibodies with variant Fc regions and methods of using same
WO2013003652A1 (en) * 2011-06-28 2013-01-03 Sea Lane Biotechnologies, Llc Multispecific stacked variable domain binding proteins
JP2013539463A (en) * 2010-08-24 2013-10-24 エフ.ホフマン−ラ ロシュ アーゲー Bispecific antibodies comprising Fv fragments stabilized by disulfides
US20130345404A1 (en) * 2011-03-28 2013-12-26 Sanofi Dual Variable Region Antibody-Like Binding Proteins Having Cross-Over Binding Region Orientation
US8629246B2 (en) 2007-09-26 2014-01-14 Ucb Pharma S.A. Dual specificity antibody fusions
EP2706069A1 (en) * 2012-09-11 2014-03-12 Samsung Electronics Co., Ltd Protein complexes and methods of manufacturing bispecific antibodies using the protein complexes
US8993730B2 (en) 2008-04-02 2015-03-31 Macrogenics, Inc. BCR-complex-specific antibodies and methods of using same
GB2519786A (en) * 2013-10-30 2015-05-06 Sergej Michailovic Kiprijanov Multivalent antigen-binding protein molecules
US9096877B2 (en) 2009-10-07 2015-08-04 Macrogenics, Inc. Fc region-containing polypeptides that exhibit improved effector function due to alterations of the extent of fucosylation, and methods for their use
WO2015138920A1 (en) 2014-03-14 2015-09-17 Novartis Ag Antibody molecules to lag-3 and uses thereof
WO2015142675A2 (en) 2014-03-15 2015-09-24 Novartis Ag Treatment of cancer using chimeric antigen receptor
WO2016014553A1 (en) 2014-07-21 2016-01-28 Novartis Ag Sortase synthesized chimeric antigen receptors
WO2016014530A1 (en) 2014-07-21 2016-01-28 Novartis Ag Combinations of low, immune enhancing. doses of mtor inhibitors and cars
WO2016014565A2 (en) 2014-07-21 2016-01-28 Novartis Ag Treatment of cancer using humanized anti-bcma chimeric antigen receptor
WO2016025880A1 (en) 2014-08-14 2016-02-18 Novartis Ag Treatment of cancer using gfr alpha-4 chimeric antigen receptor
US9266967B2 (en) 2007-12-21 2016-02-23 Hoffmann-La Roche, Inc. Bivalent, bispecific antibodies
WO2016044605A1 (en) 2014-09-17 2016-03-24 Beatty, Gregory Targeting cytotoxic cells with chimeric receptors for adoptive immunotherapy
WO2016061142A1 (en) 2014-10-14 2016-04-21 Novartis Ag Antibody molecules to pd-l1 and uses thereof
WO2016090034A2 (en) 2014-12-03 2016-06-09 Novartis Ag Methods for b cell preconditioning in car therapy
US9382323B2 (en) 2009-04-02 2016-07-05 Roche Glycart Ag Multispecific antibodies comprising full length antibodies and single chain fab fragments
US9441049B2 (en) 2010-03-04 2016-09-13 Macrogenics, Inc. Antibodies reactive with B7-H3 and uses thereof
WO2016156466A1 (en) 2015-03-31 2016-10-06 Vhsquared Limited Peptide construct having a protease-cleavable linker
WO2016164731A2 (en) 2015-04-08 2016-10-13 Novartis Ag Cd20 therapies, cd22 therapies, and combination therapies with a cd19 chimeric antigen receptor (car) - expressing cell
WO2016172583A1 (en) 2015-04-23 2016-10-27 Novartis Ag Treatment of cancer using chimeric antigen receptor and protein kinase a blocker
US9487587B2 (en) 2013-03-05 2016-11-08 Macrogenics, Inc. Bispecific molecules that are immunoreactive with immune effector cells of a companion animal that express an activating receptor and cells that express B7-H3 and uses thereof
WO2017019894A1 (en) 2015-07-29 2017-02-02 Novartis Ag Combination therapies comprising antibody molecules to lag-3
WO2017019897A1 (en) 2015-07-29 2017-02-02 Novartis Ag Combination therapies comprising antibody molecules to tim-3
WO2017025698A1 (en) * 2015-08-11 2017-02-16 Queen Mary University Of London Bispecific, cleavable antibodies
US9593166B2 (en) 2013-03-14 2017-03-14 Bayer Healthcare Llc Monoclonal antibodies against antithrombin β
EP2532676B1 (en) 2007-08-15 2017-03-22 Bayer Pharma Aktiengesellschaft Protease-regulated antibody
US9676845B2 (en) 2009-06-16 2017-06-13 Hoffmann-La Roche, Inc. Bispecific antigen binding proteins
WO2017106810A2 (en) 2015-12-17 2017-06-22 Novartis Ag Combination of c-met inhibitor with antibody molecule to pd-1 and uses thereof
WO2017106656A1 (en) 2015-12-17 2017-06-22 Novartis Ag Antibody molecules to pd-1 and uses thereof
US9688758B2 (en) 2012-02-10 2017-06-27 Genentech, Inc. Single-chain antibodies and other heteromultimers
US9714296B2 (en) 2010-03-04 2017-07-25 Macrogenics, Inc. Antibodies reactive with B7-H3, immunologically active fragments thereof and uses thereof
WO2017125897A1 (en) 2016-01-21 2017-07-27 Novartis Ag Multispecific molecules targeting cll-1
WO2017149515A1 (en) 2016-03-04 2017-09-08 Novartis Ag Cells expressing multiple chimeric antigen receptor (car) molecules and uses therefore
WO2017165683A1 (en) 2016-03-23 2017-09-28 Novartis Ag Cell secreted minibodies and uses thereof
WO2017181119A2 (en) 2016-04-15 2017-10-19 Novartis Ag Compositions and methods for selective protein expression
WO2017210617A2 (en) 2016-06-02 2017-12-07 Porter, David, L. Therapeutic regimens for chimeric antigen receptor (car)- expressing cells
CN107454905A (en) * 2015-03-31 2017-12-08 韦斯夸尔德有限公司 Polypeptide
WO2018013918A2 (en) 2016-07-15 2018-01-18 Novartis Ag Treatment and prevention of cytokine release syndrome using a chimeric antigen receptor in combination with a kinase inhibitor
WO2018023025A1 (en) 2016-07-28 2018-02-01 Novartis Ag Combination therapies of chimeric antigen receptors adn pd-1 inhibitors
WO2018026819A2 (en) 2016-08-01 2018-02-08 Novartis Ag Treatment of cancer using a chimeric antigen receptor in combination with an inhibitor of a pro-m2 macrophage molecule
US9890204B2 (en) 2009-04-07 2018-02-13 Hoffmann-La Roche Inc. Trivalent, bispecific antibodies
WO2018067992A1 (en) 2016-10-07 2018-04-12 Novartis Ag Chimeric antigen receptors for the treatment of cancer
US9975956B2 (en) 2011-12-22 2018-05-22 I2 Pharmaceuticals, Inc. Surrogate binding proteins which bind DR4 and/or DR5
US9982036B2 (en) 2011-02-28 2018-05-29 Hoffmann-La Roche Inc. Dual FC antigen binding proteins
US9994646B2 (en) 2009-09-16 2018-06-12 Genentech, Inc. Coiled coil and/or tether containing protein complexes and uses thereof
US10017561B2 (en) 2009-05-13 2018-07-10 I2 Pharmaceuticals, Inc. Neutralizing molecules to influenza viruses
WO2018140725A1 (en) 2017-01-26 2018-08-02 Novartis Ag Cd28 compositions and methods for chimeric antigen receptor therapy
WO2018160731A1 (en) 2017-02-28 2018-09-07 Novartis Ag Shp inhibitor compositions and uses for chimeric antigen receptor therapy
US10106612B2 (en) 2012-06-27 2018-10-23 Hoffmann-La Roche Inc. Method for selection and production of tailor-made highly selective and multi-specific targeting entities containing at least two different binding entities and uses thereof
US10106600B2 (en) 2010-03-26 2018-10-23 Roche Glycart Ag Bispecific antibodies
WO2018201056A1 (en) 2017-04-28 2018-11-01 Novartis Ag Cells expressing a bcma-targeting chimeric antigen receptor, and combination therapy with a gamma secretase inhibitor
WO2018201051A1 (en) 2017-04-28 2018-11-01 Novartis Ag Bcma-targeting agent, and combination therapy with a gamma secretase inhibitor
US10138293B2 (en) 2007-12-21 2018-11-27 Hoffmann-La Roche, Inc. Bivalent, bispecific antibodies
WO2018237157A1 (en) 2017-06-22 2018-12-27 Novartis Ag Antibody molecules to cd73 and uses thereof
WO2019006007A1 (en) 2017-06-27 2019-01-03 Novartis Ag Dosage regimens for anti-tim-3 antibodies and uses thereof
WO2019018730A1 (en) 2017-07-20 2019-01-24 Novartis Ag Dosage regimens of anti-lag-3 antibodies and uses thereof
US10214580B2 (en) 2007-03-27 2019-02-26 I2 Pharmaceuticals, Inc. Constructs and libraries comprising antibody surrogate light chain sequences
WO2019089798A1 (en) 2017-10-31 2019-05-09 Novartis Ag Anti-car compositions and methods
WO2019099838A1 (en) 2017-11-16 2019-05-23 Novartis Ag Combination therapies
US10300140B2 (en) 2011-07-28 2019-05-28 I2 Pharmaceuticals, Inc. Sur-binding proteins against ERBB3
US10323099B2 (en) 2013-10-11 2019-06-18 Hoffmann-La Roche Inc. Multispecific domain exchanged common variable light chain antibodies
EP3514179A1 (en) 2014-01-24 2019-07-24 Dana-Farber Cancer Institute, Inc. Antibody molecules to pd-1 and uses thereof
WO2019152660A1 (en) 2018-01-31 2019-08-08 Novartis Ag Combination therapy using a chimeric antigen receptor
US10407513B2 (en) 2008-09-26 2019-09-10 Ucb Biopharma Sprl Biological products
