WO2024028282A1 - Modification spécifique d'un site par une navette bhe d'entités à base d'anticorps pour traverser la barrière hémato-encéphalique - Google Patents

Modification spécifique d'un site par une navette bhe d'entités à base d'anticorps pour traverser la barrière hémato-encéphalique Download PDF

Info

Publication number
WO2024028282A1
WO2024028282A1 PCT/EP2023/071199 EP2023071199W WO2024028282A1 WO 2024028282 A1 WO2024028282 A1 WO 2024028282A1 EP 2023071199 W EP2023071199 W EP 2023071199W WO 2024028282 A1 WO2024028282 A1 WO 2024028282A1
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
group
bond
shuttle
disulfide
Prior art date
Application number
PCT/EP2023/071199
Other languages
English (en)
Inventor
Macarena SÁNCHEZ NAVARRO
Meritxell TEIXIDÓ TURÀ
Ernest GIRALT LLEDÓ
Original Assignee
Gate2Brain, S.L.
Universitat De Barcelona
Fundació Institut De Recerca Biomèdica (Irb Barcelona)
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 Gate2Brain, S.L., Universitat De Barcelona, Fundació Institut De Recerca Biomèdica (Irb Barcelona) filed Critical Gate2Brain, S.L.
Publication of WO2024028282A1 publication Critical patent/WO2024028282A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/44Antibodies bound to carriers
    • 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
    • A61K47/68Medicinal 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
    • A61K47/68037Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates the drug being a camptothecin [CPT] or derivatives
    • 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
    • A61K47/6889Conjugates 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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
    • C07K16/22Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against growth factors ; against growth regulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2863Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for growth factors, growth regulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/32Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against translation products of oncogenes
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/04Linear peptides containing only normal peptide links
    • C07K7/08Linear peptides containing only normal peptide links having 12 to 20 amino acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/33Fusion polypeptide fusions for targeting to specific cell types, e.g. tissue specific targeting, targeting of a bacterial subspecies

Definitions

  • the present invention relates to the field of conjugates with antibodies, and its use to the treatment and diagnosis of diseases that need that antibody cross the blood-brain barrier.
  • Monoclonal antibodies have revolutionized the treatment of several diseases, especially in oncology.
  • Antibody-based treatments target leukemia and solid tumors in many organs.
  • brain tumors remain practically intractable with biotherapeutics and most small molecules.
  • BBB blood-brain barrier
  • BTB blood-tumor barrier
  • BM Brain metastasis
  • BM Brain metastasis
  • HER-2 human epidermal growth factor receptor
  • mAbs to treat BM or primary brain tumors remains elusive, mainly due to the lack of antibody penetration across the BBB and BTB.
  • WO2015/001015A1 describes specific apamin-derived peptides comprising the KAPETAL fragment, which are useful as shuttles as they have the ability to cross the BBB and are capable of facilitating the transport into the brain of drugs or other diagnostically useful substances which by themselves cannot cross the BBB.
  • Example 22 (Cetuximab-V3-Ap3-NH2) and Example 23: (Cetuximab-V2-Ap5a-NH2), the AP5 peptide corresponds to MiniAp4 peptide, a cyclic peptidomimetic derived from bee venom which displays high resistance to proteolysis and negligible toxicity and immunogenicity, being V2and V 3 as follows:
  • V3 linker bonds 1 and 2 are connected to the side chain of a lysine of the antibody.
  • all the previous methods based on modifications of lysine such as the ones disclosed in the previous documents do not allow to control where the peptides are located when they are incorporated in a monoclonal antibody.
  • the present inventors have developed a conjugate of a mAb antibody and selected peptide shuttles linked to the antibody through a bromomaleimide linker at specific sites of the antibody, and which effectively crosses the blood-brain barrier.
  • the linker used in the present invention allows a peptide incorporation strategy in which the incorporation of the shuttle peptides into the monoclonal antibody is controlled and allows knowing where they are located.
  • This strategy is based on incorporating the peptides through the -SH groups of the reduced antibody.
  • this strategy may be applied to any monoclonal antibody providing homogeneous conjugates by site-specific modification of the antibody, a highly desirable feature for a pharmacological product, enhancing therapeutic index and facilitating product manufacturing and profiling.
  • Trastuzumab (Herceptin®) is an FDA-approved antibody against HER2, which is widely used in clinics to treat breast cancer.
  • the conjugation has been achieved by reducing interchain disulfide bridges and rebridging them using dibromomaleimide, which enables high control over the number of peptides anchored to every antibody molecule.
  • the conjugate shows enhanced transport across the blood-brain barrier in an in vitro cell model with respect to trastuzumab alone or with respect to a conjugate of trastuzumab with the Angiopep-2 peptide disclosed by Anthony Regina et al., in “ANG4043, a novel brain-penetrant Peptide-mAb conjugate, is efficacious against HER2-positive Intracranial Tumors in Mice”, mct.aacrjournals.org.
