US20120308584A1 - Protein-active agent conjugates and method for preparing the same - Google Patents

Protein-active agent conjugates and method for preparing the same Download PDF

Info

Publication number
US20120308584A1
US20120308584A1 US13/466,875 US201213466875A US2012308584A1 US 20120308584 A1 US20120308584 A1 US 20120308584A1 US 201213466875 A US201213466875 A US 201213466875A US 2012308584 A1 US2012308584 A1 US 2012308584A1
Authority
US
United States
Prior art keywords
protein
active agent
amino acid
compound
mmol
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US13/466,875
Other languages
English (en)
Inventor
Yongzu Kim
Taekyo Park
Sungho Woo
Hyangsook Lee
Sunyoung Kim
Jongun Cho
Doohwan Jung
Youngun Kim
Hyunjin Kwon
Kyuman Oh
Yunseo Chung
Yun-hee Park
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Legochem Biosciences Inc
Original Assignee
Legochem Biosciences Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Legochem Biosciences Inc filed Critical Legochem Biosciences Inc
Priority to US13/466,875 priority Critical patent/US20120308584A1/en
Assigned to LEGOCHEM BIOSCIENCES, INC. reassignment LEGOCHEM BIOSCIENCES, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CHO, JONGUN, MR., CHUNG, YUNSEO, MR., JUNG, DOOHWAN, MR., KIM, SUNYOUNG, MR., KIM, YONGZU, MR., KIM, YOUNGUN, MR., KWON, HYUNJIN, MR., LEE, HYANGSOOK, MR., OH, KYUMAN, MR., PARK, TAEKYO, MR., PARK, YUN-HEE, MR., WOO, SUNGHO
Publication of US20120308584A1 publication Critical patent/US20120308584A1/en
Priority to US14/181,648 priority patent/US20140161829A1/en
Priority to US14/181,649 priority patent/US20140187756A1/en
Priority to US14/517,616 priority patent/US9669107B2/en
Priority to US15/054,257 priority patent/US20160213786A1/en
Priority to US15/612,766 priority patent/US10583197B2/en
Priority to US16/812,690 priority patent/US20200289662A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • 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
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • 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
    • A61K47/54Medicinal 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 organic compound
    • A61K47/549Sugars, nucleosides, nucleotides or nucleic acids
    • 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
    • A61K47/54Medicinal 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 organic compound
    • A61K47/55Medicinal 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 organic compound the modifying agent being also a pharmacologically or therapeutically active agent, i.e. the entire conjugate being a codrug, i.e. a dimer, oligomer or polymer of pharmacologically or therapeutically active compounds
    • 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
    • A61K47/62Medicinal 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 a protein, peptide or polyamino acid
    • A61K47/65Peptidic linkers, binders or spacers, e.g. peptidic enzyme-labile linkers
    • 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
    • A61K47/6811Drugs 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
    • 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
    • A61K47/6849Medicinal 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 receptor, a cell surface antigen or a cell surface determinant
    • 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
    • A61K47/6855Medicinal 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 breast cancer 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
    • A61K47/6859Medicinal 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 liver or pancreas cancer 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
    • A61K47/6889Conjugates wherein the antibody being the modifying agent and wherein the linker, binder or spacer confers particular properties to the conjugates, e.g. peptidic enzyme-labile linkers or acid-labile linkers, providing for an acid-labile immuno conjugate wherein the drug may be released from its antibody conjugated part in an acidic, e.g. tumoural or environment
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/10Antimycotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2863Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for growth factors, growth regulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/32Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against translation products of oncogenes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/48Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving transferase