WO2019200229A1 (en) 2018-04-13 2019-10-17 Novartis Ag Dosage regimens for anti-pd-l1 antibodies and uses thereof
US10457748B2 (en) 2012-12-21 2019-10-29 Ucb Biopharma Sprl Single linker FabFv antibodies and methods of producing same
WO2019210153A1 (en) 2018-04-27 2019-10-31 Novartis Ag Car t cell therapies with enhanced efficacy
WO2019227003A1 (en) 2018-05-25 2019-11-28 Novartis Ag Combination therapy with chimeric antigen receptor (car) therapies
WO2019226617A1 (en) 2018-05-21 2019-11-28 Compass Therapeutics Llc Compositions and methods for enhancing the killing of target cells by nk cells
WO2019226658A1 (en) 2018-05-21 2019-11-28 Compass Therapeutics Llc Multispecific antigen-binding compositions and methods of use
WO2019232244A2 (en) 2018-05-31 2019-12-05 Novartis Ag Antibody molecules to cd73 and uses thereof
WO2019230866A1 (en) * 2018-05-30 2019-12-05 Chugai Seiyaku Kabushiki Kaisha Polypeptide comprising il-1r1 binding domain and carrying moiety
WO2019241426A1 (en) 2018-06-13 2019-12-19 Novartis Ag Bcma chimeric antigen receptors and uses thereof
WO2019246293A2 (en) 2018-06-19 2019-12-26 Atarga, Llc Antibody molecules to complement component 5 and uses thereof
WO2020012337A1 (en) 2018-07-10 2020-01-16 Novartis Ag 3-(5-amino-1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and their use in the treatment of i karos family zinc finger 2 (ikzf2)-dependent diseases
WO2020021465A1 (en) 2018-07-25 2020-01-30 Advanced Accelerator Applications (Italy) S.R.L. Method of treatment of neuroendocrine tumors
US10611825B2 (en) 2011-02-28 2020-04-07 Hoffmann La-Roche Inc. Monovalent antigen binding proteins
US10633453B2 (en) 2013-05-28 2020-04-28 Kaohsiung Medical University Antibody locker for the inactivation of protein drug
US10633457B2 (en) 2014-12-03 2020-04-28 Hoffmann-La Roche Inc. Multispecific antibodies
EP3660042A1 (en) 2014-07-31 2020-06-03 Novartis AG Subset-optimized chimeric antigen receptor-containing t-cells
WO2020128972A1 (en) 2018-12-20 2020-06-25 Novartis Ag Dosing regimen and pharmaceutical combination comprising 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives
WO2020128898A1 (en) 2018-12-20 2020-06-25 Novartis Ag Pharmaceutical combinations
WO2020165833A1 (en) 2019-02-15 2020-08-20 Novartis Ag 3-(1-oxo-5-(piperidin-4-yl)isoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof
WO2020165834A1 (en) 2019-02-15 2020-08-20 Novartis Ag Substituted 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof
WO2020165868A1 (en) 2019-02-15 2020-08-20 Perkinelmer Cellular Technologies Germany Gmbh Low-power microscope-objective pre-scan and high-power microscope-objective scan in x,y and z-direction for imaging objects such as cells using a microscope
WO2020172553A1 (en) 2019-02-22 2020-08-27 Novartis Ag Combination therapies of egfrviii chimeric antigen receptors and pd-1 inhibitors
EP3712171A1 (en) 2014-08-19 2020-09-23 Novartis AG Treatment of cancer using a cd123 chimeric antigen receptor
WO2020205523A1 (en) 2019-03-29 2020-10-08 Atarga, Llc Anti fgf23 antibody
EP3722316A1 (en) 2014-07-21 2020-10-14 Novartis AG Treatment of cancer using a cd33 chimeric antigen receptor
US10961311B2 (en) 2016-04-15 2021-03-30 Macrogenics, Inc. B7-H3 binding molecules, antibody drug conjugates thereof and methods of use thereof
WO2021079188A1 (en) 2019-10-21 2021-04-29 Novartis Ag Combination therapies with venetoclax and tim-3 inhibitors
WO2021079195A1 (en) 2019-10-21 2021-04-29 Novartis Ag Tim-3 inhibitors and uses thereof
WO2021108661A2 (en) 2019-11-26 2021-06-03 Novartis Ag Chimeric antigen receptors and uses thereof
WO2021123902A1 (en) 2019-12-20 2021-06-24 Novartis Ag Combination of anti tim-3 antibody mbg453 and anti tgf-beta antibody nis793, with or without decitabine or the anti pd-1 antibody spartalizumab, for treating myelofibrosis and myelodysplastic syndrome
US11046769B2 (en) 2018-11-13 2021-06-29 Compass Therapeutics Llc Multispecific binding constructs against checkpoint molecules and uses thereof
WO2021144657A1 (en) 2020-01-17 2021-07-22 Novartis Ag Combination comprising a tim-3 inhibitor and a hypomethylating agent for use in treating myelodysplastic syndrome or chronic myelomonocytic leukemia
WO2021146636A1 (en) 2020-01-17 2021-07-22 Becton, Dickinson And Company Methods and compositions for single cell secretomics
WO2021173995A2 (en) 2020-02-27 2021-09-02 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
US11168139B2 (en) 2016-11-28 2021-11-09 Chugai Seiyaku Kabushiki Kaisha Antigen-binding domain, and polypeptide including conveying section
WO2021260528A1 (en) 2020-06-23 2021-12-30 Novartis Ag Dosing regimen comprising 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives
WO2022013787A1 (en) 2020-07-16 2022-01-20 Novartis Ag Anti-betacellulin antibodies, fragments thereof, and multi-specific binding molecules
US11230610B2 (en) 2016-12-09 2022-01-25 Seagen Inc. Bivalent antibodies masked by coiled coils
WO2022026592A2 (en) 2020-07-28 2022-02-03 Celltas Bio, Inc. Antibody molecules to coronavirus and uses thereof
WO2022029573A1 (en) 2020-08-03 2022-02-10 Novartis Ag Heteroaryl substituted 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof
WO2022043557A1 (en) 2020-08-31 2022-03-03 Advanced Accelerator Applications International Sa Method of treating psma-expressing cancers
WO2022043558A1 (en) 2020-08-31 2022-03-03 Advanced Accelerator Applications International Sa Method of treating psma-expressing cancers
WO2022097065A2 (en) 2020-11-06 2022-05-12 Novartis Ag ANTIBODY Fc VARIANTS
WO2022104061A1 (en) 2020-11-13 2022-05-19 Novartis Ag Combination therapies with chimeric antigen receptor (car)-expressing cells
US11345760B2 (en) 2014-06-25 2022-05-31 UCB Biopharma SRL Multispecific antibody constructs
WO2022162569A1 (en) 2021-01-29 2022-08-04 Novartis Ag Dosage regimes for anti-cd73 and anti-entpd2 antibodies and uses thereof
US11421022B2 (en) 2012-06-27 2022-08-23 Hoffmann-La Roche Inc. Method for making antibody Fc-region conjugates comprising at least one binding entity that specifically binds to a target and uses thereof
WO2022215011A1 (en) 2021-04-07 2022-10-13 Novartis Ag USES OF ANTI-TGFβ ANTIBODIES AND OTHER THERAPEUTIC AGENTS FOR THE TREATMENT OF PROLIFERATIVE DISEASES
WO2022243846A1 (en) 2021-05-18 2022-11-24 Novartis Ag Combination therapies
US11512335B2 (en) 2014-04-29 2022-11-29 Novartis Ag Vertebrate cells and methods for recombinantly expressing a polypeptide of interest
WO2023044483A2 (en) 2021-09-20 2023-03-23 Voyager Therapeutics, Inc. Compositions and methods for the treatment of her2 positive cancer
US11618790B2 (en) 2010-12-23 2023-04-04 Hoffmann-La Roche Inc. Polypeptide-polynucleotide-complex and its use in targeted effector moiety delivery
US11623952B2 (en) 2019-06-21 2023-04-11 Sorriso Pharmaceuticals, Inc. IL-23 and TNF-alpha binding bi-specific heavy chain polypeptides
WO2023092004A1 (en) 2021-11-17 2023-05-25 Voyager Therapeutics, Inc. Compositions and methods for the treatment of tau-related disorders
US11667719B2 (en) 2019-06-21 2023-06-06 Sorriso Pharmaceuticals, Inc. VHH immunoglobulin chain variable domain that binds to IL-7R and methods of use thereof for treating autoimmune and/or inflammatory diseases
US11684677B2 (en) 2016-09-30 2023-06-27 Sorriso Pharmaceuticals, Inc. Compositions
US11718679B2 (en) 2017-10-31 2023-08-08 Compass Therapeutics Llc CD137 antibodies and PD-1 antagonists and uses thereof
WO2023150778A1 (en) 2022-02-07 2023-08-10 Visterra, Inc. Anti-idiotype antibody molecules and uses thereof
US11752207B2 (en) 2017-07-11 2023-09-12 Compass Therapeutics Llc Agonist antibodies that bind human CD137 and uses thereof
WO2023220695A2 (en) 2022-05-13 2023-11-16 Voyager Therapeutics, Inc. Compositions and methods for the treatment of her2 positive cancer
US11851497B2 (en) 2017-11-20 2023-12-26 Compass Therapeutics Llc CD137 antibodies and tumor antigen-targeting antibodies and uses thereof
WO2024030976A2 (en) 2022-08-03 2024-02-08 Voyager Therapeutics, Inc. Compositions and methods for crossing the blood brain barrier
EP4324518A2 (en) 2014-01-31 2024-02-21 Novartis AG Antibody molecules to tim-3 and uses thereof
US11970538B2 (en) 2021-05-20 2024-04-30 Compass Therapeutics Llc Multispecific binding constructs against checkpoint molecules and uses thereof