  • a first aspect of the present invention relates to an antibody shuttle conjugate of formula (I) or a pharmaceutically acceptable salt thereof, which comprises from 1-6 peptides of formula P inserted in the disulfide bonds of the antibody in the form of a -P-(W)s-Y and joined to the sulfides through a linker -[(l_i)-(l_2)- (L3)m-; wherein:
  • Z represents the structure of a monoclonal antibody or a monoclonal antibody fragment thereof;
  • the disulfide bonds are any disulfide bond initially present in the antibody and that is capable of structurally retaining sulfide bonds in the structure upon reduction conditions, the disulfide bond being selected from the group consisting of a naturally present interchain disulfide bond of the antibody, a naturally present intrachain disulfide bond of the antibody, and a disulfide bond introduced in the antibody by genetic engineering;
  • Li is a linker selected from the group consisting of Li a and L ; q is an integer from 1 to 6;
  • L2 is a biradical composed from 2 to 8 biradicals selected from the group consisting of LA, LB, LC and has the formula -LA-(LB) U -LC-
  • D is a substance attached to the linker L3 selected from a biologically active substance, a substance for use in a diagnostic method; and a radioligand for radiotherapy;
  • P is a biradical of a single peptide, equal or different, which is selected from the group consisting of: (a) a peptide which comprises the amino acid sequence X1KAPETALX2 with an intrapeptide bond between the Xi and X2 which is an amide bond; wherein Xi is selected from the group consisting of Dap (2,3-diaminopropionic acid) and Dab (2,4- diaminobutanoic acid); and X2 is selected from the group consisting of D (aspartic acid) and E (glutamic acid); i.e.
  • SEQ ID N0:1 X1KAPETALX2
  • a peptide having 12-20 amino acids residues in length having at least an intrapeptide bond which is a disulfide or diselenide bond, and comprises an amino acid sequence which is: X3KAPETALX4AAA; having at least an intrapeptide disulfide or diselenide bond between X3 and X4, wherein X3 and X4 are equal and are selected from the group consisting of C (cysteines), Sec (selenocysteines), and Pen (penicillamines); i.e.
  • peptide which comprises the amino acid sequence XI KAPETALX 2 wherein Xi is selected from the group consisting of Dap and Dab; and X 2 is selected from the group consisting of D (aspartic acid) and E (glutamic acid) (SEQ ID NO:7) being a linear peptide;
  • S indicates sulfide
  • a second aspect of the present invention relates to a process for preparing an antibody shuttle conjugate as defined above, comprising: a) reducing the disulfide bridges of the antibody; b) rebridging the disulfide bridges by reacting the -SH groups of the antibody with a dibromomaleimide-peptide of formula (II), and c) optionally, carrying out a hydrolysis; wherein q; L2, L3, D, P, m, W, s and Y are as defined in the antibody shuttle conjugate of formula (I);
  • a third aspect of the present invention relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a therapeutically effective amount of the antibody shuttle conjugate of the present invention, together with appropriate amounts of pharmaceutically acceptable carriers or excipients.
  • a four aspect of the present invention relates to the antibody shuttle conjugate of the present invention as defined above, for use as a medicament.
  • a fifth aspect of the present invention relates to the antibody shuttle conjugate of the present invention as defined above for use in the treatment of Central Nervous System (CNS) disorders in a mammal, including a human.
  • CNS Central Nervous System
  • a sixth aspect of the present invention relates to the antibody shuttle conjugate of the present invention as defined above for use as a diagnostic agent.
  • FIG 1. H NMR of DBM (3,4-dibromo-2,5-dioxo-2,5-dihydro-1 H-pyrrol-1-yl)acetic acid).
  • FIG. 13 C NMR of DBM (3,4-dibromo-2,5-dioxo-2,5-dihydro-1 H-pyrrol-1-yl)acetic acid).
  • Example 1 FIG 3. LIPLC traces and MS spectra of the Comparative Example 2 and Example 3. LIPLC chromatograms are recorded at 220 nm in a 2-min linear gradient from 0 to 100% of MeCN (0.036% TFA) in H 2 O (0.045% TFA).
  • FIG 4. The site-specific Trastuzumab-DBM-TTDS-SEQ ID NO: 1 (Example 4) and Trastuzumab-DBM-TTDS-SEQ ID NO: 15 (Comparative Example 3) were generated and characterized, a) ASC synthetic scheme; b) Mass characterization of Trastuzumab-DBM- TTDS-SEQ ID NO: 15 (also named Tz-Ang2, Comparative Example 3) (top) and Trastuzumab-DBM-TTDS-SEQ ID NO: 1 (also named Tz-MiniAp4, Example 4) (down) by LCT-Premier. Deconvoluted spectra are shown.
