US20220023433A1 - Saponin Conjugates - Google Patents

Saponin Conjugates Download PDF

Info

Publication number
US20220023433A1
US20220023433A1 US17/312,104 US201917312104A US2022023433A1 US 20220023433 A1 US20220023433 A1 US 20220023433A1 US 201917312104 A US201917312104 A US 201917312104A US 2022023433 A1 US2022023433 A1 US 2022023433A1
Authority
US
United States
Prior art keywords
saponin
bond
effector moiety
cell
antibody
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.)
Pending
Application number
US17/312,104
Other languages
English (en)
Inventor
Ruben Postel
Hendrik Fuchs
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.)
Sapreme Technologies BV
Original Assignee
Charite Universitaetsmedizin Berlin
Sapreme Technologies BV
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 Charite Universitaetsmedizin Berlin, Sapreme Technologies BV filed Critical Charite Universitaetsmedizin Berlin
Assigned to CHARITÉ - UNIVERSITÄTSMEDIZIN BERLIN, SAPREME TECHNOLOGIES B.V. reassignment CHARITÉ - UNIVERSITÄTSMEDIZIN BERLIN ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: FUCHS, HENDRIK, POSTEL, Ruben
Publication of US20220023433A1 publication Critical patent/US20220023433A1/en
Assigned to SAPREME TECHNOLOGIES B.V. reassignment SAPREME TECHNOLOGIES B.V. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CHARITÉ - UNIVERSITÄTSMEDIZIN BERLIN
Pending 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
    • 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
    • A61K47/6883Polymer-drug antibody conjugates, e.g. mitomycin-dextran-Ab; DNA-polylysine-antibody complex or conjugate used for therapy
    • A61K47/6885Polymer-drug antibody conjugates, e.g. mitomycin-dextran-Ab; DNA-polylysine-antibody complex or conjugate used for therapy the conjugate or the polymer being a starburst, a dendrimer, a cascade
    • 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
    • A61K47/54Medicinal 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
    • A61K47/554Medicinal 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 a steroid plant sterol, glycyrrhetic acid, enoxolone or bile acid
    • 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
    • A61K47/56Medicinal 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 macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
    • A61K47/59Medicinal 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 macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes
    • A61K47/593Polyesters, e.g. PLGA or polylactide-co-glycolide
    • 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
    • A61K47/56Medicinal 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 macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
    • A61K47/59Medicinal 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 macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes
    • A61K47/595Polyamides, e.g. nylon
    • 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
    • A61K47/56Medicinal 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 macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
    • A61K47/59Medicinal 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 macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes
    • A61K47/60Medicinal 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 macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes the organic macromolecular compound being a polyoxyalkylene oligomer, polymer or dendrimer, e.g. PEG, PPG, PEO or polyglycerol
    • 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
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • 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
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • A61K47/6415Toxins or lectins, e.g. clostridial toxins or Pseudomonas exotoxins
    • 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
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • A61K47/642Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent the peptide or protein in the drug conjugate being a cytokine, e.g. IL2, chemokine, growth factors or interferons being the inactive part of the conjugate
    • 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
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • A61K47/643Albumins, e.g. HSA, BSA, ovalbumin or a Keyhole Limpet Hemocyanin [KHL]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6801Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
    • A61K47/6803Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates
    • A61K47/6807Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates the drug or compound being a sugar, nucleoside, nucleotide, nucleic acid, e.g. RNA antisense
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6801Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
    • A61K47/6803Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates
    • A61K47/6811Drugs 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
    • A61K47/6801Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
    • A61K47/6803Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates
    • A61K47/6811Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates the drug being a protein or peptide, e.g. transferrin or bleomycin
    • A61K47/6817Toxins
    • A61K47/6819Plant toxins
    • A61K47/6825Ribosomal inhibitory proteins, i.e. RIP-I or RIP-II, e.g. Pap, gelonin or dianthin
    • 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
    • A61K47/6845Medicinal 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 cytokine, e.g. growth factors, VEGF, TNF, a lymphokine or an interferon
    • 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
    • A61K47/6857Medicinal 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 lung 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
    • A61K47/6863Medicinal 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 stomach or intestines 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
    • 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
    • A61K47/69Medicinal 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 conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
    • A61K47/6905Medicinal 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 conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a colloid or an emulsion
    • A61K47/6911Medicinal 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 conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a colloid or an emulsion the form being a liposome
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H15/00Compounds containing hydrocarbon or substituted hydrocarbon radicals directly attached to hetero atoms of saccharide radicals
    • C07H15/20Carbocyclic rings
    • C07H15/24Condensed ring systems having three or more rings
    • C07H15/256Polyterpene radicals
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • A61K2039/507Comprising a combination of two or more separate antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2300/00Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07JSTEROIDS
    • C07J63/00Steroids in which the cyclopenta(a)hydrophenanthrene skeleton has been modified by expansion of only one ring by one or two atoms
    • C07J63/008Expansion of ring D by one atom, e.g. D homo steroids
    • 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
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/11Antisense
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2320/00Applications; Uses
    • C12N2320/30Special therapeutic applications