Families Citing this family (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
NZ588554A (en) * 2008-04-29 2013-03-28 Abbott Lab Dual variable domain immunoglobulins and uses thereof
WO2009137666A2 (en) * 2008-05-08 2009-11-12 The Trustees Of The University Of Pennsylvania Chemiluminescence enhanced detection
CN102112494A (en) 2008-06-03 2011-06-29 雅培制药有限公司 Dual variable domain immunoglobulins and uses thereof
AR072001A1 (en) 2008-06-03 2010-07-28 Abbott Lab IMMUNOGLOBULIN WITH DUAL VARIABLE DOMAIN AND USES OF THE SAME
WO2010006060A2 (en) 2008-07-08 2010-01-14 Abbott Laboratories Prostaglandin e2 dual variable domain immunoglobulins and uses thereof
RU2011127198A (en) * 2008-12-04 2013-01-10 Эбботт Лэборетриз IMMUNOGLOBULINS WITH DOUBLE VARIABLE DOMAINS AND THEIR APPLICATION
EP2424566A4 (en) * 2009-05-01 2013-07-31 Abbvie Inc Dual variable domain immunoglobulins and uses thereof
EP2488658A4 (en) * 2009-10-15 2013-06-19 Abbvie Inc Dual variable domain immunoglobulins and uses thereof
UY32979A (en) 2009-10-28 2011-02-28 Abbott Lab IMMUNOGLOBULINS WITH DUAL VARIABLE DOMAIN AND USES OF THE SAME
EP3252072A3 (en) 2010-08-03 2018-03-14 AbbVie Inc. Dual variable domain immunoglobulins and uses thereof
KR20130139884A (en) 2010-08-26 2013-12-23 애브비 인코포레이티드 Dual variable domain immunoglobulins and uses thereof
CN104159920A (en) 2011-12-30 2014-11-19 艾伯维公司 Dual specific binding proteins directed against il-13 and/or il-17
GB201203442D0 (en) 2012-02-28 2012-04-11 Univ Birmingham Immunotherapeutic molecules and uses
AU2013240279A1 (en) * 2012-03-30 2014-10-16 Bayer Healthcare Llc Protease-regulated antibodies
KR20180008921A (en) 2012-11-01 2018-01-24 애브비 인코포레이티드 Anti-vegf/dll4 dual variable domain immunoglobulins and uses thereof
US20160009817A1 (en) * 2013-03-15 2016-01-14 Bayer Healthcare Llc Pro-drug antibodies against tissue factor pathway inhibitor
CN105324396A (en) 2013-03-15 2016-02-10 艾伯维公司 Dual specific binding proteins directed against il-1 beta and il-17
EP3164417A1 (en) 2014-07-01 2017-05-10 Pfizer Inc. Bispecific heterodimeric diabodies and uses thereof
US10093733B2 (en) 2014-12-11 2018-10-09 Abbvie Inc. LRP-8 binding dual variable domain immunoglobulin proteins
TW201710286A (en) 2015-06-15 2017-03-16 艾伯維有限公司 Binding proteins against VEGF, PDGF, and/or their receptors
JP7073258B2 (en) 2015-11-19 2022-05-23 レビトープ リミテッド Mutual complementation of functional antibody fragments for a two-component system against undesired cell redirection killing
WO2019016625A1 (en) * 2017-07-18 2019-01-24 Avx Corporation Board-to-board contact bridge system
SG11202000820PA (en) * 2017-07-31 2020-02-27 Tizona Therapeutics Anti-cd39 antibodies, compositions comprising anti-cd39 antibodies and methods of using anti-cd39 antibodies
AU2018328291B2 (en) 2017-09-08 2022-10-27 Takeda Pharmaceutical Company Limited Constrained conditionally activated binding proteins
CN109957026A (en) * 2017-12-22 2019-07-02 成都恩沐生物科技有限公司 Covalent multi-specificity antibody
EP3934762A1 (en) 2019-03-05 2022-01-12 Takeda Pharmaceutical Company Limited Constrained conditionally activated binding proteins