  • FIG. Mass characterization of Anti- Trastuzumab (Anti- Idiotype) Alexa Fluor® 647- conjugated Antibody (AF647 modified Trastuzumab (also named Tz) (a), Trastuzumab- DBM-TTDS-SEQ ID NO: 15 (also named Tz-Ang2, Comparative Example 3) (b) and Trastuzumab-DBM-TTDS-SEQ ID NO: 1 (also named Tz-MiniAp4, Example 4) (c) by LCT-Premier. Antibodies are deglycosylated. Deconvoluted spectra are shown.
  • FIG. Mass characterization of Trastuzumab (also named Tz) (a), Trastuzumab-DBM- TTDS-SEQ ID NO: 15 (also named Tz-Ang2, Comparative Example 3) (b) and Trastuzumab-DBM-TTDS-SEQ ID NO: 1 (also named Tz-MiniAp4, Example 4) (c) after immunoprecipitation of acceptor well from HBBBCMTA by LCT-Premier. The raw data (top) and the deconvoluted spectra (down) are shown.
  • FIG 7. Stability studies in mouse serum of the BBB-shuttles peptides present in Example 4 and Comparative Example 3.
  • BT474 and SKBR3 cells were aliquoted and incubated with Tz, Tz-Ang2 or Tz-MiniAp4 at 4°C for 2 hours.
  • Anti-human dylight 650 was used to detect I g 1 .
  • FIG 11. Cell cycle arrest analysis of Tz, Tz-DBM-TTDS-SEQ ID NO: 1 (Example 4) and Tz-DBM-TTDS-SEQ ID NO: 15 (Comparative Example 3) treated cells.
  • SKBR3, BT-474 or MDA-MB-231 cells were serum starved and stimulated with Tz, Tz-DBM-TTDS-SEQ ID NO: 1 (Example 4) and Tz-DBM-TTDS-SEQ ID NO: 15 (Comparative Example 3) (100 nM) for 5 days.
  • amino acids cited herein are L-amino acids.
  • the 1-letter code and the 3-letter code have been used indistinctly.
  • diaminopropionic acid (Dap) diaminopropionic acid (Dap), Diaminobutiric (Dab), Selenocystein (Sec) and Penicillamine (Pen).
  • Dap diaminopropionic acid
  • Dab Diaminobutiric
  • Dbu Dbu
  • an antibody shuttle conjugate of formula (I) or a pharmaceutically acceptable salt thereof which comprises from 1-6 peptides of formula P inserted in the disulfide bonds of the antibody in the form of a -P-(W)s-Y and joined to the sulfides through a linker -[(Li )-(l_2)- (l_3)m- ; wherein, Z, Li , L2, L3, D, m, P, W, S, Y and n have the meaning mentioned above.
  • the antibody shuttle conjugate of the present invention is that where the disulfide bonds initially present in the antibody are capable of structurally retaining the sulfide bonds in the structure upon reduction conditions and of forming between them new bridges by reaction of the sulfide groups of the reduced disulfide bonds of the antibody with the linker Li, to incorporate the biradical below between the sulfide groups of the antibody.
  • the antibody shuttle conjugate of formula (I) or a pharmaceutically acceptable salt thereof may be in the form of a formulation comprising the antibody or their salts and excipients.
  • appropriate excipients for Bevacizumab (Avastin) are trehalose dihydrate, sodium phosphate, polysorbate 20, and water for injectable preparations
  • appropriate excipients for Pertuzumab (Perjeta) are Glacial acetic acid, L-histidine, Sucrose, Polysorbate 20, and water for injectable preparations
  • appropriate excipients for are Cetuximab (Erbitux): Sodium Chloride, Glycine, Polysorbate 80, Citric Acid Monohydrate, Sodium Hydroxide, and water for Injection Preparations
  • appropriate excipients for trastuzumab (Herceptin) are for instance: L-histidine hydrochloride monohydrate, L- histidine, a,a-trehalose, or polysorbate 20 dihydrate.
  • the antibody shuttle conjugate of formula (I) or a pharmaceutically acceptable salt thereof is that wherein the linker L2 is a biradical selected from the group consisting of:
  • the antibody shuttle conjugate of formula (I) or a pharmaceutically acceptable salt thereof is that wherein n is an integer from 1 to 4.
  • the antibody shuttle conjugate of formula (I) or a pharmaceutically acceptable salt thereof is that wherein n is 4, i.e. , it has four peptides incorporated within the antibody structure.
  • all the peptides are equal.
  • the disulfide bond of the antibody is an interchain bond. In another particular embodiment, either alone or in combination with any of the embodiments of the invention, the disulfide bond of the antibody is an intrachain bond. In another particular embodiment, either alone or in combination with any of the embodiments of the invention, the disulfide bond of the antibody is a disulfide bond formed by genetic engineering.