Definitions

  • the current invention relates to an effector moiety capable of inducing an intracellular effect when present inside a mammalian cell, the effector moiety conjugated with at least one saponin.
  • the current invention relates to an effector moiety capable of inducing an intracellular effect when present inside a mammalian cell, the effector moiety comprising a payload conjugated with at least one saponin.
  • the invention also relates to an antibody-drug conjugate comprising the effector moiety according to the invention, or a ligand-drug conjugate comprising the effector moiety of the invention, the effector moiety comprising covalently coupled saponin.
  • the invention also relates to a therapeutic combination comprising: (a) the effector moiety of the invention, comprising at least one saponin, and optionally a pharmaceutically acceptable excipient; and (b) an antibody-drug conjugate or a ligand-drug conjugate, and optionally a pharmaceutically acceptable excipient.
  • a pharmaceutical composition comprising the effector moiety of the invention or the antibody-drug conjugate of the invention or the ligand-drug conjugate of the invention, comprising at least one saponin covalently linked to the effector molecule, and optionally a pharmaceutically acceptable excipient.
  • the invention also relates to the effector moiety of the invention or the antibody-drug conjugate of the invention or the therapeutic combination of the invention or the ligand-drug conjugate of the invention or the pharmaceutical composition of the invention, for use as a medicament.
  • the invention also relates to the effector moiety of the invention or the antibody-drug conjugate of the invention or the therapeutic combination of the invention or the ligand-drug conjugate of the invention or the pharmaceutical composition of the invention, for use in the treatment or prevention of a cancer or an autoimmune disease.
  • Molecules with a therapeutic biological activity are in many occasions in theory suitable for application as an effective therapeutic drug for the treatment of a disease such as a cancer in human patients in need thereof.
  • a typical example are small-molecule biologically active moieties.
  • therapeutically active molecules may exert off-target effects, in addition to the biologically activity directed to an aspect underlying a to-be-treated disease or health problem. Such off-target effects are undesired and bear a risk for induction of health- or even life-threatening side effects of the administered molecule.
  • the administered drug molecule should reach the targeted site in the human patient within a certain time frame and should remain at the targeted site for a certain time frame), and/or (6) have sufficiently long lasting therapeutic activity in the patient's body, amongst others.
  • ‘ideal’ therapeutics with many or even all of the beneficial characteristics here above outlined, are not available to the patients, despite already long-lasting and intensive research and despite the impressive progress made in several areas of the individually addressed encountered difficulties and drawbacks.
  • Chemotherapy is one of the most important therapeutic options for cancer treatment. However, it is often associated with a low therapeutic window because it has no specificity towards cancer cells over dividing cells in healthy tissue.
  • the invention of monoclonal antibodies offered the possibility of exploiting their specific binding properties as a mechanism for the targeted delivery of cytotoxic agents to cancer cells, while sparing normal cells. This can be achieved by chemical conjugation of cytotoxic effectors (also known as payloads or warheads) to antibodies, to create antibody—drug conjugates (ADCs).
  • cytotoxic effectors also known as payloads or warheads
  • ADCs antibody—drug conjugates
  • very potent payloads such as emtansine (DM1) are used which have a limited therapeutic index (a ratio that compares toxic dose to efficacious dose) in their unconjugated forms.
  • Mylotarg was however, withdrawn from the market at the request of the Federal Drug Administration (FDA) due to a number of concerns including its safety profile. Patients treated with Mylotarg were more often found to die than patients treated with conventional chemotherapy. Mylotarg was admitted to the market again in 2017 with a lower recommended dose, a different schedule in combination with chemotherapy or on its own, and a new patient population. To date, only five ADCs have been approved for clinical use, and meanwhile clinical development of approximately fifty-five ADCs has been halted. However, interest remains high and approximately eighty ADCs are still in clinical development in nearly six-hundred clinical trials at present.
  • FDA Federal Drug Administration
  • a low therapeutic index (a ratio that compares toxic dose to efficacious dose) is a major problem accounting for the discontinuance of many ADCs in clinical development, which can be caused by several mechanisms such as off-target toxicity on normal cells, development of resistance against the cytotoxic agents and premature release of drugs in the circulation.
  • a systematic review by the FDA of ADCs found that the toxicity profiles of most ADCs could be categorized according to the payload used, but not the antibody used, suggesting that toxicity is mostly determined by premature release of the payload. Of the approximately fifty-five ADCs that were discontinued, it is estimated that at least twenty-three were due to a poor therapeutic index.
  • trastuzumab tesirine conjugate ADCT-502, HER-2 targeted, ADC therapeutics
  • ADCT-502, HER-2 targeted, ADC therapeutics were recently discontinued due to a narrow therapeutic index, possibly due to an on-target, off-tissue effect in pulmonary tissue which expresses considerable levels of HER2.
  • ADCs in phase 3 trials have been discontinued due to missing primary endpoint.
  • ABT-414 EGFR targeted, AbbVie
  • IMGN853 folate receptor alpha
  • FRa folate receptor alpha
  • ImmunoGen immunogen
  • ADCs are mainly composed of an antibody, a cytotoxic moiety such as a payload, and a linker.
  • new clinical and translational strategies are also being deployed to maximize the therapeutic index, such as, change dosing schedules through fractionated dosing; perform biodistribution studies; include biomarkers to optimize patient selection, to capture response signals early and monitor the duration and depth of response, and to inform combination studies.
  • ADCs with clinical potential are those ADCs such as brentuximab vedotin, inotuzumab ozogamicin, moxetumomab pasudotox, and polatuzumab vedotin, which are evaluated as a treatment option for lymphoid malignancies and multiple myeloma.
  • Polatuzumab vedotin, binding to CD79b on (malignant) B-cells, and pinatuzumab vedotin, binding to CD22 are tested in clinical trials wherein the ADCs each were combined with co-administered rituximab, a monoclonal antibody binding to CD20 and not provided with a payload [B. Yu and D. Liu, Antibody - drug conjugates in clinical trials for lymphoid malignancies and multiple myeloma; Journal of Hematology & Oncology (2019) 12:94]. Combinations of monoclonal antibodies such as these examples are yet a further approach and attempt to arrive at the ‘magic bullet’ which combines many or even all of the aforementioned desired characteristics of ADCs.
  • nucleic acid-based therapeutics are under development.
  • Therapeutic nucleic acids can be based on deoxyribonucleic acid (DNA) or ribonucleic acid (RNA), Anti-sense oligonucleotides (ASOs, AONs), and short interfering RNAs (siRNAs), MicroRNAs, and DNA and RNA aptamers, for approaches such as gene therapy, RNA interference (RNAi).
  • RNAi RNA interference
  • Many of them share the same fundamental basis of action by inhibition of either DNA or RNA expression, thereby preventing expression of disease-related abnormal proteins.
  • the largest number of clinical trials is being carried out in the field of gene therapy, with almost 2600 ongoing or completed clinical trials worldwide but with only about 4% entering phase 3.
  • ASOs peptide nucleic acid
  • PMO phosphoramidate morpholino oligomer
  • LNA locked nucleic acid
  • BNA bridged nucleic acid
  • ASOs as potential therapeutic agents
  • the application of ASOs as potential therapeutic agents requires safe and effective methods for their delivery to the cytoplasm and/or nucleus of the target cells and tissues.
  • inefficient cellular uptake both in vitro and in vivo, limit the efficacy of ASOs and has been a barrier to therapeutic development.
  • Cellular uptake can be ⁇ 2% of the dose resulting in too low ASO concentration at the active site for an effective and sustained outcome. This consequently requires an increase of the administered dose which induces off-target effects.
  • Most common side-effects are activation of the complement cascade, the inhibition of the clotting cascade and toll-like receptor mediated stimulation of the immune system.
  • Chemotherapeutics are most commonly small molecules, however, their efficacy is hampered by the severe off-target side toxicity, as well as their poor solubility, rapid clearance and limited tumor exposure.
  • Scaffold-small-molecule drug conjugates such as polymer-drug conjugates (PDCs) are macromolecular constructs with pharmacologically activity, which comprises one or more molecules of a small-molecule drug bound to a carrier scaffold (e.g. polyethylene glycol (PEG)).
  • PDCs polymer-drug conjugates
  • PK1 N-(2-hydroxypropyl)methacrylamide (HPMA) copolymer doxorubicin; development by Pharmacia, Pfizer
  • HPMA 2-hydroxypropyl)methacrylamide copolymer doxorubicin
  • scaffold-small-molecule drug conjugates is at least partially attributed to its poor accumulation at the tumor site.
  • PK1 showed 45-250 times higher accumulation in the tumor than in healthy tissues (liver, kidney, lung, spleen, and heart), accumulation in tumor was only observed in a small subset of patients in the clinical trial.
  • Liposomes are sphere-shaped vesicles consisting of one or more phospholipid bilayers, which are spontaneously formed when phospholipids are dispersed in water.
  • the amphiphilicity characteristics of the phospholipids provide it with the properties of self-assembly, emulsifying and wetting characteristics, and these properties can be employed in the design of new drugs and new drug delivery systems.
  • Drug encapsulated in a liposomal delivery system may convey several advantages over a direct administration of the drug, such as an improvement and control over pharmacokinetics and pharmacodynamics, tissue targeting property, decreased toxicity and enhanced drug activity.
  • doxorubicin a small molecule chemotherapy agent doxorubicin
  • Doxil a pegylated liposome-encapsulated form of doxorubicin
  • Myocet a non-pegylated liposomal doxorubicin
  • At least one of the above objectives of embodiments of the invention is achieved by providing an effector moiety capable of inducing an intracellular effect when present inside a mammalian cell, the effector moiety conjugated with at least one saponin, wherein the at least one saponin is covalently bound to the effector moiety via at least one linker, or is covalently bound directly to said effector moiety.
  • An aspect of the invention relates to an effector moiety capable of inducing an intracellular effect when present inside a mammalian cell, the effector moiety conjugated with at least one saponin, wherein the at least one saponin is covalently bound to the effector moiety via at least one linker, or is covalently bound directly to said effector moiety.
  • An aspect of the invention relates to an antibody-drug conjugate comprising the effector moiety according to the invention, or a ligand-drug conjugate comprising the effector moiety of the invention, the effector moiety comprising covalently coupled saponin.
  • the effector moiety of the invention is a conjugate comprising at least one saponin and at least one effector molecule, covalently coupled to each other, either directly, or via at least one linker, optionally comprising a cleavable linker, and optionally via an oligomeric or polymeric scaffold to which the at least one saponin and/or the at least one effector moiety are covalently bound.
  • An aspect of the invention relates to a therapeutic combination comprising: (a) the effector moiety of the invention, comprising at least one saponin, and optionally a pharmaceutically acceptable excipient; and (b) an antibody-drug conjugate or a ligand-drug conjugate, and optionally a pharmaceutically acceptable excipient.
  • An aspect of the invention relates to a pharmaceutical composition
  • a pharmaceutical composition comprising the effector moiety of the invention or the antibody-drug conjugate of the invention or the ligand-drug conjugate of the invention, comprising at least one saponin covalently linked to the effector molecule, and optionally a pharmaceutically acceptable excipient.
  • An aspect of the invention relates to the effector moiety of the invention or the antibody-drug conjugate of the invention or the therapeutic combination of the invention or the ligand-drug conjugate of the invention or the pharmaceutical composition of the invention, for use as a medicament.
  • An aspect of the invention relates to the effector moiety of the invention or the antibody-drug conjugate of the invention or the therapeutic combination of the invention or the ligand-drug conjugate of the invention or the pharmaceutical composition of the invention, for use in the treatment or prevention of a cancer or an autoimmune disease.
  • linker has its regular scientific meaning, and here refers to a chemical moiety or a linear stretch of amino-acid residues complexed through peptide bonds, which attaches a molecule or an atom to another molecule, e.g. to a ligand or to an effector molecule or to a scaffold.
  • the linker comprises a chain of atoms linked by chemical bonds.
  • Any linker molecule or linker technology known in the art can be used in the present disclosure.
  • the linker is a linker for covalently binding of molecules through a chemical group on such a molecule suitable for forming a covalent linkage or bond with the linker.
  • the linker may be a non-cleavable linker, e.g., the linker is stable in physiological conditions.
  • the linker may be a cleavable linker, e.g. a linker that is cleavable, in the presence of an enzyme or at a particular pH range or value, or under physiological conditions such as intracellular conditions in the endosomes such as the late endosomes and the lysosomes of mammalian cells such as human cells.
  • linkers that can be used in the context of the present disclosure includes, but is not limited to, N- ⁇ -maleimidocaproic acid hydrazide (EMCH), succinimidyl 3-(2-pyridyldithio)propionate or 3-(2-Pyridyldithio)propionic acid N-hydroxysuccinimide ester (SPDP), and 1-[Bis(dimethylamino)methylene]-1H-1,2,3-triazolo[4,5-b]pyridinium 3-oxid hexafluorophosphate (HATU).
  • EMCH N- ⁇ -maleimidocaproic acid hydrazide
  • SPDP succinimidyl 3-(2-pyridyldithio)propionate or 3-(2-Pyridyldithio)propionic acid N-hydroxysuccinimide ester
  • HATU 1-[Bis(dimethylamino)methylene]-1H-1,2,3-triazolo[4,5-b
  • tri-functional linker has its regular scientific meaning, and here refers to a linker which attaches three molecules via a chemical group on each of the three molecules.
  • the skilled person is able to design such tri-functional linkers, based on the present disclosure and the common general knowledge.
  • Such tri-functional linker can exhibit, for instance, a maleimido group that can be used for conjugation to targeting ligands that exhibit thiol groups to perform a thiol-ene reaction.
  • the tri-functional linker could exhibit a dibenzocyclooctyne (DBCO) group to perform the so-called strain-promoted alkyne-azide cycloaddition (SPAAC, click chemistry) with an azido bearing saponin.
  • DBCO dibenzocyclooctyne
  • SPAAC strain-promoted alkyne-azide cycloaddition
  • the tri-functional linker could obtain a third functional group such as a trans-cyclooctene (TCO) group to perform the so-called inverse electron demand Diels-Alder (IEDDA) reaction with a tetrazine (Tz) bearing effector molecule.
  • TCO trans-cyclooctene
  • IEDDA inverse electron demand Diels-Alder
  • the chemical groups of the tri-functional linker can be all three the same, or different, or the linker may comprise two of the same chemical groups for linking a molecule to the tri-functional linker.
  • the formed bonds between the tri-functional linker can be covalent or non-covalent, and covalent bonds are preferred.
  • the formed bonds between the tri-functional linker and the one or two or three bound molecules via respective chemical groups can be cleavable (labile) bonds, such as cleavable under acidic conditions inside cells such as endosomes and lysosomes of mammalian cells such as human cells, or can be non-cleavable bonds.
  • the tri-functional linker may encompass one or two chemical groups for forming covalent bonds while the further two or one chemical group(s), respectively, are/is for forming a non-covalent bond.
  • the tri-functional linker may encompass one or two chemical groups for forming cleavable bonds while the further two or one chemical group(s), respectively, are/is for forming a non-cleavable bond.
  • cleavable such as used in the term “cleavable linker” or “cleavable bond” has its regular scientific meaning, and here refers to being subject to cleavage under acidic conditions, reductive conditions, enzymatic conditions or light-induced conditions.
  • a cleavable linker may be subject to cleavage under acidic conditions, preferably said cleavable linker is subject to cleavage in vivo under acidic conditions as present in endosomes and/or lysosomes of mammalian cells, preferably human cells, preferably at pH 4.0-6.5, and more preferably at pH 5.5.
  • a cleavable linker may be subject to cleavage by an enzyme, e.g. by cathepsin.
  • an example of a covalent bond cleavable under reductive conditions is a disulphide bond.
  • oligomer and “polymer” in the context of an oligomeric or polymeric scaffold has its regular scientific meaning.
  • a polymer here refers to a substance which has a molecular structure built up chiefly or completely from a large number of equal or similar units bonded together; an oligomer here refers to a polymer whose molecules consist of relatively few repeating units.
  • a structure comprising 5-10 or less equal or similar units may be called an oligomeric structure
  • a structure comprising 10-50 monomeric units or more may be called a polymeric structure
  • a structure of 10 monomeric units may be called either oligomeric or polymeric.
  • binding site has its regular scientific meaning, and here refers to a region or an epitope on a molecule, e.g. a protein, DNA or RNA, to which another molecule can bind.
  • scaffold has its regular scientific meaning, and here refers to an oligomeric or polymeric template or a carrier or a base (base molecule or base structure), to which one or more molecules, e.g. ligand molecule, saponin of the invention, glycoside, effector molecule, can be covalently bound, either directly, or via a linker, such as a cleavable linker.
  • ligand molecule e.g. ligand molecule, saponin of the invention, glycoside, effector molecule
  • a scaffold may have a structurally ordered formation such as a polymer, oligomer, dendrimer, dendronized polymer, or dendronized oligomer or have an assembled polymeric structure such as a hydrogel, microgel, nanogel, stabilized polymeric micelle or liposome, but excludes structures that are composed of non-covalent assemblies of monomers such as cholesterol/phospholipid mixtures.
  • a scaffold may comprise a polymeric or oligomeric structure, such as poly- or oligo(amines), e.g., polyethylenimine and poly(amidoamine); or structures such as polyethylene glycol, poly- or oligo(esters), such as poly(lactids), poly(lactams), polylactide-co-glycolide copolymers; or poly(dextrin), poly- or oligosaccharides, such as cyclodextrin or polydextrose; or structures such as natural and/or artificial poly- or oligoamino acids such as poly-lysine or a peptide or a protein, DNA oligo- or polymers, stabilized RNA polymers or PNA (peptide nucleic acid) polymers.
  • polymeric or oligomeric structure such as poly- or oligo(amines), e.g., polyethylenimine and poly(amidoamine); or structures such as polyethylene glycol, poly- or
  • the polymeric or oligomeric structures are biocompatible, wherein biocompatible means that the polymeric or oligomeric structure does not show substantial acute or chronic toxicity in organisms and can be either excreted as it is or fully degraded to excretable and/or physiological compounds by the body's metabolism.
  • ligand has its regular scientific meaning, and here refers to any molecule or molecules which may selectively bind to a target cell-surface molecule or target cell-surface receptor expressed at target cells, e.g. target cancer cells or target auto-immune cells.
  • the ligand may bind to an epitope comprised by receptors or other antigens on the target cells.
  • the cell-binding ligands are antibodies.
  • antibody as used herein is used in the broadest sense, which may refer to an immunoglobulin (Ig) defined as a protein belonging to the class IgG, IgM, IgE, IgA, or IgD (or any subclass thereof), or a functional binding fragment or binding domain of an immunoglobulin.
  • Ig immunoglobulin
  • a “binding fragment” or a “binding domain” of an immunoglobulin is defined as antigen-binding fragment or -domain or other derivative of a parental immunoglobulin that essentially maintains the antigen binding activity of such parental immunoglobulin.
  • Functional fragments and functional domains are antibodies in the sense of the present invention even if their affinity to the antigen is lower than that of the parental immunoglobulin.
  • “Functional fragments and -domains” in accordance with the invention include, but are not limited to, F(ab′)2 fragments, Fab′ fragments, Fab fragments, scFv, dsFv, single-domain antibody (sdAb), monovalent IgG, scFv-Fc, reduced IgG (rlgG), minibody, diabodies, triabodies, tetrabodies, Fc fusion proteins, nanobodies, variable V domains such as VHH, Vh, and other types of antigen recognizing immunoglobulin fragments and domains.
  • the fragments and domains may be engineered to minimize or completely remove the intermolecular disulphide interactions that occur between the CH1 and CL domains.
  • Functional fragment and -domains offer the advantage of greater tumor penetration because of their smaller size. In addition, the functional fragment or -domain can be more evenly distributed throughout the tumor mass as compared to whole immunoglobulin.
  • the antibodies (immunoglobulins) of the present invention may be bi- or multifunctional.
  • a bifunctional antibody has one arm having a specificity for one receptor or antigen, while the other arm recognizes a different receptor or antigen.
  • each arm of the bifunctional antibody may have specificity for a different epitope of the same receptor or antigen of the target cell.
  • the antibodies (immunoglobulins) of the present invention may be, but are not limited to, polyclonal antibodies, monoclonal antibodies, human antibodies, humanized antibodies, chimeric antibodies, resurfaced antibodies, anti-idiotypic antibodies, mouse antibodies, rat antibodies, rat/mouse hybrid antibodies, llama antibodies, llama heavy-chain only antibodies, heavy-chain only antibodies, and veterinary antibodies.
  • the antibody (immunoglobulin) of the present invention is a monoclonal antibody.