Definitions

  • the present disclosure relates to a protein-active agent conjugate.
  • the protein e.g., an oligopeptide, a polypeptide, an antibody, or the like
  • the active agent e.g., a drug, a toxin, a ligand, a detection probe, and the like
  • the disclosure also relates to methods for preparing the conjugate.
  • the disclosure further relates to methods of using the conjugate to deliver an active agent to a target cell in a subject, as well as methods for treating a subject in need of the active agent (e.g., a subject having cancer).
  • Targeted delivery of an antibody-drug conjugate can kill a particular cancer cell.
  • the antibody or antibody fragment
  • the drug is delivered to the target cancer cell.
  • Targeted delivery of the drug ensures that the drug acts on the target cancer cell instead of normal host cells, thereby minimizing the side effects resulting from damage to normal cells.
  • Antibody conjugates can be used to deliver chemical and/or biological molecules.
  • exemplary chemical and/or biological molecules include a drug conventionally used in chemical treatment, a bacterial protein toxin (e.g., diphtheria toxin), a plant protein toxin (e.g., ricin), a small molecule toxin (e.g., auristatin, geldanamycin, maytansinoid, calicheamycin, daunomycin, methotrexate, vindesine, and tubulysin), an affinity ligand, a detection probe (e.g., fluorescent probe, radioactive probe), and the like (including combinations thereof).
  • a bacterial protein toxin e.g., diphtheria toxin
  • a plant protein toxin e.g., ricin
  • a small molecule toxin e.g., auristatin, geldanamycin, maytansinoid, calicheamycin, daunomycin, met
  • Antibody-drug conjugates that have been proposed thus far are prepared by bonding a drug moiety with a plurality of lysine groups of an antibody.
  • antibody-drug conjugates are prepared by reducing all or part of the interchain disulfide groups of an antibody or reducing all the interchain disulfide groups followed by partial oxidation to thereby give free cysteine thiol groups, and then bonding the free cysteine thiol groups with a drug moiety.
  • the electric charge of the lysine groups may be lost, thereby causing the antibody to lose its unique antigen specificity.
  • the tertiary or quaternary structure of the antibody may not be maintained when preparing antibody-drug conjugates by reducing disulfide groups, thereby causing the antibody to be inactivated or become a non-specific antibody.
  • the drug may be cleaved (non specifically) from the conjugates via, e.g., a reverse reaction.
  • an alternative method was proposed in which amino acid groups in particular positions of an antibody are replaced with cysteine groups. Although this method shows better result than the prior preparation methods in terms of toxicity, activity, and safety, this method still involves thiol-maleimide bonding and thus suffers from the diastereomer and instability problems associated with thiol-maleimide bonding.
  • Another alternative method was proposed in which selenocysteine groups are attached to the carboxy terminals of an antibody.
  • Ambrx Technology http://www.ambrx.com
  • Ambrx's expression systems contain tRNA synthetases that aminoacylate the original tRNA with a non-natural amino acid, thereby inserting a non-natural amino acid whenever the amber stop is encountered.
  • Redwood Bioscience's (http://www.redwoodbioscience.com) technology employs genetically encoded aldehyde tags and aims to exploit a specific sequence that is posttranslationally recognized and modified by an enzyme, i.e., a formyl glycine-generating enzyme, to produce a so-called aldehyde chemical handle.
  • an enzyme i.e., a formyl glycine-generating enzyme
  • the present invention generally features protein-active agent conjugates and methods for making the protein-active agent conjugates.
  • the invention also features methods for delivering the protein-active agent conjugate to a target cell in a subject, as well as methods for treating a subject in need of the active agent.
  • the protein-active agent conjugates of the invention can be produced homogeneously and advantageously used for targeted treatment of a disease.
  • the invention provides protein-active agent conjugates.
  • the protein has an amino acid motif that can be recognized by an isoprenoid transferase.
  • the active agent is covalently linked to the protein at the amino acid motif.
  • the protein has a deletion in the carboxy terminus of the protein.
  • the modification is attached to the amino acid motif.
  • the protein has an oligopeptide or polypeptide addition in the carboxy terminus of the protein.
  • the modification is attached to the amino acid motif.
  • the protein has a deletion in the carboxy terminus of the protein and an oligopeptide or polypeptide addition in the carboxy terminus of the protein.
  • the modification is attached to the amino acid motif.
  • the protein is an antibody or a fragment of an antigenic polypeptide.
  • the protein is a monoclonal antibody.
  • at least one light chain and/or at least one heavy chain of the monoclonal antibody comprises an amino acid region having the amino acid motif.
  • the isoprenoid transferase is FTase or GGTase.
  • the active agent is a drug, a toxin, an affinity ligand, a detection probe, or a combination thereof.
  • the amino acid motif is CAAX, XXCC, XCXC, or CXX, wherein C represents cysteine, A represents an aliphatic amino acid, and X represents an amino acid that determines a substrate specificity of the isoprenoid transferase.
  • the amino acid motif is covalently linked to the active agent via at least one linker.
  • the linker is an isoprenyl derivative that can be recognized by the isoprenoid transferase.
  • the linker is represented by the following formula (I):
  • P 1 and Y is independently a group containing a first functional group (FG1), the FG1 being selected from the group consisting of: acetylene, azide, aldehyde, hydroxylamine, hydrazine, ketone, nitrobenzofurazan (NBD), dansyl, fluorescein, biotin, and Rhodamin,
  • FG1 first functional group
  • L 1 is (CH 2 ) r X q —CH 2 ) p ,
  • X is oxygen, sulfur, —NR 1 —, —C(O)NR 1 —, —NR 1 C(O)—, —NR 1 SO 2 —, —SO 2 NR 1 —, —(CH ⁇ CH)—, or acetylene,
  • R 1 is hydrogen, C 1-6 alkyl, C 1-6 alkyl aryl, or C 1-6 alkyl heteroaryl
  • r and p is independently an integer of 0 to 6
  • q is an integer of 0 to 1
  • n is an integer of 1 to 4.
  • the active agent is attached to a group containing a second functional group (FG2) that can react with the FG1.
  • FG2 is an acetylene, hydroxylamine, azide, aldehyde, hydrazine, ketone, or amine.
  • the active agent is attached to the group containing an FG2 via —(CH 2 ) r X q (CH 2 ) p — or —[ZCH 2 CH 2 O(CH 2 CH 2 O) w CH 2 CH 2 Z]—, in which
  • X is oxygen, sulfur, —NR 1 —, —C(O)NR 1 —, —NR 1 C(O)—, SO 2 —, or —SO 2 NR 1 —,
  • Z is oxygen, sulfur or NR 1 ,
  • R 1 is hydrogen, C 1-6 alkyl, C 1-6 alkyl aryl, or C 1-6 alkyl heteroaryl
  • r and p is independently an integer of 0 to 6
  • q is an integer of 0 to 1
  • n is an integer of 0 to 6.
  • the —(CH 2 ) r X q (CH 2 ) p — or —[ZCH 2 CH 2 O(CH 2 CH 2 O) w CH 2 CH 2 Z]- is attached to (i) a peptide(s) that can be cleaved by cathepsin B or (ii) a glucuronide that can be cleaved by ⁇ -glucuronidase.
  • the peptide that can be cleaved by cathepsin B is
  • the glucuronide that can be cleaved by ⁇ -glucuronidase is
  • the invention provides methods for preparing any of the protein-active agent conjugate described herein.
  • the methods involve expressing a protein having an amino acid motif that can be recognized by an isoprenoid transferase.
  • the methods involve enzymatically reacting, with the isoprenoid transferase, the expressed protein and at least one isosubstrate having a first functional group (FG1), thereby producing a functionalized protein.
  • the methods involve attaching a second functional group (FG2) to an active agent, thereby producing a functionalized active agent.
  • the methods involve reacting the functionalized protein with the functionalized active agent, thereby producing the protein-active agent conjugate.
  • the amino acid motif is in the carboxy terminus of the protein.
  • the amino acid motif is CAAX, XXCC, XCXC, or CXX, wherein C represents cysteine, A represents an aliphatic amino acid, and X represents an amino acid that determines the substrate specificity of the isoprenoid transferase.
  • the amino acid motif is CAAX
  • the method further comprises removing AAX from the amino acid motif after step (b).
  • the FG2 is attached to the active agent by at least one linker.
  • the reaction between the functionalized protein and the functionalized active agent is click chemistry reaction or a hydrazone and/or oxime formation.
  • the FG1 is an azide group and the FG2 is an acetylene group. In embodiments, the FG1 is an acetylene group and the FG2 is an azide group. In embodiments, the FG1 is an aldehyde or ketone group and the FG2 is a hydrazine or hydroxylamine. In embodiments, the FG1 is hydrazine or hydroxylamine and the FG2 is an aldehyde or ketone.
  • the invention provides methods for preparing any of the protein-active agent conjugate described herein, and the methods involve expressing a protein having an amino acid motif that can be recognized by an isoprenoid transferase.
  • the methods involve attaching an isosubstrate of an isoprenoid transferase to an active agent.
  • the methods involve enzymatically reacting, with the isoprenoid transferase, the expressed protein and the active agent attached to the isosubstrate.
  • the amino acid motif is in the carboxy terminus of the protein.
  • the amino acid motif is CAAX, XXCC, XCXC, or CXX, wherein C represents cysteine, A represents an aliphatic amino acid, and X represents an amino acid that determines the substrate specificity of the isoprenoid transferase.
  • the isosubstrate is attached to the active agent by at least one linker.
  • the invention provides compositions containing any of the protein-active agent conjugates described herein.
  • the composition is a homogeneous mixture of the protein-active agent conjugate.
  • the protein is an antibody or a fragment of an antigenic polypeptide.
  • the invention provides methods for delivering an active agent to a target cell in a subject.
  • the methods involve administering at least one of the protein-active agent conjugates or compositions described herein.
  • the target cell is a cancer cell.
  • the invention provides methods for treating a subject in need thereof (i.e., in need of the active agent).
  • the methods involve administering at least one of the protein-active agent conjugates or compositions described herein.
  • the subject has cancer.
  • the subject has an infection with a pathogenic agent.
  • the pathogenic agent may be a virus, bacteria, fungus, or parasite.
  • the active agent may be an immunomodulatory compound, an anti-cancer agent, an anti-viral agent, an anti-bacterial agent, an anti-fungal agent, or an anti-parasitic agent.
  • FIG. 1 shows an amino acid sequence of a modified Herceptin antibody (Herceptin-HC-GCVIM) prepared by inserting GCVIM to the C-terminus of the heavy chain of Herceptin.
  • Herceptin-HC-GCVIM modified Herceptin antibody
  • FIG. 2 shows an amino acid sequence of a modified Herceptin antibody (Herceptin-LC-GCVIM) prepared by inserting GCVIM to the C-terminus of the light chain of Herceptin.
  • Herceptin-LC-GCVIM modified Herceptin antibody
  • FIG. 3 shows an amino acid sequence of a modified Herceptin antibody (Herceptin-HC-G 5 CVIM) prepared by inserting G 5 CVIM to the C-terminus of the heavy chain of Herceptin.
  • Herceptin-HC-G 5 CVIM modified Herceptin antibody
  • FIG. 4 shows an amino acid sequence of a modified Herceptin antibody (Herceptin-LC-G 5 CVIM) prepared by inserting G 5 CVIM to the C-terminus of the light chain of Herceptin.
  • Herceptin-LC-G 5 CVIM modified Herceptin antibody
  • FIG. 5 shows an amino acid sequence of a modified Herceptin antibody (Herceptin-HC-G 7 CVIM) prepared by inserting G 7 CVIM to the C-terminus of the heavy chain of Herceptin.
  • Herceptin-HC-G 7 CVIM modified Herceptin antibody
  • FIG. 6 shows an amino acid sequence of a modified Herceptin antibody (Herceptin-LC-G 7 CVIM) prepared by inserting G 7 CVIM to the C-terminus of the light chain of Herceptin.
  • Herceptin-LC-G 7 CVIM modified Herceptin antibody
  • FIG. 7 shows an amino acid sequence of a modified Herceptin antibody (Herceptin-HC-G 10 CVIM) prepared by inserting G 10 CVIM to the C-terminus of the heavy chain of Herceptin.
  • Herceptin-HC-G 10 CVIM modified Herceptin antibody
  • FIG. 8 shows an amino acid sequence of a modified Herceptin antibody (Herceptin-LC-G 10 CVIM) prepared by inserting G 10 CVIM to the C-terminus of the light chain of Herceptin.
  • Herceptin-LC-G 10 CVIM modified Herceptin antibody
  • FIG. 9 shows an amino acid sequence of a modified Herceptin antibody (Herceptin-HC-G 10 CVLL) prepared by inserting G 10 CVLL to the C-terminus of the heavy chain of Herceptin.
  • Herceptin-HC-G 10 CVLL modified Herceptin antibody
  • FIG. 10 shows an amino acid sequence of a modified Herceptin antibody (Herceptin-LC-G 10 CVLL) prepared by inserting G 10 CVLL to the C-terminus of the light chain of Herceptin.
  • Herceptin-LC-G 10 CVLL modified Herceptin antibody
  • FIG. 11 shows an SDS-PAGE gel analyzing a modified anti cMET antibody (anti cMET-HC-G 7 CVIM) prepared by inserting G 7 CVIM to the C-terminus of the heavy chain of anti cMET antibody, a modified anti cMET antibody (anti cMET-LC-G 7 CVIM) prepared by inserting G 7 CVIM to the C-terminus of the light chain of anti cMET antibody, a modified anti cMET antibody (anti cMET-HC-G 10 CVIM) prepared by inserting G 10 CVIM to the C-terminus of the heavy chain of anti cMET antibody, and a modified anti cMET antibody (anti cMET-LC-G 10 CVIM) prepared by inserting G 10 CVIM to the C-terminus of the light chain of anti cMET antibody.
  • a modified anti cMET antibody anti cMET-HC-G 7 CVIM
  • FIG. 12 shows an SDS-PAGE gel analyzing prenylation of Herceptin-HC-G n CVIM by using FTase and NBD-GPP.
  • FIG. 13 shows an SDS-PAGE gel analyzing prenylation of Herceptin-LC-G n CVIM by using FTase and NBD-GPP.
  • FIG. 14 shows an SDS-PAGE gel analyzing prenylation of cMET-HC-G n CVIM by using FTase and NBD-GPP.
  • FIG. 15 shows an SDS-PAGE gel analyzing prenylation of cMET-LC-G n CVIM by using FTase and NBD-GPP.
  • FIG. 16 shows an SDS-PAGE gel analyzing prenylation of Herceptin-HC-G 10 CVLL and Herceptin-LC-G 10 CVLL by using FTase/NBD-GPP or GGTase I/NBD-FPP.
  • FIG. 17 shows the results from LC/MS analysis of a prenylated Herceptin-LC-G 7 CVIM.
  • FIG. 18 shows the results from LC/MS analysis of a prenylated Herceptin-LC-G 10 CVIM.
  • FIG. 19 shows the results from LC/MS and deconvoluted mass spectra analysis of LCB14-0104 (Herceptin-LC-G 7 CVIM-NC-MMAF-Ome).
  • FIG. 20 shows the HIC-HPLC chromatograms of Herceptin-LC-G 7 CVIM, prenylated Herceptin-LC-G 7 CVIM, and LCB14-0101 (Herceptin-LC-G 7 CVIM-BG-MMAF).
  • FIG. 21 shows the results from an anti-proliferation assay of LCB14-0101 (Herceptin-LC-G 7 CVIM-BG-MMAF) with breast cancer cell lines MCF-7, MDA-MB-468, and SK-BR-3.
  • FIG. 22 shows the results from an anti-proliferation assay of LCB14-0102 (Herceptin-LC-G 7 CVIM-VC-MMAF-OMe) with breast cancer cell lines MCF-7 and SK-BR-3.
  • FIG. 23 shows the results from an anti-proliferation assay of LCB14-0103 (Herceptin-LC-G 7 CVIM-BG-MMAE) with breast cancer cell lines MCF-7 and SK-BR-3.
  • FIG. 24 shows a process of posttranslational modification of a protein (C-terminal CVIM).
  • FIG. 25 shows a mechanism of release of active drugs (except non-cleavable linker).
  • FIG. 26 shows the chemical structures of antibody-drug conjugates LCB14-0101, LCB14-0102, LCB14-0103, and LCB14-0104.
  • FIG. 27 is a schematic diagram depicting a process for preparing a protein-active agent conjugate by using an isoprenoid transferase and an isosubstrate thereof in which cysteine of the CAAX motif is alkylated.
  • agent or “active agent” is meant any small molecule chemical compound, antibody, nucleic acid molecule, or polypeptide, or fragments thereof. Examples include, but are not limited to, a drug, a toxin, an affinity ligand, a detection probe, or a combination thereof.
  • an analog is meant a molecule that is not identical, but has analogous functional or structural features.
  • a polypeptide analog retains the biological activity of a corresponding naturally-occurring polypeptide, while having certain biochemical modifications that enhance the analog's function relative to a naturally occurring polypeptide. Such biochemical modifications could increase the analog's protease resistance, membrane permeability, or half-life, without altering, for example, ligand binding.
  • An analog may include an unnatural amino acid.
  • Contacting a cell is understood herein as providing an agent to a cell e.g., a cell to be treated in culture, ex vivo, or in an animal, such that the agent can interact with the cell (e.g., cell to be treated), potentially be taken up by the cell, and have an effect on the cell.
  • the agent e.g., an adjuvant
  • the agent can be delivered to the cell directly (e.g., by addition of the agent to culture medium or by injection into the cell or tissue of interest), or by delivery to the organism by a topical or parenteral route of administration for delivery to the cell by vascular, lymphatic, or other means.
  • administration of the protein-active agent conjugates of the invention to a subject involves contacting the protein-active agent conjugate with a cell of the subject.
  • disease is meant any condition or disorder that damages or interferes with the normal function of a cell, tissue, or organ.
  • an effective amount refers to that amount of an agent to produce the intended pharmacological, therapeutic, or preventive result.
  • the pharmacologically effective amount results in the prevention or delay of onset of disease, either in an individual or in the frequency of disease in a population. More than one dose may be required to provide an effective dose. It is understood that an effective dose in one population may or may not be sufficient in all populations.
  • the agent or immunogenic composition is “effective against” a disease or condition when administration in a clinically appropriate manner results in a beneficial effect for at least a statistically significant fraction of subjects, such as a prevention of disease onset, improvement of symptoms, a cure, a reduction in disease signs or symptoms, extension of life, improvement in quality of life, or other effect generally recognized as positive by medical doctors familiar with treating the particular type of disease or condition.
  • fragment is meant a portion of a polypeptide or nucleic acid molecule. This portion contains, preferably, at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90% of the entire length of the reference nucleic acid molecule or polypeptide.
  • a fragment may contain 10, 20, 30, 40, 50, 60, 70, 80, 90, or 100, 200, 300, 400, 500, 600, 700, 800, 900, or 1000 nucleotides or amino acids.
  • Hybridization means hydrogen bonding, which may be Watson-Crick, Hoogsteen or reversed Hoogsteen hydrogen bonding, between complementary nucleotide bases.
  • adenine and thymine are complementary nucleotide bases that pair through the formation of hydrogen bonds.
  • phrases “pharmaceutically acceptable carrier, excipient, or diluent” is art recognized and includes a pharmaceutically acceptable material, composition or vehicle, suitable for administering compounds of the present invention to mammals.
  • pharmaceutically acceptable means being approved by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopia, European Pharmacopia or other generally recognized pharmacopia for use in mammals, e.g., humans.
  • reduces is meant a negative alteration of at least 10%, 25%, 50%, 75%, 100%, or any number therebetween.
  • sample refers to a biological material that is isolated from its environment (e.g., blood or tissue from an animal, cells, or conditioned media from tissue culture).
  • the sample is suspected of containing, or known to contain an analyte, such as a protein of interest (e.g., antibody, cytokine, and the like).