  • the antibody shuttle conjugate is that wherein P is a biradical of a peptide selected from the group consisting of:
  • TIHCO ⁇ (b) a peptide having 9-20 ammo acids residues in length having at least an intrapeptide bond which is a disulfide bond, and comprises an amino acid sequence which is: CKAPETALCAAA having at least an intrapeptide disulfide bond between cysteines 1 and 9, that is SEQ ID NO:9: CKAPETALCAAA
  • a peptide having 9-11 amino acids residues in length having at least an intrapeptide bond which is a disulfide bond and consists of an amino acid sequence selected from the group consisting of CKAPETALC; CKAPETALCA; and CKAPETALCAA having at least an intrapeptide disulfide bond between cysteines 1 and 9, that are
  • SEQ ID NO: 10 CKAPETALC
  • SEQ ID NO: 12 CKAPETALCAA
  • the lines between two amino acids of the sequences above or below represent the intrapeptide bond between the side chains of the two amino acids.
  • the lines between two amino acids of the sequences above or below represent the intrapeptide bond between the side chains of the two amino acids.
  • the antibody shuttle conjugate is that wherein, P is a biradical of a peptide selected from the group consisting of: (a) the peptide having the amino acid sequence DapKAPETALD with an intrapeptide bond between the Dap and D which is an amide bond (SEQ ID NO:7); (b) the peptide having the amino acid sequence CKAPETALC having at least an intrapeptide disulfide bond between cysteines in position 1 and 9 (SEQ ID NO: 10; and (c) the peptide having the amino acid sequence DapKAPETALD (SEQ ID NO:14).
  • P is a biradical of a peptide selected from the group consisting of: (a) the peptide having the amino acid sequence DapKAPETALD with an intrapeptide bond between the Dap and D which is an amide bond (SEQ ID NO:7); (b) the peptide having the amino acid sequence CKAPETALC having at least an intrapeptide disulfide bond between cysteines in position 1 and 9 (
  • the antibody shuttle conjugate is that wherein P is a biradical of the peptide DapKAPETALD with an intrapeptide bond between the Dap and D which is an amide bond (SEQ ID NO:7).
  • the antibody shuttle conjugate is that wherein P is a biradical of a peptide having one intrapeptide bond. In another particular embodiment, the antibody shuttle conjugate is that wherein P is a biradical of a peptide having two intrapeptide bonds.
  • Antibodies generally consist of two heavy chains (HCs) and two light chains (LCs) folded into constant and variable domains, although some antibodies, such as camelid antibodies, contain only 2 heavy chains. They can occur individually as monomers (e.g. IgG), or in multimers of two (e.g. IgA) to five units (e.g. IgM). They have disulfide intrachain bonds.
  • the antibodies for the purposes of the present invention may be chimeric, humanized or fully human antibodies. In the sense of the present invention the antibodies can be natural antibodies or recombinant antibodies.
  • the antibody is a therapeutical antibody.
  • the antibody is for diagnosis.
  • the antibody is an antibody-drug conjugate (ADC).
  • the antibody can also be an antibody fragment, provided it contains at least one disulfide bond and provided that it maintains the function of the antibody from which it is derived In particular, its therapeutic or diagnostic activity.
  • the antibody may be an antibody fragment such as Fab, scFV, (Fab)2, diabody, triabody, tetrabody, minibody, or nanobody.
  • disulfide bonds can be incorporated for instance into the antibodies or fragments thereof by incorporating cys by mutagenizing a nucleic acid sequence of an antibody and replacing one or more amino acid residues by cysteines to encode the cysteine engineered antibody (see CA2957354A1).
  • Antibody fragments can offer several advantages compared to full-length antibodies.
  • the antibody shuttle conjugate is that wherein the antibody comprises a heavy chain constant domain of the type of IgA, IgD, I g E, IgG or IgM.
  • Antibodies of the IgA type can be divided into two isotopes: I gA1 or lgA2.
  • Antibodies of the IgG class can be divided into four isotypes: lgG1 , lgG2, lgG3 and lgG4. classes of antibodies are encompassed within the scope of the present invention.
  • the antibody shuttle conjugate is that wherein the antibody comprises a light chain constant domain, e.g., of the type of kappa or lambda.
  • the antibody shuttle conjugate is that wherein the antibody or antibody fragment according to the present invention comprises:(a) an immunoglobulin constant region; (b) an IgG 1 constant region; or (c) a human IgG 1 constant region.
  • I gG 1 is preferred as it is the most widely used isotype for anticancer mAbs, as well as the most effective IgG isotype in mediating ADCC (antibody-dependent cellular cytotoxicity).
  • the antibody is a monoclonal chimeric, humanized, or fully human antibody. Antibody humanization allows reducing immunogenicity by decreasing the mouse content of monoclonal antibodies.
  • Common techniques developed for the production of fully human monoclonal antibodies can be used such as phage display, where a library of human antibodies is expressed on the surface of phage and subsequently selected and amplified in E. coli, and transgenic mice expressing a human antibody repertoire.
  • the antibody shuttle conjugate is that wherein the antibody is: (a) is multispecific.
  • Multispecific antibodies is a class of engineered antibody and antibody-like proteins that combine multiple specific antigen binding elements in a single construct.