  • the resurfaced, chimeric, humanized and fully human antibodies are also more preferred because they are less likely to cause immunogenicity in humans.
  • the antibodies of the ADC of the present invention preferably specifically binds to an antigen expressed on the surface of a cancer cell, an autoimmune cell, a diseased cell, an aberrant cell, while leaving any healthy cell essentially unaltered (e.g. by not binding to such normal cell, or by binding to a lesser extent in number and/or affinity to such healthy cell).
  • anti-HER2 monoclonal antibody such as trastuzumab and pertuzumab
  • anti-CD20 monoclonal antibody such as rituximab, ofatumumab, tositumomab and ibritumomab
  • anti-CA125 monoclonal antibody such as oregovomab
  • anti-EpCAM (17-1A) monoclonal antibody such as edrecolomab
  • anti-EGFR monoclonal antibody such as cetuximab, panitumumab and nimotuzumab
  • anti-CD30 monoclonal antibody such brentuximab
  • anti-CD33 monoclonal antibody such as gemtuzumab and huMy9-6
  • anti-vascular integrin alpha-v beta-3 monoclonal antibody such as etaracizumab
  • anti-CD52 monoclonal antibody such as alemtuzumab
  • An embodiment is the protein-toxin conjugate of Stemline: ELZONRISTM (tagraxofusp, SL-401)—ELZONRIS is a novel targeted therapy directed to the interleukin-3 (IL-3) receptor- ⁇ (CD123), a target present on a wide range of malignancies.
  • ELZONRISTM tagraxofusp, SL-401
  • IL-3 interleukin-3 receptor- ⁇
  • any other molecules than antibodies that bind to a cell receptor or antigen of a target cell can also be used as the cell-binding ligand for the ligand-drug conjugates of the present invention and the ligands provided with covalently bound saponin according to the invention.
  • These ligands include, but are not limited to, proteins, polypeptides, peptides, small molecules. Examples of these non-antibody ligands are interferons (e.g.
  • IFN- ⁇ , IFN- ⁇ , and IFN- ⁇ transferrins, lectins, epidermal growth factors (EGF) and EGF-like domains, gastrin-releasing peptides (GRP), platelet-derived growth factors (PDGF), transforming growth factors (TGF), vaccinia growth factor (VGF), insulin and insulin-like growth factors (IGF, e.g. IGF-1 and IGF-2), other suitable hormones such as thyrotropin releasing hormones (TRH), melanocyte-stimulating hormones (MSH), steroid hormones (e.g. estrogen and androgen), somatostatin, lymphokines (e.g.
  • CSF colony-stimulating factors
  • M-CSF and GM-CSF colony-stimulating factors
  • aptamers e.g. AS-1411, GBI-10, RNA aptamers against HIV glycoprotein
  • small molecules e.g. folate, anisamide phenylboronic acid
  • vitamins e.g., vitamin D
  • carbohydrates e.g. hyaluronic acid, galactose
  • effector molecule or “effector moiety” or “payload” has its regular scientific meaning and in the context of this invention is any substance that affects the metabolism of a cell by interaction with an intracellular effector molecule target, wherein this effector molecule target is any molecule or structure inside cells excluding the lumen of compartments and vesicles of the endocytic and recycling pathway but including the membranes of these compartments and vesicles.
  • Said structures inside cells thus include the nucleus, mitochondria, chloroplasts, endoplasmic reticulum, Golgi apparatus, other transport vesicles, the inner part of the plasma membrane and the cytosol.
  • the effector molecule or -moiety is a pharmaceutically active substance, such as a toxin such as a proteinaceous toxin, a drug, a polypeptide or a polynucleotide.
  • a pharmaceutically active substance in this invention is an effector molecule or -moiety that is used to achieve a beneficial outcome in an organism, preferably a vertebrate, more preferably a mammal such as non-human subjects or a human being/subject. Benefits include diagnosis, prognosis, treatment, cure and prevention (prophylaxis) of diseases and/or symptoms and/or health problems.
  • the pharmaceutically active substance may also lead to undesired and sometimes even harmful side effects (adverse events such as observed during clinical trials).
  • pros and cons must be weighed to decide whether the pharmaceutically active substance is suitable in the particular case. If the effect of the pharmaceutically active substance inside a cell is predominantly beneficial for the organism as a whole, the cell is called a target cell. If the effect inside a cell is predominantly harmful for the organism as a whole, the cell is called an off-target cell. In artificial systems such as cell cultures and bioreactors, target cells and off-target cells depend on the purpose and are defined by the user. Examples of effector molecules and -moieties are a drug, a toxin, a polypeptide (such as an enzyme), a polynucleotide (including polypeptides and polynucleotides that comprise non-natural amino acids or nucleic acids), and any combination thereof.
  • an effector molecule or effector moiety that is a drug may include, but not limited to, anti-cancer agents, anti-inflammatory agents, and anti-infective (e.g., anti-fungal, antibacterial, anti-parasitic, anti-viral) agents.
  • the drug molecule of the present invention is an anti-cancer agent or an anti-auto-immune agent.
  • Suitable anti-cancer agents include, but are not limited to, alkylating agents, antimetabolites, spindle poison plant alkaloids, cytotoxic/antitumor antibiotics, topoisomerase inhibitors, photosensitizers, and kinase inhibitors. Also included in the definition of “anti-cancer agent” are: e.g.
  • anti-hormonal agents that act to regulate or inhibit hormone action on tumors such as anti-estrogens and selective estrogen receptor modulators;
  • aromatase inhibitors that inhibit the enzyme aromatase, which regulates estrogen production in the adrenal glands;
  • anti-androgens iv
  • protein kinase inhibitors iii
  • lipid kinase inhibitors iii) antisense oligonucleotides, particularly those which inhibit expression of genes in signaling pathways implicated in aberrant cell proliferation
  • ribozymes such as VEGF expression inhibitors and HER2 expression inhibitors;
  • vaccines such as gene therapy vaccines; topoisomerase 1 inhibitors;
  • anti-angiogenic agents such as anti-angiogenic agents; and pharmaceutically acceptable salts, acids, solvates and derivatives of any of the above.
  • An effector molecule or -moiety that is a toxin may include, but is not limited to, proteinaceous toxins (e.g. bacterial-derived toxins, and plant-derived toxins), toxins targeting tubulin filaments, toxins targeting DNA, toxins targeting RNA.
  • proteinaceous toxins are saporin, dianthin, ricin, modeccin, abrin, volkensin, viscumin, shiga toxin, shiga-like toxin, pseudomonas exotoxin (PE, also known as exotoxin A), diphtheria toxin (DT), and cholera toxin.
  • tubulin filaments-targeting toxins are maytansinoids (e.g.
  • DM1 and DM4 auristatins (e.g. Monomethyl auristatin E (MMAE) and Monomethyl auristatin F (MMAF)), toxoids, tubulysins, cryptophycins, rhizoxin.
  • DNA-targeting toxins are calicheamicins: N-Acetyl- ⁇ -calicheamicin, CC-1065 analogs, duocarmycins, doxorubicin, methotrexate, benzodiazepines, camptothecin analogues, and anthracyclines.
  • DNA-targeting toxins are amanitins, spliceostatins, and thailanstatins.
  • a toxin is defined as a pharmaceutically active substance that is able to kill or inactivate a cell.
  • a targeted toxin is a toxin that is only, or at least predominantly, toxic for target cells but not for off-target cells. The net effect of the targeted toxin is preferably beneficial for the organism as a whole.
  • An effector molecule or -moiety that is a polypeptide may be, e.g., a polypeptide that recover a lost function, such as for instance enzyme replacement, gene regulating functions, or a toxin.
  • polypeptides as effector molecules are, e.g., Cas9; toxins (e.g. saporin, dianthin, gelonin, (de)bouganin, agrostin, ricin (toxin A chain); pokeweed antiviral protein, apoptin, diphtheria toxin, pseudomonas exotoxin) metabolic enzymes (e.g.
  • argininosuccinate lyase argininosuccinate synthetase
  • enzymes of the coagulation cascade repairing enzymes
  • enzymes for cell signaling cell cycle regulation factors
  • gene regulating factors transcription factors such as NF- ⁇ B or gene repressors such as methionine repressor
  • An effector molecule or an effector moiety that is a polynucleotide may, e.g., be a polynucleotide that comprises coding information, such as a gene or an open reading frame encoding a protein. It may also comprise regulatory information, e.g. promotor or regulatory element binding regions, or sequences coding for micro RNAs.
  • Such polynucleotide may comprise natural and artificial nucleic acids. Artificial nucleic acids include, e.g. peptide nucleic acid (PNA), Morpholino and locked nucleic acid (LNA), as well as glycol nucleic acid (GNA) and threose nucleic acid (TNA).
  • nucleotides as effector molecules are, but not limited to, e.g., DNA: single stranded DNA (e.g. DNA for adenine phosphoribosyltransferase); linear double stranded DNA (e.g. clotting factor IX gene); circular double stranded DNA (e.g. plasmids); RNA: mRNA (e.g. TAL effector molecule nucleases), tRNA, rRNA, siRNA, miRNA, antisense RNA; anti-sense oligonucleotides (ASOs, AONs e.g. PNA, PMO, LNA and BNA).
  • DNA single stranded DNA
  • linear double stranded DNA e.g. clotting factor IX gene
  • circular double stranded DNA e.g. plasmids
  • RNA mRNA (e.g. TAL effector molecule nucleases), tRNA, rRNA, siRNA,
  • proteinaceous used in e.g. “proteinaceous molecule” and “proteinaceous toxin”, are molecules and toxins comprising at least a string of amino acid residues that can be obtained as an expression product from a single mRNA.
  • Such a molecule or toxin may further comprise any post-translational modifications, a carbohydrate such as an N- or O-linked carbohydrate, disulphide bonds, phosphorylations, sulphatations, etc., as a result of any post-translational modification, and/or may further comprise any other modification such as those resulting from chemical modifications (e.g., linking of effector moieties, saponin, scaffolds, ligands, etc., either directly to e.g.
  • proteinaceous also encompasses and includes assemblies of such molecules, e.g. homodimers, heterotrimers, heterohexamers or complex assemblies such as ribosomes.
  • the number of receptors or molecular targets is considered, of a cell-surface receptor or molecular target on the surface of a first type of cell such as a tumor cell, autoimmune cell, diseased cell, aberrant cell, relative to the extent of expression of the same receptor or molecular target at a second type of cell such as a healthy cell, etc., wherein expression at the second type of cell can be fully absent or very low, relative to any extent of expression on the tumor cell, etc.
  • the term “specific”, for example in “specific binding”, has its normal scientific meaning known in the art, and here has the meaning of indicating a molecule that can have an interaction with another molecule with higher binding affinity than background interactions between molecules.
  • binding molecules such as immunoglobulins bind via their binding site such as immunoglobulin variable regions of the immunoglobulin, to binding sites on molecules, such as epitopes, cell-surface receptors, etc., with a higher binding affinity than background interactions between molecules.
  • background interactions are typically interactions with an affinity lower than a K D of 10E-4 M.
  • specific binding domains are domains that preferentially bind to binding sites on molecules, such as epitopes, cell-surface receptors, etc., with a higher binding affinity than background interactions between molecules.
  • background interactions are typically interactions with an affinity lower than a K D of 10E-4 M.
  • specific binding domains bind with an affinity higher than a K D of about 10E-5 M.
  • binding is defined as interactions between molecules that can be distinguished from background interactions.
  • fragment refers to an amino acid sequence which is part of a protein domain or which builds up an intact protein domain. Binding fragments according to the invention must have binding specificity for the respective target such as a cell-surface receptor, e.g. on the surface of a diseased cell such as a tumor cell.
  • ADC or “antibody-drug conjugate” has its regular scientific meaning known to the skilled person, and here refers to a class of biopharmaceutical drugs designed as a targeted therapy for treating e.g. cancer. Unlike chemotherapy, ADCs are intended to target and kill tumor cells while sparing healthy cells. ADCs are composed of an antibody linked to a biologically active cytotoxic (anticancer) payload or drug. ADCs combine the targeting capabilities of monoclonal antibodies with the cancer-killing ability of cytotoxic drugs. They are designed with the intention to discriminate between healthy cells and diseased tissue such as tumor cells in a tumor.
  • Saponinum album has its normal meaning and here refers to a mixture of saponins produced by Merck KGaA (Darmstadt, Germany) containing saponins from Gypsophila paniculata and Gypsophila arostii , containing SA1657 and mainly SA1641.
  • Quillaja saponin has its normal meaning and here refers to the saponin fraction of Quillaja saponaria and thus the source for all other QS saponins, mainly containing QS-18 and QS-21.
  • QS-21 or “QS21” has its regular scientific meaning and here refers to a mixture of QS-21 A-apio ( ⁇ 63%), QS-21 A-xylo ( ⁇ 32%), QS-21 B-apio ( ⁇ 3.3%), and QS-21 B-xylo ( ⁇ 1.7%).
  • QS-21A has its regular scientific meaning and here refers to a mixture of QS-21 A-apio ( ⁇ 65%) and QS-21 A-xylo ( ⁇ 35%).
  • QS-21B has its regular scientific meaning and here refers to a mixture of QS-21 B-apio ( ⁇ 65%) and QS-21 B-xylo ( ⁇ 35%).
  • Quil-A refers to a commercially available semi-purified extract from Quillaja saponaria and contains variable quantities of more than 50 distinct saponins, many of which incorporate the triterpene-trisaccharide substructure Gal-(1 ⁇ 2)-[Xyl-(1 ⁇ 3)]-GlcA- at the C-3beta-OH group found in QS-7, QS-17, QS18, and QS-21.
  • the saponins found in Quil-A are listed in van Setten (1995), Table 2 [Dirk C. van Setten, Gerrit van de Maschinenen, Gijsbert Zomer and Gideon F. A.
  • Quil-A and also Quillaja saponin are fractions of saponins from Quillaja saponaria and both contain a large variety of different saponins with largely overlapping content. The two fractions differ in their specific composition as the two fractions are gained by different purification procedures.
  • QS1861 and the term “QS1862” refer to QS-7 and QS-7 api.
  • QS1861 has a molecular mass of 1861 Dalton
  • QS1862 has a molecular mass of 1862 Dalton.
  • QS1862 is described in Fleck et al. (2019) in Table 1, row no.
  • a method comprising step A and step B should not be limited to a method consisting only of steps A and B, rather with respect to the present invention, the only enumerated steps of the method are A and B, and further the claim should be interpreted as including equivalents of those steps.
  • indefinite article “a” or “an” does not exclude the possibility that more than one of the features such as for example a component, excipient, saponin, etc. are present, unless the context clearly requires that there is one and only one of the features.
  • the indefinite article “a” or “an” thus usually means “at least one”.
  • FIG. 1 HSP27BNA gene silencing activity of HSP27BNA, HSP27BNA-SO1861 and HSP27BNA-dendron-(SO1861) 4 in A431 cancer cell lines.
  • FIG. 2 A. MALDI-TOF-MS spectrum of BSA-SO1861.
  • FIG. 3 A. H-NMR spectrum of SO1861.
  • B. H-NMR spectrum of SO1861-EMCH ((EMCH N- ⁇ -maleimidocaproic acid hydrazide) conjugate.
  • FIG. 4 A. MALDI-TOF-MS spectrum of SO1861-EMCH conjugate.
  • FIG. 5 A. synthesis scheme of SO1861-EMCH.
  • TFA trifluoroacetic acid, r.t: room temperature, h: hours, and MW: molecular weight.
  • TFA trifluoroacetic acid, r.t: room temperature, h: hours, and MW: molecular weight.
  • FIG. 6 Reaction scheme of Dendron(-L-SO1861) 4 synthesis.
  • FIG. 7 Reaction scheme of Dendron(-L-SO1861) 8 synthesis.
  • FIG. 8 A. MALDI-TOF-MS spectrum of Cy3-PAMAM.
  • FIG. 9 A. MALDI-TOF-MS spectrum of Cy3-PAMAM-SO1861 with 5 equivalents feed SO1861-EMCH. B. MALDI-TOF-MS spectrum of Cy3-PAMAM-SO1861 with 30 equivalents feed SO1861-EMCH.
  • FIG. 11 A. Reaction scheme of the thiolation of PAMAM using the thiolation reagent 2-iminothiolane.
  • FIG. 12 A. Reaction scheme of the PEGylation of PAMAM using the PEGylating reagent mPEG 2k -NHS.
  • FIG. 13 A. Reaction scheme of Cy3-PAMAM-NC-SO1861-Dibenzocyclooctyne (DBCO). B. MALDI-TOF MS spectrum of Cy3-PAMAM-NC-SO1861-Dibenzocyclooctyne (DBCO). C. MALDI-TOF-MS spectrum of Cy3-PAMAM-(SO1861) 5 -DBCO. D. MALDI-TOF-MS spectrum of Cy3-PAMAM-(SO1861) 27 -DBCO.
  • FIG. 14 A. Reaction scheme of dianthin-EGF-Alexa488.
  • B Reaction scheme of dianthin-EGF-Alexa488-SS-PEG-N 3 .
  • D. MALDI-TOF-MS spectrum of dianthin-EGF-Alexa488.
  • E. MALDI-TOF-MS spectrum of dianthin-EGF-Alexa488-SS-PEG-N 3 .
  • FIG. 15 A. Reaction scheme of dianthin-Alexa488.
  • B Reaction scheme of dianthin-Alexa488-SS-PEG-N 3 .
  • C MALDI-TOF-MS spectrum of Dianthin.
  • D MALDI-TOF-MS spectrum of dianthin-Alexa488.
  • E MALDI-TOF-MS spectrum of dianthin-Alexa488-SS-PEG-N 3 .
  • FIG. 16 Fluorescence images of SDS-PAGE gel performed on a VersaDoc imaging system.
  • M marker
  • P Cy3-PAMAM-(SO1861)27-DBCO
  • D dianthin-EGF-Alexa488-SS-PEG-N3
  • C1 Cy3-PAMAM-(SO1861)5-Dianthin-EGF-Alexa488,
  • C2 Cy3-PAMAM-NC-SO1861-Dianthin-EGF-Alexa488, and
  • C3 Cy3-PAMAM-(SO1861)27-Dianthin-EGF-Alexa488.
  • FIG. 17 A. Synthesis scheme of Cy3-PAMAM-NC-SO1861 via reductive amination.
  • FIG. 18 A. MALDI-TOF-MS spectrum of native peptide. B. MALDI-TOF-MS spectrum of peptide-SO1861 conjugate.
  • FIG. 19 SO1861 structure with highlighted chemical groups for conjugation of endosomal escape enhancing saponins to a polymeric structure. Highlighted groups are aldehyde (black circle), carboxylic acid (dashed circle), alkene (dashed pentagon), and alcohol (dashed box).
  • FIG. 20 A. Schematic representation of the production of stable ‘ready-to conjugate’ endosomal escape enhancer saponins.
  • B Schematic representation of the production of cleavable ‘ready-to conjugate’ endosomal escape enhancer saponins.
  • FIG. 21 FIG. 26 : Hydrolysis of the hydrazone bond of SO1861-EMCH under acidic conditions.
  • FIG. 22 A. Standard molecular structure of SO-1861-EMCH conjugate. Maleimide group is marked with a circle. B. 3D model of SO1861-EMCH conjugate. Maleimide group is marked with a circle.
  • FIG. 23 A. MALDI-TOF-MS spectrum of SO1861-EMCH before hydrolysis in HCl solution at pH 3.
  • B MALDI-TOF-MS spectrum of SO1861-EMCH after hydrolysis in HCl solution at pH 3.
  • FIG. 24 Reaction scheme of SO1861-EMCH conjugation to any amine-bearing polymeric structure.
  • FIG. 25 A. Reaction scheme of SO1861-EMCH conjugation to a cyanine 3 dye labeled polyamidoamine (PAMAM) G5 dendrimer.
  • FIG. 26 Molecular structure of G4-dendron with protected amino groups.
  • FIG. 27 Synthesis scheme for the generation of dendron based scaffolds.
  • FIG. 28 A. Reaction scheme for partial dye labeling and deprotection of the G4-dendron. B. MALDI-TOF-MS spectrum of deprotected and partially dye labeled G4-dendron.
  • FIG. 29 A. MALDI-TOF-MS spectrum of G4-dendron-SO1861 scaffold with 22 feed equivalents of SO1861-EMCH.
  • FIG. 30 A. EGFR cell surface expression as determined by FACS analyses of HeLa cells.
  • the molecule In order for a bioactive molecule to work, the molecule must be able to engage with its target, e.g. in the blood serum, on the outside of the cell surface or inside a cell or an organelle.
  • the active moiety of almost all protein-based targeted toxins e.g., must enter the cytosol of the target cell to mediate its target modulatory effect.
  • the toxin remains ineffective since (1) the targeting moiety is poorly internalized and remains bound to the outside of the cells, (2) is recycled back to the cell surface after internalization or (3) transported to the endolysosomes where it is degraded.
  • An aspect of the invention relates to an effector moiety capable of inducing an intracellular effect when present inside a mammalian cell, the effector moiety conjugated with at least one saponin, wherein the at least one saponin is covalently bound to the effector moiety via at least one linker, or is covalently bound directly to said effector moiety.
  • the inventors established that the therapeutic window of a conjugate of a saponin and an effector moiety, increases when administered to a tumor-bearing mammal (mouse) to whom the conjugate is administered, wherein said conjugate comprises at least one covalently bound saponin.
  • the conjugate of the invention has at least one glycoside such as a saponin bound thereto, preferably covalently, more preferably via a cleavable linker.
  • the saponin augments the therapeutic efficacy of the effector moiety bound to the saponin, likely by enhancing the endosomal escape of the effector moiety into the cytosol where the activity of the effector moiety is desired.
  • the target cell is for example a diseased cell such as a tumor cell or an auto-immune cell or a B-cell disease related B-cell, etc.
  • the effector moiety is for example a toxin such as a toxin which is part of an ADC, or an oligonucleotide such as an antisense BNA which is part of an AOC, according to the invention.
  • An embodiment is the effector moiety of the invention, comprising or consisting of at least one oligonucleotide, a nucleic acid, a xeno nucleic acid, preferably selected from any one or more of a vector, a gene, a cell suicide inducing transgene, deoxyribonucleic acid (DNA), ribonucleic acid (RNA), anti-sense oligonucleotide (ASO, AON), short interfering RNA (siRNA), microRNA (miRNA), DNA aptamer, RNA aptamer, mRNA, mini-circle DNA, peptide nucleic acid (PNA), phosphoramidate morpholino oligomer (PMO), locked nucleic acid (LNA), bridged nucleic acid (BNA), 2′-deoxy-2′-fluoroarabino nucleic acid (FANA), 2′-O-methoxyethyl-RNA (MOE), 2′-0,4′-amino
  • a conjugate of antisense BNA such as for silencing HSP27 gene expression, and a saponin such as SO1861, covalently linked together effectively silence HSP27 expression in rapidly growing/dividing tumor cells, such as mouse A431 cancer cell line cells, compared to control.
  • the BNA-saponin conjugate resulted in an about threefold lower HSP27 expression in cells, compared to HSP27 expression in cells exposed to the same concentration of the free unconjugated BNA in the absence of saponin. See also FIG. 1 of the Examples.
  • contacting tumor cells with an antisense oligonucleotide such as antisense BNA results in gene silencing which is improved or enhanced when saponin such as SO1861 is covalently linked to the oligonucleotide.
  • saponin such as SO1861
  • the therapeutic window with regard to the oligonucleotide is enlarged, since the same gene silencing effect in the tumor cells can now be obtained with a threefold lower dose of the antisense BNA when a conjugate of BNA and saponin according to the invention is applied.
  • An embodiment is the effector moiety of the invention, comprising at least one proteinaceous molecule, the proteinaceous molecule preferably selected from any one or more of a peptide, a protein, an enzyme such as urease and Cre-recombinase, a ribosome-inactivating protein, a proteinaceous toxin such as any one or more of a protein toxin selected from Table A5 and/or a bacterial toxin or plant toxin, more preferably selected from any one or more of a viral toxin such as apoptin; a bacterial toxin such as Shiga toxin, Shiga-like toxin, Pseudomonas aeruginosa exotoxin (PE) or exotoxin A of PE, full-length or truncated diphtheria toxin (DT), cholera toxin; a fungal toxin such as alpha-sarcin; a plant toxin including ribosome-