Family Cites Families (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CH652145A5 (en) 1982-01-22 1985-10-31 Sandoz Ag METHOD FOR IN VITRO PRODUCTION OF HYBRID OMEN WHAT human monoclonal antibodies GENERATE.
US4634666A (en) 1984-01-06 1987-01-06 The Board Of Trustees Of The Leland Stanford Junior University Human-murine hybridoma fusion partner
US4745181A (en) 1986-10-06 1988-05-17 Ciba Corning Diagnostics Corp. Polysubstituted aryl acridinium esters
DE3856559T2 (en) * 1987-05-21 2004-04-29 Micromet Ag Multifunctional proteins with predetermined objectives
US5427908A (en) 1990-05-01 1995-06-27 Affymax Technologies N.V. Recombinant library screening methods
US5298608A (en) 1990-05-17 1994-03-29 Mitsubishi Kasei Corporation Metal chelate of a monoazo compound and optical recording medium using it
GB9015198D0 (en) 1990-07-10 1990-08-29 Brien Caroline J O Binding substance
US5395752A (en) 1993-03-19 1995-03-07 Ciba Corning Diagnostics Corp. Long emission wavelength chemiluminescent compounds and their use in test assays
EP1177441A1 (en) * 1999-05-10 2002-02-06 Prolinx, Inc. Cell separation device and methods for use
WO2001091798A2 (en) 2000-06-01 2001-12-06 Universite Catholique De Louvain Tumor activated prodrug compounds
JP2004503512A (en) 2000-06-09 2004-02-05 アメリカ合衆国 PEGylation of a linker that improves the antitumor activity of the immunoconjugate and reduces toxicity
AUPR729801A0 (en) * 2001-08-27 2001-09-20 Nucleics Pty Ltd A method of sequestering and/or purifying a polypeptide
KR100708398B1 (en) 2002-03-22 2007-04-18 (주) 에이프로젠 Humanized antibody and process for preparing same
US20040109855A1 (en) 2002-07-23 2004-06-10 Herman Waldmann Therapeutic antibodies with reduced side effect
GB0230203D0 (en) 2002-12-27 2003-02-05 Domantis Ltd Fc fusion
WO2005035753A1 (en) 2003-10-10 2005-04-21 Chugai Seiyaku Kabushiki Kaisha Double specific antibodies substituting for functional protein
US20060252096A1 (en) * 2005-04-26 2006-11-09 Glycofi, Inc. Single chain antibody with cleavable linker
WO2007024715A2 (en) 2005-08-19 2007-03-01 Abbott Laboratories Dual variable domain immunoglobin and uses thereof
CA2696263C (en) 2007-08-15 2017-06-13 Bing Liu Monospecific and multispecific antibodies and method of use
EP3492488A1 (en) 2007-08-22 2019-06-05 The Regents of The University of California Activatable binding polypeptides and methods of identification and use thereof