  • the antibody shuttle conjugate is that wherein the antibody is: heterodimeric bispecific antibodies which are traditional IgG molecule with one arm targeting one antigen and the other targeting a second antigen.
  • the antibody shuttle conjugate is that wherein the antibody is a bispecific antibody fusion which is a non-standard IgG molecule. IgG is elongated at its N-terminus on the corresponding heavy and light chains by an additional variable domain of a second antibody.
  • the antibody shuttle conjugate is that wherein the antibody is a trispecific antibody which is also a non-standard IgG molecule.
  • the same technologies used to generate bispecific antibodies can also be combined to generate trispecific antibodies with varying valences.
  • the antibody shuttle conjugate is that wherein the antibody is fusion of a scFv to an IgG via the attachment of the scFv to the N or C-terminus of the heavy or light chain.
  • the antibody shuttle conjugate of the invention is that wherein the antibody is selected from the group consisting of Trastuzumab, Bevacizumab, Cetuximab, Pertuzumab, Aducanumab, Bapineuzumab, Nimotuzumab, and Necitumumab.
  • the antibody shuttle conjugate of the invention is that where the antibody is selected from the group consisting of Trastuzumab, Bevacizumab, Cetuximab and Pertuzumab.
  • the antibody shuttle conjugate of the invention is that where the antibody is Trastuzumab.
  • Trastuzumab (Herceptin®), an FDA-approved antibody against HER2, which is widely used in clinics to treat breast cancer.
  • the antibody shuttle conjugate of the invention is that which is Trastuzumab-DBM-TTDS-Dap-Lys-Ala- Pro-Glu-Thr-Ala-Leu-Asp-NH2 (Tz-DBM-TTDS-SEQ ID NO: 1) with an amide bond between Dap side-chain amino group and Asp side-chain carboxylic acid.
  • the antibody shuttle conjugate of the invention is that where the antibody is Cetuximab.
  • Cetuximab Erbitux®
  • an FDA-approved antibody against EGFR which is widely used in clinics to treat colorectal cancer and head and neck cancer.
  • the antibody shuttle conjugate of the invention is that which is Cetuximab-DBM-TTDS-Dap-Lys-Ala-Pro-Glu-Thr-Ala-Leu-Asp-NH2 (Cx-DBM-TTDS- SEQ ID NO: 1) with an amide bond between Dap side-chain amino group and Asp sidechain carboxylic acid.
  • DBM corresponds to linker Li a .
  • TTDS corresponds to linker l_2a.
  • DBM Lia has the formula:
  • the antibody shuttle conjugate of the invention is that where the antibody is Bevacizumab.
  • Bevacizumab (Avastin ®), an FDA-approved antibody against VEGF-A, which is widely used in clinics to treat colon cancer, lung cancer, glioblastoma and renal-cell carcinoma.
  • the antibody shuttle conjugate of the invention is that which is Bevacizumab-DBM-TTDS-Dap-Lys-Ala-Pro-Glu- Thr-Ala-Leu-Asp-NH2 (Bv-DBM-TTDS-SEQ ID NO: 1) with an amide bond between Dap side-chain amino group and Asp side-chain carboxylic acid.
  • the antibody shuttle conjugate of the invention is that where the antibody is Pertuzumab.
  • Pertuzumab Perjeta ®
  • an FDA-approved antibody against HER2 which is widely used in clinics to treat breast cancer.
  • the antibody shuttle conjugate of the invention is that which is Pertuzumab-DBM-TTDS- Dap-Lys-Ala-Pro-Glu-Thr-Ala-Leu-Asp-NH2 (Pt-DBM-TTDS-SEQ ID NO: 1) with an amide bond between Dap side-chain amino group and Asp side-chain carboxylic acid.
  • the antibody shuttle conjugate of the invention is that where the antibody is Aducanumab, a lgG1 antibody against an epitope of the p-amyloid protein.
  • the antibody shuttle conjugate of the invention is that where the antibody is Bapineuzumab, an anti-Ap-amyloid lgG1 antibody. Both antibodies are useful for the treatment of Alzheimer disease.
  • the antibody shuttle conjugate of the invention is that where the antibody is Nimotuzumab, a lgG1 anti-EGFR antibody. In another particular embodiment, either alone or in combination with any of the embodiments of the invention, the antibody shuttle conjugate of the invention is that where the antibody is Necitumumab, IgG 1 anti-EGFR antibody. Both antibodies are useful for the treatment of brain metastases.
  • the formulation comprising the antibody shuttle conjugate of the invention is that where D is present, yielding to an antibody-drug shuttle conjugate, an antibodyradioligand shuttle conjugate, or an antibody-diagnostic agent shuttle conjugate.
  • the biologically active substance is a pharmaceutical active ingredient.
  • the antibody-drug shuttle conjugate is that where the antibody is cetuximab and D is SN38.