  • dianthin-30 or dianthin-32 saporin e.g. saporin-S3 or saporin-S6, bouganin or de-immunized derivative debouganin of bouganin, shiga-like toxin A, pokeweed antiviral protein, ricin, ricin A chain, modeccin, modeccin A chain, abrin, abrin A chain, volkensin, volkensin A chain, viscumin, viscumin A chain; or an animal or human toxin such as frog RNase, or granzyme B or angiogenin from humans, or any fragment or derivative thereof; preferably the protein toxin is dianthin and/or saporin.
  • saporin e.g. saporin-S3 or saporin-S6, bouganin or de-immunized derivative debouganin of bouganin, shiga-like toxin A, pokeweed antiviral protein, ricin, ricin A chain, modeccin, modeccin A
  • An embodiment is the effector moiety of the invention, comprising at least one payload, the payload preferably selected from any one or more of a toxin targeting ribosomes, a toxin targeting elongation factors, a toxin targeting tubulin, a toxin targeting DNA and a toxin targeting RNA, more preferably any one or more of emtansine, pasudotox, maytansinoid derivative DM1, maytansinoid derivative DM4, monomethyl auristatin E (MMAE, vedotin), monomethyl auristatin F (MMAF, mafodotin), a Calicheamicin, N-Acetyl- ⁇ -calicheamicin, a pyrrolobenzodiazepine (PBD) dimer, a benzodiazepine, a CC-1065 analogue, a duocarmycin, Doxorubicin, paclitaxel, docetaxel, cisplatin
  • An embodiment is the effector moiety of the invention, wherein the at least one saponin is a triterpenoid saponin and/or a bisdesmosidic triterpene saponin belonging to the type of a 12,13-dehydrooleanane with an aldehyde function in position C-23 and optionally comprising a glucuronic acid function in a carbohydrate substituent at the C-3beta-OH group of the saponin, and/or a saponin isolated from a Gypsophila species and/or a Saponaria species and/or an Agrostemma species and/or a Quillaja species such as Quillaja saponaria.
  • the at least one saponin is a triterpenoid saponin and/or a bisdesmosidic triterpene saponin belonging to the type of a 12,13-dehydrooleanane with an aldehyde function in position C-23 and optionally comprising a glucuronic acid
  • An embodiment is the effector moiety of the invention, wherein the at least one saponin is a single specific saponin or is a mixture of two or more different saponins.
  • an embodiment is the effector moiety of the invention, wherein the saponin is one or more of the saponins in Table A1 or Scheme I, SO1861, SA1657, GE1741, SA1641, QS-21, QS-21A, QS-21 A-api, QS-21 A-xyl, QS-21B, QS-21 B-api, QS-21 B-xyl, QS-7-xyl, QS-7-api, QS-17-api, QS-17-xyl, QS1861, QS1862, Quillaja saponin , Saponinum album, QS-18, Quil-A, Gyp1, gypsoside A, AG1, AG2, SO1542, SO1584, SO1658, SO1674, SO1832, or any of their stereomers and/or any combinations thereof, preferably the saponin is SO1861 and/or GE1741 and/or SA1641 and/or QS-21 and/or saponin with a quillaic acid aglycon core, a Gal-
  • a water-soluble saponin fraction from Quillaja saponaria comprising QS-21 and its family members QS-21A, QS-21 A-api, QS-21 A-xyl, QS-21B, QS-21 B-api, QS-21 B-xyl, QS-7-xyl, QS-7-api, QS-17-api, QS-17-xyl, QS1861, QS1862, QS-18 and Quil-A, also exhibits the ability to potentiate a biological effect in vitro of e.g.
  • a nucleic acid bound to a monoclonal antibody or a protein toxin bound to a monoclonal antibody when administered to tumor cells of a mammalian species (human) in the form of a covalent conjugate comprising a monoclonal antibody and the at least one glycoside such as the QS-21 and its family member saponins encompassed by such QS-21 preparation (e.g. water soluble fraction of Quillaja saponaria ), herein the effector molecule and the glycoside, e.g.
  • saponin fraction of Quillaja saponaria QS-21, SO1861, SA1641, GE1741, are covalently bound to for example a proteinaceous molecules directly or via a linker or via a polymeric or oligomeric scaffold, either directly or via at least one linker.
  • the observed stimulation or potentiation of for example antisense BNA mediated reduction of tumor-cell HSP27 expression (HSP27 gene silencing) in the presence of saponins derived from Quillaja saponaria in vitro may (also) relate to activation of the inflammasome in the tumor cell by the saponins, for example resulting in tumor cell pyroptosis.
  • an antibody conjugated to for example antisense BNA or dianthin or saporin exerted any anti-tumor cell activity in vitro at all or improved anti-tumor cell activity when contacted with cells in bio-based cell assays, when in the presence of saponin coupled to a targeting antibody, and targeted to the same (tumor) cells, whereas in the absence of the saponin, no such activity towards the tumor cell was observed.
  • QS-21 and also the water-soluble saponins fraction comprising QS-21 from Quillaja saponaria is already for a long time known and previously intensively applied for its immune-potentiating abilities, e.g. as an adjuvant in e.g. sub-unit vaccines.
  • QS-21 is applied in two phase III clinical trials with human patients, who were vaccinated with a sub-unit vaccine mixed with an adjuvant comprising QS-21 (Glaxo-Smith-Kline, MAGRIT trial, DERMA study), wherein the sub-unit was MAGE-A3 protein, which is specifically expressed and presented by tumor cells.
  • the anti-tumor vaccinations potentiated with QS-21, aimed for extension of disease-free survival of the cancer patients (melanoma; non-small cell lung cancer).
  • QS-21 has been tested as an adjuvant in clinical trials for developing anti-cancer vaccine treatment, for vaccines for HIV-1 infection, in development of a vaccine against hepatitis B, and for anti-malaria vaccine development using QS-21 comprising adjuvants AS01 and AS02 of Glaxo-Smith-Kline.
  • Previous studies revealed an immune response elicited against MAGE-A3 peptides presented at the cancer cell surface, under influence of the QS-21 saponin comprising adjuvant (AS15; GSK).
  • the saponin fraction of Quillaja saponaria potentiates the anti-tumor cell activity of e.g. a payload such as a protein toxin (dianthin), bound to the second proteinaceous molecule (e.g. the ligand EGF).
  • a payload such as a protein toxin (dianthin)
  • the second proteinaceous molecule e.g. the ligand EGF
  • an embodiment is the effector moiety of the invention, wherein the saponin is a bisdesmosidic saponin having a molecular mass of at least 1.500 Dalton and comprising an oleanan-type triterpene containing an aldehyde group at the C-23 position and optionally a hydroxyl group at the C-16 position, with a first branched carbohydrate side chain at the C-3 position which first branched carbohydrate side chain optionally contains glucuronic acid, wherein the saponin contains an ester group with a second branched carbohydrate side chain at the C-28 position which second branched carbohydrate chain preferably comprises at least four carbohydrate units and optionally contains at least one acetyl residue such as two acetyl residues and/or optionally comprises one or more deoxy carbohydrates and/or quinovose and/or glucose and/or 4-methoxycinnamic acid and/or optionally comprising 5-O-[5-O-Ara/Api
  • a tumor-cell targeting monoclonal antibody provided with covalently coupled antisense BNA such as BNA(HSP27), and contacted with the tumor cells together with an antibody with couopled saponin (e.g. SO1861, QS-21), both the BNA and the saponin coupled to the respective antibody (e.g. cetuximab) via a cleavable bond, is capable of silencing HSP27 in vivo in tumors, compared to control and compared to AOC only, without presence of an antibody with coupled saponin.
  • Co-administering an ADC or an antibody-oligonucleotide conjugate (AOC), such as an antibody-BNA conjugate, with a an antibody with linked saponin thus endows the ADC or AOC with anti-tumor cell activity not seen with only the ADC or only the AOC at the same dose.
  • the AOC and the monoclonal antibody with covalently coupled saponin increase HSP27 expression in tumor cells, when administered to tumor-bearing mice separately in separate groups of mice, compared to a control group (vehicle administered, only). Only co-administration of the AOC comprising the effector moiety and an antibody with covalently coupled saponin, displays reduced HSP27 expression when compared to controls.
  • the antisense BNA was BNA with oligo nucleic acid sequence 5′-GGCacagccagtgGCG-3′ according to Zhang et al. (2011) [Y Zhang, Z Qu, S Kim, V Shi, B Liao1, P Kraft, R Bandaru, Y Wu, L M Greenberger and I D Horak, Down - modulation of cancer targets using locked nucleic acid ( LNA )- based antisense oligonucleotides without transfection, Gene Therapy (2011) 18, 326-333].
  • LNA locked nucleic acid
  • BNA is designed for application as a free nucleic acid.
  • the antisense BNA can be covalently coupled through a (non-)cleavable linker with a ligand or an antibody, in a way that gene-silencing activity is retained in vitro and more importantly in vivo in the tumor cells of a tumor-bearing animal.
  • This approach of providing BNA based AOCs opens new ways to administer targeted BNA to human (cancer) patients in need thereof.
  • the inventors disclose here that covalently coupling saponins such as saponins in the water-soluble fraction of Quillaja saponaria , QS-21, SA1641, SO1861, Table A1, Scheme I, to for example an antisense oligonucleotide such as a BNA, or for example to a proteinaceous molecule, such as via a tri-functional linker, e.g.
  • the tri-functional linker of Scheme II results in improved cell toxicity exerted by the effector moiety such as a toxin, comprised by the effector moiety of the invention, under influence of the covalently coupled saponin in the effector moiety of the invention.
  • the saponin is a saponin listed in Table A1, Scheme I. It has been proven beneficial for the activity of the saponin, e.g. the endosomal escape enhancing activity inside cells when the entry into the cell and the accumulation inside the cytosol of an effector moiety of the invention comprising coupled saponin, is considered, when the saponin is covalently coupled to the payload comprised by the effector moiety (e.g. a BNA) involving a hydrazone bond, and/or a hydrazide bond, and/or a disulphide bond.
  • the effector moiety e.g. a BNA
  • Such bond types readily cleave under the acidic conditions inside (late) endosomes and lysosomes of mammalian cells, e.g.
  • the inventors demonstrate that covalent coupling of saponin to the payload comprised by the effector moiety of the invention via a bond that is not readily cleavable under the physiological conditions inside cells, e.g. (late) endosomes, lysosomes, cytosol, is also beneficial to the potentiating activity of the saponin on the biological effect of e.g. an effector moiety such as a nucleic acid (e.g. BNA silencing HSP27) and a proteinaceous toxin such as saporin.
  • an effector moiety such as a nucleic acid (e.g. BNA silencing HSP27) and a proteinaceous toxin such as saporin.
  • cleavable linker ‘cleavable bond’, etc.
  • L labile linker
  • labile bond for example in the context of cleavage of such a bond or linker in the (late) endosome and/or lysosome when a conjugate of the invention, e.g.
  • an effector moiety such as BNA-SO1861, or antisense oligonucleotide-saponin conjugate (the saponin being any of the saponins in Table A1, Scheme I), optionally comprising a scaffold with saponins coupled to payload through a linker and/or via the scaffold via hydrazone bonds or disulphide bonds, is referred to.
  • the inventors showed the in vivo HSP27 gene silencing in human tumors in mice.
  • the tumor-bearing mice were treated with an antibody consisting of monoclonal antibody with saponin bound thereto via a labile linker (hydrazone bond) according to the invention, whereas a different portion of the same antibody comprised bound antisense BNA for silencing the HSP27 gene in the tumor cells, covalently coupled to the monoclonal antibody (being same type of antibody as the first monoclonal antibody) via a a disulphide bond.
  • the hydrazone bond and the disulphide bond are cleaved in the (late) endosomes and/or lysosomes of the targeted tumor cells that express the epitope on the targeted cell-surface molecule, here the EGFR, at the cell surface, once the therapeutic combination of the invention is internalized by e.g. endocytosis.
  • Cleavage of the bonds likely contributes to the endosomal escape enhancing activity of the saponin when the entry of the BNA from the endosome and/or lysosome into the cytosol is considered, although such cleavage is not a necessity for observing the gene silencing effect of the combination of the cetuximab-SO1861 conjugate and the cetuximab-BNA conjugate of the invention.
  • a tri-functional linker is a scaffold of the invention suitable for covalently coupling one, two or three saponin moieties.
  • the second and/or third binding site is for example suitable for covalent coupling an payload, such that an effector moiety of the invention is provided.
  • the second or third binding site of the tri-functional linker is suitable for covalent coupling of an immunoglobulin such as a monoclonal antibody, preferably a tumor-cell specific molecule, more preferably a tumor cell receptor that is specifically (over-)expressed at the surface of the tumor cell.
  • the immunoglobulin, or any fragment(s) and/or domain(s) thereof which encompass the binding specificity of the immunoglobulin is suitable for binding to a cell surface molecule such as a receptor, expressed at the surface of an autoimmune cell.
  • the tri-functional linker then comprises a covalently coupled saponin and a covalently coupled payload, therewith providing the effector moiety of the invention, and in addition a covalently coupled e.g. antibody, for targeting the effector moiety to a selected cell such as a tumor cell expressing the tumor-cell specific binding site for the coupled antibody, e.g. a specific tumor-cell receptor.
  • the effector moiety of the invention comprises the tri-functional linker, said linker comprises or consists of a covalently bound saponin and effector moiety, e.g. QS-21, SO1861 and e.g. antisense BNA, protein toxin, and the covalently bound antibody for (specific) binding to a tumor cell, an auto-immune cell, a diseased cell, an aberrant cell, a non-healthy cell, a B-cell disease.
  • a covalently bound saponin and effector moiety e.g. QS-21, SO1861 and e.g. antisense BNA, protein toxin
  • the covalently bound antibody for (specific) binding to a tumor cell, an auto-immune cell, a diseased cell, an aberrant cell, a non-healthy cell, a B-cell disease.
  • an embodiment is the effector moiety of the invention, wherein the at least one saponin is a bisdesmosidic triterpene saponin belonging to the type of a 12,13-dehydrooleanane with an aldehyde function in position C-23, wherein the at least one saponin is covalently coupled to an amino-acid residue, when present, of the effector moiety via an aldehyde function in the saponin, preferably said aldehyde function in position C-23, preferably via at least one linker, more preferably via at least one cleavable linker, wherein the amino-acid residue preferably is selected from cysteine and lysine.
  • the at least one saponin is a bisdesmosidic triterpene saponin belonging to the type of a 12,13-dehydrooleanane with an aldehyde function in position C-23, wherein the at least one saponin is covalently coupled to an amino-acid residue, when present
  • an embodiment is the effector moiety of the invention, wherein the at least one saponin is a bisdesmosidic triterpene saponin belonging to the type of a 12,13-dehydrooleanane with an aldehyde function in position C-23 and comprising a glucuronic acid function in a carbohydrate substituent at the C-3beta-OH group of the saponin, wherein the at least one saponin is covalently coupled to an amino-acid residue, when present, of the effector moiety via the glucuronic acid function in the carbohydrate substituent at the C-3beta-OH group of the saponin, preferably via at least one linker, wherein the amino-acid residue preferably is selected from cysteine and lysine.
  • BNAoligo, antisense BNA oligo targeting the mRNA transcript of the cancer target (upregulated in cancer cells), heat shock protein 27 (HSP27BNA) was conjugated to SO1861-EMCH (HSP27BNA-L-SO1861) or dendron(-L-SO1861) 4 (HSP27BNA-dendron(-L-SO1861) 4 ) and co-administrated to an A431 cancer cell line, according to the invention.
  • a conjugate of antisense BNA such as for silencing HSP27 gene expression, and a saponin such as SO1861, covalently linked together effectively silence HSP27 expression in rapidly growing/dividing tumor cells, such as mouse A431 cancer cell line cells, compared to control.
  • Covalently conjugating any of the saponins of the invention with an oligonucleotide, such as an antisense oligonucleotide, for example a BNA such as an antisense BNA results in increased gene silencing efficacy at the same dose of BNA when compared to free BNA in the absence of conjugated saponin and in the absence of free saponin.
  • the BNA-saponin conjugate resulted in an about threefold lower HSP27 expression in cells, compared to HSP27 expression in cells exposed to the same concentration of the free unconjugated BNA in the absence of saponin. See also FIG. 1 of the Examples.
  • an antisense oligonucleotide such as antisense BNA results in gene silencing which is improved or enhanced when saponin such as SO1861 is covalently linked to the oligonucleotide.
  • the therapeutic window with regard to the oligonucleotide is enlarged, since the same gene silencing effect in the tumor cells can now be obtained with a threefold lower dose of the antisense BNA when a conjugate of BNA and saponin according to the invention is applied.
  • the inventors established that the conjugate of BNA with one saponin is more effective when gene silencing is considered than the gene silencing activity obtainable with free BNA administered to the cells.
  • the conjugate of BNA with four saponins is increasingly effective when gene silencing is considered and when compared to the gene silencing activity obtainable with free BNA administered to the cells. That is to say, improved gene silencing with BNA(-saponin)4 is even higher than improved gene silencing obtainable with BNA-saponin, according to the invention.
  • the linker(s) is/are cleavable linkers.
  • an embodiment is the effector moiety of the invention, wherein the at least one linker comprises at least one non-cleavable linker and/or at least one cleavable linker, wherein optionally said cleavable linker is subject to cleavage under acidic, reductive, enzymatic or light-induced conditions, and preferably the cleavable linker comprises a cleavable bond selected from a hydrazone bond and a hydrazide bond subject to cleavage under acidic conditions, and/or a bond susceptible to proteolysis, for example proteolysis by Cathepsin B, and/or a bond susceptible for cleavage under reductive conditions such as a disulphide bond.
  • an embodiment is the effector moiety of the invention, wherein the at least one linker comprises at least one cleavable linker which is subject to cleavage in vivo under acidic conditions as present in endosomes and/or in lysosomes of mammalian cells, preferably of human cells, preferably at pH 4.0-6.5, and more preferably at pH 5.5.
  • An embodiment is the effector moiety of the invention, wherein the at least one saponin is covalently bound to a lysine side chain, forming an amide bond, and/or to a cysteine side chain, forming a thio-ether linkage, or a disulphide bond, wherein the lysine and/or cysteine is/are comprised by the effector moiety, and wherein the saponin is bound either directly to the effector moiety, or bound via at least one linker.
  • An embodiment is the effector moiety of the invention, wherein the at least one saponin is covalently bound to the effector moiety via at least one linker, wherein the linker is or comprises a scaffold comprising a polymeric or oligomeric structure and a chemical group for covalently coupling of the scaffold to the effector moiety.
  • An embodiment is the effector moiety of the invention, wherein the at least one saponin is covalently bound to the polymeric or oligomeric structure of the scaffold via a cleavable bond.
  • An embodiment is the effector moiety of the invention, wherein the cleavable bond is subject to cleavage under any of acidic conditions, reductive conditions, enzymatic conditions and light-induced conditions, more preferably the cleavable bond is a hydrazone bond or a hydrazide bond subject to cleavage under acidic conditions, and/or is a bond susceptible to proteolysis, for example proteolysis by Cathepsin B, and/or is a bond susceptible for cleavage under reductive conditions such as a disulphide bond.
  • An embodiment is the effector moiety of the invention, wherein the cleavable bond is subject to cleavage in vivo under acidic conditions as present in endosomes and/or in lysosomes of mammalian cells, preferably of human cells, preferably at pH 4.0-6.5, and more preferably at pH 5.5.
  • An embodiment is the effector moiety of the invention, wherein the at least one saponin is covalently bound to the polymeric or oligomeric structure of the scaffold via an imine bond, a hydrazone bond, a hydrazide bond, an oxime bond, a 1,3-dioxolane bond, a disulphide bond, a thio-ether bond, an amide bond, a peptide bond and/or an ester bond, preferably via at least one linker, preferably an amide bond, a hydrazide bond, a thio-ether bond and/or a hydrazone bond.
  • An embodiment is the effector moiety of the invention, wherein the at least one saponin is covalently bound to the polymeric or oligomeric structure of the scaffold, involving in the covalent bond the aldehyde function in position C-23 of the at least one saponin, when present, the covalent bond being preferably an imine bond or a hydrazone bond or an amide bond or a thio-ether bond or a disulphide bond, and/or involving in the covalent bond the glucuronic acid function in the carbohydrate substituent at the C-3beta-OH group of the at least one saponin, when present, wherein preferably the covalent bond is an amide bond or a disulphide bond or a thio-ether bond.
  • An embodiment is the effector moiety of the invention, wherein the aldehyde function in position C-23 of the at least one saponin is covalently coupled to linker N- ⁇ -maleimidocaproic acid hydrazide, which linker is covalently coupled via a thio-ether bond to a sulfhydryl group in the polymeric or oligomeric structure of the scaffold, such as a sulfhydryl group of a cysteine.
  • An embodiment is the effector moiety of the invention, wherein the glucuronic acid function in the carbohydrate substituent at the C-3beta-OH group of the at least one saponin is covalently coupled to linker 1-[Bis(dimethylamino)methylene]-1H-1,2,3-triazolo[4,5-b]pyridinium 3-oxid hexafluorophosphate, which linker is covalently coupled via an amide bond to an amine group in the polymeric or oligomeric structure of the scaffold, such as an amine group of a lysine or an N-terminus of the polymeric or oligomeric structure of the scaffold.
  • linker 1-[Bis(dimethylamino)methylene]-1H-1,2,3-triazolo[4,5-b]pyridinium 3-oxid hexafluorophosphate which linker is covalently coupled via an amide bond to an amine group in the polymeric or oligomeric structure of the scaffold, such as an amine group
  • an embodiment is the effector moiety of the invention, wherein the chemical group of the scaffold, for covalently coupling of the scaffold to the effector moiety, is a click chemistry group, preferably selected from a tetrazine, an azide, an alkene or an alkyne, or a cyclic derivative of these groups, more preferably the click chemistry group is an azide.