  • a sample can also be a partially purified fraction of a tissue or bodily fluid.
  • a reference sample can be a “normal” sample, from a donor not having the disease or condition fluid, or from a normal tissue in a subject having the disease or condition, or an untreated subject (e.g., a subject not treated with the vaccine).
  • a reference sample can also be taken at a “zero time point” prior to contacting the cell or subject with the agent or therapeutic intervention to be tested.
  • telomere binding By “specifically binds” is meant recognition and binding to a target (e.g., polypeptide, cell, and the like), but which does not substantially recognize and bind other molecules in a sample, for example, a biological sample.
  • a target e.g., polypeptide, cell, and the like
  • a “subject” as used herein refers to a living organism.
  • the living organism is an animal.
  • the subject is a mammal.
  • the subject is a domesticated mammal or a primate including a non-human primate. Examples of subjects include, but are not limited to, humans, monkeys, dogs, cats, mice, rats, cows, horses, swine, goats, sheep, and birds.
  • a subject may also be referred to as a patient.
  • a subject “suffering from or suspected of suffering from” a specific disease, condition, or syndrome has a sufficient number of risk factors or presents with a sufficient number or combination of signs or symptoms of the disease, condition, or syndrome such that a competent individual would diagnose or suspect that the subject was suffering from the disease, condition, or syndrome.
  • Methods for identification of subjects suffering from or suspected of suffering from a disease or condition is within the ability of those in the art.
  • Subjects suffering from, and suspected of suffering from, a specific disease, condition, or syndrome are not necessarily two distinct groups.
  • a subject in need of an active agent may also be a subject suffering from or suspected of suffering from a specific disease, condition, or syndrome.
  • the terms “treat,” “treating,” “treatment,” and the like refer to reducing or ameliorating a disorder and/or symptoms associated therewith (e.g., cancer or cancer associated symptoms). It will be appreciated that, although not precluded, treating a disorder or condition does not require that the disorder, condition or symptoms associated therewith be completely eliminated.
  • Ranges provided herein are understood to be shorthand for all of the values within the range.
  • a range of 1 to 50 is understood to include any number, combination of numbers, or sub-range from the group consisting 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, or 50.
  • the term “about” is understood as within a range of normal tolerance in the art, for example within 2 standard deviations of the mean. About can be understood as within 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, 0.5%, 0.1%, 0.05%, or 0.01% of the stated value. Unless otherwise clear from context, all numerical values provided herein can be modified by the term about.
  • compositions or methods provided herein can be combined with one or more of any of the other compositions and methods provided herein.
  • a method for preparing a protein-active agent conjugate comprises: (a) expressing a protein having an amino acid motif that can be recognized by an isoprenoid transferase; (b) enzymatically reacting, using the isoprenoid transferase, the expressed protein and at least one isosubstrate having a first functional group (FG1), thereby producing a functionalized protein; (c) attaching a second functional group (FG2) to an active agent, thereby producing a functionalized active agent; and (d) reacting the functionalized protein with the functionalized active agent, thereby producing the protein-active agent conjugate.
  • FG1 first functional group
  • FG2 second functional group
  • protein used herein is understood as two or more independently selected natural or non-natural amino acids joined by a covalent bond (e.g., a peptide bond).
  • a peptide can include 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, or more natural or non-natural amino acids joined by peptide bonds.
  • Polypeptides as described herein include full length proteins (e.g., fully processed proteins) as well as shorter amino acids sequences (e.g., fragments of naturally occurring proteins or synthetic polypeptide fragments).
  • a protein refers to an oligopeptide or polypeptide containing at least one C-terminus and at least one N-terminus.
  • the term is used herein to include an intact oligopeptide or polypeptide, a modified form thereof, a fragment thereof, and analogs thereof.
  • the term can refer to an oligopeptide or polypeptide, or an oligopeptide or polypeptide modified by attaching thereto an amino acid sequence that can be recognized by an isoprenoid transferase.
  • fragment used herein refers to a portion of the amino acid sequence consisting of an oligopeptide or polypeptide.
  • the term is used herein to include a portion of the amino acid sequence that has the substrate specificity of the oligopeptide or polypeptide.
  • the term “analog” refers to an oligopeptide or polypeptide having a sequence identity of at least 70% or 75%, at least 80% or 85%, at least 90%, 91%, 92%, 93%, 94%, or 95%, or at least 96, 97%, 98%, or 9% with a reference oligopeptide or polypeptide.
  • protein used herein also includes an antibody a fragment of an antigenic polypeptide, or an analog or derivative thereof.
  • antibody means an immunoglobulin molecule that recognizes and specifically binds to a target, such as a protein, polypeptide, peptide, carbohydrate, polynucleotide, lipid, or combinations of the foregoing through at least one antigen recognition site within the variable region of the immunoglobulin molecule.
  • antibody encompasses intact polyclonal antibodies, intact monoclonal antibodies, antibody fragments (such as Fab, Fab′, F(ab′) 2 , Fd, and Fv fragments), single chain Fv (scFv) mutants, multispecific antibodies such as bispecific antibodies generated from at least two intact antibodies, chimeric antibodies, humanized antibodies, human antibodies, fusion proteins comprising an antigen determination portion of an antibody, and any other modified immunoglobulin molecule comprising an antigen recognition site so long as the antibodies exhibit the desired biological activity.
  • antibody fragments such as Fab, Fab′, F(ab′) 2 , Fd, and Fv fragments
  • scFv single chain Fv mutants
  • multispecific antibodies such as bispecific antibodies generated from at least two intact antibodies, chimeric antibodies, humanized antibodies, human antibodies, fusion proteins comprising an antigen determination portion of an antibody, and any other modified immunoglobulin molecule comprising an antigen recognition site so long as the antibodies exhibit the desired biological activity.
  • An antibody can be of any the five major classes of immunoglobulins: IgA, IgD, IgE, IgG, and IgM, or subclasses (isotypes) thereof (e.g., IgG1, IgG2, IgG3, IgG4, IgA1 and IgA2), based on the identity of their heavy-chain constant domains referred to as alpha, delta, epsilon, gamma, and mu, respectively.
  • the different classes of immunoglobulins have different and well known subunit structures and three-dimensional configurations.
  • antibody fragment refers to a portion of an intact antibody and refers to the antigenic determining variable regions of an intact antibody.
  • antibody fragments include, but are not limited to Fab, Fab′, F(ab′) 2 , Fd, and Fv fragments, linear antibodies, single chain antibodies, and multispecific antibodies formed from antibody fragments.
  • a “monoclonal antibody” refers to homogenous antibody population involved in the highly specific recognition and binding of a single antigenic determinant, or epitope. This is in contrast to polyclonal antibodies that typically include different antibodies directed against different antigenic determinants.
  • the term “monoclonal antibody” encompasses both intact and full-length monoclonal antibodies as well as antibody fragments (such as Fab, Fab′, F(ab′)2, Fd, Fv), single chain (scFv) mutants, fusion proteins comprising an antibody portion, and any other modified immunoglobulin molecule comprising an antigen recognition site.
  • “monoclonal antibody” refers to such antibodies made in any number of manners including but not limited to by hybridoma, phage selection, recombinant expression, and transgenic animals.
  • humanized antibody refers to forms of non-human (e.g., murine) antibodies that are specific immunoglobulin chains, chimeric immunoglobulins, or fragments thereof that contain minimal non-human (e.g., murine) sequences.
  • humanized antibodies are human immunoglobulins in which residues from the complementary determining region (CDR) are replaced by residues from the CDR of a non-human species (e.g., mouse, rat, rabbit, hamster) that have the desired specificity, affinity, and capability (Jones et al., 1986 , Nature, 321:522-525; Riechmann et al., 1988 , Nature, 332:323-327; Verhoeyen et al., 1988 , Science, 239:1534-1536).
  • CDR complementary determining region
  • the Fv framework region (FR) residues of a human immunoglobulin are replaced with the corresponding residues in an antibody from a non-human species that has the desired specificity, affinity, and capability.
  • the humanized antibody can be further modified by the substitution of additional residue either in the Fv framework region and/or within the replaced non-human residues to refine and optimize antibody specificity, affinity, and/or capability.
  • the humanized antibody will comprise substantially all of at least one, and typically two or three, variable domains containing all or substantially all of the CDR regions that correspond to the non-human immunoglobulin whereas all or substantially all of the FR regions are those of a human immunoglobulin consensus sequence.
  • the humanized antibody can also comprise at least a portion of an immunoglobulin constant region or domain (Fc), typically that of a human immunoglobulin. Examples of methods used to generate humanized antibodies are described in U.S. Pat. No. 5,225,539.
  • human antibody means an antibody produced by a human or an antibody having an amino acid sequence corresponding to an antibody produced by a human made using any technique known in the art. This definition of a human antibody includes intact or full-length antibodies, fragments thereof, and/or antibodies comprising at least one human heavy and/or light chain polypeptide such as, for example, an antibody comprising murine light chain and human heavy chain polypeptides.
  • chimeric antibodies refers to antibodies wherein the amino acid sequence of the immunoglobulin molecule is derived from two or more species.
  • the variable region of both light and heavy chains corresponds to the variable region of antibodies derived from one species of mammals (e.g., mouse, rat, rabbit, etc) with the desired specificity, affinity, and capability while the constant regions are homologous to the sequences in antibodies derived from another (usually human) to avoid eliciting an immune response in that species.
  • epitopes or “antigenic determinant” are used interchangeably herein and refer to that portion of an antigen capable of being recognized and specifically bound by a particular antibody.
  • the antigen is a polypeptide
  • epitopes can be formed both from contiguous amino acids and noncontiguous amino acids juxtaposed by tertiary folding of a protein. Epitopes formed from contiguous amino acids are typically retained upon protein denaturing, whereas epitopes formed by tertiary folding are typically lost upon protein denaturing.
  • An epitope typically includes at least 3, at least 5, or at least 8-10 amino acids in a unique spatial conformation.
  • an antibody “specifically binds” to an epitope or antigenic molecule means that the antibody reacts or associates more frequently, more rapidly, with greater duration, with greater affinity, or with some combination of the above to an epitope or antigenic molecule than with alternative substances, including unrelated proteins.
  • “specifically binds” means, for instance, that an antibody binds to a protein with a K D of about 0.1 mM or less, but more usually less than about 1 ⁇ M.
  • “specifically binds” means that an antibody binds to a protein at times with a K D of at least about 0.1 ⁇ M or less, and at other times at least about 0.01 ⁇ M or less.
  • specific binding can include an antibody that recognizes a particular protein in more than one species. It is understood that an antibody or binding moiety that specifically binds to a first target may or may not specifically bind to a second target. As such, “specific binding” does not necessarily require (although it can include) exclusive binding, i.e. binding to a single target. Generally, but not necessarily, reference to binding means specific binding.
  • the antibodies can be obtained using known methods in the art. (See McCafferty et al., Nature 348:552-554 (1990); Clackson et al., Nature 352:624-628; Marks et al., J. Mol. Biol. 222:581-597 (1991); Marks et al., Bio/Technology 10:779-783 (1992); Waterhouse et al., Nucleic. Acids Res.
  • Non-limiting examples of the antibody include, but not limited to, Muromonab-CD3 Abciximab, Rituximab, Daclizumab, Palivizumab, lnfliximab, Trastuzumab, Etanercept, Basiliximab, Gemtuzumab ozogamicin, Alemtuzumab, Ibritumomab tiuxetan, Adalimumab, Alefacept, Omalizumab, Efalizumab, Tositumomob-1131, Cetuximab, Bevacizumab, Natalizumab, Ranibizumab, Panitumumab, Ecolizumab, Rilonacept, Certolizumab pegol, Romiplostim, AMG-531, CNTO-148, CNTO-1275, ABT-874, LEA-29Y, Belimumab, TACl-Ig, 2nd gen.
  • anti-CD20 ACZ-885, Tocilizumab (Atlizumab), Mepolizumab, Pertuzumab, Humax CD20, CP-675, 206 (Ticilimumab), MDX-010, IDEC-114, Inotuzumab ozogamycin, HuMax EGFR, Aflibercept, VEGF Trap-Eye, HuMax-CD4, Ala-Ala, ChAgIyCD3; TRX4, Catumaxomab, IGN101, MT-201, Pregovomab, CH-14.18, WX-G250, AMG-162, AAB-001, Motavizumab; MEDI-524, efumgumab, Aurograb®, Raxibacumab, 3rd gen. anti-CD20, LY2469298, Veltuzumab.
  • At least one light chain of the monoclonal antibody, at least one heavy chain of the monoclonal antibody, or both may comprise an amino acid region having an amino acid motif that can be recognized an isoprenoid transferase.
  • the C-terminus of the light or heavy chain is modified.
  • the CH2 regions of the Fc region may be glycosylated.
  • a C-terminus of a protein (a fragment, analog, or derivative thereof) can be attached to an amino acid motif that can be recognized by isoprenoid transferase.
  • the C-terminus can be modified.
  • the modification can be (i) a deletion in the carboxy terminus of the protein, (ii) an oligopeptide or polypeptide addition in the carboxy terminus of the protein, or (iii) a deletion in the carboxy terminus of the protein and an oligopeptide or polypeptide addition in the carboxy terminus of the protein.
  • the modification can be attached to the amino acid motif.
  • isoprenoid transferase refers to an enzyme that can recognize a certain amino acid motif at or near a C-terminus of a protein and perform selective alkylation at thiol position(s) of cysteine residue(s) of the certain amino acid motif by adding an isoprenoid unit(s) to the protein bearing the certain amino acid motif.
  • isoprenoid transferase examples include farnesyltransferase (FTase) and geranylgeranyltransferase (GGTase), which involve the transfer of a farnesyl or a geranyl-geranyl moiety to C-terminal cysteine(s) of the target protein, respectively.
  • GGTase can be classified into GGTase I and GGTase II.
  • FTase and GGTase I can recognize a CAAX motif and GGTase II can recognize a XXCC, XCXC, or CXX motif, in which C represents cysteine, A represents an aliphatic amino acid, and X represents an amino acid that determines the substrate specificity of the isoprenoid transferases (Nature Rev. Cancer 2005, 5(5), pp. 405-12; Nature Chemical Biology, 2010, 17, pp. 498-506; Lane K T, Bees L S, Structural Biology of Protein of Farnesyltransferase and Geranylgeranyltransferase Type 1, Journal of Lipid Research, 47, pp. 681-699 (2006); Patrick J. Kasey, Miguel C. Seabra; Protein Prenyltransferases, The Journal of Biological Chemistry, Vol. 271, No. 10, Issue of March 8, pp. 5289-5292 (1996), the contents of these references are hereby incorporated by reference in their entirety).
  • isoprenoid transferases from a variety of sources, e.g., humans, animals, plants, bacteria, virus, and the like can be used. In some embodiments, naturally occurring isoprenoid transferases can be used. In some other embodiments, naturally or artificially modified isoprenoid transferases can be used.
  • an isoprenoid transferase having at least one amino acid sequence naturally changed including post-translational modification
  • a naturally or artificially truncated form of a naturally occurring isoprenoid transferase an isoprenoid transferase that has been modified by at least one of (His)-tag, GST, GFP, MBP, CBP, lospeptag, BCCP, Myc-tag, Calmodulin-tag, FLAG-tag, HA-tag, Maltose binding protein-tag, Nus-tag, Glutathione-S-transferase-tag, Green fluorescent protein-tag, Thioredoxin-tag, S-tag, Softag 1, Softag 3, Strep-tag, SBP-tag, Ty tag, and the like.
  • Isoprenoid transferases can recognize an isosubstrate as well as a substrate.
  • the isosubstrate refers to a substrate analog which has a modification in the substrate.
  • Isoprenoid transferases alkylate a certain amino acid motif (e.g., CAAX motif) at a C-terminus of a protein (Benjamin P. Duckworth et al, ChemBioChem 2007, 8, 98; Uyen T. T. Nguyen et al, ChemBioChem 2007, 8, 408; Guillermo R. Labadie et al, J. Org. Chem. 2007, 72(24), 9291; James W.
  • a certain amino acid motif e.g., CAAX motif
  • a functionalized protein can be produced using an isoprenoid transferase and an isosubstrate through alkylation at a C-terminal cysteine(s).
  • cysteine residue of a C-terminal CAAX motif can be reacted with an isosubstrate using an isoprehoid transferase.
  • AAX can then be removed by a protease.
  • the resulting cysteine can then be methylated at the carboxy terminus by an enzyme.
  • cysteinylation and gluthathionylation through disulfide bond formation can occur due to post-translational modification.
  • Such a disulfide bond can be reduced when such alkylation occurs by isoprenoid transferases.
  • the proteins of the present invention can be made using any molecular biology or cell biology method well known in the art. For example, transient transfection methods can be used. Genetic sequences encoding a certain amino acid motif that can be recognized by an isoprenoid transferase can be inserted into a known plasmid vector using standard PCR technologies so as to express a protein (a fragment or analog thereof) having the certain amino acid motif at a C-terminus thereof. As such, a protein having at least one amino acid motif that can be recognized by an isoprenoid transferase can be expressed.
  • the expressed protein can then be enzymatically reacted with an isosubstrate of an isoprenoid transferase using the isoprenoid transferase to produce a functionalized protein.
  • the isosubstrate contains a functional group.
  • a protein having an amino acid motif that can be recognized by an isoprenoid transferase may be enzymatically reacted, using an isoprenoid transferase and at least one isosubstrate having a first functional group (FG1), thereby producing a functionalized protein.
  • FG1 first functional group
  • the term “functional group” used herein refers to a group that can lead to, e.g., 1,3-dipolar cycloaddition reactions, hetero-diels reactions, nucloephilic substitution reactions (e.g., of a ring opening reaction of a heterocyclic electophile such as epoxide, aziridine, cyclic sulfate, and aziridium), non-aldol type carbonyl reactions (e.g., formation of oxime ethers, ureas, thioureas, aromatic heterocycles, hydrazones and amides), additions to carbon-carbon multiple bonds, oxidation reactions (e.g., epoxidation, aziridination, and sulfenyl halide addition), and click chemistry.