Non-Patent Citations (1)

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

Cited By (183)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8217147B2 (en) 2005-08-10 2012-07-10 Macrogenics, Inc. Identification and engineering of antibodies with variant Fc regions and methods of using same
US10214580B2 (en) 2007-03-27 2019-02-26 I2 Pharmaceuticals, Inc. Constructs and libraries comprising antibody surrogate light chain sequences
EP2532676B1 (en) 2007-08-15 2017-03-22 Bayer Pharma Aktiengesellschaft Protease-regulated antibody
US8629246B2 (en) 2007-09-26 2014-01-14 Ucb Pharma S.A. Dual specificity antibody fusions
US11427650B2 (en) 2007-09-26 2022-08-30 UCB Biopharma SRL Dual specificity antibody fusions
US9309327B2 (en) 2007-09-26 2016-04-12 Ucb Pharma S.A. Dual specificity antibody fusions
US9828438B2 (en) 2007-09-26 2017-11-28 Ucb Pharma S.A. Dual specificity antibody fusions
US10100130B2 (en) 2007-09-26 2018-10-16 Ucb Biopharma Sprl Dual specificity antibody fusions
US10138293B2 (en) 2007-12-21 2018-11-27 Hoffmann-La Roche, Inc. Bivalent, bispecific antibodies
US9266967B2 (en) 2007-12-21 2016-02-23 Hoffmann-La Roche, Inc. Bivalent, bispecific antibodies
US10927163B2 (en) 2007-12-21 2021-02-23 Hoffmann-La Roche, Inc. Bivalent, bispecific antibodies
US9695236B2 (en) 2008-04-02 2017-07-04 Macrogenics, Inc. BCR-complex-specific antibodies and methods of using same
US10479831B2 (en) 2008-04-02 2019-11-19 Macrogenics, Inc BCR-complex-specific antibodies and methods of using same
US8993730B2 (en) 2008-04-02 2015-03-31 Macrogenics, Inc. BCR-complex-specific antibodies and methods of using same
US10407513B2 (en) 2008-09-26 2019-09-10 Ucb Biopharma Sprl Biological products
US9382323B2 (en) 2009-04-02 2016-07-05 Roche Glycart Ag Multispecific antibodies comprising full length antibodies and single chain fab fragments
US9890204B2 (en) 2009-04-07 2018-02-13 Hoffmann-La Roche Inc. Trivalent, bispecific antibodies
US10017561B2 (en) 2009-05-13 2018-07-10 I2 Pharmaceuticals, Inc. Neutralizing molecules to influenza viruses
US11673945B2 (en) 2009-06-16 2023-06-13 Hoffmann-La Roche Inc. Bispecific antigen binding proteins
US9676845B2 (en) 2009-06-16 2017-06-13 Hoffmann-La Roche, Inc. Bispecific antigen binding proteins
US10640555B2 (en) 2009-06-16 2020-05-05 Hoffmann-La Roche Inc. Bispecific antigen binding proteins
WO2011030107A1 (en) * 2009-09-10 2011-03-17 Ucb Pharma S.A. Multivalent antibodies
US9994646B2 (en) 2009-09-16 2018-06-12 Genentech, Inc. Coiled coil and/or tether containing protein complexes and uses thereof
US9096877B2 (en) 2009-10-07 2015-08-04 Macrogenics, Inc. Fc region-containing polypeptides that exhibit improved effector function due to alterations of the extent of fucosylation, and methods for their use
WO2011086091A1 (en) * 2010-01-12 2011-07-21 Ucb Pharma S.A. Multivalent antibodies
US9441049B2 (en) 2010-03-04 2016-09-13 Macrogenics, Inc. Antibodies reactive with B7-H3 and uses thereof
US10683364B2 (en) 2010-03-04 2020-06-16 Macrogenics, Inc. Antibodies reactive with B7-H3, immunologically active fragments thereof and uses thereof
US9714296B2 (en) 2010-03-04 2017-07-25 Macrogenics, Inc. Antibodies reactive with B7-H3, immunologically active fragments thereof and uses thereof
US9714295B2 (en) 2010-03-04 2017-07-25 Macrogenics, Inc. Antibodies reactive with B7-H3, immunologically active fragments thereof and uses thereof
US10730945B2 (en) 2010-03-04 2020-08-04 Macrogenics, Inc. Antibodies reactive with B7-H3 and users thereof
US9896508B2 (en) 2010-03-04 2018-02-20 Macrogenics, Inc. Antibodies reactive with B7-H3 and uses thereof
US10106600B2 (en) 2010-03-26 2018-10-23 Roche Glycart Ag Bispecific antibodies
CN103068847A (en) * 2010-08-24 2013-04-24 罗切格利卡特公司 Activatable bispecific antibodies
US9879095B2 (en) 2010-08-24 2018-01-30 Hoffman-La Roche Inc. Bispecific antibodies comprising a disulfide stabilized-Fv fragment
EP2609112B1 (en) * 2010-08-24 2017-11-22 Roche Glycart AG Activatable bispecific antibodies
EP2609111B1 (en) * 2010-08-24 2017-11-01 F. Hoffmann-La Roche AG Bispecific antibodies comprising a disulfide stabilized-fv fragment
WO2012025525A1 (en) 2010-08-24 2012-03-01 Roche Glycart Ag Activatable bispecific antibodies
JP2013539463A (en) * 2010-08-24 2013-10-24 エフ.ホフマン−ラ ロシュ アーゲー Bispecific antibodies comprising Fv fragments stabilized by disulfides
KR101612999B1 (en) * 2010-08-24 2016-04-15 로슈 글리카트 아게 Activatable bispecific antibodies
JP2013538204A (en) * 2010-08-24 2013-10-10 ロシュ グリクアート アーゲー Activable bispecific antibody
US11618790B2 (en) 2010-12-23 2023-04-04 Hoffmann-La Roche Inc. Polypeptide-polynucleotide-complex and its use in targeted effector moiety delivery
US10793621B2 (en) 2011-02-28 2020-10-06 Hoffmann-La Roche Inc. Nucleic acid encoding dual Fc antigen binding proteins
US10611825B2 (en) 2011-02-28 2020-04-07 Hoffmann La-Roche Inc. Monovalent antigen binding proteins
US9982036B2 (en) 2011-02-28 2018-05-29 Hoffmann-La Roche Inc. Dual FC antigen binding proteins
US20140056895A1 (en) * 2011-03-28 2014-02-27 Sanofi Dual Variable Region Antibody-Like Binding Proteins Having Cross-Over Binding Region Orientation
US9181349B2 (en) * 2011-03-28 2015-11-10 Sanofi Dual variable region antibody-like binding proteins having cross-over binding region orientation
US20140011238A1 (en) * 2011-03-28 2014-01-09 Sanofi Dual Variable Region Antibody-Like Binding Proteins Having Cross-Over Binding Region Orientation
US20130345404A1 (en) * 2011-03-28 2013-12-26 Sanofi Dual Variable Region Antibody-Like Binding Proteins Having Cross-Over Binding Region Orientation
US9221917B2 (en) * 2011-03-28 2015-12-29 Sanofi Dual variable region antibody-like binding proteins having cross-over binding region orientation
WO2013003652A1 (en) * 2011-06-28 2013-01-03 Sea Lane Biotechnologies, Llc Multispecific stacked variable domain binding proteins
US10300140B2 (en) 2011-07-28 2019-05-28 I2 Pharmaceuticals, Inc. Sur-binding proteins against ERBB3
US9975956B2 (en) 2011-12-22 2018-05-22 I2 Pharmaceuticals, Inc. Surrogate binding proteins which bind DR4 and/or DR5
US9688758B2 (en) 2012-02-10 2017-06-27 Genentech, Inc. Single-chain antibodies and other heteromultimers
US11407836B2 (en) 2012-06-27 2022-08-09 Hoffmann-La Roche Inc. Method for selection and production of tailor-made highly selective and multi-specific targeting entities containing at least two different binding entities and uses thereof
US10106612B2 (en) 2012-06-27 2018-10-23 Hoffmann-La Roche Inc. Method for selection and production of tailor-made highly selective and multi-specific targeting entities containing at least two different binding entities and uses thereof
US11421022B2 (en) 2012-06-27 2022-08-23 Hoffmann-La Roche Inc. Method for making antibody Fc-region conjugates comprising at least one binding entity that specifically binds to a target and uses thereof
EP2706069A1 (en) * 2012-09-11 2014-03-12 Samsung Electronics Co., Ltd Protein complexes and methods of manufacturing bispecific antibodies using the protein complexes
US9522961B2 (en) 2012-09-11 2016-12-20 Samsung Electronics Co., Ltd. Protein complexes and methods of manufacturing bispecific antibodies using the protein complexes
US10457748B2 (en) 2012-12-21 2019-10-29 Ucb Biopharma Sprl Single linker FabFv antibodies and methods of producing same
US11401349B2 (en) 2012-12-21 2022-08-02 UCB Biopharma SRL Single linker FabFv antibodies and methods of producing same