  • the antibody-drug shuttle conjugate according to the invention is that where the antibody is cetuximab, the drug is SN38, the peptide is MiniAp4, and the linker is TTDS-DBM.
  • D is biotin-Ytrium 90 or Biotin- Iodine 131 or 123. They can be joined through streptavidin to the antibody shuttle conjugate of the present invention, where the streptavidin is attached to a lateral chain of the amino acids of the biradical Lswhen present in the antibody shuttle conjugate of the present invention.
  • D is a contrast agent for a magnetic resonance imaging method (MRI), such as Gadolinium, in particular, Gd-DTPA (Magnevist®).
  • MRI magnetic resonance imaging method
  • Gadolinium may be complexed with DOTA which is attached to a lateral chain of the amino acids of the biradical Lswhen present in the antibody shuttle conjugate of the present invention or to an amino acid derivative by a feasible bond.
  • the antibody shuttle conjugates of the present invention may be prepared by a process comprising: a) reducing the disulfide bridges of the antibody; b) rebridging the disulfide bridges, i.e. forming new bridges, by reacting the -SH groups of the antibody with a dibromomaleimide-peptide (DBM-peptide) of formula (II), DBM-P-(W) S -(Y), in which the dibromomaleimide is attached to the peptide via the N-terminus of the peptide, wherein DBM, P, W, Y and s is as defined for the antibody shuttle conjugate of formula (I).
  • DBM-peptide dibromomaleimide-peptide
  • the binding of the DBM-peptide to the antibody is possible thanks to the reactivity of the cysteines forming the disulfide bridges. This method allows a high control of the number of peptides anchored to each antibody molecule.
  • the antibody is trastuzumab.
  • the antibody shuttle conjugate of the present invention may be defined by its preparation process. Accordingly, an antibody shuttle conjugate obtainable by the process defined above is also considered part of the present invention.
  • the antibody shuttle conjugate as defined above may be obtainable by reacting the hydrosulfide groups of the reduced disulfide bonds of the corresponding antibody or of a fragment thereof with a dibromomaleimide-peptide of formula (II) as defined above to rebridge the disulfide bond with the biradical below incorporated between the disulfide bridges of the antibody.
  • the antibody shuttle conjugate as defined above may be obtainable by a process comprising: a) reducing disulfide bridges of the antibody; b) rebridging the disulfide bridges by reacting the -SH groups of the antibody with a dibromomaleimide-peptide of formula (II), c) optionally, carrying out a hydrolysis; wherein q; L2, L3, D, P, m, W, s, and Y are as defined in the antibody shuttle conjugate of formula (I).
  • the disulfide bonds are interchain bonds. All the embodiments defined above for the conjugate of formula (I) as product per se are also embodiments of the process for its preparation.
  • the shuttle peptide used in the conjugates of the present invention can be generated wholly or partly by chemical synthesis.
  • the amino acids required for the preparation of compounds of formula (I) are commercially available.
  • the compounds of formula (I) can be prepared easily, for example by synthesis in liquid-phase or, preferably, by solid-phase peptide synthesis, for which there are a number of procedures published (see M. Amblard, et al., "Methods and protocols of modern solid-phase peptide synthesis. Molecular Biotechnology 2006, Vol. 33, p. 239-254).
  • the compounds of formula (I) can also be prepared by any combination of liquid-phase synthesis and/or solid-phase synthesis.
  • linker dibromomaleimide For example, by synthesizing the body of the peptide P through solid-phase synthesis and subsequently removing protecting groups in solution.
  • the binding to the linker dibromomaleimide can be performed in solid-phase or in solution.
  • the construction of the linker can also be prepared by any combination of liquid-phase synthesis and/or solidphase synthesis.
  • the linker used is 3,4-dibromo-2,5-dioxo-2,5-dihydro-1 H-pyrrol-1- yl)acetic acid or similar derivatives thereof.
  • the compound of formula (II) can be prepared by a process comprising: reacting a peptide derivatized with the linkers with 3,4-dibromo-2,5-dioxo-2,5-dihydro-1 H-pyrrol-1- yl)acetic acid by solid phase peptide synthesis or in solution to yield the DBM-peptide.
  • the compound of formula (II) with a linker L can be obtained by the corresponding compound of formula (II) with a linker Li a by hydrolysis.
  • the hydrolysis can be carried out in mild pH basic conditions,
  • the DBM peptide of formula (II) has the following formula:
  • DBM peptide of formula (II) has the following formula:
  • the antibody shuttle conjugate of the invention is that where the linker Li is L .
  • This conjugate may be prepared by hydrolysis in mild basic pH conditions from the corresponding antibody shuttle conjugate having as Li linker the linker Lla.
  • the antibody shuttle conjugates of the present invention may be in the form of a pharmaceutically acceptable salt thereof.