  • nucloephilic substitution reactions e.g., of a ring opening reaction of a heterocyclic electophile such as epoxide, aziridine, cyclic sulfate
  • the functional group can include, but not limited to, a fluorescent tag, a triazole, a maleimide, and a radio isotope (Angew. Chem. Int. Ed. 2001, 40, 2004-2021; Drug Discovery Today, 2003, 8(24), 1128-1137; Chem. Rev. 2008, 108, 2952-3015, the contents of which are incorporated herein by reference.)
  • the functional group can be an acetylene group and an azide group.
  • the functional group can be attached to a protein or an active agent via at least one linker.
  • the linker is a linear linker.
  • the linker is a branched linker.
  • active agents can be attached to all of the branches. Each branch can have the same or different active agents.
  • the linker can be cleavable. In some other embodiments, it can be non-cleavable.
  • a functionalized active agent is produced by attaching a second functional group (FG2) to an active agent.
  • active agents include, but are not limited to, a drug, a toxin, an affinity ligand, a detection probe, or a combination thereof.
  • Exemplary drugs include, but are not limited to, erlotinib (TARCEVA; Genentech/OSI Pharm.), bortezomib (VELCADE; MilleniumPharm.), fulvestrant (FASLODEX; AstraZeneca), sutent (SU11248; Pfizer), letrozole (FEMARA; Novartis), imatinib mesylate (GLEEVEC; Novartis), PTK787/ZK 222584 (Novartis), oxaliplatin (Eloxatin; Sanofi), 5-fluorouracil (5-FU, leucovorin, rapamycin (Sirolimus, RAPAMUNE; Wyeth), lapatinib (TYKERB, GSK572016; GlaxoSmithKline), lonafarnib (SCH 66336), sorafenib (BAY43-9006; Bayer Labs.), gefitinib (IRESSA; Astrazeneca), AG
  • Additional drugs include, but are not limited to, (i) anti-hormonal agents that act to regulate or inhibit hormone action on tumors such as anti-estrogens and selective estrogen receptor modulators (SERMs), including, for example, tamoxifen (including NOLVADEX® tamoxifen), raloxifene, droloxifene, 4-hydroxytamoxifen, trioxifene, keoxifene, LY117018, onapristone, and FAREATON® toremifene; (ii) aromatase inhibitors that inhibit the enzyme aromatase, which regulates estrogen production in the adrenal glands, such as, for example, 4(5)-imidazoles, aminoglutethimide, MEGASE® megestrol acetate, AROMASIN® exemestane, FEMARA® letrozole, and ARIMIDEX® anastrozole; (iii) anti-androgens such as flutamide, nilutamide, b
  • cytokines can be used as the drug.
  • Cytokines are small cell-signaling protein molecules that are secreted by numerous cells and are a category of signaling molecules used extensively in intercellular communication. They include monokines, lympokines, traditional polypeptide hormones, and the like.
  • cytokines include, but are not limited to, growth hormone such as human growth hormone, N-methionyl human growth hormone, and bovine growth hormone; parathyroid hormone; thyroxine; insulin; proinsulin; relaxin; prorelaxin; glycoprotein hormones such as follicle stilumating hormone (FSH), thyroid stimulating hormone (TSH), and luteinizing hormone (LH); hepatic growth factor fibroblast growth factor; prolactin; placental lactogen; tumor necrosis factor- ⁇ and - ⁇ ; mullerian-inhibiting substance; mouse gonadotropin-associated peptide; inhibin; activin; vascular endothelial growth factor; integrin; thrombopoietin (TPO); nerve growth factors such as NGF- ⁇ ; platelet-growth factor; transforming growth factors (TGFs) such TGF- ⁇ and TGF- ⁇ ; insulin-like growth factor-I and -II; erythropoietin (EPO); osteo in
  • Toxin refers to a poisonous substance produced within living cells or organisms.
  • Toxins can be small molecules, peptides or proteins that are capable of causing disease on contact with or absorption by body tissue interacting with biological macromolecules such as enzyme or cellular receptors.
  • Toxins include plant toxins and animal toxins.
  • animal toxins include, but are not limited to, diphtheria antitoxin, botulium toxin, tetanus antitoxin, dysentery toxin, cholera toxin, tetrodotoxin, brevetoxin, ciguatoxin.
  • plant toxins include, but are not limited to, ricin and AM-toxin.
  • small molecule toxins include, but are not limited to, auristatin, geldanamycin (Kerr et al., 1997, Bioconjugate Chem. 8(6):781-784), maytansinoids (EP 1391213, ACR 2008, 41, 98-107), calicheamycin (US 2009105461, Cancer Res. 1993, 53, 3336-3342), daunomycin, doxorubicin, methotrexate, vindesine, SG2285 (Cancer Res.
  • Toxins can exhibit cytotoxicity and cell growth inhibiting activity by tubulin binding, DNA binding, topoisomerase suppression, and the like.
  • ligand refers to a molecule that can form a complex with a target biomolecule.
  • An example of a ligand is a molecule that is attached to a predetermined position of a targeted protein and transmits a signal. It can be a substrate, an inhibitor, a stimulating agent, a neurotransmitter, or a radioisotope.
  • Detectable moiety refers to a composition detectable by spectroscopic, photochemical, biochemical, immunochemical, radioactive, or chemical means.
  • useful labels include 32 P, 35 S, fluorescent dyes, electron-dense reagents, enzymes (e.g., as commonly used in an ELISA), biotin-streptavidin, dioxigenin, haptens and proteins for which antisera or monoclonal antibodies are available, or nucleic acid molecules with a sequence complementary to a target.
  • the detectable moiety often generates a measurable signal, such as a radioactive, chromogenic, or fluorescent signal, that can be used to quantify the amount of bound detectable moiety in a sample.
  • Quantitation of the signal is achieved by, e.g., scintillation counting, densitometry, flow cytometry, ELISA, or direct anlaysis by mass spectreometry of intact or subsequently digested peptides (one or more peptide can be assessed).
  • Persons of skill in the art are familiar with techniques for labeling compounds of interest, and means for detection. Such techniques and methods are conventional and well-known in the art.
  • probe refers to a material that can (i) provide a detectable signal, (ii) can interact a first probe or a second probe to modify a detectable signal provided by the first or second probe, such as fluorescence resonance energy transfer (FRET), (iii) stabilize the interaction with an antigen or a ligand or increase the binding affinity; (iv) affect electrophoresis mobility or cell-intruding activity by a physical parameter such as charge, hydrophobicity, etc., or (v) control ligand affinity, antigen-antibody binding, or ionic complex formation.
  • FRET fluorescence resonance energy transfer
  • the reaction between the functionalized protein and the functionalized active agent may be a click chemistry reaction or via a hydrazone and/or oxime formation.
  • the FG1 is an azide group and the FG2 is an acetylene group, or vice versa.
  • the FG1 may be an aldehyde or ketone group and the FG2 is a hydrazine or hydroxylamine, or vice versa.
  • Click chemistry reactions are conducted in a mild condition, making it possible to handle proteins easily. Click chemistry reaction shows very high reaction specificity. Thus, even if a protein has other functional groups (e.g., side chain residue or at a C-terminus or N-terminus), these functional groups are not affected by the click chemistry reaction. For example, a click chemistry reaction between an acetylene group and an azide group of a protein can occur while other functional groups of the protein are not affected by the click chemistry reaction. In addition, a click chemistry reaction can specifically occur without being affected by the kind of ligand involved. In some cases, the ligand can be selected to improve overall reaction efficiency.
  • the ligand can be selected to improve overall reaction efficiency.
  • azide-acetylene click chemistry can produce a triazole at a high yield (Rhiannon K. Iha et al, Chem. Rev. 2009, 109, 5620; Morten Meldal and Christian Wenzel Tornoe, Chem. Rev., 2008, 108, 2952; Hartmuth C. Kolb et al, Angew. Chemie Int. Ed. Engl., 2001, 40, 2004, all of which are incorporated herein by reference.)
  • Azide and acetylene groups are functional groups that do not exist in amino acid sequences of naturally occurring proteins. If a conjugation reaction occurs using these functional groups, none of the side chain residues and none of the N-terminal or C-terminal functional groups are affected by the click chemistry reaction. Accordingly, a protein-active agent conjugate in which an active agent is conjugated at a targeted position(s) can be produced.
  • the protein when the protein is an antibody, all or a part of the antibody can be reduced to a single chain during alkylation by an isoprenoid transferase.
  • the single chain can be oxidized to form a H 2 L 2 -form antibody due to an oxidizer used in the click chemistry reaction.
  • alkylation can be made at 1-4 positions per antibody.
  • the number of the active agents can be more than 4 since a plurality of the active agents can be attached to a linker.
  • the method may further include removing AAX.
  • the method may further include adding a methyl group at the C-terminus after removing AAX (Journal of Lipid Research, 2006, 47, 681-699, which is incorporated herein by reference.).
  • a method for preparing a protein-active agent conjugate comprises: (a) expressing a protein having an amino acid motif that can be recognized by an isoprenoid transferase; (b) attaching an isosubstrate of an isoprenoid transferase to an active agent; and (c) enzymatically reacting, using the isoprenoid transferase, the expressed protein with the active agent attached to the isosubstrate.
  • the protein is reacted with an active agent attached to an isosubstrate of the isoprenoid transferase.
  • an active agent attached to an isosubstrate of the isoprenoid transferase.
  • thiol-maleimide conjugation may occur.
  • the active agents are conjugated at the targeted positions only according to the present invention. Accordingly, a problem associated with the prior art that a non-homogeneous mixture is produced is avoided.
  • the present invention provides a protein-active agent conjugate comprising a protein having an amino acid motif that can be recognized by an isoprenoid transferase, wherein the active agent is covalently linked to the protein at the amino acid motif.
  • a protein that selectively binds a target of interest e.g., a target cell in a subject.
  • exemplary proteins include, but are not limited to antibodies or fragments of an antigenic that specifically bind to the target of interest.
  • CAAX Protein (CAAX Antibody)
  • a protein-active agent conjugate prepared by a method of the present invention is represented by the following formula (I), in which the protein is an antibody (fragment or analog thereof) (Ab), the active agent is a drug (D), and the amino acid motif that can be recognized by an isoprenoid transferase is CAAX.
  • formula (I) in which the protein is an antibody (fragment or analog thereof) (Ab), the active agent is a drug (D), and the amino acid motif that can be recognized by an isoprenoid transferase is CAAX.
  • Ab(M) represents that the antibody or fragment thereof, which can comprise a modification.
  • the modification can be (i) a deletion in the carboxy terminus of the antibody or fragment thereof; (ii) an oligopeptide or polypeptide addition in the carboxy terminus of the antibody or fragment thereof; and (iii) a deletion in the carboxy terminus of the antibody or fragment thereof and an oligopeptide or polypeptide addition in the carboxy terminus of the antibody or fragment thereof.
  • Q represents a linker.
  • the linker can be a linear linker or a branched linker.
  • the linker can include a first functional group (FG1).
  • n 1 , n 2 , and m can be appropriately determined depending on the antibody, the amino acid motif, linker, active agent, etc.
  • n 1 and n 2 are independently an integer of 1 to 4 and m is an integer of 1 to 16.
  • the linker can be represented by the following formula (II):
  • P 1 and Y is independently a group containing a first functional group (FG1).
  • the FG 1 can be selected from the group consisting of: acetylene, azide, aldehyde, hydroxylamine, hydrazine, ketone, nitrobenzofurazan (NBD), dansyl, fluorescein, biotin, and Rhodamin.
  • L 1 is (CH 2 ) r X q (CH 2 ) p , in which X is oxygen, sulfur, —NR 1 —, —C(O)NR 1 —, —NR 1 C(O)—, —SO 2 NR 1 —, —(CH ⁇ CH)—, or acetylene;
  • R 1 is hydrogen, C 1-6 alkyl, C 1-6 alkyl aryl, or C 1-6 alkyl heteroaryl;
  • r and p is independently an integer of 0 to 6;
  • q is an integer of 0 to 1; and
  • n is an integer of 1 to 4.
  • the drug (D) can be attached to the linker via a group containing a second functional group (FG2) that can react with the FG1.
  • FG2 can be selected from the group consisting of acetylene, hydroxylamine, azide, aldehyde, hydrazine, ketone, and amine.
  • the drug (D) can be attached to the group containing an FG2 via —(CH 2 ) r X q (CH 2 ) p — or —[ZCH 2 CH 2 O(CH 2 CH 2 O) w CH 2 CH 2 Z]—, in which X is oxygen, sulfur, —NR 1 —, —C(O)NR 1 —, —NR 1 C(O)—, —NR 1 SO 2 —, or —SO 2 NR 1 —; Z is oxygen, sulfur or NR 1 ; R 1 is hydrogen, C 1-6 alkyl, C 1-6 alkyl aryl, or C 1-6 alkyl heteroaryl; r and p is independently an integer of 0 to 6; q is an integer of 0 to 1; and w is an integer of 0 to 6.
  • a peptide(s) that can be cleaved by cathepsin B or (ii) a glucuronide that can be cleaved by ⁇ -glucuronidase can be attached to the —(CH 2 ) r X q (CH 2 ) p — or —[ZCH 2 CH 2 O(CH 2 CH 2 O) w CH 2 CH 2 Z]—.
  • a non self-immolative group or a self-immolative group can be attached to the (i) peptide(s) that can be cleaved by cathepsin B or (ii) glucuronide that can be cleaved by ⁇ -glucuronidase.
  • Non-limiting examples of the self-immolative group may be aminophenylmethyloxycarbonyl and hydroxyphenylmethyloxycarbonyl.
  • the peptide that can be cleaved by cathepsin B is represented by the following formula (III):
  • the glucuronide that can be cleaved by ⁇ -glucuronidase is represented by the following formula (IV):
  • the present invention provides compositions comprising a protein-active agent conjugate described herein.
  • the compositions are used for delivering an active agent to a target cell in a subject.
  • the compositions are used to treat a subject in need thereof (i.e., in need of the active agent).
  • compositions are known to one skilled in the art, and such compositions can be delivered in vivo to a subject.
  • compositions are prepared in an injectable form, either as a liquid solution or as a suspension.
  • Solid forms suitable for injection may also be prepared as emulsions, or with the polypeptides encapsulated in liposomes.
  • the protein-active agent conjugates can be combined with a pharmaceutically acceptable carrier, which includes any carrier that does not induce the production of antibodies harmful to the subject receiving the carrier.
  • Suitable carriers typically comprise large macromolecules that are slowly metabolized, such as proteins, polysaccharides, polylactic acids, polyglycolic acids, polymeric amino acids, amino acid copolymers, lipid aggregates, and the like. Such carriers are well known to those skilled in the art.
  • compositions of the invention can also contain diluents, such as water, saline, glycerol, ethanol. Auxiliary substances may also be present, such as wetting or emulsifying agents, pH buffering substances, and the like. Proteins may be formulated into the vaccine as neutral or salt forms.
  • the compositions can be administered parenterally, by injection; such injection may be either subcutaneously or intramuscularly. Additional formulations are suitable for other forms of administration, such as by suppository or orally. Oral compositions may be administered as a solution, suspension, tablet, pill, capsule, or sustained release formulation.
  • compositions are administered in a manner compatible with the dose formulation.
  • the composition comprises an therapeutically effective amount of the protein-active agent conjugate.
  • a therapeutically effective amount is meant a single dose, or a composition administered in a multiple dose schedule, that is effective for the treatment or prevention of a disease or disorder.
  • the dose administered will vary, depending on the subject to be treated, the subject's health and physical condition, the degree of protection desired, and other relevant factors. Precise amounts of the active ingredient required will depend on the judgment of the practitioner.
  • the present invention provides a method for delivering an active agent to a target cell in a subject, the method comprising administering the protein-active agent conjugate or the composition.
  • the present invention provides a method of treating a subject in need thereof (i.e., a subject in need of the active agent), the method comprising administering an effective amount of the protein-active agent conjugate or a composition comprising the conjugate to the subject.
  • a protein-active agent conjugate e.g., antibody-drug conjugate
  • a composition comprising the conjugate in a therapeutically effective amount can be administered to a patient suffering from a cancer or tumor to treat the cancer or tumor.
  • a protein-active agent conjugate e.g., antibody-drug conjugate
  • a composition comprising the conjugate in a therapeutically effective amount can be administered to a patient to treating or preventing an infection by a pathogenic agent (e.g., a virus, a bacteria, a fungus, a parasite, and the like).
  • a pathogenic agent e.g., a virus, a bacteria, a fungus, a parasite, and the like.
  • Such methods include the step of administering to the mammal a therapeutic or prophylactic amount of an amount of the conjugate sufficient to treat the disease or disorder or symptom thereof, under conditions such that the disease or disorder is prevented or treated.
  • the protein-active agent conjugate or composition can be administered in the form of a pharmaceutically acceptable salt or solvate thereof. In some embodiments, it can be administered with a pharmaceutically acceptable carrier, a pharmaceutically acceptable expient, and/or a pharmaceutically acceptable additive.
  • a pharmaceutically acceptable carrier e.g., a pharmaceutically acceptable sulfate, a pharmaceutically acceptable sulfate, a pharmaceutically acceptable sulfate, a pharmaceutically acceptable additive.
  • the pharmaceutically effective amount and the type of the pharmaceutically acceptable salt or solvate, expient and additive can be determined using standard methods (Remington's Pharmaceutical Sciences, Mack Publishing Co., Easton, Pa., 18 th edition, 1990).
  • terapéuticaally effective amount with regard to a cancer or tumor means an amount that can decrease the number of cancer cells; decrease the size of cancer cells; prohibit cancer cells from intruding peripheral systems or decrease the intrusion; prohibit cancer cells from being spreading to other systems or decrease the spreading; prohibit cancer cells from growing; and/or ameliorate at least one symptoms related to the cancer.
  • TTP time to tumor progression
  • RR response rate
  • terapéuticaally effective amount with regard to infection by a pathogenic agent means an amount that can prevent, treat, or reduce the symptoms associated with infection.
  • salts used herein includes organic salts and inorganic salts. Examples thereof include, but are not limited to, hydrochloride, hydrobromide, hydroiodide, sulfate, citrate, acetate, oxalate, chloride, bromide, iodide, nitrate, bisulfate, phosphate, acidic phosphate, isonicotinate, lactate, salicylate, acidic citrate, tartrate, oleate, tannate, pantonate, bitartrate, ascorbate, succinate, maleate, gentisinate, fumarate, gluconate, glucoronate, saccharate, formate, benzoate, glutamate, methane sulfonate, ethane sulfonate, benzene sulfonate, p-toluene sulfonate, and pamoate (i.e., 1,1′-methylenebis-
  • Exemplary solvates that can be used to pharmaceutical acceptable solvates of the compounds according to the present invention include, but not limited to, water, isopropanol, ethanol, methanol, DMSO, ethylacetate, acetic acid, and ethanol amine.
  • Herceptin antibodies were generated using standard recombinant DNA technology and PCR cloning protocols with pNATABH::Herceptin HC plasmid or pNATABL::Herceptin LC plasmid. Recombinant plasmids were expressed in an HEK293E cell line by transient transfection. The antibodies were separated and purified by protein A column chromatography.