US9487587B2 (en) 2013-03-05 2016-11-08 Macrogenics, Inc. Bispecific molecules that are immunoreactive with immune effector cells of a companion animal that express an activating receptor and cells that express B7-H3 and uses thereof
US10144784B2 (en) 2013-03-14 2018-12-04 Bayer Healthcare Llc Monoclonal antibodies against antithrombin beta
US9593166B2 (en) 2013-03-14 2017-03-14 Bayer Healthcare Llc Monoclonal antibodies against antithrombin β
US9908942B2 (en) 2013-03-14 2018-03-06 Bayer Healthcare, Llc Monoclonal antibodies against antithrombin β
US10633453B2 (en) 2013-05-28 2020-04-28 Kaohsiung Medical University Antibody locker for the inactivation of protein drug
US10323099B2 (en) 2013-10-11 2019-06-18 Hoffmann-La Roche Inc. Multispecific domain exchanged common variable light chain antibodies
GB2519786A (en) * 2013-10-30 2015-05-06 Sergej Michailovic Kiprijanov Multivalent antigen-binding protein molecules
WO2015063187A1 (en) * 2013-10-30 2015-05-07 Sergej Michailovic Kiprijanov Multivalent antigen-binding proteins
EP3514179A1 (en) 2014-01-24 2019-07-24 Dana-Farber Cancer Institute, Inc. Antibody molecules to pd-1 and uses thereof
EP4324518A2 (en) 2014-01-31 2024-02-21 Novartis AG Antibody molecules to tim-3 and uses thereof
EP3660050A1 (en) 2014-03-14 2020-06-03 Novartis AG Antibody molecules to lag-3 and uses thereof
WO2015138920A1 (en) 2014-03-14 2015-09-17 Novartis Ag Antibody molecules to lag-3 and uses thereof
WO2015142675A2 (en) 2014-03-15 2015-09-24 Novartis Ag Treatment of cancer using chimeric antigen receptor
US11512335B2 (en) 2014-04-29 2022-11-29 Novartis Ag Vertebrate cells and methods for recombinantly expressing a polypeptide of interest
US11345760B2 (en) 2014-06-25 2022-05-31 UCB Biopharma SRL Multispecific antibody constructs
EP3722316A1 (en) 2014-07-21 2020-10-14 Novartis AG Treatment of cancer using a cd33 chimeric antigen receptor
WO2016014553A1 (en) 2014-07-21 2016-01-28 Novartis Ag Sortase synthesized chimeric antigen receptors
WO2016014530A1 (en) 2014-07-21 2016-01-28 Novartis Ag Combinations of low, immune enhancing. doses of mtor inhibitors and cars
WO2016014565A2 (en) 2014-07-21 2016-01-28 Novartis Ag Treatment of cancer using humanized anti-bcma chimeric antigen receptor
EP3660042A1 (en) 2014-07-31 2020-06-03 Novartis AG Subset-optimized chimeric antigen receptor-containing t-cells
EP4205749A1 (en) 2014-07-31 2023-07-05 Novartis AG Subset-optimized chimeric antigen receptor-containing cells
WO2016025880A1 (en) 2014-08-14 2016-02-18 Novartis Ag Treatment of cancer using gfr alpha-4 chimeric antigen receptor
EP3712171A1 (en) 2014-08-19 2020-09-23 Novartis AG Treatment of cancer using a cd123 chimeric antigen receptor
WO2016044605A1 (en) 2014-09-17 2016-03-24 Beatty, Gregory Targeting cytotoxic cells with chimeric receptors for adoptive immunotherapy
EP3967709A1 (en) 2014-09-17 2022-03-16 Novartis AG Targeting cytotoxic cells with chimeric receptors for adoptive immunotherapy
EP4245376A2 (en) 2014-10-14 2023-09-20 Novartis AG Antibody molecules to pd-l1 and uses thereof
WO2016061142A1 (en) 2014-10-14 2016-04-21 Novartis Ag Antibody molecules to pd-l1 and uses thereof
US10633457B2 (en) 2014-12-03 2020-04-28 Hoffmann-La Roche Inc. Multispecific antibodies
WO2016090034A2 (en) 2014-12-03 2016-06-09 Novartis Ag Methods for b cell preconditioning in car therapy
CN107427577A (en) * 2015-03-31 2017-12-01 韦斯夸尔德有限公司 Peptidic constructs with the cleavable joint of protease
CN107454905A (en) * 2015-03-31 2017-12-08 韦斯夸尔德有限公司 Polypeptide
WO2016156466A1 (en) 2015-03-31 2016-10-06 Vhsquared Limited Peptide construct having a protease-cleavable linker
US10633438B2 (en) 2015-03-31 2020-04-28 Vhsquared Limited Polypeptides
JP2018515596A (en) * 2015-03-31 2018-06-14 ブイエイチスクエアード リミテッド Peptide construct having a protease-cleavable linker
CN107454905B (en) * 2015-03-31 2023-04-28 索里索制药公司 Polypeptides
JP7064670B2 (en) 2015-03-31 2022-05-11 ソリッソ ファーマシューティカルズ,インク. Peptide construct with a linker that can be cleaved by a protease
WO2016164731A2 (en) 2015-04-08 2016-10-13 Novartis Ag Cd20 therapies, cd22 therapies, and combination therapies with a cd19 chimeric antigen receptor (car) - expressing cell
EP4056588A1 (en) 2015-04-08 2022-09-14 Novartis AG Cd20 therapies, cd22 therapies, and combination therapies with a cd19 chimeric antigen receptor (car)- expressing cell
WO2016172583A1 (en) 2015-04-23 2016-10-27 Novartis Ag Treatment of cancer using chimeric antigen receptor and protein kinase a blocker
WO2017019894A1 (en) 2015-07-29 2017-02-02 Novartis Ag Combination therapies comprising antibody molecules to lag-3
EP3878465A1 (en) 2015-07-29 2021-09-15 Novartis AG Combination therapies comprising antibody molecules to tim-3
EP3964528A1 (en) 2015-07-29 2022-03-09 Novartis AG Combination therapies comprising antibody molecules to lag-3
WO2017019897A1 (en) 2015-07-29 2017-02-02 Novartis Ag Combination therapies comprising antibody molecules to tim-3
WO2017025698A1 (en) * 2015-08-11 2017-02-16 Queen Mary University Of London Bispecific, cleavable antibodies
WO2017106810A2 (en) 2015-12-17 2017-06-22 Novartis Ag Combination of c-met inhibitor with antibody molecule to pd-1 and uses thereof
WO2017106656A1 (en) 2015-12-17 2017-06-22 Novartis Ag Antibody molecules to pd-1 and uses thereof
EP3851457A1 (en) 2016-01-21 2021-07-21 Novartis AG Multispecific molecules targeting cll-1
WO2017125897A1 (en) 2016-01-21 2017-07-27 Novartis Ag Multispecific molecules targeting cll-1
WO2017149515A1 (en) 2016-03-04 2017-09-08 Novartis Ag Cells expressing multiple chimeric antigen receptor (car) molecules and uses therefore
WO2017165683A1 (en) 2016-03-23 2017-09-28 Novartis Ag Cell secreted minibodies and uses thereof
US10961311B2 (en) 2016-04-15 2021-03-30 Macrogenics, Inc. B7-H3 binding molecules, antibody drug conjugates thereof and methods of use thereof
EP4219721A2 (en) 2016-04-15 2023-08-02 Novartis AG Compositions and methods for selective protein expression
WO2017181119A2 (en) 2016-04-15 2017-10-19 Novartis Ag Compositions and methods for selective protein expression
US11591400B2 (en) 2016-04-15 2023-02-28 Macrogenics, Inc. B7-H3 directed antibody drug conjugates
WO2017210617A2 (en) 2016-06-02 2017-12-07 Porter, David, L. Therapeutic regimens for chimeric antigen receptor (car)- expressing cells
WO2018013918A2 (en) 2016-07-15 2018-01-18 Novartis Ag Treatment and prevention of cytokine release syndrome using a chimeric antigen receptor in combination with a kinase inhibitor
WO2018023025A1 (en) 2016-07-28 2018-02-01 Novartis Ag Combination therapies of chimeric antigen receptors adn pd-1 inhibitors
WO2018026819A2 (en) 2016-08-01 2018-02-08 Novartis Ag Treatment of cancer using a chimeric antigen receptor in combination with an inhibitor of a pro-m2 macrophage molecule
US11684677B2 (en) 2016-09-30 2023-06-27 Sorriso Pharmaceuticals, Inc. Compositions
WO2018067992A1 (en) 2016-10-07 2018-04-12 Novartis Ag Chimeric antigen receptors for the treatment of cancer
US11932697B2 (en) 2016-11-28 2024-03-19 Chugai Seiyaku Kabushiki Kaisha Antigen-binding domain, and polypeptide including conveying section
US11168139B2 (en) 2016-11-28 2021-11-09 Chugai Seiyaku Kabushiki Kaisha Antigen-binding domain, and polypeptide including conveying section
US11230610B2 (en) 2016-12-09 2022-01-25 Seagen Inc. Bivalent antibodies masked by coiled coils
EP4043485A1 (en) 2017-01-26 2022-08-17 Novartis AG Cd28 compositions and methods for chimeric antigen receptor therapy
WO2018140725A1 (en) 2017-01-26 2018-08-02 Novartis Ag Cd28 compositions and methods for chimeric antigen receptor therapy