  • a click chemistry group preferably selected from a tetrazine, an azide, an alkene or an alkyne, or a cyclic derivative of these groups, more preferably the click chemistry group is an azide.
  • polymeric or oligomeric structure of the scaffold comprises a linear, branched and/or cyclic polymer, oligomer, dendrimer, dendron, dendronized polymer, dendronized oligomer, a DNA, a polypeptide, poly-lysine, a poly-ethylene glycol, or an assembly of these polymeric or oligomeric structures which assembly is preferably built up by covalent cross-linking.
  • An embodiment is the effector moiety of the invention, wherein the at least one saponin is a defined number of saponins or a defined range of saponins, preferably 1-128 saponins or at least 2, 3, 4, 5, 6, 8, 10, 16, 32, 64 or 128 saponins, or any number of saponins therein between, such as 7, 9, 12 saponins.
  • an embodiment is the effector moiety of the invention, wherein the effector moiety comprises more than one saponin, preferably 2, 3, 4, 5, 6, 8, 10, 16, 32, 64 or 1-100 saponins, or any number of saponins therein between, such as 7, 9, 12 saponins, covalently bound directly to an amino-acid residue of the effector moiety, preferably to a cysteine and/or to a lysine, and/or covalently bound via at least one linker and/or via at least one cleavable linker and/or via at least one polymeric or oligomeric scaffold of any one of the claims 14 - 23 , preferably 1-8 of such scaffolds or 2-4 of such scaffolds, wherein 1-32 saponins, preferably 2, 3, 4, 5, 6, 8, 10, 16 or 32 saponins, are covalently bound to the at least one scaffold.
  • the effector moiety comprises more than one saponin, preferably 2, 3, 4, 5, 6, 8, 10, 16, 32, 64 or 1-100 saponins, or any
  • beneficial anti-tumor activity in various in vitro mammalian cell-based bioassays can be achieved by treating the animals with conjugates according to the invention, optionally comprising a scaffold according to the invention such as a G4-dendron.
  • the scaffold for example being a tri-functional linker with at least one covalently bound saponin (e.g. SO1861, QS-21) via a cleavable or non-cleavable linkage, and/or with a covalently bound effector moiety (e.g.
  • HSP27 silencing BNA
  • the scaffold being a dendron, such as a dendron to which for example four moieties can bind such as four saponin molecules, or a dendron for binding for example two saponins and two effector molecules.
  • the glycoside molecule preferably a saponin of the invention, increases the functional efficacy of that effector moiety (e.g. the therapeutic index of a toxin or a drug or an oligonucleotide such as a BNA; the metabolic efficacy of a modifier in biotechnological processes; the transfection efficacy of genes in cell culture research experiments), preferably by enabling or improving its target engagement. Acceleration, prolongation, or enhancement of antigen-specific immune responses are preferably not included. Therapeutic efficacy includes but is not limited to a stronger therapeutic effect, preferably with lower dosing and/or with less side effects.
  • “Improving an effect of an effector moiety” can also mean that an effector moiety, which could not be used because of lack of effect (and was e.g. not known as being an effector moiety), becomes effective when used in combination with the present invention. Any other effect, which is beneficial or desired and can be attributed to the combination of effector moiety and the second or third proteinaceous molecule, as provided by the invention is considered to be “an improved effect”.
  • the scaffold comprising bound saponin(s) and comprised by the effector moiety enhances an effect of the payload/effector moiety which effect is intended and/or desired.
  • a number of preferred features can be formulated for endosomal escape enhancers comprised by effecor moiety, i.e. a glycoside or saponin, preferably a saponin according to the invention: (1) they are preferably not toxic and do not invoke an immune response, (2) they preferably do not mediate the cytosolic uptake of the effector moiety into off-target cells, (3) their presence at the site of action is preferably synchronized with the presence of the effector moiety, (4) they are preferably biodegradable or excretable, and (5) they preferably do not substantially interfere with biological processes of the organism unrelated to the biological activity of the effector molecule with which the endosomal escape enhancer is combined with, e.g. interact with hormones.
  • effecor moiety i.e. a glycoside or saponin, preferably a saponin according to the invention: (1) they are preferably not toxic and do not invoke an immune response, (2) they preferably do not mediate the cytosolic uptake of the effect
  • glycoside molecules such as saponins of the invention that fulfill the before mentioned criteria, at least to some extent, are bisdesmosidic triterpenes, preferably bisdesmosidic triterpene saponins, such as 801861, SA1641, QS-21, GE1741, and the saponins in Table A1, Scheme I.
  • the uptake of extracellular substances such as the effector moiety of the invention, into a cell by vesicle budding is called endocytosis.
  • Said vesicle budding can be characterized by (1) receptor-dependent ligand uptake mediated by the cytosolic protein clathrin, (2) lipid-raft uptake mediated by the cholesterol-binding protein caveolin, (3) unspecific fluid uptake (pinocytosis), or (4) unspecific particle uptake (phagocytosis).
  • endocytic pathways All types of endocytosis run into the following cellular processes of vesicle transport and substance sorting called the endocytic pathways.
  • the endocytic pathways are complex and not fully understood.
  • organelles may be formed de novo and mature into the next organelle along the endocytic pathway. It is however, now hypothesized that the endocytic pathways involve stable compartments that are connected by vesicular traffic.
  • a compartment is a complex, multifunctional membrane organelle that is specialized for a particular set of essential functions for the cell.
  • Vesicles are considered to be transient organelles, simpler in composition, and are defined as membrane-enclosed containers that form de novo by budding from a preexisting compartment.
  • vesicles can undergo maturation, which is a physiologically irreversible series of biochemical changes.
  • Early endosomes and late endosomes represent stable compartments in the endocytic pathway while primary endocytic vesicles, phagosomes, multivesicular bodies (also called endosome carrier vesicles), secretory granules, and even lysosomes represent vesicles.
  • the endocytic vesicle which arises at the plasma membrane, most prominently from clathrin-coated pits, first fuses with the early endosome, which is a major sorting compartment of approximately pH 6.5.
  • Lysosomes are vesicles that can store mature lysosomal enzymes and deliver them to a late endosomal compartment when needed.
  • the resulting organelle is called the hybrid organelle or endolysosome. Lysosomes bud off the hybrid organelle in a process referred to as lysosome reformation. Late endosomes, lysosomes, and hybrid organelles are extremely dynamic organelles, and distinction between them is often difficult.
  • Endosomal escape is the active or passive release of a substance from the inner lumen of any kind of compartment or vesicle from the endocytic pathway, preferably from clathrin-mediated endocytosis, or recycling pathway into the cytosol. Endosomal escape thus includes but is not limited to release from endosomes, endolysosomes or lysosomes, including their intermediate and hybrid organelles.
  • endosome or “endosomal escape” is used herein, it also includes the endolysosome and lysosome, and escape from the endolysosome and lysosome, respectively. After entering the cytosol, said substance might move to other cell units such as the nucleus.
  • a glycoside is any molecule in which a sugar group is bound through its anomeric carbon to another group via a glycosidic bond.
  • Glycoside molecules, such as saponins, in the context of the invention are such molecules that are further able to enhance the effect of an effector moiety, without wishing to be bound by any theory, in particular by facilitating the endosomal escape of the effector moiety.
  • the glycoside molecules interact with the membranes of compartments and vesicles of the endocytic and recycling pathway and make them leaky for said effector moieties resulting in augmented endosomal escape.
  • the scaffold is able to augment endosomal escape of the effector moiety
  • the at least one saponin (glycoside molecule) which is coupled to the polymeric or oligomeric structure of the scaffold, is able to enhance endosomal escape of a payload comprised by an effector moiety of the invention when both molecules are within an endosome, e.g.
  • a late endosome optionally and preferably after the at least one glycoside such as a saponin is released from the effector moiety such as from a linker or polymeric or oligomeric structure comprised by said effector moiety, e.g., by cleavage of a cleavable bond between the at least one glycoside (saponin) and the payload (for example via a polymeric or oligomeric structure of a scaffold and/or via a linker).
  • the effector moiety such as from a linker or polymeric or oligomeric structure comprised by said effector moiety, e.g., by cleavage of a cleavable bond between the at least one glycoside (saponin) and the payload (for example via a polymeric or oligomeric structure of a scaffold and/or via a linker).
  • a bond between the at least one glycoside such as a saponin according to the invention and the payload comprised by the effector moiety of the invention, optionally via a linker or a scaffold may be a “stable bond”, that does not mean that such bond cannot be cleaved in the endosomes by, e.g., enzymes.
  • the glycoside or saponin optionally together with a linker or a part of the oligomeric or polymeric structure of a scaffold, may be cleaved off from the remaining linker fragment or oligomeric or polymeric structure.
  • a protease cuts a (proteinaceous) linker or proteinaceous polymeric structure, e.g., albumin, thereby releasing the at least one glycoside, saponin.
  • the glycoside molecule preferably saponin
  • the glycoside molecule is released in an active form, preferably in the original form that it had before it was (prepared to be) coupled to the payload comprised by the effector moiety of the invention, optionally via a linker and/or an oligomeric or polymeric scaffold; thus the glycoside (saponin) has its natural structure after such cleavage or the glycoside (saponin) has (part of) a chemical group or linker bound thereto, after such cleavage, while glycoside biological activity (saponin biological activity), e.g.
  • endosomal/lysosomal escape enhancing activity towards a payload present in the same endosome or lysosome is maintained or restored upon said cleavage of the bond between the glycoside (saponin) and e.g. the payload or a carrier molecule comprised by the effector moiety of the invention, optionally comprising a linker and/or a scaffold of the invention.
  • saponins and amino-acid residues of the payload, a linker, a polymeric or oligomeric structures (of the scaffold), ligands, (monoclonal) immunoglobulins or binding domains or -fragments thereof, and/or furher effectors (effector moieties, effector molecules), is meant that the bond is not readily broken or at least not designed to be readily broken by, e.g., pH differences, salt concentrations, or UV-light, reductive conditions.
  • saponins and the (proteinaceous) payload comprised by the effector moiety of the invention linkers, amino-acid residues, polymeric or oligomeric structures of the scaffold, ligands, antibodies and/or further effectors, is meant that the bond is designed to be readily broken by, e.g., pH differences, salt concentrations, under reductive conditions, and the like.
  • the skilled person is well aware of such cleavable bonds and how to prepare them.
  • the effector moiety of the invention as a semi-finished product for the manufacture of an ADC-saponin conjugate or an AOC-saponin conjugate, it has now become possible to guide one or multiple glycoside molecules (saponin) to a (target) cell, together with the ADC carrying a payload or together with a (monoclonal) antibody conjugated with an oligonucleotide such as a BNA according to the invention, thus the antibody being provided with covalently bound effector moiety of the invention which comprises covalently coupled saponin of the invention.
  • the effector moiety of the invention can be covalently coupled to a monoclonal antibody for binding to an aberrant cell specific epitope such as a tumor-cell receptor that is specifically exposed on the surface of said tumor cell, such that the effector moiety of the invention is applied as a semi-finished product for providing an ADC-saponin conjugate or an AOC-saponin conjugate, according to the invention.
  • a solution provided for by the invention comprises the covalent binding of at least one saponin to a payload, therewith providing an effector moiety of the invention.
  • a further solution provided for by the invention comprises (first) polymerizing the glycoside molecules (saponins) using an oligomeric or polymeric scaffold, and providing the payload comprised by the effector moiety of the invention with a cluster of covalently bound saponins, enabling re-monomerization of the one or more saponins at the intracellular site where the mode of action of the saponin is desired, e.g. after endocytosis.
  • “Polymerizes” in this context means the reversible and/or irreversible multiple conjugation of saponin molecules to the payload molecule, either via linker, or directly or via a polymeric or oligomeric structure to form a scaffold or the reversible and/or irreversible multiple conjugation of (modified) saponins thereby forming a polymeric or oligomeric structure to form a scaffold.
  • “Re-monomerization” in this context means the cleavage of the saponins from the payload molecule (e.g.
  • a protein toxin, an antsense BNA from the linker linking the saponin(s) to the payload molecule in the effector moiety of the invention or from the scaffold, for example after endocytosis, and regaining the (native) chemical state of the unbound saponins, which unbound saponins may or may not comprise additional chemical groups such as a chemical group for linking the saponin to a linker, an amino-acid residue of the payload or to the scaffold, and/or a (chemical) linker bound to a chemical group of the saponin such as an aldehyde group or carboxylic acid group.
  • additional chemical groups such as a chemical group for linking the saponin to a linker, an amino-acid residue of the payload or to the scaffold, and/or a (chemical) linker bound to a chemical group of the saponin such as an aldehyde group or carboxylic acid group.
  • the chemical reactions used for providing the linkers and the scaffold comprising covalently linked glycosides for covalent binding to the payload e.g. triterpenoid saponins (polymerization of the glycosides)
  • triterpenoid saponins polymerization of the glycosides
  • saponins and for example biocompatible polymers applied as a scaffold for bearing bound saponins are water-soluble molecules.
  • the at least one saponin, and the effector moiety preferably a toxin or an oligonucleotide, in compartments or vesicles of the endocytic pathway of the target cell, e.g. a tumor cell or an auto-immune cell.
  • the target cell e.g. a tumor cell or an auto-immune cell.
  • BNA and free saponin synchronizing the presence of the molecules in the late endosomes, in order to obtain the synergistic effects in vivo cannot be beneficially obtained.
  • the invention preferably solves at least the following problem with respect to combining the effector moiety comprised by the conjugate of the invention and the saponins comprised by the conjugate of the invention: without wishing to be bound by any theory the only reasonable chemical group within, e.g., the saponins that can be used for (covalent), in particular single and cleavable, retainable coupling is required for the endosomal escape activity.
  • saponins previously applied for their immune-potentiating activity in the vaccination context involving saponins as adjuvant component are now also suitably for (covalent) coupling to ADCs or AOCs of the invention, for anti-tumor activity in vitro and in vivo.
  • An aspect of the invention relates to an antibody-drug conjugate comprising the effector moiety according to the invention, or a ligand-drug conjugate comprising the effector moiety of the invention, the effector moiety comprising covalently coupled saponin.
  • the effector moiety of the invention is a conjugate comprising at least one saponin and at least one effector molecule, covalently coupled to each other, either directly, or via at least one linker, optionally comprising a cleavable linker, and optionally via an oligomeric or polymeric scaffold to which the at least one saponin and/or the at least one effector moiety are covalently bound.
  • An embodiment is the antibody-drug conjugate or ligand-drug conjugate of the invention, wherein the antibody can bind to any one of CD71, CA125, EpCAM(17-1A), CD52, CEA, CD44v6, FAP, EGF-IR, integrin, syndecan-1, vascular integrin alpha-V beta-3, HER2, EGFR, CD20, CD22, Folate receptor 1, CD146, CD56, CD19, CD138, CD27L receptor, PSMA, CanAg, integrin-alphaV, CA6, CD33, mesothelin, Cripto, CD3, CD30, CD239, CD70, CD123, CD352, DLL3, CD25, ephrinA4, MUC1, Trop2, CEACAMS, CEACAM6, HER3, CD74, PTK7, Notch3, FGF2, C4.4A, FLT3, CD38, FGFR3, CD7, PD-L1, CTLA4, CD52, PDGFRA, VEGFR1, VEGFR2,
  • An aspect of the invention relates to a therapeutic combination comprising: (a) the effector moiety of the invention, comprising at least one saponin, and optionally a pharmaceutically acceptable excipient; and (b) an antibody-drug conjugate or a ligand-drug conjugate, and optionally a pharmaceutically acceptable excipient.
  • an embodiment is the therapeutic combination of the invention, wherein the antibody-drug conjugate can bind to any one of tumor-cell receptors CD71, CA125, EpCAM(17-1A), CD52, CEA, CD44v6, FAP, EGF-IR, integrin, syndecan-1, vascular integrin alpha-V beta-3, HER2, EGFR, CD20, CD22, Folate receptor 1, CD146, CD56, CD19, CD138, CD27L receptor, PSMA, CanAg, integrin-alphaV, CA6, CD33, mesothelin, Cripto, CD3, CD30, CD239, CD70, CD123, CD352, DLL3, CD25, ephrinA4, MUC1, Trop2, CEACAM5, CEACAM6, HER3, CD74, PTK7, Notch3, FGF2, C4.4A, FLT3, CD38, FGFR3, CD7, PD-L1, CTLA4, CD52, PDGFRA, VEGFR1, VEGFR2, preferably CD71
  • An effector moiety useful in the present invention preferably relies on late endosomal escape for exerting its effect.
  • Some effectors such as, e.g., a pseudomonas exotoxin, are rerouted to other organelles prior to the “late endosomal stage” and, thus, would normally not benefit from coupling to the second proteinaceous molecule according to the present invention.
  • toxin may be adapted for use with the present invention, e.g., by deleting the signal peptide responsible rerouting.
  • toxins that are highly toxic and would require only one molecule to escape the endosomes to kill a cell maybe modified to be less potent.
  • a conjugate of the invention comprising linked effector moiety—saponin comprises a covalently conjugated functionalized scaffold, i.e. a scaffold comprising covalently bound effector moietie(s) for delivering the scaffold comprising the bound effector moietie(s) at a target cell such as a tumor cell or an auto-immune cell, together with the covalently bound saponin.
  • a target cell such as a tumor cell or an auto-immune cell
  • ligand as used in this invention has its ordinary meaning and preferably means a molecule or structure that is able to bind another molecule or structure on the cell surface of a target cell, wherein said molecule or structure on the cell surface can be endocytosed and is preferably absent or less prominent on off-target cells.
  • said molecule or structure on the cell surface is constitutively endocytosed.
  • a ligand in this invention induces endocytosis of said molecule or structure on the cell surface of target cells after binding to said molecule or structure. This is for instance the case for Epidermal Growth Factor Receptor (EGFR), present on the surface of a variety of cancer cells.
  • EGFR Epidermal Growth Factor Receptor
  • a ligand can, e.g., be an antibody, a growth factor or a cytokine.
  • an effector moiety of the invention e.g. a toxin coupled to a saponin
  • a ligand is one possibility to create a targeted toxin.
  • a toxin that is only toxic in a target cell because it interferes with processes that occur in target cells only can also be seen as a targeted toxin (as in off-target cells it cannot exert its toxic action, e.g. apoptin).
  • a targeted toxin is a toxin that is combined with a ligand or e.g. a monoclonal antibody in order to be active in target cells and not in off-target cells (as it is only bound to and endocytosed by target cells).
  • a functionalized scaffold comprising a carrier molecule comprising a ligand and an effector moiety of the invention
  • the ligand or the monoclonal antibody guides the effector moiety with the saponin bound thereto, and scaffold to the target cells.
  • the at least one glycoside, preferably a saponin comprised by the effector moiety of the invention mediates the endosomal escape of the effector moiety.
  • the saponin is typically a saponin listed in Table A1 and Scheme I, and preferably the saponin is SO1861 and/or QS-21, and/or SA1641 and/or GE1741.
  • a payload, and an effector molecule, or effector moiety of the invention, in the context of this invention is any substance that affects the metabolism of a cell by interaction with an intracellular effector molecule target, wherein this effector molecule target is any molecule or structure inside cells excluding the lumen of compartments and vesicles of the endocytic and recycling pathway but including the membranes of these compartments and vesicles.
  • Said structures inside cells thus include the nucleus, mitochondria, chloroplasts, endoplasmic reticulum, Golgi apparatus, other transport vesicles, the inner part of the plasma membrane and the cytosol.
  • Cytosolic delivery of a payload or an effector moiety in the context of the invention preferably means that the payload or the effector moiety is able to escape the endosome (and/or lysosome), which, as defined previously, also includes escaping the endolysosome and the lysosome, and is preferably able to reach thepayload or the effector moiety target as described herein.
  • the invention also encompasses a new type of molecule, referred to as scaffold that serves to bring both a payload and at least one glycoside molecule such as a saponin, or an effector moiety of the invention in an endosome at the same time in a pre-defined ratio, when the effector moiety comprises the payload and the saponin.
  • the polymeric or oligomeric structure of the scaffold is a structurally ordered formation such as a polymer, oligomer, dendrimer, dendronized polymer, or dendronized oligomer or it is an assembled polymeric structure such as a hydrogel, microgel, nanogel, stabilized polymeric micelle or liposome, but excludes structures that are composed of non-covalent assemblies of monomers such as cholesterol/phospholipid mixtures.
  • polymer, oligomer, dendrimer, dendronized polymer, or dendronized oligomer have their ordinary meaning.