  • Herceptin-HC-GCVIM and Herceptin-LC-GCVIM plasmids were constructed by inserting a DNA sequence encoding a CAAX motif (e.g., GCVIM, G 5 CVIM, GCVIM, G 10 CVIM, or G 10 CVLL), to the C-terminus of the heavy chain or light chain encoded in the pNATABH::Herceptin HC or pNATABH::Herceptin LC plasmid.
  • a CAAX motif e.g., GCVIM, G 5 CVIM, GCVIM, G 10 CVIM, or G 10 CVLL
  • a SacII recognition sequence is present at amino acid 172 in the C-terminus of the human IgG1-Fc region.
  • a forward primer was designed to bind the SacII site in the Fc region.
  • the DNA sequence to be inserted e.g., the 15-mer encoding GCVIM 5-mer sequence
  • the forward and reverse primers were used to amplify a PCR product, and the resultant product was purified using a PCR purification kit.
  • the reverse primer contained a XhoI site, the PCR product was digested with SacII and XhoI.
  • the pNATABH::Herceptin HC plasmid was digested with SacII and XhoI.
  • the digested backbone was purified using a gel purification kit and ligated with the digested PCR product. Ligation was performed by appropriately adjusting the ratio of the vector and the insert, and the ligation product was transformed into competent bacterial cells for screening.
  • Herceptin-HC-GCVIM and Herceptin-LC-GCVIM plasmids were prepared from sequenced clones.
  • FIGS. 1-10 The amino acid sequences from the resultant plasmids are shown in FIGS. 1-10 . Sections 1-4 and 1-7 below provide a detailed description of each of the constructs.
  • Herceptin-HC-GCVIM Herceptin-LC-GCVIM
  • HEK293 E cells were cultured in DMEM/10% FBS media on 150 mm plates (#430599, Corning USA) until 70 ⁇ 80% confluency. 13 ⁇ g of DNA and 26 ⁇ g of PEI (#23966, Polysciences, USA) were mixed in a ratio of 1:2, incubated at RT for about 20 minutes, and then added to the HEK193E cells. After 16-20 hours, the media was replaced with serum free media (No FBS DMEM (#SH30243.01, Hyclone Thermo., USA)) and supernatant was collected every two or three days.
  • the supernatants were filtered with a 0.22 um top-filter (#PRO2890, Millipore, USA) and then bound to 500 ⁇ l of protein A bead (#17-1279-03, GE healthcare Sweden) packed in a 5 mL column. Using a peristaltic pump, overnight binding was performed at 0.9 mL/min at 4° C. The column was washed with 100 mL or greater of PBS (#70011, Gibco, USA). Bound protein was then eluted with 0.1M Glycine-HCl (#G7126, Sigma, USA) into 6 fractions and neutralized with 1M Tris (#T-1503, Sigma, USA) (pH 9.0). The protein was quantified.
  • Herceptin-HC-GCVIM 2 or 3 fractions containing the protein were collected and concentrated with Amicon Ultra filter units (#UFC805024, Millipore, USA). Buffer was changed about 10 times with 1 ⁇ PBS (#70011, Gibco, USA).
  • the protein product was confirmed to be Herceptin-HC-GCVIM or Herceptin-LC-GCVIM by Western blot.
  • ImmunoPure peroxidase conjugated goat anti-human IgG Fc (#31413, Pierce, USA) was used.
  • 1-2 mg of Herceptin-HC-CGVIM or Herceptin-LC-GCVIM was obtained from IL of cell culture medium.
  • Herceptin-HC-CGVIM and Herceptin-LC-GCVIM products were also analyzed with an Agilent bioanalyzer. Briefly, 8 ⁇ l of purified protein sample (approx. 1 mg/ml) was analyzed using the Agilent Protein 230 Kit (5067-1515 Agilent Technologies, USA). The protein sample was separated into 2 fractions (4 ⁇ l each). 4 ⁇ l of non-reducing buffer or reducing buffer was added to each sample. The sample was heated at 95-100° C. for 5 minutes and cooled with ice to 4° C. After spin-down, 84 ⁇ l of deionized water was added to the sample and ladder and vortexed. Thereafter, the sample was loaded and analyzed with the kit per manufacturer's instructions.
  • Modified anti cMET-CAAX antibodies were also prepared by the above-described methods.
  • modified anti cMET-CAAX antibodies were generated using standard recombinant DNA technology and PCR cloning protocols with pPMC-C1A5 plasmid. Recombinant plasmids were expressed in an HEK293T cell line by transient transfection. The antibodies were separated and purified by protein A column chromatography.
  • Herceptin-HC-GCVIM, Herceptin-HC-G 5 CVIM, Herceptin-HC-G 7 CVIM, and Herceptin-HC-G 10 CVIM antibodies were prepared.
  • the antibodies respectively, have a 5-mer(GCVIM), a 9-mer(G 5 CVIM), an 11-mer(G 7 CVIM), or a 14-mer(G 10 CVIM) sequence at the C-terminus of the heavy chain ( FIGS. 1 , 3 , 5 , and 7 ).
  • Herceptin-LC-GCVIM, Herceptin-LC-G 5 CVIM, Herceptin-LC-G 7 CVIM, and Herceptin-LC-G 10 CVIM antibodies were prepared.
  • the antibodies respectively, have a 5-mer(GCVIM), a 9-mer(G 5 CVIM), an 1′-mer(G 7 CVIM), or a 14-mer(G 10 CVIM) sequence at the C-terminus of the light chain ( FIGS. 2 , 4 , 6 , and 8 ).
  • a Herceptin-HC-G 10 CVLL antibody was prepared.
  • the antibody has a 14-mer(G 10 CVLL) sequence at the C-terminus of the heavy chain ( FIG. 9 ).
  • a Herceptin-LC-G 10 CVLL antibody was prepared.
  • the antibody has a 14-mer(G 10 CVLL) sequence at the C-terminus of the light chain ( FIG. 10 ).
  • Anti cMET-HC-G 7 CVIM and anti cMET-HC-G 10 CVIM antibodies were prepared.
  • the antibodies respectively, have an 11-mer(G 7 CVIM), or a 14-mer(G 10 CVIM) sequence at the C-terminus of the heavy chain (not shown).
  • FIG. 11 shows an SDS-PAGE gel analyzing the anti cMET-HC-G 7 CVIM and anti cMET-HC-G 10 CVIM antibodies
  • Anti cMET-LC-G 7 CVIM and anti cMET-LC-G 10 CVIM antibodies were prepared.
  • the antibodies respectively, have an 11-mer(G 7 CVIM), or a 14-mer(G 10 CVIM) sequence at the C-terminus of the light chain (not shown).
  • FIG. 11 shows an SDS-PAGE gel analyzing the anti cMET-LC-G 7 CVIM and anti cMET-LC-G 10 CVIM antibodies.
  • Acetoxydecadienyl aldehyde (C) was prepared from farnesol in 5 steps. From the compound (C), the compounds (D) and (E) were prepared in 6 steps and 5 steps, respectively. From the compounds (D) and (E), the above-referenced compounds (F) and (G) were prepared by a method similar to the method described in the section 2-1 above. The compounds (C), (D), and (E) were prepared by a method similar to the method described in JOC 2007, 72(24), 9291-9297, the contents of which are hereby incorporated by reference in their entirety.
  • Tris-ammonium[3,7-dimethyl-8-(7-nitro-benzo[1,2,5]oxadiazol-4-ylamino)-octa-2,6-diene-1]pyrophosphate (NBD-GPP) was prepared by a method similar to the method described in JACS 2006, 128, 2822-2835, the contents of which are hereby incorporated by reference in their entirety.
  • the compound 2 (5 g, 13.28 mmol) was added to a solution of 33% HBr in AcOH (20 mL) at 0° C. The resulting mixture was stirred for 2 hours at room temperature. After the reaction was completed, the resulting mixture was diluted by toluene (50 mL). The resulting mixture was concentrated under reduced pressure. Ethyl acetate (100 mL) and saturated NaHCO 3 solution (100 mL) were added to extract an organic layer. The thus-obtained organic layer was dried with anhydrous sodium sulfate to give the compound 3 (5.27 g, 100%).
  • MMAF-OMe was prepared according to the methods described in U.S. No. 61/483,698, ChemPharmBull, 1995, 43(10), 1706-1718, U.S. Pat. No. 7,423,116, U.S. Pat. No. 7,498,298, and WO2002/088172, the contents of each of these references are hereby incorporated by reference in their entirety.
  • the compound 2 (470 mg, 0.53 mM) was dissolved in tert-butanol ( t BuOH, 8 mL) and distilled water (0.8 mL). At 0° C., 10% Pd/C (50 mg) was added. The resulting mixture was stirred in H 2 gas for 2 hours. After the reaction was completed, the Pd/C was filtered using celite. The resulting filtered solution was concentrated under reduced pressure to give the compound LCB14-0601 (340 mg, 85%).
  • Azide-Linker-drug LCB14-0505, -0531, and -0510
  • the compound 1 was prepared with reference to the method described in ChemPharmBull, 1995, 43(10), 1706-1718, the contents of which are hereby incorporated by reference in their entirety.
  • a solution of the compound 1 (0.50 g, 0.57 mmol) in tert-butanol (6 mL) and water (0.6 mL) was stirred for 4 hours under hydrogen atmosphere with Pd/C (6 mg, 0.06 mmol).
  • the reactant solution was filtered through a celite pad and the filtrate was concentrated under reduced pressure to give the compound 2 (0.42 g) as a white solid.
  • the compound LCB14-0531 (65%) was prepared in a similar method to the above-described method.
  • the compound 6 was prepared using the methods described in BioconjugateChem. 2002, 13, 855-869 and US2005238649, the contents of each of these references are hereby incorporated by reference in their entirety.
  • a solution of the compound 6 (69 mg, 0.15 mmol) and compound 2 (100 mg, 0.13 mmol) in DMF (2 mL) was treated with DIPEA (0.04 mL, 0.2 mmol) and PyBOP (0.09 g, 0.17 mmol) at 0° C. The resulting mixture was stirred for 3 hours. Ethyl acetate (100 mL) and water (30 mL) were used to extract an organic layer, which was concentrated under reduced pressure. The residue was subjected to column chromatography with methylenechloride and methanol to give the compound LCB14-0510 (94 mg, 64%) as a brown solid.
  • Acetylene-Linker-NBD LCB14-0532
  • Fmoc-Val-Cit-OH was prepared according to the method described in WO2007/008603, the contents of which are hereby incorporated by reference in their entirety.
  • dichloromethane 50 mL
  • methanol 20 mL
  • para-aminobenzylalcohol 2.43 g, 19.70 mmol
  • 1-ethoxycarbonyl-2-ethoxy-1,2-dihydroquinoline 1.98 g, 19.7 mmol
  • the compound 4 was prepared according to the methods described in WO2006/111759 A1, WO2010/043880 A1, and WO2010/010347 A1, the contents of each of these references are hereby incorporated by reference in their entirety.
  • the compound 4 (83 mg, 0.059 mmol), sodium carbonate (10 mg, 0.089 mmol), and Pd(TPP) 4 (3.4 mg, 0.003 mmol) were sequentially dissolved in a mixture solvent of ethanol/toluene/distilled water (0.3 mL/0.3 mL/0.3 mL).
  • a solution of the compound 3 (31.6 mg, 0.065 mmol) in toluene (3 mL) was added.
  • the resulting mixture was stirred at room temperature for 1 hour.
  • ethyl acetate (50 mL) and distilled water (50 mL) were added.
  • the thus-obtained organic layer was dried with anhydrous sodium sulfate and concentrated under reduced pressure. The residue was subjected to column chromatography to give the compound 5 (79 mg, 74%).
  • the compound 4 (245 mg, 0.175 mmol), sodium carbonate (28 mg, 0.262 mmol), and Pd(TPP) 4 (10 mg, 0.009 mmol) were sequentially dissolved in a mixture solution of ethanol/toluene/distilled water (1.5 mL/1.5 mL/1.5 mL). A solution of the compound 3 (94 mg, 0.192 mmol) in toluene (1.5 mL) was added. The resulting mixture was stirred at room temperature for 12 hours. After the reaction was completed, ethyl acetate (100 mL) and distilled water (100 mL) were added. The thus-obtained organic layer was dried with anhydrous sodium sulfate and concentrated under reduced pressure. The residue was subjected to column chromatography to give the compound 5 (100 mg, 35.5%).
  • the compound LCB14-0606 was prepared according to the similar method described in JACS, 2010, 132(12), 4281, the contents of which are incorporated herein by reference in their entirety.
  • a solution of the compound 7 (320 mg, 1.40 mmol) in 7 mL of acetonitrile at room temperature was slowly added a solution of tris(tetrabutylammonium) hydrogen pyrophosphate (2.25 g, 2.80 mmol) in acetonitrile (7 ml).
  • the resulting mixture was stirred for 1 hour. After the reaction was completed, the resulting mixture was concentrated under reduced pressure below at 25° C.
  • Prenylation of Ab(M)-CAAX was performed using NBD-GPP (Tris-ammonium[3,7-dimethyl-8-(7-nitro-benzo[1,2,5]oxadiazol-4-ylamino)-octa-2,6-diene-1]pyrophosphate) and FTase (#344146, Calbiochem, USA) or NBD-FPP (#LI-013, Jena Bioscience, Germany) and GGTase I (#345852, Calbiochem, USA).
  • NBD-GPP Tris-ammonium[3,7-dimethyl-8-(7-nitro-benzo[1,2,5]oxadiazol-4-ylamino)-octa-2,6-diene-1]pyrophosphate
  • FTase #344146, Calbiochem, USA
  • NBD-FPP NBD-FPP
  • GGTase I #345852, Calbiochem, USA
  • the prenylation reaction was conducted at 30° C. for 3 hours by using a 50 mM Tris-HCl (pH 7.4) buffer solution containing 5 mM MgCl 2 , 10 ⁇ M ZnCl 2 , and 5 mM DTT. After the reaction was completed, SDS-PAGE analysis was made. An image analyzer (ChemiDoc XRS + , BioRad, USA) was used to identify fluorescent protein band(s) to confirm that the prenylation reaction occurred.
  • Herceptin-HC-GCVIM, Herceptin-HC-G 5 CVIM (not shown), Herceptin-HC-G 7 CVIM, and Herceptin-HC-G 10 CVIM antibodies were prenylated using NBD-GPP and FTase in the method described above. Fluorescence was detected on protein band(s) corresponding to the heavy chain(s) (about 50K dalton) of the respective antibodies. This result confirmed that Herceptin-HC-CAAX antibodies, each having a spacer with various lengths, could be prenylated ( FIG. 12 ).
  • Herceptin-LC-GCVIM, Herceptin-LC-G 5 CVIM, Herceptin-LC-G 7 CVIM, and Herceptin-LC-G 10 CVIM antibodies were prenylated using NBD-GPP and FTase in the method described above. Fluorescence was detected on protein band(s) corresponding to the light chain(s) (about 25K dalton) of the respective antibodies. This result confirmed that Herceptin-LC-CAAX antibodies, each having a spacer with various lengths, could be prenylated ( FIG. 13 ).
  • Anti cMET-HC-G 7 CVIM and anti cMET-HC-G 10 CVIM antibodies were prenylated using NBD-GPP and FTase in the method described above. Fluorescence was detected on protein band(s) corresponding to the heavy chain(s) (about 50K dalton) of the respective antibodies. This result confirmed that anti cMET-HC-CAAX antibodies, each having a spacer with various lengths, could be prenylated ( FIG. 14 ).
  • Anti cMET-LC-G 7 CVIM and anti cMET-LC-G 10 CVIM antibodies were prenylated using NBD-GPP and FTase in the method described above. Fluorescence was detected on protein band(s) corresponding to the light chain(s) (about 25K dalton) of the respective antibodies. This result confirmed that anti cMET-LC-CAAX antibodies, each having a spacer with various lengths, could be prenylated ( FIG. 15 ).
  • Herceptin-HC-G 10 CVLL antibody was prenylated using NBD-FPP and GGTase I in the method described above. Fluorescence was detected on a protein band corresponding to the heavy chain(s) (about 50K dalton) of the antibody that is connected with the CAAX-motif at the C-terminus via the G 10 spacer. This result confirmed that Herceptin-HC-CAAX antibodies could be prenylated by GGTase I ( FIG. 16 ).
  • Herceptin-LC-G 10 CVLL antibody was prenylated using NBD-FPP and GGTase I in the method described above. Fluorescence was detected on a protein band corresponding to the light chain(s) (about 25K dalton) of the antibody that is connected with the CAAX-motif at the C-terminus via the G 10 spacer. This result confirmed that Herceptin-LC-CAAX antibodies could be prenylated by GGTase I ( FIG. 16 ).
  • a Herceptin-LC-G 7 CVIM antibody was prenylated using LCB14-0512 and FTase in the method described above.
  • the prenylated Herceptin-LC-G 7 CVIM antibody was subjected to LC/MS analysis in a reduction condition without treating PNGase F, it was predicted that the theoretical molecular weights of the heavy chain and the light chain would be 50,597 daltons and 24,480 daltons, respectively.
  • the experimental molecular weights of the heavy chain and the light chain were measured to be 50,600 daltons and 24,479 daltons, respectively. The difference between the theoretical molecular weight values and the experimental molecular weight values was within a standard error range. This result confirmed that the Herceptin-LC-G 7 CVIM antibody was prenylated by FTase and an isosubstrate (LCB14-0512).
  • a Herceptin-LC-G 10 CVIM antibody was prenylated using LCB14-0512 and FTase in the method described above.
  • the prenylated Herceptin-LC-G 10 CVIM antibody was subjected to LC/MS analysis in a reduction condition without treating PNGase F, it was predicted that the theoretical molecular weights of the heavy chain and the light chain would be 50,596 daltons and 24,651 daltons, respectively.
  • the experimental molecular weights of the heavy chain and the light chain were measured to be 50,601 daltons and 24,651 daltons, respectively.
  • the difference between the theoretical molecular weight values and the experimental molecular weight values was within a standard error range. This result confirmed that the Herceptin-LC-G 10 CVIM antibody was prenylated by FTase and an isosubstrate (LCB14-0512).
  • Diafiltration was performed to remove excess reagents in the prenylated Herceptin-LC-G 7 CVIM prepared according to the above described method.
  • the antibody was reoxidized using CuSO 4 .
  • Diafiltration was performed to remove CuSO 4 .
  • the conjugate LCB14-0101 was subjected to hydrophobic interaction chromatography-high performance liquid chromatography with Ether-5PW column (7.5 ⁇ 75 mm, 10 ⁇ m, Tosoh Bioscience, USA). 50 mM potassium phosphate buffer (pH 7.0) containing 1.5M ammonium sulfate was used as buffer A and 50 mM potassium phosphate buffer (pH 7.0) containing 20% isopropyl alcohol was used as buffer B. 90% A/10% B was held for 5 minutes. Elution was conducted using a linear gradient from 90% A/10% B to 10% A/90% B for the next 30 minutes. The flow rate and temperature were set as 0.8 mL/min and 25° C., respectively.
  • Unmodified Herceptin-LC-G 7 CVIM and prenylated Herceptin-LC-G 7 CVIM were used as controls.
  • the retention times of the unmodified Herceptin-LC-G 7 CVIM, the prenylated Herceptin-LC-G 7 CVIM, and the conjugate LCB14-0101 were 9.6, 11.7, and 12.4 minutes ( FIG. 20 ), respectively.
  • MCF-7 HER2 negative to normal
  • MDA-MB-468 HER2 negative
  • SK-BR-3 HER2 positive
  • LCB14-0537 MMAF
  • LCB14-0508 MMAF-OMe
  • LCB14-0562 MMAE
  • protein-active agent conjugate LCB14-0101, LCB14-0102, and LCB14-0103 ( FIG. 26 ) were used.
  • the Herceptin-LC-G 7 CVIM was prenylated using LCB14-0512.
  • the prenylated Herceptin-LC-G 7 CVIM was subjected to click reaction using LCB14-0592 to conjugate ⁇ -glucuronide linker(BG)-MMAF, thereby preparing LCB14-0101.
  • the prenylated Herceptin-LC-G 7 CVIM was subjected to click reaction using LCB14-0589 to conjugate Val-Cit linker(VC)-MMAF-OMe, thereby preparing LCB14-0102.
  • the prenylated Herceptin-LC-G 7 CVIM was subjected to click reaction by using LCB14-0598 to conjugate ⁇ -glucuronide linker(BG)-MMAE, thereby preparing LCB14-0103.
  • Anti-proliferation activities of the antibodies, drugs, and conjugates with regard to the cancer cell lines were measured.
  • the cells were plated in 96-well, tissue culture plates at 1 ⁇ 10 4 cells per well. After 24 hour incubation, the antibodies, drugs, and conjugates were added in various concentrations. The number of viable cells after 72 hours were counted using SRB dye. Absorbance was measured at 540 nm using SpectraMax 190 (Molecular Devices, USA).
  • Herceptin-LC-G 7 CVIM had an IC 50 of 10 ⁇ g/mL or higher with MCF-7, MDA-MB-468, and SK-BR-3.
  • LCB14-0101 (MMAF conjugate) had an IC 50 of 8.09 ⁇ g/mL and 4.18 ⁇ g/mL with MCF-7 and MDA-MB-468, respectively, which expresses no or low level of HER2, whereas it had an IC 50 of 0.11 ⁇ g/mL with SK-BR-3, which overexpresses HER2.
  • LCB14-0101 is about 40-80 times more selective than Herceptin-LC-G 7 CVIM. Accordingly, it is confirmed that LCB14-0101 has both cytotoxic drug potency and anti HER2 selectivity ( FIG. 21 ).
  • Herceptin-LC-G 7 CVIM had an IC 50 of 10 ⁇ g/mL with MCF-7 and SK-BR-3.
  • LCB14-0102 (MMAF-OMe conjugate) had an IC 50 of 4.38 ⁇ g/mL with MCF-7, whereas had an IC 50 of 0.15 ⁇ g/mL with SK-BR-3.
  • LCB14-0102 is about 30 times more selective than Herceptin-LC-G 7 CVIM. Accordingly, it is confirmed that LCB14-0102 has both cytotoxic drug potency and anti HER2 selectivity ( FIG. 22 ).
  • LCB14-0103 (MMAE conjugate) had an IC 50 of 7.25 ⁇ g/mL with MCF-7, whereas it had an IC 50 of 0.072 ⁇ g/mL with SK-BR-3. In addition to its excellent inhibitory activity, LCB14-0103 is about 100 times more selective than Herceptin-LC-G 7 CVIM. Accordingly, it is confirmed that LCB14-0103 has both cytotoxic drug potency and anti HER2 selectivity ( FIG. 23 ).