WO2018160731A1 (en) 2017-02-28 2018-09-07 Novartis Ag Shp inhibitor compositions and uses for chimeric antigen receptor therapy
WO2018201051A1 (en) 2017-04-28 2018-11-01 Novartis Ag Bcma-targeting agent, and combination therapy with a gamma secretase inhibitor
WO2018201056A1 (en) 2017-04-28 2018-11-01 Novartis Ag Cells expressing a bcma-targeting chimeric antigen receptor, and combination therapy with a gamma secretase inhibitor
WO2018237157A1 (en) 2017-06-22 2018-12-27 Novartis Ag Antibody molecules to cd73 and uses thereof
WO2019006007A1 (en) 2017-06-27 2019-01-03 Novartis Ag Dosage regimens for anti-tim-3 antibodies and uses thereof
US11752207B2 (en) 2017-07-11 2023-09-12 Compass Therapeutics Llc Agonist antibodies that bind human CD137 and uses thereof
WO2019018730A1 (en) 2017-07-20 2019-01-24 Novartis Ag Dosage regimens of anti-lag-3 antibodies and uses thereof
WO2019089798A1 (en) 2017-10-31 2019-05-09 Novartis Ag Anti-car compositions and methods
US11718679B2 (en) 2017-10-31 2023-08-08 Compass Therapeutics Llc CD137 antibodies and PD-1 antagonists and uses thereof
WO2019099838A1 (en) 2017-11-16 2019-05-23 Novartis Ag Combination therapies
US11851497B2 (en) 2017-11-20 2023-12-26 Compass Therapeutics Llc CD137 antibodies and tumor antigen-targeting antibodies and uses thereof
WO2019152660A1 (en) 2018-01-31 2019-08-08 Novartis Ag Combination therapy using a chimeric antigen receptor
WO2019200229A1 (en) 2018-04-13 2019-10-17 Novartis Ag Dosage regimens for anti-pd-l1 antibodies and uses thereof
WO2019210153A1 (en) 2018-04-27 2019-10-31 Novartis Ag Car t cell therapies with enhanced efficacy
WO2019226658A1 (en) 2018-05-21 2019-11-28 Compass Therapeutics Llc Multispecific antigen-binding compositions and methods of use
WO2019226617A1 (en) 2018-05-21 2019-11-28 Compass Therapeutics Llc Compositions and methods for enhancing the killing of target cells by nk cells
WO2019227003A1 (en) 2018-05-25 2019-11-28 Novartis Ag Combination therapy with chimeric antigen receptor (car) therapies
WO2019230866A1 (en) * 2018-05-30 2019-12-05 Chugai Seiyaku Kabushiki Kaisha Polypeptide comprising il-1r1 binding domain and carrying moiety
WO2019232244A2 (en) 2018-05-31 2019-12-05 Novartis Ag Antibody molecules to cd73 and uses thereof
WO2019241426A1 (en) 2018-06-13 2019-12-19 Novartis Ag Bcma chimeric antigen receptors and uses thereof
WO2019246293A2 (en) 2018-06-19 2019-12-26 Atarga, Llc Antibody molecules to complement component 5 and uses thereof
WO2020012337A1 (en) 2018-07-10 2020-01-16 Novartis Ag 3-(5-amino-1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and their use in the treatment of i karos family zinc finger 2 (ikzf2)-dependent diseases
WO2020021465A1 (en) 2018-07-25 2020-01-30 Advanced Accelerator Applications (Italy) S.R.L. Method of treatment of neuroendocrine tumors
US11046769B2 (en) 2018-11-13 2021-06-29 Compass Therapeutics Llc Multispecific binding constructs against checkpoint molecules and uses thereof
WO2020128972A1 (en) 2018-12-20 2020-06-25 Novartis Ag Dosing regimen and pharmaceutical combination comprising 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives
WO2020128898A1 (en) 2018-12-20 2020-06-25 Novartis Ag Pharmaceutical combinations
WO2020165868A1 (en) 2019-02-15 2020-08-20 Perkinelmer Cellular Technologies Germany Gmbh Low-power microscope-objective pre-scan and high-power microscope-objective scan in x,y and z-direction for imaging objects such as cells using a microscope
WO2020165833A1 (en) 2019-02-15 2020-08-20 Novartis Ag 3-(1-oxo-5-(piperidin-4-yl)isoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof
WO2020165834A1 (en) 2019-02-15 2020-08-20 Novartis Ag Substituted 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof
WO2020172553A1 (en) 2019-02-22 2020-08-27 Novartis Ag Combination therapies of egfrviii chimeric antigen receptors and pd-1 inhibitors
WO2020205523A1 (en) 2019-03-29 2020-10-08 Atarga, Llc Anti fgf23 antibody
US11667719B2 (en) 2019-06-21 2023-06-06 Sorriso Pharmaceuticals, Inc. VHH immunoglobulin chain variable domain that binds to IL-7R and methods of use thereof for treating autoimmune and/or inflammatory diseases
US11623952B2 (en) 2019-06-21 2023-04-11 Sorriso Pharmaceuticals, Inc. IL-23 and TNF-alpha binding bi-specific heavy chain polypeptides
WO2021079188A1 (en) 2019-10-21 2021-04-29 Novartis Ag Combination therapies with venetoclax and tim-3 inhibitors
WO2021079195A1 (en) 2019-10-21 2021-04-29 Novartis Ag Tim-3 inhibitors and uses thereof
WO2021108661A2 (en) 2019-11-26 2021-06-03 Novartis Ag Chimeric antigen receptors and uses thereof
WO2021123996A1 (en) 2019-12-20 2021-06-24 Novartis Ag Uses of anti-tgf-beta antibodies and checkpoint inhibitors for the treatment of proliferative diseases
WO2021123902A1 (en) 2019-12-20 2021-06-24 Novartis Ag Combination of anti tim-3 antibody mbg453 and anti tgf-beta antibody nis793, with or without decitabine or the anti pd-1 antibody spartalizumab, for treating myelofibrosis and myelodysplastic syndrome
WO2021144657A1 (en) 2020-01-17 2021-07-22 Novartis Ag Combination comprising a tim-3 inhibitor and a hypomethylating agent for use in treating myelodysplastic syndrome or chronic myelomonocytic leukemia
WO2021146636A1 (en) 2020-01-17 2021-07-22 Becton, Dickinson And Company Methods and compositions for single cell secretomics
WO2021173995A2 (en) 2020-02-27 2021-09-02 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
WO2021260528A1 (en) 2020-06-23 2021-12-30 Novartis Ag Dosing regimen comprising 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives
WO2022013787A1 (en) 2020-07-16 2022-01-20 Novartis Ag Anti-betacellulin antibodies, fragments thereof, and multi-specific binding molecules
WO2022026592A2 (en) 2020-07-28 2022-02-03 Celltas Bio, Inc. Antibody molecules to coronavirus and uses thereof
WO2022029573A1 (en) 2020-08-03 2022-02-10 Novartis Ag Heteroaryl substituted 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof
WO2022043557A1 (en) 2020-08-31 2022-03-03 Advanced Accelerator Applications International Sa Method of treating psma-expressing cancers
WO2022043558A1 (en) 2020-08-31 2022-03-03 Advanced Accelerator Applications International Sa Method of treating psma-expressing cancers
WO2022097065A2 (en) 2020-11-06 2022-05-12 Novartis Ag ANTIBODY Fc VARIANTS
WO2022104061A1 (en) 2020-11-13 2022-05-19 Novartis Ag Combination therapies with chimeric antigen receptor (car)-expressing cells
WO2022162569A1 (en) 2021-01-29 2022-08-04 Novartis Ag Dosage regimes for anti-cd73 and anti-entpd2 antibodies and uses thereof
WO2022215011A1 (en) 2021-04-07 2022-10-13 Novartis Ag USES OF ANTI-TGFβ ANTIBODIES AND OTHER THERAPEUTIC AGENTS FOR THE TREATMENT OF PROLIFERATIVE DISEASES
WO2022243846A1 (en) 2021-05-18 2022-11-24 Novartis Ag Combination therapies
US11970538B2 (en) 2021-05-20 2024-04-30 Compass Therapeutics Llc Multispecific binding constructs against checkpoint molecules and uses thereof
WO2023044483A2 (en) 2021-09-20 2023-03-23 Voyager Therapeutics, Inc. Compositions and methods for the treatment of her2 positive cancer
WO2023092004A1 (en) 2021-11-17 2023-05-25 Voyager Therapeutics, Inc. Compositions and methods for the treatment of tau-related disorders
WO2023150778A1 (en) 2022-02-07 2023-08-10 Visterra, Inc. Anti-idiotype antibody molecules and uses thereof
WO2023220695A2 (en) 2022-05-13 2023-11-16 Voyager Therapeutics, Inc. Compositions and methods for the treatment of her2 positive cancer
WO2024030976A2 (en) 2022-08-03 2024-02-08 Voyager Therapeutics, Inc. Compositions and methods for crossing the blood brain barrier