  • a polymer is a substance which has a molecular structure built up chiefly or completely from a large number of equal or similar units bonded together and an oligomer is a polymer whose molecules consist of relatively few repeating units.
  • an oligomer is a polymer whose molecules consist of relatively few repeating units.
  • the scaffold may comprise a polymeric or an oligomeric structure, or both, the full range of numbers of similar units bonded together applies to such structure. i.e. from 2 monomeric units to 100 monomeric units, 1000 monomeric units, and more.
  • a structure of 10 monomeric units maybe called either oligomeric or polymeric.
  • a scaffold as defined herein, further comprises at least one glycoside molecule such as a saponin of the invention.
  • a scaffold preferably includes a polymeric or oligomeric structure such as poly- or oligo(amines), e.g., polyethylenimine and poly(amidoamine), and biocompatible structures such as polyethylene glycol, poly- or oligo(esters), such as poly(lactids), poly(lactams), polylactide-co-glycolide copolymers, and poly(dextrin), poly- or oligosaccharides, such as cyclodextrin or polydextrose, and poly- or oligoamino acids, such as poly-lysine or a peptide or a protein, or DNA oligo- or polymers.
  • polymeric or oligomeric structure such as poly- or oligo(amines), e.g., polyethylenimine and poly(amidoamine), and biocompatible structures such as polyethylene glycol, poly- or oligo(esters), such as poly(lactids), poly(lactams
  • An assembled polymeric structure as defined herein comprises at least one scaffold and, optionally, other individual polymeric or oligomeric structures.
  • Other individual polymeric or oligomeric structures of said assembly may be (a) scaffolds (thus comprising at least one glycoside molecule such as a saponin of the invention), (b) functionalized scaffolds (thus comprising at least one glycoside molecule such as a saponin, and a ligand, antibody, etc., (c) polymeric or oligomeric structures without a glycoside molecule such as a saponin of the invention (See Table A1 for example), without a ligand, antibody, etc.
  • a functionalized assembled polymeric structure is an assembled polymeric structure that contains (a) at least one functionalized scaffold or (b) at least one scaffold and at least one polymeric structure comprising at least one ligand, antibody, etc.
  • Polymeric or oligomeric structures within an assembled polymeric structure that do not comprise any of the above mentioned molecules i.e. no glycosides such as saponins, no first proteinaceous molecule such as ligands, antibodies
  • the acidic environment seems to be a prerequisite for the synergistic action between glycoside (saponin) and effector moiety.
  • a pharmaceutically active substance in this invention is an effector moiety that is used to achieve a beneficial outcome in an organism, preferably a vertebrate, more preferably a human being such as a cancer patient or an auto-immune patient.
  • Benefit includes diagnosis, prognosis, treatment, cure and/or prevention of diseases and/or symptoms.
  • the pharmaceutically active substance may also lead to undesired harmful side effects.
  • pros and cons must be weighed to decide whether the pharmaceutically active substance is suitable in the particular case. If the effect of the pharmaceutically active substance inside a cell is predominantly beneficial for the whole organism, the cell is called a target cell. If the effect inside a cell is predominantly harmful for the whole organism, the cell is called an off-target cell. In artificial systems such as cell cultures and bioreactors, target cells and off-target cells depend on the purpose and are defined by the user.
  • An effector moiety that is a polypeptide may be, e.g., a polypeptide that recover a lost function, such as for instance enzyme replacement, gene regulating functions, or a toxin.
  • the effector moiety which effect is enhanced by the saponins bound thereto, detaches from the saponin and/or e.g. an antibody, when endocytosed. This can be achieved by a cleavable bond that breaks, e.g. under acidic, reductive, enzymatic or light-induced conditions.
  • An aspect of the invention relates to a pharmaceutical composition
  • a pharmaceutical composition comprising the effector moiety of the invention or the antibody-drug conjugate of the invention or the ligand-drug conjugate of the invention, comprising at least one saponin covalently linked to the effector molecule, and optionally a pharmaceutically acceptable excipient.
  • An aspect of the invention relates to the effector moiety of the invention or the antibody-drug conjugate of the invention or the therapeutic combination of the invention or the ligand-drug conjugate of the invention or the pharmaceutical composition of the invention, for use as a medicament.
  • An aspect of the invention relates to the effector moiety of the invention or the antibody-drug conjugate of the invention or the therapeutic combination of the invention or the ligand-drug conjugate of the invention or the pharmaceutical composition of the invention, for use in the treatment or prevention of a cancer or an autoimmune disease.
  • the inventors disclose here that covalently coupling saponins such as saponins in the water-soluble fraction of Quillaja saponaria , QS-21, SA1641, SO1861, Table A1, Scheme I, to an effector moiety such as an oligonucleotide, e.g. an antisense BNA, such as via a tri-functional linker, e.g. the tri-functional linker of Scheme II, or via an oligomeric or polymeric structure of a scaffold, such as a dendron, e.g.
  • a G4-dendron comprising covalently bound saponins
  • the effector moiety of the invention is an antisense oligonucleotide covalently conjugated with a dendron, which has multiple saponins covalently bound to it, such as 2, 4, 8, 16 saponins, preferably 4 saponins.
  • An embodiment is the conjugate of the invention comprising the effector moiety and a saponin comprising one or several or all of the indicated structural features of the saponin of Structure A in Scheme I, the saponin of structure A referred to as a saponin with an ‘ideal’ structure when endosomal escape enhancing activity towards an effector moiety present in the endosome of a cell contacted with conjugate of the invention, and/or a saponin selected from any one or more of the further saponins in Scheme I:
  • a glycoside, such as a saponin according to the invention, bound to the effector moiety comprised by the conjugate of the invention, which has the ‘ideal’ structure for the purpose of enhancing endosomal escape of an effector molecule comprised by the conjugate of the invention is a bisdesmosidic saponin according to Structure A of Scheme I, having a molecular mass of at least 1.500 Dalton and comprising an oleanan-type triterpene containing an aldehyde group at the C-23 position and optionally a hydroxyl group at the C-16 position, with a first branched carbohydrate side chain at the C-3 position which first branched carbohydrate side chain optionally contains glucuronic acid, wherein the saponin contains an ester group with a second branched carbohydrate side chain at the C-28 position which second branched carbohydrate chain preferably comprises at least four carbohydrate units, optionally containing at least one acetyl residue such as two acet
  • SO1861 is different from the “ideal structure” displayed in Scheme I, Structure A, only in having only one acetyl residue at the quinovose and having an additional xylose.
  • the “ideal structure” of a saponin for enhancing endosomal escape of an effector molecule or effector moiety, such as an antisense oligonucleotide such as a BNA, is a saponin which preferably has the Structure A of Scheme I, and saponins which display the endosomal escape enhancing activity have one or more of the structural features displayed in Structure A of Scheme I.
  • the QS-21 saponin and some of the saponins in the water soluble fraction of Quillaja saponaria differ in the carbohydrate modification at C-28 when the ideal structure of Structure A in Scheme I is considered: presence of an acyl chain in QS-21 for example.
  • saponins such as QS-7, QS1862, are similar to the ideal Structure A, and are similar to SO1861.
  • Tumor-specific cell-surface receptor targets which can be targeted by antibodies that can be used for the AOC-saponin conjugates comprising the conjugate of a payload and a saponin, i.e. the effecor moiety of the invention, ADC-saponin conjugates comprising the effector moiety of the invention comprising the conjugate of a payload and a saponin of the invention (not presented as a limitation; further immunoglobulins are equally suitable for the invention)
  • Target cell-surface receptor Example monoclonal antibodies HER2 anti-HER2 monoclonal antibody such as trastuzumab and pertuzumab CD20 anti-CD20 monoclonal antibody such as rituximab, ofatumumab, tositumomab and ibritumomab CA125 anti-CA125 monoclonal antibody such as oregovomab EpCAM (17-1A) anti-EpCAM (17-1A) monoclonal antibody such as edrecolo
  • Nigrin b SNA-V
  • Nigrin f SNA-Vf
  • tristis Yucca leaf protein YLP RIP 1 Carrière [Syn.: Yucca recurvifolia Salisb.] Basellaceae Basella rubra L. Basella RIP 2a, Basella RIP 2b, Basella RIP 3 RIP 1 Agrostemma githago L. Agrostin 2, Agrostin 5, Agrostin 6, Agrostin RIP 1 Dianthus barbatus L. Dianthin 29 RIP 1 Dianthus caryophyllus L. Dianthin 30, Dianthin 32 RIP 1 Dianthus chinensis L. [Syn.: D. sinensis RIP RIP 1 Dianthus sinensis Link] Gypsophila elegans M. Bieb.
  • moschata RIP RIP 1 Duchesne
  • PRIP 1 Cucurbita Moschatin
  • PRIP 2 moschata Duchesne ex ⁇ -moschin, ⁇ -moschin sRIP 1 candidate Lam.
  • Duchesne ex Poir. Cucurbita pepo L.
  • Pepocin RIP 1 Cucurbita pepo var. texana Texanin RIP 1 (Scheele)
  • D. S. Decker [Syn.: Cucurbita texana (Scheele) A. Gray] Gynostemma pentaphyllum Gynostemmin RIP 1 (Thunb.) Makino Lagenaria siceraria (Molina) Lagenin RIP 1 candidate Standl.
  • Trichokirin, Trichomislin TCM
  • Trichokirin S1, S-Trichokirin, Trichosanthrip sRIP 1 TKL-1 Trichosanthes kirilowii lectin-1 lectin/RIP 2 candidate TK-I, TK-II, TK-III, Trichosanthes kirilowii lectin lectin Trichosanthes kirilowii Karasurin-A, Karasurin-B, Karasurin-C RIP 1 Maximovicz var.
  • Trichomaglin RIP 1 Trichosanthes dioica Roxb.
  • TDSL lectin/RIP 2 candidate Trichosanthes sp.
  • E. characias lectin lectin Suregada multiflora Gelonin GAP 31 RIP 1 (A. Juss.) Baill. [Syn.: Gelonium multiflorum A.
  • Boerhaavia inhibitor RIP 1 candidate Bougainvillea spectabilis BAP I, Bouganin Bougainvillea RIP I RIP 1 Willd. Bougainvillea ⁇ buttiana cv. BBP-24, BBP-28 RIP 1 Enid Lancester Bougainvillea ⁇ buttiana cv. BBAP1 RIP 1 Mahara Mirabilis expansa (Ruiz & ME1, ME2 RIP 1 Pav.) Standl. Mirabilis jalapa L. MAP, MAP-2, MAP-3, MAP-4, MAP-S RIP 1 Olacaceae Malania oleifera Chun & Malanin lectin/RIP 2 S. K. Lee candidate Ximenia americana L.
  • keramanthus lectin RIP 2 candidate Adenia lanceolata Engl. Lanceolin RIP 2 Adenia racemosa W. J. de A. racemosa lectin lectin Wilde Adenia spinosa Burtt Davy A. spinosa lectin RIP 2 candidate Adenia stenodactyla Harms Stenodactylin RIP 2 Adenia venenata Forssk.
  • Phytolacca heterotepala Heterotepalin 4 Heterotepalin 5b RIP 1 H.
  • the at least one saponin that is comprised by the effector moiety according to the invention increases the efficacy of at least current and new effector moieties as defined in this invention. Potential side-effects will be decreased due to lowering of dosing of the effector moiety comprised by the conjugate formed with the saponin, without lowering the efficacy. Therefore, the invention provides an effector moiety according to the invention for use in medicine or for use as a medicament.
  • an aspect of the invention relates to an effector moiety of the invention, for use as a medicament.
  • an effector moiety according to the invention for manufacturing a medicament. Especially cancer medicines, and in particular the classical chemotherapy medicaments, are notorious for their side effects.
  • a therapeutic composition according to the invention is especially valuable for use as a medicament, in particular for use in a method of treating cancer.
  • the invention thus provides a therapeutic composition comprising an effector moiety according to the invention for use in a method of treating cancer.
  • the invention also provides a therapeutic composition comprising an effector moiety according to the invention for use in a method of treating acquired or hereditary disorders, in particular monogenic deficiency disorders.
  • the therapeutic composition thus comprises the effector moiety, e.g. a payload such as an antisense oligonucleotide.
  • an aspect of the invention relates to a therapeutic composition comprising an effector moiety according to the invention, wherein payload comprises a covalently bound saponin, for use in a method for the treatment of a cancer or an auto-immune disease.
  • MALDI-TOF spectra were recorded on a MALDI-Mass Spectrometer (Bruker Ultrafex III). Typically, the sample dissolved in MilliQ water in nanomolar to micromolar range was spotted on the target (MTP 384 target plate polished steel T F, Bruker Daltons) using either super-DHB (99%, Fluka) or sinapinic acid (SA, 99%, Sigma-Aldrich) as the matrix dissolved in acetonitrile (MADLI-TOF-MS tested, Sigma)/0.1% TFA (7:3 v/v) via the dried-droplet-method.
  • RP mode refers to reflector positive mode.
  • RN mode refers to reflector negative mode.
  • LP mode refers to linear positive mode.
  • Size exclusion chromatography was performed with Sephadex G 25 Superfine from GE Healthcare and on prepacked PD10 columns (GE Healthcare, Sephadex G 25 M). The material was activated by swelling in the respective eluent prior to performing chromatography.
  • MALDI-TOF-MS (RP mode) ( FIG. 4 A): m/z 2124 Da ([M+K] + , saponin-EMCH), m/z 2109 Da ([M+K] + , SO1861-EMCH), m/z 2094 Da ([M+Na] + , SO1861-EMCH)
  • MALDI-TOF-MS (RN mode) ( FIG. 5 C): m/z 2275 Da ([M ⁇ H] ⁇ , saponin-EMCH conjugate), 2244 Da ([M ⁇ H] ⁇ , saponin-EMCH conjugate), 2222 Da ([M ⁇ H] ⁇ , saponin-EMCH conjugate), 2178 Da ([M ⁇ H] ⁇ , saponin-EMCH conjugate), 2144 Da ([M ⁇ H] ⁇ , saponin-EMCH conjugate), 2122 Da ([M ⁇ H] ⁇ , saponin-EMCH conjugate), 2092 Da ([M ⁇ H] ⁇ , saponin-EMCH conjugate), 2070 Da ([M ⁇ H] ⁇ , SO1861-EMCH), 2038 Da ([M ⁇ H] ⁇ , SO1832-EMCH), 1936 Da ([M ⁇ H] ⁇ , SO1730-EMCH), 1861 Da ([M ⁇ H] ⁇ , SO1861).
  • 2-iminothiolane (231 ⁇ g, 1.1 ⁇ mol) dissolved in 47 ⁇ L PBS was added to a BSA-RhodB solution (10 mg, 0.15 ⁇ mol) in 200 ⁇ L PBS and the mix was shaken for 40 min at 800 rpm and room temperature on a ThermoMixer C (Eppendorf). After shaking for 40 min, the reaction mix was immediately run through a Sephadex G25 superfine size exclusion column (16 mL column volume) and SO1861-EMCH (1 mg, 0.5 ⁇ mol) dissolved in 100 ⁇ L PBS was added to the collected BSA-SH fraction.
  • the reaction mixture was shaken for 12 h at 800 rpm and room temperature on a ThermoMixer C (Eppendorf). After shaking for 12 h the BSA-SO1861 concentrated using centrifugal filtration at 4,000 rpm (15° C.) via Amicon Ultra 15 filters with a MWCO of 3 kDa. The conjugate was stored as solution in the fridge and aliquots were taken for analysis. Yield: not determined.
  • MALDI-TOF-MS ( FIG. 2 A) (LP mode): m/z 74.2 kDa ([M+H] + , BSA-SO1861 with 4 SO1861 attached), 72.2 kDa ([M+H] + , BSA-SO1861 with 3 SO1861 attached), 70.2 kDa ([M+H] + , BSA-SO1861 with 2 SO1861 attached), 37.0 kDa ([M+H] 2+ , BSA-SO1861 with 4 SO1861 attached), 35.9 kDa ([M+H] 2+ , BSA-SO1861 with 3 SO1861 attached), 34.7 kDa ([M+H] 2+ , BSA-SO1861 with 2 SO1861 attached).
  • 6-azidohexanoic acid (0.943 g, 6.00 mmol), EDCI.HCl (1.21 g, 6.30 mmol) and Oxyma Pure (0.938 g, 6.60 mmol) were dissolved in DMF (10.0 mL) and the mixture was stirred for 5 min.
  • a solution of di-tert-butyl (azanediylbis(ethane-2,1-diyl))dicarbamate (1.82 g, 6.00 mmol) in DMF (5.00 mL) was added and the reaction mixture was stirred at room temperature. After 5 hours the reaction mixture was evaporated in vacuo and the residue was dissolved in ethyl acetate (50 mL).
  • N,N-bis(2-aminoethyl)-6-azidohexanamide dihydrochloride (1.19 g, 3.76 mmol) in DMF (30.0 mL) and DIPEA (2.62 mL, 15.1 mmol) was added Boc-Lys(Boc)-ONp (3.69 g, 7.90 mmol) and the mixture was stirred at room temperature overnight. The reaction mixture was evaporated in vacuo and the residue was dissolved in ethyl acetate (100 mL).
  • N,N′-((9S,19S)-14-(6-aminohexanoyl)-1-mercapto-9-(3-mercaptopropanamido)-3,10,18-trioxo-4,11,14,17-tetraazatricosane-19,23-diyl)bis(3-mercaptopropanamide) formate (2.73 mg, 3.13 ⁇ mol) was dissolved in a mixture of 20 mM NH 4 HCO3 with 0.5 mM TCEP/acetonitrile (3:1, v/v, 3.00 mL). Next, SO1861-EMCH (29.2 mg, 0.014 mmol) was added and the reaction mixture was stirred at room temperature.
  • Dendron(SO1861) 4 -amine (6.81 mg, 0.748 ⁇ mol) and 2,5-dioxopyrrolidin-1-yl 1-azido-3,6,9,12-tetraoxapentadecan-15-oate (2.90 mg, 7.48 ⁇ mol) were dissolved in DMF(1.00 mL).
  • DIPEA 1.302 ⁇ L, 7.48 ⁇ mol
  • 3C Fractions corresponding to the product were immediately pooled together, frozen and lyophilized overnight to give the title compound (5.86 mg, 84%) as a white fluffy solid. Purity based on LC-MS 90%.
  • the resulting residue was dissolved in a mixture of 20 mM NH 4 HCO 3 with 0.5 mM TCEP/acetonitrile (3:1, v/v, 3.242 mL). From this solution, directly, 1000 ⁇ L was added to SO1861-EMCH (14.4 mg, 6.94 ⁇ mol, 4.5 equiv. compared to the scaffold) and the mixture was shaken for 1 min and left standing at room temperature. After 10 min the reaction mixture was lyophilized overnight.
  • HSP27 BNA oligo disulfide (1.10 mg, 0.187 ⁇ mol) was dissolved in 20 mM NH 4 HCO 3 with 1.0 mM TCEP (500 ⁇ L) and the mixture was shaken for 1 min and left standing at room temperature. After 1 hour the reaction mixture was filtered by using a centrifugal filter with a molecular weight cut-off of 3000 Da (14000 ⁇ g for 30 min). The residue solution was diluted with 20 mM NH 4 HCO 3 with 1.0 mM TCEP (500 ⁇ L) and the resulting mixture was filtered again under the same conditions described above.
  • HSP27 BNA oligo disulfide (1.1 mg, 0.187 ⁇ mol) was dissolved in 20 mM NH4HCO3 with 1.0 mM TCEP (500 ⁇ L) and the mixture was shaken for 1 min and left standing at room temperature. After 1 hour the reaction mixture was filtered by using a centrifugal filter with a molecular weight cut-off of 3000 Da (14000 ⁇ g for 30 min). The residue solution was diluted with 20 mM NH 4 HCO 3 with 1.0 mM TCEP (500 ⁇ L) and the resulting mixture was filtered again under the same conditions described above.
  • HSP27 primers F: R:
  • HSP27BNA(-thiol) oligos (sequence 5′-GGCacagccagtgGCG-3′) (Zhang et al., 2011) were ordered at Bio-synthesis Inc. (Lewisville, Tex.)
  • HSP27BNA heat shock protein 27
  • SO1861-EMCH SO1861-EMCH
  • HSP27BNA-L-SO1861 dendron(-L-SO1861) 4
  • A431 cancer cell line As readout, gene silencing of HSP27 mRNA in A431 cells was determined.
  • HSP27BNA-L-SO1861 treatment resulted in an improvement of HSP27 gene silencing activity compared to the HSP27BNA alone, whereas the activity of HSP27BNA-dendron(-L-SO1861) 4 (4 SO1861 molecules/BNA) is even stronger (3-fold) compared to the gene silencing activity of HSP27BNA alone ( FIG. 1 ).
  • conjugation of 1 or more SO1861 molecules improves the gene silencing activity of the therapeutic BNA oligo nucleotide due to the enhancement of SO1861-mediated endosomal escape and cytoplasmic delivery of the antisense BNA.
  • MALDI-TOF spectra were recorded on a MALDI-Mass Spectrometer (Bruker Ultrafex III). Typically, the sample dissolved in MilliQ water in nanomolar to micromolar range was spotted on the target (MTP 384 target plate polished steel T F, Bruker Daltons) using either super-DHB (99%, Fluka) or sinapinic acid (SA, 99%, Sigma-Aldrich) as the matrix dissolved in acetonitrile (MADLI-TOF-MS tested, Sigma)/0.1% TFA (7:3 v/v) via the dried-droplet-method.
  • RP mode refers to reflector positive mode.
  • RN mode refers to reflector negative mode.
  • LP mode refers to linear positive mode.
  • UV-Vis measurements were performed on a NanoDrop ND-1000 spectrophotometer in the spectral range of 200-750 nm.
  • Size exclusion chromatography was performed with Sephadex G 25 Superfine from GE Healthcare and on prepacked PD10 columns (GE Healthcare, Sephadex G 25 M). The material was activated by swelling in the respective eluent prior to performing chromatography.
  • Freeze-drying was performed on an Alpha 1-2 LD plus (Martin Christ Gefriertrocknungsanlagen GmbH). Typically, samples were frozen with liquid nitrogen and placed into the freeze-dryer at high vacuum.
  • MALDI-TOF-MS (RP mode) ( FIG. 4 A): m/z 2124 Da ([M+K] + , saponin-EMCH), m/z 2109 Da ([M+K] + , SO1861-EMCH), m/z 2094 Da ([M+Na] + , SO1861-EMCH)
  • MALDI-TOF-MS (RN mode) ( FIG. 5 C): m/z 2275 Da ([M ⁇ H] ⁇ , saponin-EMCH conjugate), 2244 Da ([M ⁇ H] ⁇ , saponin-EMCH conjugate), 2222 Da ([M ⁇ H] ⁇ , saponin-EMCH conjugate), 2178 Da ([M ⁇ H] ⁇ , saponin-EMCH conjugate), 2144 Da ([M ⁇ H] ⁇ , saponin-EMCH conjugate), 2122 Da ([M ⁇ H] ⁇ , saponin-EMCH conjugate), 2092 Da ([M ⁇ H] ⁇ , saponin-EMCH conjugate), 2070 Da ([M ⁇ H] ⁇ , SO1861-EMCH), 2038 Da ([M ⁇ H] ⁇ , S01832-EMCH), 1936 Da ([M ⁇ H] ⁇ , S01730-EMCH), 1861 Da ([M ⁇ H] ⁇ , SO1861).
  • the HATU-Cy3 solution was added to the stirring PAMAM solution and the reaction mix was stirred for 12 h at room temperature. After stirring for 12 h, the reaction mix was diluted with MilliQ water and dialyzed extensively for 24 h against MilliQ water using regenerated cellulose membrane tubes (Spectra/Por 6) with a MWCO of 2 kDa. After dialysis, the volume of the conjugate solution was reduced via a rotary evaporator (20 mbar, 60° C.) and the concentrated conjugate solution was run through a Sephadex G25 superfine size exclusion column (16 mL column volume). The first fraction was collected and lyophilized to obtain the viscous pink PAMAM-Cy3 conjugate.
  • PAMAM-Cy3 conjugate formation was confirmed by chromatography on thin layer chromatography (methanol/water, v/v 1:1), and the appearance of a faster band on a Sephadex G 25 superfine column. Yield 21.3 mg (63%).
  • the dye per PAMAM molar ratio determined by UV-Vis spectrophotometry was 0.43.
  • FIG. 8 A (LP mode): m/z 28.0 kDa ([M+H] + , Cy3-PAMAM).
  • the reaction mixture was shaken for 12 h at 800 rpm and room temperature on a ThermoMixer C (Eppendorf). After shaking for 12 h, the reaction mix was diluted with MilliQ water and dialyzed extensively for 24 h against MilliQ water using regenerated cellulose membrane tubes (ZelluTrans, Carl Roth) with a MWCO of 12-14 kDa. After dialysis, the Cy3-PAMAM-SO1861 solution was concentrated using centrifugal filtration at 4000 rpm (15° C.) via Amicon Ultra 15 filters with a MWCO of 3 kDa. The conjugate was stored as solution in the fridge and aliquots were taken for analysis. Yield: 0.5 mg (75%).
  • MALDI-TOF-MS spectra are illustrated in FIGS. 8 B-D, and FIG. 9 .
  • MALDI-TOF-MS of Cy3-PAMAM-(SO1861) 6 ( FIG. 8 B) (LP mode): m/z 38.4 kDa ([M+H] + , Cy3-PAMAM-SO1861), 17.9 kDa ([M+H] 2+ , Cy3-PAMAM-SO1861).
  • Cy3-PAMAM (0.5 mg, 18 nmol), SO1861 (2.3 mg, 1.24 ⁇ mol), and HATU (64.6 mg, 170 ⁇ mol) were dissolved separately in 200 ⁇ L DMSO.
  • SO1861 and HATU solutions were mixed and shaken for 20 min at 800 rpm and room temperature on a ThermoMixer C (Eppendorf).
  • Cy3-PAMAM solution was added to the shaking SO1861-HATU solution and the reaction mixture was allowed to shake for 12 h at 800 rpm and room temperature on a ThermoMixer C (Eppendorf).
  • the reaction mix was diluted with MilliQ water and dialyzed extensively for 24 h against MilliQ water using regenerated cellulose membrane tubes (ZelluTrans, Carl Roth) with a MWCO of 12-14 kDa.