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Immunology (AREA)
  • Epidemiology (AREA)
  • Organic Chemistry (AREA)
  • Molecular Biology (AREA)
  • Cell Biology (AREA)
  • Oncology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Biochemistry (AREA)
  • Genetics & Genomics (AREA)
  • Biophysics (AREA)
  • Communicable Diseases (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Microbiology (AREA)
  • Biotechnology (AREA)
  • Physics & Mathematics (AREA)
  • General Engineering & Computer Science (AREA)
  • Analytical Chemistry (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Toxicology (AREA)
  • Mycology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Virology (AREA)
  • Peptides Or Proteins (AREA)
  • Medicinal Preparation (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
US13/466,875 2011-05-08 2012-05-08 Protein-active agent conjugates and method for preparing the same Abandoned US20120308584A1 (en)

Priority Applications (7)

Application Number Priority Date Filing Date Title
US13/466,875 US20120308584A1 (en) 2011-05-08 2012-05-08 Protein-active agent conjugates and method for preparing the same
US14/181,648 US20140161829A1 (en) 2011-05-08 2014-02-15 Antibody-active agent conjugates and methods of use
US14/181,649 US20140187756A1 (en) 2011-05-08 2014-02-15 Methods of preparing antibody-active agent conjugates
US14/517,616 US9669107B2 (en) 2011-05-08 2014-10-17 Antibody-active agent conjugates and methods of use
US15/054,257 US20160213786A1 (en) 2011-05-08 2016-02-26 Protein-active agent conjugates and methods of use
US15/612,766 US10583197B2 (en) 2011-05-08 2017-06-02 Antibody-active agent conjugates and methods of use
US16/812,690 US20200289662A1 (en) 2011-05-08 2020-03-09 Antibody-active agent conjugates and methods of use

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201161483698P 2011-05-08 2011-05-08
US13/466,875 US20120308584A1 (en) 2011-05-08 2012-05-08 Protein-active agent conjugates and method for preparing the same

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US14/181,649 Continuation US20140187756A1 (en) 2011-05-08 2014-02-15 Methods of preparing antibody-active agent conjugates
US14/181,648 Division US20140161829A1 (en) 2011-05-08 2014-02-15 Antibody-active agent conjugates and methods of use

Publications (1)

Publication Number Publication Date
US20120308584A1 true US20120308584A1 (en) 2012-12-06

Family

ID=47139740

Family Applications (7)