Also Published As

Publication number Publication date
EP2178914A2 (en) 2010-04-28
JP5485152B2 (en) 2014-05-07
US20110229476A1 (en) 2011-09-22
US20170210825A1 (en) 2017-07-27
JP2014129374A (en) 2014-07-10
CA2696263C (en) 2017-06-13
WO2009021754A3 (en) 2009-06-18
US9624309B2 (en) 2017-04-18
JP2010535832A (en) 2010-11-25
EP2532676B1 (en) 2017-03-22
EP2532676A1 (en) 2012-12-12
CA2696263A1 (en) 2009-02-19
ES2628395T3 (en) 2017-08-02
JP5863201B2 (en) 2016-02-16

Similar Documents

Publication Publication Date Title
US9624309B2 (en) Monospecific and multispecific antibodies and method of use
US8168758B2 (en) Anti-MN antibodies and methods of using same
KR20190112040A (en) CD47 antigen binding unit and its use
EP1394253A1 (en) Anti-human ovarian cancer -anti-cd3 bispecific antibody
WO2017023780A1 (en) Antibody-drug conjugates for targeting cd56-positive tumors
WO2016022939A1 (en) Human monoclonal antibodies specific for 5t4 and methods of their use
AU2021247323A1 (en) Platform for constructing multispecific antibody
Elter et al. Protease-activation of fc-masked therapeutic antibodies to alleviate off-tumor cytotoxicity
Büyükköroğlu et al. Engineering monoclonal antibodies: Production and applications
CN112979790B (en) Antibodies and use in detecting novel coronaviruses
AU2021387127A1 (en) Her2 targeting agent
CN114641503A (en) anti-CD 371 antibodies and uses thereof
CN113461820B (en) anti-CD 3 humanized antibodies
AU2016200917A1 (en) Anti-MN antibodies and methods of using same
WO2023029089A1 (en) Anti-cd3 humanized antibody
US20230057263A1 (en) Single-and multi-chain polypeptides that bind specifically to cd3 epsilon
Wei et al. Expression and characterisation of recombinant human CD48 and isolation of a human anti‐CD48 monoclonal antibody by phage display
WO2023175117A1 (en) Antibodies against lypd3
CN115368457A (en) anti-TIGIT antibodies and uses thereof
WO2023086897A1 (en) Siglec-6 antibodies, derivative compounds and related uses
AU2012251927A1 (en) Anti-MN antibodies and methods of using same

Legal Events

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

Ref document number: 08785592

Country of ref document: EP

Kind code of ref document: A2

WWE Wipo information: entry into national phase

Ref document number: 2008785592

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2696263

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2010520502

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 12671942

Country of ref document: US