  • the Cy3-PAMAM-NC-SO1861 solution was concentrated using centrifugal filtration at 4,000 rpm (15° C.) via Amicon Ultra 15 filters with a MWCO of 3 kDa.
  • the Cy3-PAMAM-NC-(SO1861) 17 conjugate was stored as solution in the fridge and aliquots were taken for analysis. Yield: 0.77 mg (69%).
  • MALDI-TOF-MS ( FIG. 10 ) (LP mode): m/z 62.3 kDa ([M+H] + , Cy3-PAMAM-NC-SO1861), 35.7 kDa ([M+H] 2+ , Cy3-PAMAM-NC-SO1861).
  • MALDI-TOF-MS spectra are illustrated in FIG. 11 .
  • MALDI-TOF-MS of PAMAM-(SH) 108 ( FIG. 11 C) (LP mode): m/z 41.5 kDa ([M+H] + , PAMAM-[SH] 108 ).
  • the batch was concentrated via centrifugal filtration using Amicon Ultra 15 mL centrifugal filters (10 kDa MWCO). The concentrated batch was run through a PD10 size exclusion column followed by lyophilization to obtain a white fluffy powder. Yield was not determined.
  • MALDI-TOF-MS spectra are illustrated in FIG. 12 .
  • MALDI-TOF-MS of PAMAM-(mPEG 2k ) 3 ( FIG. 12 C) (LP mode): m/z 33.46 kDa ([M+H] + , PAMAM-[mPEG 2k ] 3 ).
  • Cy3-PAMAM-(SO1861) 27 -(DBCO) 10 Procedure is described exemplary for Cy3-PAMAM-(SO1861) 27 -(DBCO) 10 .
  • Cy3-PAMAM-(SO1861) 27 (0.41 mg, 4.71 nmol) was freeze-fried and dissolved in 100 ⁇ L DMSO.
  • DBCO-PEG13-NHS ester (0.197 mg, 188 nmol) dissolved in DMSO was added to the Cy3-PAMAM-SO1861 solution and the mixture was shaken at 800 rpm and room temperature on a ThermoMixer C (Eppendorf).
  • the reaction mix was diluted with MilliQ water and dialyzed extensively for 24 h against MilliQ water using regenerated cellulose membrane tubes (ZelluTrans, Carl Roth) with a MWCO of 12-14 kDa.
  • the Cy3-PAMAM-SO1861-DBCO solution was concentrated using centrifugal filtration at 4,000 rpm (15° C.) via Amicon Ultra 15 filters with a MWCO of 3 kDa.
  • the conjugate was stored as solution in the fridge and aliquots were taken for analysis. Yield: 0.1 mg (22%).
  • MALDI-TOF-MS ( FIG. 13 D) (LP mode): m/z 92.5 kDa ([M+H] + , Cy3-PAMAM-SO1861-DBCO), 53.0 kDa ([M+H] 2+ , Cy3-PAMAM-SO1861-DBCO).
  • Cy3-PAMAM-NC-(SO1861) 17 (0.3 mg, 4.8 nmol) was freeze-fried and dissolved in 100 ⁇ L DMSO.
  • DBCO-PEG13-NHS ester (0.202 mg, 194 nmol) dissolved in DMSO was added to the Cy3-PAMAM-NC-SO1861 solution and the mixture was shaken at 800 rpm and room temperature on a ThermoMixer C (Eppendorf). After shaking for 3 h, the reaction mix was diluted with MilliQ water and dialyzed extensively for 24 h against MilliQ water using regenerated cellulose membrane tubes (ZelluTrans, Carl Roth) with a MWCO of 12-14 kDa.
  • MALDI-TOF-MS ( FIG. 13 B) (LP mode): m/z 93.2 kDa ([M+H] + , Cy3-PAMAM-NC-SO1861-DBCO), 49.6 kDa ([M+H] 2+ , Cy3-PAMAM-NC-SO1861-DBCO).
  • Plasmid-DNA (His-dianthin-EGF-pET11d or His-dianthin-pET11d) [20] was transformed into chemically competent Escherichia coli NiCo21 (DE3) (New England Biolabs®, Inc.) and grown in 3 mL lysogeny broth supplemented with 50 ⁇ g/mL ampicillin at 37° C. for 5 h at 200 rpm. These bacteria were used to inoculate 500 mL lysogeny broth supplemented with 50 ⁇ g/mL ampicillin for overnight culture at 37° C. Subsequently, the culture volume was scaled up to 2 L and bacteria were grown until an optical density (A600) of 0.9.
  • A600 optical density
  • IPTG isopropyl ⁇ -D-1-thiogalactopyranoside
  • Cells were further grown for 3 h at 37° C. and 200 rpm. After centrifugation (5 min, 5,000 g, 4° C.) cell pellets were resuspended in 20 mL phosphate buffered saline (Dulbecco's phosphate-buffered saline (PBS) with Ca 2+ and Mg 2+ , pH 7.4) and stored at ⁇ 20° C. After thawing, proteins were released by ultrasound device (Branson Sonifier 250, G. Heinemann).
  • PBS phosphate buffered saline
  • the solution was centrifuged (15,800 ⁇ g, 30 min, 4° C.) and adjusted to 20 mM imidazole concentration.
  • the construct contained an N-terminal His-tag and was purified by nickel nitrilotriacetic acid chromatography (Ni-NTA Agarose, Qiagen, Hilden, Germany). After elution with imidazole (20-250 mM) the eluates were analyzed by sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) (12%).
  • Fractions containing dianthin-EGF or dianthin were dialyzed against 2 L chitin binding domain buffer (20 mM tris(hydroxymethyl)-aminomethane/HCl, 500 mM NaCl, 1 mM EDTA, 0.1% Tween-20, pH 8.0) at 4° C. Further purification by chitin column affinity chromatography served to remove bacterial proteins with binding activity for Ni-NTA agarose. After elution with chitin binding domain buffer, the fractions were analyzed by SDS-PAGE (12%). Fractions containing dianthin-EGF or dianthin were dialyzed against 5 L PBS at 4° C. Purified proteins were concentrated by Amicon centrifugal filter devices (10 kDa, Millipore, Eschborn, Germany). The protein concentration was determined by a bicinchoninic acid assay (Pierce, Rockford, USA).
  • Dianthin-EGF (240 ⁇ g, 6.7 nmol) solution in PBS was placed into an Amicon Ultra 15 filter with a MWCO of 3 kDa and centrifuged at 4,000 rpm and 4° C. for 30 min three times. After each cycle, the Amicon filter was refilled with 0.1 M sodium carbonate buffer at pH 9. After the third centrifugation cycle, the volume was reduced to 0.5 mL via centrifugation. The dianthin-EGF sodium carbonate solution was placed into a 2 mL reaction tube and Alexa FluorTM 488 5-TFP (50 ⁇ g, 56 nmol) dissolved in 10 ⁇ L DMSO was added to the protein solution.
  • Alexa FluorTM 488 5-TFP 50 ⁇ g, 56 nmol
  • the mix was shaken at 800 rpm and room temperature on a ThermoMixer C (Eppendorf) for 80 min. After shaking, the mix was run through a Sephadex G25 M size exclusion column (GE Healthcare, PD10 column). The dianthin-EGF-Alexa488 conjugate was stored in solution in 0.1 M sodium carbonate buffer at pH 9 in the fridge and aliquots were taken for analysis. Yield: 210 ⁇ g (85%).
  • MALDI-TOF-MS ( FIG. 14 D) (LP mode): m/z 36.8 kDa ([M+H] + , dianthin-EGF-Alexa488), m/z 33.6 kDa ([M+H] + , dianthin-EGF-Alexa488), 18.8 kDa ([M+H] 2+ , dianthin-EGF-Alexa488), 16.6 kDa ([M+H] 2+ , dianthin-EGF-Alexa488).
  • Dianthin (184 ⁇ g, 6.2 nmol) solution in PBS was placed into an Amicon Ultra 15 filter with a MWCO of 3 kDa and centrifuged at 4,000 rpm and 4° C. for 30 min three times. After each cycle, the Amicon filter was refilled with 0.1 M sodium carbonate buffer at pH 9. After the third centrifugation cycle, the volume was reduced to 0.5 mL via centrifugation. The dianthin sodium carbonate solution was placed into a 2 mL reaction tube and Alexa FluorTM 488 5-TFP (16.7 ⁇ g, 19 nmol) dissolved in 3.5 ⁇ L DMSO was added to the protein solution.
  • the mix was shaken at 800 rpm and room temperature on a ThermoMixer C (Eppendorf) for 80 min. After shaking, the mix was run through a Sephadex G25 M size exclusion column (GE Healthcare, PD 10 column).
  • the dianthin-Alexa488 conjugate was stored in solution in 0.1 M sodium carbonate buffer at pH 9 in the fridge and aliquots were taken for analysis. Yield: not determined MALDI-TOF-MS ( FIG. 15 D) (LP mode): m/z 30.7 kDa ([M+H] + , dianthin-Alexa488).
  • MALDI-TOF-MS ( FIG. 14 E) (LP mode): m/z 40.8 kDa ([M+H] + , dianthin-EGF-Alexa488-S-S-PEG-N3), m/z 37.5 kDa ([M+H] + , dianthin-EGF-Alexa488-S-S-PEG-N 3 ).
  • Dianthin-Alexa488 (24.5 ⁇ g, 0.8 nmol) sodium carbonate solution was placed into a 2 mL reaction tube and azido-PEG3-S-S-NHS (34 ⁇ g, 78 nmol) dissolved in 9 ⁇ L DMSO was added to the protein solution.
  • the mix was shaken at 800 rpm and 15° C. on a ThermoMixer C (Eppendorf) for 12 h. After shaking, the reaction mix was diluted with PBS and was washed with PBS via centrifugal filtration at 4,000 rpm and 4° C. using Amicon Ultra 15 filter with a MWCO of 3 kDa.
  • FIG. 15 E (LP mode): m/z 32.9 kDa ([M+H] + , dianthin-Alexa488-S-S-PEG-N3).
  • Cy3-PAMAM-(SO1861) 27 -DBCO Procedure is described exemplary for Cy3-PAMAM-(SO1861) 27 -DBCO.
  • Cy3-PAMAM-(SO1861) 27 -DBCO (17 ⁇ g, 0.184 nmol) solution in MilliQ water was mixed with a dianthin-EGF-Alexa488-SS-PEG3-N 3 (3.6 ⁇ g, 0.089 nmol) solution in PBS in a 1.5 mL reaction tube and the reaction mix was shaken at 800 rpm and 15° C. on a ThermoMixer C (Eppendorf) for 2 h. After shaking, small aliquots were taken out for analysis via SDS-PAGE and fluorescence imaging on a Molecular Imager® VersaDocTM MP 4000 imaging system (Bio-Rad) ( FIG. 16 ).
  • Cy3-PAMAM (0.19 mg, 13 nmol) and SO1861 (0.73 mg, 0.39 ⁇ mol) were dissolved separately in 200 ⁇ L 0.1 M acetate buffer at pH 5.
  • SO1861 and Cy3-PAMAM solutions were mixed and shaken for 20 min at 800 rpm and room temperature on a ThermoMixer C (Eppendorf).
  • NaCNBH 3 5 mg, 81 ⁇ mol was added to the shaking reaction solution and the reaction mixture was allowed to shake for 12 h at 800 rpm and room temperature on a ThermoMixer C (Eppendorf).
  • the reaction mix was diluted with MilliQ water and dialyzed extensively for 24 h against MilliQ water using regenerated cellulose membrane tubes (ZelluTrans, Carl Roth) with a MWCO of 12-14 kDa.
  • the Cy3-PAMAM-NC-SO1861 solution was concentrated using centrifugal filtration at 4,000 rpm (15° C.) via Amicon Ultra 15 filters with a MWCO of 3 kDa.
  • the conjugate was stored as solution in the fridge and aliquots were taken for analysis. Yield: not determined
  • MALDI-TOF-MS ( FIG. 17 B, C) (LP mode): m/z 88.7 kDa ([M+H] + , Cy3-PAMAM-NC-SO1861), 49.2 kDa ([M+H] 2+ , Cy3-PAMAM-NC-SO1861).
  • Customized peptide with the sequence SESDDAMFCDAMDESDSK (0.6 mg, 0.3 ⁇ mol) and SO1861-EMCH (0.8 mg, 0.39 ⁇ mol) were dissolved separately in 200 ⁇ L PBS.
  • SO1861-EMCH and peptide solutions were mixed and shaken for 12 h at 800 rpm and room temperature on a ThermoMixer C (Eppendorf). After shaking small aliquots were taken out for analysis. Yield: not determined MALDI-TOF-MS ( FIG.
  • aldehyde and alcohol groups are best suitable for reversible conjugation reactions, while the alkene and the carboxylic acid (glucuronic acid) are the groups best suitable for irreversible/stable conjugation reactions.
  • the aldehyde group within the molecule structure of SO1861, however, is the most suitable for reversible conjugation reactions over the alcohols. On the one hand, because there is only one aldehyde present in the structure that allows chemoselective reactions.
  • the aldehyde can perform reversible conjugation reactions with a variety of chemical groups such as amines, hydrazides, and hydroxylamines forming acid-cleavable moieties like imines, hydrazones, and oximes.
  • This factor enables a freedom of choice over the chemical group for the desired reversible conjugation reaction.
  • the alcohols are good candidates for reversible conjugation reaction via the formation of acetals and ketals as well, but lack in chemoselectivity since they are present in a large quantity on the glycosidic structure.
  • carboxylic acid is the most suitable since it can form amides and esters with the common tools used in peptide chemistry (e.g. reaction with amines via carbodiimide mediated amide formation).
  • an endosomal escape enhancing saponin such as SO1861
  • a methodology has been established that allows the generation of a non-cleavable and cleavable ‘ready to conjugate’ saponins ( FIG. 20 ) using the most suitable chemical groups present on SO1861.
  • the carboxylic group of SO1861 is activated via a reagent used in peptide coupling chemistry to generate an active ester (e.g. 1-[Bis(dimethylamino)methylene]-1H-1,2,3-triazolo[4,5-b]pyridinium 3-oxide hexafluorophosphate, HATU).
  • the resulting active ester of SO1861 is able to react with amines forming stable amide bonded conjugates ( FIG. 20 A).
  • the aldehyde group of SO1861 is reacted with an EMCH (E-maleimidocaproic acid hydrazide) linker.
  • EMCH E-maleimidocaproic acid hydrazide
  • the hydrazide group of EMCH forms an acid cleavable hydrazone bond with the aldehyde of SO1861.
  • the EMCH linker presents a maleimide group that is thiol (sulfhydryl group) reactive and thus can be conjugated to thiols ( FIG. 20 B).
  • the maleimide group of SO1861-EMCH performs a rapid and specific Michael addition reaction with thiols and thiol bearing polymeric structures when carried out in a pH range of 6.5-7.5 ( FIG. 20 B).
  • the acid sensitive hydrazone linkage between the SO1861 and EMCH can be utilized to perform saponin release from a scaffold in acidic environment ( FIG. 21 ).
  • the EMCH linker fulfills both the need for a pH cleavable strategy and a conjugation strategy to a polymeric structure.
  • FIG. 5 A To synthesize the SO1861-EMCH, a strategy has been developed that allows the conversion of the aldehyde group on the SO1861 to EMCH ( FIG. 5 A).
  • the SO1861-EMCH conjugate was isolated and successfully characterized via nuclear magnetic resonance spectroscopy (see materials and methods section, FIG. 3 B) and matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF-MS) as shown in FIGS. 5 B and 5 C, and FIG. 4 A.
  • MALDI-TOF-MS matrix-assisted laser desorption/ionization time-of-flight mass spectrometry
  • PAMAM-Cy3 was utilized as the polymeric structure for the conjugation to both SO1861-EMCH and SO1861-HATU and served as a model for conjugation of SO1861 to a polymeric structure ( FIG. 25 ).
  • the three different feeds corresponded to an obtained mass of m/z 38.4 kDa, m/z 53.9 kDa and m/z 133.8 kDa for the Cy3-PAMAM-SO1861 conjugates that correspond to 6, 13 and 51 SO1861 molecules attached per PAMAM dendrimer as shown on FIG. 8 B-D.
  • the carboxylic acid of SO1861 was activated with HATU and then reacted with the amines of Cy3-PAMAM forming a pH stable amide bound between Cy3-PAMAM and SO1861.
  • the saponin conjugated scaffolds were conjugated to linking points for a possible conjugation to targeted therapeutics (e.g. targeted toxins) via the so-called strain-promoted alkyne-azide cycloaddition (SPAAC, click chemistry) to obtain a functionalized scaffold.
  • targeted therapeutics e.g. targeted toxins
  • SPAAC strain-promoted alkyne-azide cycloaddition
  • the NHS N Hydroxysuccinimide
  • the resulting DBCO dibenzocyclooctyne moiety on the conjugates is able to perform SPAAC with corresponding azides on the targeted therapeutics.
  • Dianthin-EGF served as a model targeted toxin and dianthin served as a non-targeted toxin. Both toxins were labeled with Alexa FluorTM 488 using the tetrafluorophenyl ester (TFP) derivative of the dye. The dye labeled proteins were then conjugated to a heterobifunctional NHS-SS-PEGS-azide linker to obtain the corresponding chemical moiety for the SPAAC to the PAMAM-saponin conjugates. Maldi-TOF-MS measurements showed that one Alexa FluorTM 488 dye and 9 NHS-SS-PEG3-azide molecules were attached per dianthin-EGF molecule ( FIG. 14 , FIG. 15 ).
  • Alexa FluorTM 488 labeled dianthin-EGF was conjugated to a heterobifunctional NHS-PEG12-azide linker that lacked the disulfide bond and would thus generate a non-toxin-cleavable construct.
  • Cy3-PAMAM-NC-SO1861-DBCO and Cy3-PAMAM-SO1861-DBCO conjugates were reacted with Alexa FluorTM 488 labeled azido-toxins to perform a strain-promoted alkyne-azide cycloaddition.
  • the conjugation between the reacting agents was indicated via gel electrophoresis and the co-localization of the fluorescent signals of Cy3 that is only attached on the PAMAM polymer and Alexa FluorTM 488 that is only attached on the toxins on a polyacrylamide gel after gel electrophoresis ( FIG. 16 ).
  • a G4-dendron (PFd-G4-Azide-NH-BOC, Polymer Factory) with 16 functional amino end groups and an azido group at the focal point was utilized for the conjugation to SO1861 ( FIG. 26 ).
  • the advantage of using a dendron over a dendrimer is the focal point that the dendron structure is exhibiting. This focal point allows the direct conjugation to a targeted toxin without the need of its post-modification with orthogonal click functions ( FIG. 27 ).
  • the dendron underwent the same methodology as described for the PAMAM dendrimer. Briefly, after partial dye labeling and deprotection ( FIG.
  • the amino groups of the dendron were converted into thiols using the thiolating reagent 2-iminothiolane followed by conjugation to SO1861-EMCH.
  • SO1861-EMCH three different feed equivalents of SO1861-EMCH were used.
  • the dendron-SO1861 conjugates were analyzed via MALDI-TOF-MS. As expected, the conjugate species of 1 and 2 SO1861 molecules per dendron molecule were obtained when low SO1861-EMCH feed equivalents of 3 and 10 were used ( FIG. 29 B, C). Higher dendron-SO1861 conjugate species of up to 9 SO1861 molecules per dendron were obtained ( FIG. 29 A) when using a feed equivalent of 22 SO1861-EMCH molecules per dendron molecule.
  • the saponin functionalized dendron will be conjugated to targeted toxins over its focal point to yield a functionalized scaffold and will be evaluated biologically.
  • HeLa cells were seeded in DMEM (PAN-Biotech GmbH) supplemented with 10% fetal bovine serum (PAN-Biotech GmbH) and 1% penicillin/streptomycin (PAN-Biotech GmbH), in a 96 well plate at 5,000 c/w in 100 ⁇ L/well and incubated overnight at 37° C. and 5% CO 2 .
  • DMEM fetal bovine serum
  • PAN-Biotech GmbH penicillin/streptomycin
  • the media was removed from the cell culture plate and replaced by 160 ⁇ L culture media, followed by the addition of 10 ⁇ L sample/well (from the 20 ⁇ concentrated stocks) and a 45 min incubation at 37° C. During this incubation the SO1861 concentration curve was prepared.
  • the SO1861 master stock was heated for 10 min at 50° C., while shaking at 1,250 rpm.
  • a serial dilution of SO1861 was prepared in PBS.
  • the SO1861 concentration curve was prepared as 10 ⁇ concentrated stock, from which 20 ⁇ L was added/well.
  • the cells were incubated for 72 hr at 37° C. before the cell viability was determined by a MTS-assay, performed according to the manufacturer's instruction (CellTiter 96® AQueous One Solution Cell Proliferation Assay, Promega). Briefly, the MTS solution was diluted 20 ⁇ in DMEM without phenol red (PAN-Biotech GmbH) supplemented with 10% FBS. The cells were washed once with 200 ⁇ L/PBS well, after which 100 ⁇ L diluted MTS solution was added/well. The plate was incubated for approximately 20-30 minutes at 37° C. Subsequently, the OD at 492 nm was measured on a Thermo Scientific Multiskan FC plate reader (Thermo Scientific).
  • the background signal of ‘medium only’ wells was subtracted from all other wells, before the cell viability percentage of treated/untreated cells was calculated, by dividing the background corrected signal of treated wells over the background corrected signal of the untreated wells ( ⁇ 100).
  • HeLa cells were seeded in DMEM (PAN-Biotech GmbH) supplemented with 10% fetal calf serum (PAN-Biotech GmbH) and 1% penicillin/streptomycin (PAN-Biotech GmbH), at 500,000 c/plate in 10 cm dishes and incubated for 48 hrs (5% CO 2 , 37° C.), until a confluency of 90% was reached. Next, the cells were trypsinized (TrypIE Express, Gibco Thermo Scientific) to single cells. 0.75 ⁇ 10 6 Cells were transferred to a 15 mL falcon tube and centrifuged (1,400 rpm, 3 min). The supernatant was discarded while leaving the cell pellet submerged.
  • the pellet was dissociated by gentle tapping the falcon tube on a vortex shaker and the cells were washed with 4 mL cold PBS (Mg 2+ and Ca 2+ free, 2% FBS). After washing the cells were resuspended in 3 mL cold PBS (Mg 2+ and Ca 2+ free, 2% FBS) and divided equally over 3 round bottom FACS tubes (1 mL/tube). The cells were centrifuged again and resuspended in 200 ⁇ L cold PBS (Mg 2+ and Ca 2+ free, 2% FBS) or 200 ⁇ L antibody solution; containing 5 ⁇ L antibody in 195 ⁇ L cold PBS (Mg 2+ and Ca 2+ free, 2% FBS).
  • APC Mouse IgG1, K Isotype Ctrl FC (#400122, Biolegend) was used as isotype control, and APC anti-human EGFR (#352906, Biolegend) was used to stain the EGFR receptor.
  • Samples were incubated for 30 min at 4° C. on a tube roller mixer. Afterwards, the cells were washed 3 ⁇ with cold PBS (Mg 2+ and Ca 2+ free, 2% FBS) and fixated for 20 min at room temperature using a 2% PFA solution in PBS. Cells were washed 2 ⁇ with cold PBS, and resuspended in 250-350 ⁇ L cold PBS for FACS analysis. Samples were analyzed with a BD FACSCanto II flow cytometry system (BD Biosciences) and FlowJo software.
  • the amino groups in amine containing polymeric structures such as PAMAM and PEI (polyethylenimine) are able to block the acidification of the endosomes via the their intrinsic H+buffering capacity. Since the endosomal escape enhancing properties of SO1861 are only exposed at low endosomal pH ( ⁇ pH5), the scaffold or functionalized scaffold should not contain chemical groups that are able to interfere in acidification of the endosomes and thus block the activity of SO1861.
  • the amine containing polymeric structures of a G5 PAMAM (128 primary amines as well as approximately 126 tertiary amines) and G4-dendron (16 primary amines) were tested, in order to determine if these molecules inhibit the endosomal escape enhancing capacity of SO1861.
  • Co-administration experiments of PAMAM (native or thiolated) or dendron (native) in combination with dianthin-EGF and various SO1861 concentrations on HeLa cells were performed. As control for the inhibition of endosomal acidification chloroquine was used.
  • HeLa cells show sufficient EGFR cell surface levels ( FIG. 30 A). It is observed that both ‘native’ PAMAM and chloroquine inhibit the SO1861-mediated endosomal escape of the targeted toxin and subsequent cell killing in Hela cells ( FIG. 30 B). PAMAM at 500 nM inhibits even to an equal extent as the endosomal acidification inhibitor chloroquine, while 667 nM dendron has no effect at all.
  • the inhibitory activity of the ‘native’ PAMAM is due to the availability of amino groups in PAMAM, the primary amino groups of PAMAM were partially thiolated through reaction with 2-iminothiolane ( FIG. 11 ), resulting in 16 of 128 ( FIG. 11 C), 65/128 ( FIGS.
  • Scheme I displays the common molecular structure of a series of QS-21 saponins (in part adapted from: Conrado Pedebos, Laércio Pol-Fachin, Ramon Pons, Cilaine V. Teixeira Hugo Verli, Atomic Model and Micelle Dynamics of QS-21 Saponin, Molecules 2014, 19, 3744-3760).
  • a mixture of water-soluble saponins obtained from Quillaja saponaria (Sigma-Aldrich, product No. S4521; Roth, Item No. 6857;
  • product ‘Quit-A’) may be applied in the effector moiety comprising at least one saponin according to the invention, or in an endosomal/lysosomal escape enhancing conjugate, composition, combination of the invention, based on endosomal/lysosomal escape enhancing properties of at least one individual saponin present in the mixture, e.g. QS-21, or based on a combination of two or more of the saponins comprised by the mixture, such as QS-21 and QS-7.
  • the effector moiety exposed to the cells was dianthin covalently coupled to the ligand EGF: EGF-dianthin.
  • Cells tested were tumor cell lines HeLa for free saponins, and A431, MDA-MB-468, CaSki and A2058 for testing the saponins of Quillaja saponaria when covalently coupled to cetuximab.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Epidemiology (AREA)
  • Immunology (AREA)
  • Cell Biology (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Organic Chemistry (AREA)
  • Oncology (AREA)
  • Toxicology (AREA)
  • Biochemistry (AREA)
  • Biotechnology (AREA)
  • Botany (AREA)
  • Genetics & Genomics (AREA)
  • Pulmonology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Biomedical Technology (AREA)
  • General Engineering & Computer Science (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Biophysics (AREA)
  • Plant Pathology (AREA)
  • Microbiology (AREA)
  • Dispersion Chemistry (AREA)
  • Physics & Mathematics (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicinal Preparation (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
US17/312,104 2018-12-21 2019-12-09 Saponin Conjugates Pending US20220023433A1 (en)