  • pharmaceutically acceptable salts used herein encompasses any salt formed from pharmaceutically acceptable non-toxic acids or bases including inorganic or organic acids or bases. There is no limitation regarding the salts, except that if used for therapeutic purposes, they must be pharmaceutically acceptable. As some of the compounds of formula (I) are basic compounds, salts may be prepared from pharmaceutically acceptable non-toxic acids, including inorganic and organic acids.
  • Such acids include, for instance, chlorohydric, acetic, benzenesulfonic, benzoic, camphor sulfonic, citric, ethansulfonic, fumaric, gluconic, glutamic, hydrobromic, hydrochloric, lactic, maleic, malic, mandelic, methane sulfonic, phosphoric, succinic, sulfuric, tartaric, p-toluensulfonic acid, and the like.
  • antibodies in the form of a salt according to the present invention are Bevacizumab (Avastin) in the form of a salt with trehalose dihydrate, sodium phosphate, polysorbate 20 salts; Pertuzumab (Perjeta) in the form of a salt with glacial acetic acid, L- histidine, Sucrose, or Polysorbate 20; cetuximab (Erbitux) in the form of a salt formed with sodium Chloride, glycine, polysorbate 80, citric acid monohydrate, or sodium hydroxide, trastuzumab (Herceptin) in the form of a salt with L-histidine hydrochloride monohydrate, L-histidine, a,a-trehalose, or polysorbate 20 dihydrate
  • compositions of formula (I) can be carried out by methods known in the art. For instance, they can be prepared from the conjugate, which contains a basic or acidic moiety, by conventional chemical methods. Generally, such salts are, for example, prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate pharmaceutically acceptable base or acid in water or in an organic solvent or in a mixture of them.
  • a pharmaceutical composition comprising a therapeutically effective amount of the antibody shuttle conjugate as defined above, together with appropriate amounts of pharmaceutically acceptable carriers or excipients is also part of the invention.
  • pharmaceutical composition refers to a mixture of a compound described herein with other chemical components, such as diluents or carriers.
  • the pharmaceutical composition facilitates administration of the compound to an organism.
  • pharmaceutically acceptable excipients or carriers refers to pharmaceutically acceptable material, composition or vehicle. Each component must be pharmaceutically acceptable in the sense of being compatible with the other ingredients of the pharmaceutical composition. It must be also suitable for use in contact with tissues or organs of humans and animals without excessive toxicity, irritation, allergic response, immunogenicity, or other problems or complications commensurate with a benefit/risk ratio.
  • therapeutically effective amount refers to the amount of a compound that, when administered, is sufficient to prevent development of, or alleviate to some extent, one or more of the symptoms of the disease which is addressed.
  • the particular dose of compound administered according to this invention will of course be determined by the particular circumstances surrounding the case, including the compound administered, the route of administration, the particular condition being treated, and the similar considerations.
  • pharmaceutically acceptable excipient refers to pharmaceutically acceptable materials, compositions or vehicles. Each component must be pharmaceutically acceptable in the sense of being compatible with the other ingredients of the pharmaceutical composition. It must also be suitable for use in contact with the tissue or organ of humans and non-human animals without excessive toxicity, irritation, allergic response, immunogenicity or other problems or complications commensurate with a reasonable benefit/risk ratio.
  • Suitable pharmaceutically acceptable excipients are solvents, dispersion media, diluents, or other liquid vehicles, dispersion or suspension aids, surface active agents, isotonic agents, thickening or emulsifying agents, preservatives, solid binders, lubricants and the like. Except insofar as any conventional excipient medium is incompatible with a substance or its derivatives, such as by producing any undesirable biological effect or otherwise interacting in a deleterious manner with any other component(s) of the pharmaceutical composition, its use is contemplated to be within the scope of this invention.
  • compositions of the invention will vary, depending upon the identity, size, and/or condition of the subject treated and further depending upon the route by which the composition is to be administered.
  • compositions of the present invention may be administered in parenteral form suitable for injection such as intravenous bolus injections, intravenous infusion, implantation into the body, oral, intratecal, or intranasal.
  • An antibody shuttle conjugate as defined above for use as a medicament is also part of the invention. This includes the antibody shuttle conjugates as well as the antibody-drug shuttle conjugates according to the present invention.
  • the term “medicament” as used herein is synonymous of a pharmaceutical or veterinary drug (also referred to as medicine, medication, or simply drug) use to cure, treat or prevent disease in animals, including humans, as widely accepted. Drugs are classified in various ways. One key distinction is between traditional small-molecule drugs, usually derived from chemical synthesis, and biopharmaceuticals, which include recombinant proteins, vaccines, blood products used therapeutically (such as I VIG), gene therapy, monoclonal antibodies and cell therapy (for instance, stem-cell therapies).
  • An antibody shuttle conjugate as defined above for use in the treatment of CNS disorders in a mammal, including a human is also part of the invention.
  • This aspect can also be formulated as the use antibody shuttle conjugate as defined above for the preparation of a medicament for the treatment of CNS disorders in a mammal, including a human.