Application Number Title Priority Date Filing Date
US13/466,875 Abandoned US20120308584A1 (en) 2011-05-08 2012-05-08 Protein-active agent conjugates and method for preparing the same
US14/181,648 Abandoned US20140161829A1 (en) 2011-05-08 2014-02-15 Antibody-active agent conjugates and methods of use
US14/181,649 Abandoned US20140187756A1 (en) 2011-05-08 2014-02-15 Methods of preparing antibody-active agent conjugates
US14/517,616 Active US9669107B2 (en) 2011-05-08 2014-10-17 Antibody-active agent conjugates and methods of use
US15/054,257 Abandoned US20160213786A1 (en) 2011-05-08 2016-02-26 Protein-active agent conjugates and methods of use
US15/612,766 Active US10583197B2 (en) 2011-05-08 2017-06-02 Antibody-active agent conjugates and methods of use
US16/812,690 Abandoned US20200289662A1 (en) 2011-05-08 2020-03-09 Antibody-active agent conjugates and methods of use

Family Applications After (6)

Application Number Title Priority Date Filing Date
US14/181,648 Abandoned US20140161829A1 (en) 2011-05-08 2014-02-15 Antibody-active agent conjugates and methods of use
US14/181,649 Abandoned US20140187756A1 (en) 2011-05-08 2014-02-15 Methods of preparing antibody-active agent conjugates
US14/517,616 Active US9669107B2 (en) 2011-05-08 2014-10-17 Antibody-active agent conjugates and methods of use
US15/054,257 Abandoned US20160213786A1 (en) 2011-05-08 2016-02-26 Protein-active agent conjugates and methods of use
US15/612,766 Active US10583197B2 (en) 2011-05-08 2017-06-02 Antibody-active agent conjugates and methods of use
US16/812,690 Abandoned US20200289662A1 (en) 2011-05-08 2020-03-09 Antibody-active agent conjugates and methods of use

Country Status (22)

Country Link
US (7) US20120308584A1 (fr)
EP (2) EP2707031B1 (fr)
JP (2) JP6196613B2 (fr)
KR (1) KR101921068B1 (fr)
CN (1) CN103648530B (fr)
BR (2) BR122020001787A8 (fr)
CA (1) CA2835576C (fr)
CY (1) CY1122152T1 (fr)
DK (1) DK2707031T3 (fr)
ES (1) ES2744226T3 (fr)
HR (1) HRP20191586T1 (fr)
HU (1) HUE045348T2 (fr)
IL (2) IL229364B (fr)
LT (1) LT2707031T (fr)
MX (2) MX364707B (fr)
PL (1) PL2707031T3 (fr)
PT (1) PT2707031T (fr)
RS (1) RS59253B1 (fr)
RU (1) RU2613906C2 (fr)
SG (1) SG194875A1 (fr)
SI (1) SI2707031T1 (fr)
WO (1) WO2012153193A2 (fr)

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20160080832A (ko) 2014-12-29 2016-07-08 주식회사 레고켐 바이오사이언스 리피바디 유도체-약물 복합체, 그 제조방법 및 용도
EP2968495A4 (fr) * 2013-03-15 2016-11-09 Daniel J Capon Immunoglobuline hybride contenant une liaison non peptidique
US20170008970A1 (en) * 2013-12-23 2017-01-12 Zymeworks Inc. Antibodies comprising c-terminal light chain polypeptide extensions and conjugates and methods of use thereof
WO2017089890A1 (fr) 2015-11-25 2017-06-01 Legochem Biosciences, Inc. Conjugués comprenant des groupes auto-immolables et procédés associés
EP3156424A4 (fr) * 2014-05-28 2018-01-24 LegoChem Biosciences, Inc. Composé contenant un groupe auto-immolable
US10118965B2 (en) 2015-09-25 2018-11-06 Legochem Biosciences, Inc. Compositions and methods related to anti-EGFR antibody drug conjugates
CN108743968A (zh) * 2017-06-20 2018-11-06 四川百利药业有限责任公司 半胱氨酸改造的抗体-毒素偶联物(tdc)定点偶联位点筛选
US10183997B2 (en) 2015-09-25 2019-01-22 Legochem Biosciences, Inc. Compositions and methods related to anti-CD19 antibody drug conjugates
US11066459B2 (en) 2014-03-14 2021-07-20 Biomolecular Holdings Llc Hybrid immunoglobulin containing non-peptidyl linkage
US11167040B2 (en) 2015-11-25 2021-11-09 Legochem Biosciences, Inc. Conjugates comprising peptide groups and methods related thereto
US11173214B2 (en) 2015-11-25 2021-11-16 Legochem Biosciences, Inc. Antibody-drug conjugates comprising branched linkers and methods related thereto
US11654197B2 (en) 2017-03-29 2023-05-23 Legochem Biosciences, Inc. Pyrrolobenzodiazepine dimer prodrug and ligand-linker conjugate compound of the same
US11707533B2 (en) 2019-09-04 2023-07-25 Legochem Biosciences, Inc. Antibody-drug conjugate comprising antibody against human ROR1 and use for the same
WO2023166322A1 (fr) 2022-03-04 2023-09-07 Iksuda Therapeutics Limited Conjugué d'anticorps anti-canag
US11827703B2 (en) 2018-05-09 2023-11-28 Legochem Biosciences, Inc. Compositions and methods related to anti-CD19 antibody drug conjugates

Families Citing this family (33)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
PL2707031T3 (pl) 2011-05-08 2020-01-31 Legochem Biosciences, Inc. Koniugaty białko-środek czynny i sposoby i ch wytwarzania
KR102356286B1 (ko) 2011-05-27 2022-02-08 암브룩스, 인코포레이티드 비-천연 아미노산 연결된 돌라스타틴 유도체를 함유하는 조성물, 이를 수반하는 방법, 및 용도
SG11201400770SA (en) * 2011-09-20 2014-04-28 Spirogen Sarl Pyrrolobenzodiazepines as unsymmetrical dimeric pbd compounds for inclusion in targeted conjugates
CN104955485B (zh) * 2012-10-12 2018-01-30 Adc疗法责任有限公司 吡咯并苯并二氮杂卓‑抗her2抗体结合物
EP2934596A1 (fr) * 2012-12-21 2015-10-28 Glykos Finland Oy Conjugués constitués de molécules de charge utile et de lieurs
ES2759503T3 (es) 2013-05-02 2020-05-11 Glykos Finland Oy Conjugados de una glicoproteína o un glicano con una carga útil tóxica
FR3008408B1 (fr) 2013-07-11 2018-03-09 Mc Saf Nouveaux conjugues anticorps-medicament et leur utilisation en therapie
WO2015052532A1 (fr) 2013-10-11 2015-04-16 Spirogen Sàrl Conjugués anticorps-pyrrolobenzodiazépines
ES2779974T3 (es) 2014-06-13 2020-08-21 Tenboron Oy Conjugados que comprenden un anticuerpo anti-egfr1
EP3160513B1 (fr) 2014-06-30 2020-02-12 Glykos Finland Oy Dérivé de saccharide d'une charge utile toxique et conjugués d'anticorps de celui-ci
US10519248B2 (en) * 2014-07-25 2019-12-31 Memorial Sloan Kettering Cancer Center Bispecific HER2 and CD3 binding molecules
US11820832B2 (en) 2014-07-25 2023-11-21 Memorial Sloan Kettering Cancer Center Bispecific HER2 and CD3 binding molecules
CN104587487B (zh) * 2015-01-06 2018-01-16 华东师范大学 一种应用于靶向给药系统的新的支链连接体
CN104650179A (zh) * 2015-02-06 2015-05-27 东南大学 一种新型的可被溶酶体酶裂解的铂类前药
GB201521709D0 (en) * 2015-12-09 2016-01-20 Kings College London And Sec Dep For Health The PBD Antibacterial agents
RS60663B1 (sr) * 2016-05-17 2020-09-30 Abbvie Biotherapeutics Inc Konjugati anti-cmet antitelo-lek i metodi za njihovu primenu
WO2017219025A1 (fr) 2016-06-17 2017-12-21 Magenta Therapeutics, Inc. Compositions et méthodes de déplétion de cellules
CN109803673A (zh) 2016-09-13 2019-05-24 阿勒根公司 非蛋白质梭菌毒素组合物
SI3544634T1 (sl) * 2016-11-23 2021-08-31 Eli Lilly And Company Konjugati zdravila s protitelesom proti met
KR102085798B1 (ko) 2016-12-28 2020-03-06 주식회사 인투셀 베타-갈락토사이드가 도입된 자가-희생 기를 포함하는 화합물
CN108250055B (zh) * 2016-12-28 2022-08-05 重庆医药工业研究院有限责任公司 一种焦磷酸丙酮基香叶酯中间体的制备方法
WO2018134787A2 (fr) 2017-01-20 2018-07-26 Magenta Therapeutics, Inc. Compositions et procédés pour la déplétion des cellules cd137+
WO2019035649A1 (fr) * 2017-08-14 2019-02-21 주식회사 레고켐 바이오사이언스 Conjugués anticorps-médicament comprenant un anticorps contre egfrviii
KR20200084802A (ko) 2019-01-03 2020-07-13 주식회사 레고켐 바이오사이언스 안전성이 향상된 피롤로벤조디아제핀 이량체 화합물 및 이의 용도
WO2020141923A2 (fr) 2019-01-03 2020-07-09 주식회사 레고켐 바이오사이언스 Composé dimère de pyrrolobenzodiazépine présentant une sécurité améliorée et son utilisation
EP3936150A4 (fr) 2019-03-06 2023-03-29 LegoChem Biosciences, Inc. Conjugués anticorps-médicament comprenant un anticorps contre dlk1 humain et utilisation associée
JP2022530482A (ja) 2019-05-02 2022-06-29 レゴケム バイオサイエンシズ, インク. トリス構造を有するリンカーを含むリガンド―薬物複合体
KR102501394B1 (ko) * 2019-05-02 2023-02-21 주식회사 레고켐 바이오사이언스 트리스 구조를 가지는 링커를 포함하는 리간드-약물 접합체
US20230310640A1 (en) 2019-12-31 2023-10-05 Legochem Biosciences, Inc. Pyrrolobenzodiazepine derivative and ligand-linker conjugate thereof
JP2024512324A (ja) 2021-03-05 2024-03-19 ジーオー セラピューティクス,インコーポレイテッド 抗グリコcd44抗体およびその使用
WO2022211508A1 (fr) 2021-03-30 2022-10-06 주식회사 레고켐바이오사이언스 Conjugué anticorps-médicament comprenant un anticorps dirigé contre la cldn18.2 humaine et utilisation associée
CA3228178A1 (fr) 2021-08-05 2023-02-09 Go Therapeutics, Inc. Anticorps anti-glyco-muc4 et leurs utilisations
WO2023018199A1 (fr) * 2021-08-09 2023-02-16 주식회사 레고켐바이오사이언스 Nouvel antibiotique et utilisation associée

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030017149A1 (en) * 1996-10-10 2003-01-23 Hoeffler James P. Single chain monoclonal antibody fusion reagents that regulate transcription in vivo
US20070122408A1 (en) * 2005-10-20 2007-05-31 The Scripps Research Institute Fc Labeling for Immunostaining and Immunotargeting
US20070141084A1 (en) * 2003-08-06 2007-06-21 Lee Che-Hung R Polysaccharide-protein conjugate vaccines

Family Cites Families (40)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US5225539A (en) 1986-03-27 1993-07-06 Medical Research Council Recombinant altered antibodies and methods of making altered antibodies
GB8823869D0 (en) 1988-10-12 1988-11-16 Medical Res Council Production of antibodies
US5175384A (en) 1988-12-05 1992-12-29 Genpharm International Transgenic mice depleted in mature t-cells and methods for making transgenic mice
EP0814159B1 (fr) 1990-08-29 2005-07-27 GenPharm International, Inc. Souris transgéniques capables de produire des anticorps hétérologues
US5545806A (en) 1990-08-29 1996-08-13 Genpharm International, Inc. Ransgenic non-human animals for producing heterologous antibodies
US5635483A (en) 1992-12-03 1997-06-03 Arizona Board Of Regents Acting On Behalf Of Arizona State University Tumor inhibiting tetrapeptide bearing modified phenethyl amides
US20030129191A1 (en) * 1992-12-23 2003-07-10 Neorx Corporation Pretargeting methods and compounds
DE614989T1 (de) 1993-02-17 1995-09-28 Morphosys Proteinoptimierung Verfahren für in vivo Selektion von Ligandenbindende Proteine.
US6011175A (en) * 1993-05-18 2000-01-04 University Of Pittsburgh Inhibition of farnesyltransferase
DE19544393A1 (de) 1995-11-15 1997-05-22 Hoechst Schering Agrevo Gmbh Synergistische herbizide Mischungen
US7605238B2 (en) 1999-08-24 2009-10-20 Medarex, Inc. Human CTLA-4 antibodies and their uses
AU2001261409A1 (en) * 2000-05-11 2001-11-20 Oklahoma Medical Research Foundation Prevention of insulin-dependent diabetes, complications thereof, or allograft rejection by inhibition of cyclooxygenase-2 activity with NS398 or PDTC
US7256257B2 (en) 2001-04-30 2007-08-14 Seattle Genetics, Inc. Pentapeptide compounds and uses related thereto
US6884869B2 (en) 2001-04-30 2005-04-26 Seattle Genetics, Inc. Pentapeptide compounds and uses related thereto
WO2003059282A2 (fr) 2002-01-09 2003-07-24 Medarex, Inc. Anticorps monoclonaux humains contre cd30
EP1391213A1 (fr) 2002-08-21 2004-02-25 Boehringer Ingelheim International GmbH Compositions et méthodes pour le traitement du cancer en utilisant un conjugué d'un anticorps contre le CD44 avec un maytansinoide et des agents chimiothérapeutiques
SG195524A1 (en) 2003-11-06 2013-12-30 Seattle Genetics Inc Monomethylvaline compounds capable of conjugation to ligands
CA2548180C (fr) * 2003-12-04 2014-02-04 Vaccinex, Inc. Procedes d'elimination de cellules tumorales par ciblage d'antigene interne expose sur des cellules tumorales apoptotiques
AU2005205867A1 (en) 2004-01-21 2005-08-04 Novo Nordisk Health Care Ag Transglutaminase mediated conjugation of peptides
TW200539855A (en) 2004-03-15 2005-12-16 Wyeth Corp Calicheamicin conjugates
US20050266008A1 (en) 2004-03-29 2005-12-01 Medarex, Inc. Human anti-IRTA-5 antibodies
SV2007002227A (es) 2004-09-10 2007-03-20 Wyeth Corp Anticuerpos anti-5t4 humanizados y conjugados anticuerpo anti-5t4/calicheamicina ref. 040000-0317637
JP5061096B2 (ja) 2005-04-21 2012-10-31 スピロゲン リミティッド ピロロベンゾジアゼピン
ES2708763T3 (es) 2005-07-07 2019-04-11 Seattle Genetics Inc Compuestos de monometilvalina que tienen modificaciones de la cadena lateral de fenilalanina en el extremo C
EP4026840A1 (fr) * 2005-07-18 2022-07-13 Seagen Inc. Conjugués de médicaments de linker bêta-glucuronide
WO2007030642A2 (fr) * 2005-09-07 2007-03-15 Medimmune, Inc. Anticorps anti-recepteur eph conjugues a des toxines
JP2009509510A (ja) 2005-09-26 2009-03-12 メダレックス インコーポレーティッド Cd70に対するヒトモノクローナル抗体
WO2007112007A2 (fr) * 2006-03-22 2007-10-04 University Of Utah Research Foundation Protéines immobilisées et procédés et utilisations de celles-ci
JP2009530392A (ja) * 2006-03-22 2009-08-27 ザ リージェンツ オブ ザ ユニバーシティー オブ カリフォルニア タンパク質プレニルトランスフェラーゼの阻害剤
AU2007333098A1 (en) 2006-12-14 2008-06-19 Medarex, Inc. Human antibodies that bind CD70 and uses thereof
PE20090481A1 (es) * 2007-07-16 2009-05-18 Genentech Inc Anticuerpos anti-cd79b e inmunoconjugados humanizados y metodos de uso
WO2009026274A1 (fr) * 2007-08-22 2009-02-26 Medarex, Inc. Attachement spécifique d'un site de médicaments ou autres agents à des anticorps synthétisés par génie génétique avec des extensions c-terminales
WO2009038685A1 (fr) 2007-09-18 2009-03-26 The Scripps Research Institute Ligands pour des réactions de cyclo-addition azide-alkyne catalysés au cuivre
CA2720247C (fr) * 2008-03-31 2020-07-14 Pacific Biosciences Of California, Inc. Procedes et compositions de charge de molecule individuelle
GB0813432D0 (en) 2008-07-22 2008-08-27 Spirogen Ltd Pyrrolobenzodiazepines
GB0819095D0 (en) 2008-10-17 2008-11-26 Spirogen Ltd Pyrrolobenzodiazepines
CN102012425A (zh) * 2010-12-29 2011-04-13 南开大学 一种新的免疫荧光标记方法
PL2707031T3 (pl) 2011-05-08 2020-01-31 Legochem Biosciences, Inc. Koniugaty białko-środek czynny i sposoby i ch wytwarzania
KR101628872B1 (ko) * 2014-05-28 2016-06-09 주식회사 레고켐 바이오사이언스 자가-희생 기를 포함하는 화합물