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
NL2022283 2018-12-21
NL2022283 2018-12-21
NL2023468 2019-07-10
NL2023468 2019-07-10
NL2023568 2019-07-25
NL2023568 2019-07-25
PCT/EP2019/084211 WO2020126610A1 (en) 2018-12-21 2019-12-09 Saponin conjugates

Publications (1)

Publication Number Publication Date
US20220023433A1 true US20220023433A1 (en) 2022-01-27

Family

ID=71101079

Family Applications (7)

Application Number Title Priority Date Filing Date
US17/312,403 Pending US20220111066A1 (en) 2018-12-21 2019-12-09 Antibody-drug conjugate with improved therapeutic window
US17/312,104 Pending US20220023433A1 (en) 2018-12-21 2019-12-09 Saponin Conjugates
US17/415,759 Pending US20220218837A1 (en) 2018-12-21 2019-12-09 Drug conjugate
US17/312,476 Pending US20220072149A1 (en) 2018-12-21 2019-12-09 Biologically active cluster of molecules
US17/312,193 Pending US20220054643A1 (en) 2018-12-21 2019-12-09 Saponin conjugated to epitope-binding proteins
US17/312,311 Pending US20230173085A1 (en) 2018-12-21 2019-12-09 Saponin Conjugates
US17/312,019 Pending US20220313834A1 (en) 2018-12-21 2019-12-09 Improved antibody-oligonucleotide conjugate

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US17/312,403 Pending US20220111066A1 (en) 2018-12-21 2019-12-09 Antibody-drug conjugate with improved therapeutic window

Family Applications After (5)

Application Number Title Priority Date Filing Date
US17/415,759 Pending US20220218837A1 (en) 2018-12-21 2019-12-09 Drug conjugate
US17/312,476 Pending US20220072149A1 (en) 2018-12-21 2019-12-09 Biologically active cluster of molecules
US17/312,193 Pending US20220054643A1 (en) 2018-12-21 2019-12-09 Saponin conjugated to epitope-binding proteins
US17/312,311 Pending US20230173085A1 (en) 2018-12-21 2019-12-09 Saponin Conjugates
US17/312,019 Pending US20220313834A1 (en) 2018-12-21 2019-12-09 Improved antibody-oligonucleotide conjugate

Country Status (15)

Country Link
US (7) US20220111066A1 (ja)
EP (14) EP4223316A3 (ja)
JP (6) JP2022515251A (ja)
KR (8) KR20210117274A (ja)
CN (6) CN113453722A (ja)
AU (6) AU2019400630A1 (ja)
BR (2) BR112021012222A8 (ja)
CA (6) CA3124129A1 (ja)
DK (3) DK3897738T3 (ja)
ES (2) ES2899612T3 (ja)
IL (8) IL284274A (ja)
MX (2) MX2021007296A (ja)
PT (1) PT4015003T (ja)
SG (8) SG11202106606RA (ja)
WO (8) WO2020126600A1 (ja)

Families Citing this family (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3618867A4 (en) 2017-05-02 2021-01-20 Cornell University METHODS AND REAGENTS FOR AGAINST TUMOR WITH HIGHER EFFICIENCY AND LOWER TOXICITY
EP4223316A3 (en) * 2018-12-21 2023-10-04 Sapreme Technologies B.V. Improved antibody-oligonucleotide conjugate
US20230357310A1 (en) * 2019-06-24 2023-11-09 Sapreme Technologies B.V. Saponin derivatives with improved therapeutic window
CN111518212B (zh) * 2020-04-16 2021-03-12 上海洛启生物医药技术有限公司 抗Trop2纳米抗体及其应用
CN116194148A (zh) * 2020-06-24 2023-05-30 萨普雷米科技有限公司 GalNAc-寡核苷酸缀合物和皂苷的治疗性组合及其用途
AU2020454675A1 (en) * 2020-06-24 2023-02-16 Sapreme Technologies B.V. Combination comprising an ADC or an AOC comprising a VHH, and a saponin or a ligand-saponin conjugate
WO2021261992A1 (en) * 2020-06-24 2021-12-30 Sapreme Technologies B.V. Conjugate of galnac and saponin, therapeutic composition comprising said conjugate and a galnac-oligonucleotide conjugate
CN116390932A (zh) * 2020-06-24 2023-07-04 萨普雷米科技有限公司 用于在药品中使用的皂苷衍生物
IL299356A (en) * 2020-06-24 2023-02-01 Sapreme Tech Bv hydrazone derivatives of saponin
US20230248845A1 (en) * 2020-06-24 2023-08-10 Sapreme Technologies B.V. Combination of an antibody-drug conjugate and an antibody-saponin conjugate
WO2022055351A1 (en) * 2020-09-10 2022-03-17 Sapreme Technologies B.V. Conjugate of saponin, oligonucleotide and galnac
WO2022055352A1 (en) * 2020-09-10 2022-03-17 Sapreme Technologies B.V. Semicarbazone-based saponin conjugate
US10947552B1 (en) 2020-09-30 2021-03-16 Alpine Roads, Inc. Recombinant fusion proteins for producing milk proteins in plants
US10894812B1 (en) 2020-09-30 2021-01-19 Alpine Roads, Inc. Recombinant milk proteins
CA3191387A1 (en) 2020-09-30 2022-04-07 Nobell Foods, Inc. Recombinant milk proteins and food compositions comprising the same
WO2022164316A1 (en) 2021-01-26 2022-08-04 Sapreme Technologies B.V. Semicarbazone-based saponin conjugate
WO2022265493A1 (en) * 2021-06-18 2022-12-22 Sapreme Technologies B.V. Conjugate of saponin, oligonucleotide and galnac
WO2023038517A1 (en) 2021-09-09 2023-03-16 Sapreme Technologies B.V. Semicarbazone-based saponin conjugate
WO2023121445A1 (en) 2021-12-22 2023-06-29 Sapreme Technologies B.V. Compositions comprising therapeutic nucleic acid and saponin for the treatment of muscle-wasting disorders
WO2023121446A1 (en) 2021-12-22 2023-06-29 Sapreme Technologies B.V. Targeted saponin-nucleic acid conjugates for treatment of muscle wasting disorders
WO2023121447A1 (en) * 2021-12-22 2023-06-29 Sapreme Technologies B.V. Conjugate of a single domain antibody, a saponin and an effector molecule, pharmaceutical composition comprising the same, therapeutic use of said pharmaceutical composition
WO2023121444A1 (en) 2021-12-22 2023-06-29 Sapreme Technologies B.V. Compositions comprising therapeutic nucleic acid and targeted saponin for the treatment of muscle-wasting disorders
WO2023249473A1 (ko) * 2022-06-24 2023-12-28 주식회사 피노바이오 하나의 항체에 2종의 약물-링커 접합체가 연결된 항체-약물 접합체
WO2024013361A1 (en) 2022-07-15 2024-01-18 Proqr Therapeutics Ii B.V. Oligonucleotides for adar-mediated rna editing and use thereof
WO2024013360A1 (en) 2022-07-15 2024-01-18 Proqr Therapeutics Ii B.V. Chemically modified oligonucleotides for adar-mediated rna editing
WO2024046622A1 (en) 2022-09-01 2024-03-07 Sapreme Technologies B.V. Compositions of polyplexes and saponins
EP4331609A1 (en) 2022-09-01 2024-03-06 Sapreme Technologies B.V. Methods and compositions for enhanced polyplex delivery
EP4331610A1 (en) 2022-09-01 2024-03-06 Sapreme Technologies B.V. Polyplex-saponin covalent conjugate

Family Cites Families (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5583112A (en) * 1987-05-29 1996-12-10 Cambridge Biotech Corporation Saponin-antigen conjugates and the use thereof
US5591829A (en) * 1987-05-29 1997-01-07 Matsushita; Shuzo Antibodies modified with toxic substance
US5856571A (en) * 1995-06-07 1999-01-05 Cellpro, Incorporated Semicarbazide-containing linker compounds for formation of stably-linked conjugates and methods related thereto
US5977081A (en) * 1997-05-20 1999-11-02 Galenica Pharmaceuticals, Inc. Triterpene saponin analogs having adjuvant and immunostimulatory activity
AU5565599A (en) * 1998-08-14 2000-03-06 Dante J. Marciani Chemically modified saponins and the use thereof as adjuvants
AU2002240250A1 (en) * 2002-02-04 2003-09-09 Antigenics Inc. Immunostimulant compositions comprising aminoalkyl glucosaminide phosphates and saponins
US20040242502A1 (en) * 2003-04-08 2004-12-02 Galenica Pharmaceuticals, Inc. Semi-synthetic saponin analogs with carrier and immune stimulatory activities for DNA and RNA vaccines
EP1623715B1 (en) * 2004-07-13 2012-09-19 Charité - Universitätsmedizin Berlin A composition comprising a pharmacologically active agent coupled to a target cell specific component, and a saponin
JP2008510829A (ja) * 2004-08-25 2008-04-10 ザ リージェンツ オブ ザ ユニバーシティ オブ ミシガン デンドリマーに基づく組成物およびそれらの使用法
BRPI0619056A2 (pt) 2005-11-28 2011-09-20 Genmab As anticorpo monovalente, método para preparar e produzir um antcorpo monovalente, construção de ácido nucleico, célula hospedeira, imunoconjugado, uso de um anticorpo monovalente, e, composição farmacêutica
AU2007322424B2 (en) * 2006-11-20 2013-05-16 Duecom Use of lipid containing particles comprising quillaja saponins for the treatment of cancer
EP2282752A4 (en) * 2008-03-26 2011-06-15 Ca Nat Research Council SAPONIN EXTRACT FROM SAPONARIA SPP AND USES THEREOF
ES2710463T3 (es) * 2008-04-11 2019-04-25 Cedars Sinai Medical Center Acido poli(beta málico) con tripéptido colgante Leu-Leu-Leu para la administración eficaz del fármaco citoplasmático
WO2011054811A1 (en) * 2009-11-03 2011-05-12 Santaris Pharma A/S Rna antagonists targeting hsp27 combination therapy
US9127088B2 (en) * 2010-05-13 2015-09-08 Indiana University Research And Technology Corporation Glucagon superfamily peptides exhibiting nuclear hormone receptor activity
US9125835B2 (en) * 2010-11-12 2015-09-08 Rutgers, The State University Of New Jersey Synergistic combinations to reduce particle dose for targeted treatment of cancer and its metastases
CN102652836A (zh) * 2011-03-03 2012-09-05 华中科技大学同济医学院附属协和医院 靶向释药的抗癌蛋白质或多肽聚合物前药及其制备方法
BR112014014763A8 (pt) * 2011-12-14 2017-07-04 Seattle Genetics Inc novos conjugados de anticorpo-medicamento (adcs) e uso dos mesmos
WO2014082065A1 (en) * 2012-11-26 2014-05-30 President And Fellows Of Harvard College Trioxacarcins, trioxacarcin-antibody conjugates, and uses thereof
WO2014163558A1 (en) * 2013-04-01 2014-10-09 Moreinx Ab Nanoparticles, composed of sterol and saponin from quillaja saponaria molina process for preparation and use thereof as carrier for amphipatic of hydrphobic molecules in fields of medicine including cancer treatment and food related compounds
GB201400994D0 (en) * 2014-01-21 2014-03-05 Ucl Business Plc Drug delivery system
CN106892957B (zh) * 2015-12-17 2018-11-16 广州中医药大学 一种齐墩果烷型三萜皂苷类化合物及其制备方法和应用
CA3027103A1 (en) * 2016-06-08 2017-12-14 Abbvie Inc. Anti-b7-h3 antibodies and antibody drug conjugates
EP4223316A3 (en) * 2018-12-21 2023-10-04 Sapreme Technologies B.V. Improved antibody-oligonucleotide conjugate
WO2022055352A1 (en) * 2020-09-10 2022-03-17 Sapreme Technologies B.V. Semicarbazone-based saponin conjugate

Also Published As

Publication number Publication date
DK4015003T3 (da) 2023-06-12
KR20210110323A (ko) 2021-09-07
AU2019411289A1 (en) 2021-08-12
IL284279A (en) 2021-08-31
BR112021012222A8 (pt) 2023-04-25
WO2020126609A3 (en) 2020-08-13
EP3897742A2 (en) 2021-10-27
AU2019411278A1 (en) 2021-08-12
BR112021012222A2 (ja) 2021-11-30
EP3773737A1 (en) 2021-02-17
AU2019408811A2 (en) 2021-09-02
EP3897741A1 (en) 2021-10-27
US20220054643A1 (en) 2022-02-24
MX2021007449A (es) 2021-12-10
IL284280A (en) 2021-08-31
CN113474009A (zh) 2021-10-01
CN113747923A (zh) 2021-12-03
SG11202106574UA (en) 2021-07-29
EP3897743A1 (en) 2021-10-27
IL284272A (en) 2021-08-31
JP2022516044A (ja) 2022-02-24
AU2019411289A2 (en) 2021-08-26
BR112021012225A2 (ja) 2021-11-30
EP3897739A1 (en) 2021-10-27
EP3897735A1 (en) 2021-10-27
EP3773737B1 (en) 2021-09-01
AU2019408811A1 (en) 2021-08-12
EP4223316A3 (en) 2023-10-04
WO2020126620A3 (en) 2020-07-23
US20220111066A1 (en) 2022-04-14
WO2020126610A9 (en) 2020-09-24
US20220218837A1 (en) 2022-07-14
KR20210117275A (ko) 2021-09-28
CN113474010A (zh) 2021-10-01
EP3915587A1 (en) 2021-12-01
WO2020126610A1 (en) 2020-06-25
JP2022515251A (ja) 2022-02-17
IL284265A (en) 2021-08-31
JP2022516045A (ja) 2022-02-24
WO2020126620A2 (en) 2020-06-25
SG11202106672SA (en) 2021-07-29
WO2020126626A1 (en) 2020-06-25
WO2020126064A1 (en) 2020-06-25
SG11202106603UA (en) 2021-07-29
AU2019400630A1 (en) 2021-08-12
CA3124129A1 (en) 2020-06-25
EP3856254A2 (en) 2021-08-04
CN113507941A (zh) 2021-10-15
CA3124151A1 (en) 2020-06-25
CA3124406A1 (en) 2020-06-25
US20220072149A1 (en) 2022-03-10
DK3897738T3 (da) 2024-02-19
SG11202106606RA (en) 2021-07-29
IL284273A (en) 2021-08-31
AU2019407234A1 (en) 2021-08-19
AU2019411276A1 (en) 2021-08-12
KR20210117274A (ko) 2021-09-28
IL284274A (en) 2021-08-31
WO2020126627A1 (en) 2020-06-25
BR112021012225A8 (pt) 2023-04-25
WO2020126604A1 (en) 2020-06-25
SG11202106664PA (en) 2021-07-29
KR20210119976A (ko) 2021-10-06
EP4015003B1 (en) 2023-05-10
SG11202106572QA (en) 2021-07-29
SG11202106575YA (en) 2021-07-29
WO2020126600A1 (en) 2020-06-25
WO2020126609A2 (en) 2020-06-25
US20230173085A1 (en) 2023-06-08
CA3124064A1 (en) 2020-06-25
SG11202106612QA (en) 2021-07-29
EP3897738A1 (en) 2021-10-27
EP4241847A3 (en) 2023-10-04
KR20210119978A (ko) 2021-10-06
JP2022515250A (ja) 2022-02-17
EP4015003A1 (en) 2022-06-22
KR20210107073A (ko) 2021-08-31
EP3808379A1 (en) 2021-04-21
CN113453722A (zh) 2021-09-28
DK3773737T3 (da) 2021-10-04
CN113498350A (zh) 2021-10-12
KR20210117276A (ko) 2021-09-28
AU2019407234A2 (en) 2021-08-26
EP3897739B1 (en) 2024-04-17
PT4015003T (pt) 2023-07-17
EP4223316A2 (en) 2023-08-09
ES2952017T3 (es) 2023-10-26
CA3124065A1 (en) 2020-06-25
EP3897738B1 (en) 2024-01-24
ES2899612T3 (es) 2022-03-14
CA3123978A1 (en) 2020-06-25
KR20210119979A (ko) 2021-10-06
MX2021007296A (es) 2021-12-10
IL284278A (en) 2021-08-31
EP4241847A2 (en) 2023-09-13
AU2019400630A2 (en) 2021-08-26
JP2022515797A (ja) 2022-02-22
JP2022516043A (ja) 2022-02-24
IL284276A (en) 2021-08-31
US20220313834A1 (en) 2022-10-06
EP4374927A2 (en) 2024-05-29

Similar Documents

Publication Publication Date Title
EP3897739B1 (en) Saponin conjugates
US20230248845A1 (en) Combination of an antibody-drug conjugate and an antibody-saponin conjugate

Legal Events

Date Code Title Description
AS Assignment

Owner name: CHARITE - UNIVERSITAETSMEDIZIN BERLIN, GERMANY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:POSTEL, RUBEN;FUCHS, HENDRIK;SIGNING DATES FROM 20210616 TO 20210628;REEL/FRAME:056847/0577

Owner name: SAPREME TECHNOLOGIES B.V., NETHERLANDS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:POSTEL, RUBEN;FUCHS, HENDRIK;SIGNING DATES FROM 20210616 TO 20210628;REEL/FRAME:056847/0577

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

AS Assignment

Owner name: SAPREME TECHNOLOGIES B.V., NETHERLANDS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:CHARITE - UNIVERSITAETSMEDIZIN BERLIN;REEL/FRAME:061070/0229

Effective date: 20220809