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030017149A1 (en) * 1996-10-10 2003-01-23 Hoeffler James P. Single chain monoclonal antibody fusion reagents that regulate transcription in vivo
US20070141084A1 (en) * 2003-08-06 2007-06-21 Lee Che-Hung R Polysaccharide-protein conjugate vaccines
US20070122408A1 (en) * 2005-10-20 2007-05-31 The Scripps Research Institute Fc Labeling for Immunostaining and Immunotargeting

Non-Patent Citations (18)

* Cited by examiner, † Cited by third party
Title
Bowie et al. Deciphering the Message in Protein Sequences: Tolerance to Amino Acid Substitutions. Science, 247:1306-1310, 1990 *
Burgess et al. Possible dissociation of the heparin-binding and mitogenic activities of heparin-binding (acidic fibroblast) growth factor-1 from its receptor-binding activities by site-directed mutagenesis of a single lysine residue. Journal of Cell Biology 111:2129-2138, 1990 *
Burgess et al. Possible dissociation of the heparin-binding and mitogenic activities of heparin-binding (acidic fibroblast) growth factor-1 from its receptor-binding activities by site-directed mutagenesis of a single lysine residue. Journal of Cell Biology 111:2129-2138, 1990. *
Chamnongpol et al. SH3 Domain Protein-Binding Arrays. Methods Mol Biol. 2004;264:183-9. *
Dongen et al. Immuno-PET: A Navigator in Monoclonal Antibody Development and Applications. The Oncologist 2007;12:1379-1389. *
Greenspan et al. Defining epitopes: It’s not as easy as it seems. Nature Biotechnology 7: 936-937, 1999 *
Greenspan et al. Defining epitopes: It’s not as easy as it seems. Nature Biotechnology 7: 936-937, 1999. *
Jain et al. Structure of human beta-glucuronidase reveals candidate lysosomal targeting and active-site motifs. Nat Struct Biol. 1996 Apr;3(4):375-81. *
Jeffrey et al. Dipeptide-based highly potent doxorubicin antibody conjugates. Bioorg Med Chem Lett. 2006 Jan 15;16(2):358-62. *
Kolb et al. The growing impact of click chemistry on drug discovery. Drug Discov Today. 2003 Dec 15;8(24):1128-37. *
Labadie et al. Farnesyl Diphosphate Analogues with omega-Bioorthogonal Azide and Alkyne Functional Groups for PFTase-Catalyzed Ligation Reactions. J Org Chem. 2007 November 23; 72(24): 9291-9297. *
Lazar et al. Transforming Growth Factor alpha: Mutation of Aspartic Acid 47 and Leucine 48 Results in Different Biological ActivitiesMolecular Cellular Biology, 8:1247- 1252, 1988. *
Lewis et al. Targeting HER2-Positive Breast Cancer with Trastuzumab-DM1, an Antibody-Cytotoxic Drug Conjugate. Cancer Res 2008; 68: (22). November 15, 2008. *
Nguyen et al. Analysis of the eukaryotic prenylome by isoprenoid affinity tagging. Nat Chem Biol. 2009 Apr;5(4):227-35. *
Reiss et al. Sequence requirement for peptide recognition by rat brain p2lras protein farnesyltransferase. Proc Natl Acad Sci U S A. 1991 Feb 1;88(3):732-6. *
Rowinsky et al. Ras Protein Farnesyltransferase: A Strategic Target for Anticancer Therapeutic Development. J Clin Oncol. 1999 Nov;17(11):3631-52. *
Tanizawa et al. Comparison of topoisomerase I inhibition, DNA damage, and cytotoxicity of camptothecin derivatives presently in clinical trials. Journal of the National Cancer Institute. 1994; 86(11); 836-842. *
Wright et al. Thematic review series: lipid posttranslational modifications. CAAX modification and membrane targeting of Ras. J Lipid Res. 2006 May;47(5):883-91. Epub 2006 Mar 16. *

Cited By (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3611180A1 (fr) * 2013-03-15 2020-02-19 Daniel J. Capon Immunoglobuline hybride contenant une liaison non peptidyle
EP2968495A4 (fr) * 2013-03-15 2016-11-09 Daniel J Capon Immunoglobuline hybride contenant une liaison non peptidique
US11220556B2 (en) 2013-03-15 2022-01-11 Biomolecular Holdings Llc Hybrid immunoglobulin containing non-peptidyl linkage
AU2014373593B2 (en) * 2013-12-23 2020-07-16 Zymeworks Bc Inc. Antibodies comprising C-terminal light chain polypeptide extensions and conjugates and methods of use thereof
JP2017501728A (ja) * 2013-12-23 2017-01-19 ザイムワークス インコーポレイティド C末端軽鎖ポリペプチド伸長を含有する抗体、ならびにその複合体及びその使用方法
US11208497B2 (en) * 2013-12-23 2021-12-28 Zymeworks Inc. Antibodies comprising C-terminal light chain polypeptide extensions and conjugates and methods of use thereof
US20170008970A1 (en) * 2013-12-23 2017-01-12 Zymeworks Inc. Antibodies comprising c-terminal light chain polypeptide extensions and conjugates and methods of use thereof
US11066459B2 (en) 2014-03-14 2021-07-20 Biomolecular Holdings Llc Hybrid immunoglobulin containing non-peptidyl linkage
US9993568B2 (en) 2014-05-28 2018-06-12 Legochem Biosciences, Inc. Compounds comprising self-immolative group
AU2021204136B2 (en) * 2014-05-28 2023-05-11 Legochem Biosciences, Inc. Compound containing self-immolative group
US9919057B2 (en) 2014-05-28 2018-03-20 Legochem Biosciences, Inc. Compounds comprising self-immolative group
EP3156424A4 (fr) * 2014-05-28 2018-01-24 LegoChem Biosciences, Inc. Composé contenant un groupe auto-immolable
US10383949B2 (en) 2014-05-28 2019-08-20 Legochem Biosciences, Inc. Compounds comprising self-immolative group
RU2712744C2 (ru) * 2014-05-28 2020-01-30 Легокем Байосайенсез, Инк. Соединения, включающие саморасщепляющуюся группу
AU2015268300B2 (en) * 2014-05-28 2021-03-25 Legochem Biosciences, Inc. Compound containing self-immolative group
US10980890B2 (en) 2014-05-28 2021-04-20 Legochem Biosciences, Inc. Compounds comprising self-immolative group
KR20160080832A (ko) 2014-12-29 2016-07-08 주식회사 레고켐 바이오사이언스 리피바디 유도체-약물 복합체, 그 제조방법 및 용도
US10183997B2 (en) 2015-09-25 2019-01-22 Legochem Biosciences, Inc. Compositions and methods related to anti-CD19 antibody drug conjugates
US10118965B2 (en) 2015-09-25 2018-11-06 Legochem Biosciences, Inc. Compositions and methods related to anti-EGFR antibody drug conjugates
US11167040B2 (en) 2015-11-25 2021-11-09 Legochem Biosciences, Inc. Conjugates comprising peptide groups and methods related thereto
US11173214B2 (en) 2015-11-25 2021-11-16 Legochem Biosciences, Inc. Antibody-drug conjugates comprising branched linkers and methods related thereto
WO2017089890A1 (fr) 2015-11-25 2017-06-01 Legochem Biosciences, Inc. Conjugués comprenant des groupes auto-immolables et procédés associés
US11413353B2 (en) 2015-11-25 2022-08-16 Legochem Biosciences, Inc. Conjugates comprising self-immolative groups and methods related thereto
US11975076B2 (en) 2015-11-25 2024-05-07 Legochem Biosciences, Inc. Antibody-drug conjugates comprising branched linkers and methods related thereto
US11654197B2 (en) 2017-03-29 2023-05-23 Legochem Biosciences, Inc. Pyrrolobenzodiazepine dimer prodrug and ligand-linker conjugate compound of the same
CN108743968A (zh) * 2017-06-20 2018-11-06 四川百利药业有限责任公司 半胱氨酸改造的抗体-毒素偶联物(tdc)定点偶联位点筛选
US11827703B2 (en) 2018-05-09 2023-11-28 Legochem Biosciences, Inc. Compositions and methods related to anti-CD19 antibody drug conjugates
US11707533B2 (en) 2019-09-04 2023-07-25 Legochem Biosciences, Inc. Antibody-drug conjugate comprising antibody against human ROR1 and use for the same
WO2023166322A1 (fr) 2022-03-04 2023-09-07 Iksuda Therapeutics Limited Conjugué d'anticorps anti-canag

Also Published As

Publication number Publication date
JP6196613B2 (ja) 2017-09-13
JP2014519492A (ja) 2014-08-14
US20200289662A1 (en) 2020-09-17
LT2707031T (lt) 2019-10-10
RU2013154264A (ru) 2015-06-20
US20160213786A1 (en) 2016-07-28
EP2707031B1 (fr) 2019-06-26
PT2707031T (pt) 2019-09-05
CA2835576C (fr) 2021-01-05
US9669107B2 (en) 2017-06-06
HUE045348T2 (hu) 2019-12-30
JP2017206515A (ja) 2017-11-24
CY1122152T1 (el) 2020-11-25
US20140161829A1 (en) 2014-06-12
SG194875A1 (en) 2013-12-30
KR20140035393A (ko) 2014-03-21
MX2013013069A (es) 2014-01-31
IL269804A (en) 2019-11-28
US20180021451A1 (en) 2018-01-25
MX364707B (es) 2019-05-06
HRP20191586T1 (hr) 2019-12-13
KR101921068B1 (ko) 2018-11-22
BR122020001787A8 (pt) 2023-04-25
EP3610889A1 (fr) 2020-02-19
EP2707031A4 (fr) 2015-03-25
MX2019001253A (es) 2019-07-04
CA2835576A1 (fr) 2012-11-15
CN103648530B (zh) 2017-02-08
BR122020001787A2 (fr) 2017-01-31
RS59253B1 (sr) 2019-10-31
JP6385525B2 (ja) 2018-09-05
WO2012153193A3 (fr) 2013-04-11
IL229364A0 (en) 2014-01-30
RU2613906C2 (ru) 2017-03-21
US10583197B2 (en) 2020-03-10
BR112013028907A2 (pt) 2017-01-31
US20140187756A1 (en) 2014-07-03
IL229364B (en) 2021-08-31
SI2707031T1 (sl) 2019-10-30
US20150105541A1 (en) 2015-04-16
WO2012153193A2 (fr) 2012-11-15
EP2707031A2 (fr) 2014-03-19
DK2707031T3 (da) 2019-10-07
ES2744226T3 (es) 2020-02-24
PL2707031T3 (pl) 2020-01-31
CN103648530A (zh) 2014-03-19

Similar Documents

Publication Publication Date Title
US20200289662A1 (en) Antibody-active agent conjugates and methods of use
US10980890B2 (en) Compounds comprising self-immolative group
US20220218840A1 (en) Antibody-drug conjugates comprising branched linkers and methods related thereto
US11413353B2 (en) Conjugates comprising self-immolative groups and methods related thereto
US10183997B2 (en) Compositions and methods related to anti-CD19 antibody drug conjugates
US10118965B2 (en) Compositions and methods related to anti-EGFR antibody drug conjugates
US11065343B2 (en) Compound bearing beta-galactoside-introduced self-immolative linker
US20220226496A1 (en) Ligand-drug conjugate including linker having tris structure
EP4257606A1 (fr) Préparation et utilisation d'un anticorps anti-fshr et d'un conjugué anticorps-médicament de celui-ci

Legal Events

Date Code Title Description
AS Assignment

Owner name: LEGOCHEM BIOSCIENCES, INC., KOREA, DEMOCRATIC PEOP

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:KIM, YONGZU, MR.;PARK, TAEKYO, MR.;WOO, SUNGHO;AND OTHERS;REEL/FRAME:028176/0354

Effective date: 20120507

STCB Information on status: application discontinuation

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