  • the invention also relates to a method of treatment of a mammal, including a human, suffering from or being susceptible of suffering from a CNS disorder, said method comprising the administration to said patient of a therapeutically effective amount of antibody shuttle conjugate as defined above, together with pharmaceutically acceptable excipients or carriers.
  • the antibody shuttle conjugate is for use in a CNS disorder which is cancer.
  • the antibody shuttle conjugate of the present invention is for use in the treatment of a primary brain tumor.
  • the antibody shuttle conjugate of the present invention is for use in the treatment of brain metastasis (BM) is a major complication in several types of cancers, particularly in lung, melanoma, and breast cancers.
  • BM brain metastasis
  • the antibody shuttle conjugates of the present invention can be used in the same manner as other known chemotherapeutic agents, i.e. , in combination with other treatments, either simultaneously or sequentially, depending on the condition to be treated. They may be used alone or in combination with other suitable bioactive compounds.
  • the antibody shuttle conjugates of the present invention are for use in the treatment of cancer in a mammal, including a human in combination therapy with a chemotherapeutic agent.
  • the antibody-drug shuttle conjugates in combination therapy with a further chemotherapeutic agent can be used for instance for specific treatment regimens.
  • the antibody shuttle conjugate of the present invention is for use in combination with radioimmunotherapy.
  • Both the antibody shuttle conjugates as the antibody-radioligand shuttle conjugates according to the invention can be used.
  • the combination of radiation therapy and immunotherapy can be used to treat non-Hodgkin lymphoma and other types of cancer including brain tumors.
  • the antibodyradioligand shuttle conjugates according to the invention can bind to cancer cells and deliver a high dose of radiation directly to the tumor.
  • an antibody shuttle conjugate as defined above for use as a diagnostic agent is also part of the invention.
  • the antibody shuttle conjugate according to the invention is for use in a method of diagnostic of CNS disorders.
  • Cell culture-treated plates and flasks were purchased from Corning Costar. Culture medium was acquired from Lonza. XTT cell proliferation kit was purchased from Biological Industries (Cromwell, CT). Pierce® iodination beads were obtained from Pierce. Desalting columns (MiniTrap and MidiTrap G-25) were obtained from GE-Healthcare.
  • Microwave assisted solid-phase peptide synthesis was carried out on a Liberty Blue Automated Microwave Peptide Synthesizer using H-Rink amide Protide resin (loading: 0.56 mmols/g). Linear peptide was synthesized on a 0.5 mmol scale using a 5 excess of Fmoc-amino acid (0.2M) relative to the resin.
  • the test used for the identification and control of the synthesis was the following: A) Kaiser colorimetric assay for the detection of solid-phase bound primary amines (E. Kaiser et al., Anal. Biochem. 1970, vol. 34, pp. 595-598); B) p-nitro phenyl ester test for secondary amines bound to solid-phase (A. Madder et al., Eur. J. Org. Chem. 1999, pp. 2787-2791).
  • Protocols used durinq the manual is of the com The compounds were synthesized at a 100 pmol scale using the following methods and protocols: The resin for the manual synthesis was selected depending on the group Y: If Y is a OH, the terminus will be COOH, 2-chlorotrytil chloride resin will be chosen among others available. If Y is a NH2, the terminus will be CONH2, Rink amide MBHA resin will be chosen among others available.
  • Resin initial The resin was conditioned by washing with MeOH (5 x 30 s),
  • DMF (5 x 30 s), DCM (5 x 30 s), 1% TFA in DCM (1 x 30 s and 2 x 10 min), DCM (5 x 30 s), DMF (5 x 30 s), DCM (5 x 30s), 5 % DIEA in DCM (1 x 30 s, 2 x 10 min), DCM (5 x 30 s), DMF (5 x 30 s).
  • Fmoc removal Removal of the 9-fluorenylmethyloxycarbonyl (Fmoc) protecting group was done with 20% (v/v) piperidine in DMF using a treatment of 30 s followed by two treatments of 10 minutes each. Two additional treatments with DBU, toluene, piperidine, DMF (5%, 5%, 20%, 70%) (2 x 5 min) were performed to ensure the removal of the Fmoc group from secondary amines (proline).
  • the solvent was removed by suction and the resin washed with DMF (5 x 30 s) and DCM (5 x 30 s).
  • the coupling reaction was carried out twice under the same conditions. The extent of coupling was checked by the Kaiser colorimetric assay.
  • the Fmoc group was removed with a 20% solution of piperidine in DMF (v/v) using 30 s treatments and two treatments of 10 minutes. If the amino acid to be deprotected was a proline, an additional treatment with DBU, toluene, piperidine, DMF (5%, 5 %, 20%, 70%) (2 x 5 min) was applied to ensure the removal of the Fmoc group.
  • the Fmoc group was removed with a 20% solution of piperidine in DMF (v/v) using 30 s treatments and two treatments of 10 minutes. If the amino acid to be deprotected was a proline, an additional treatment with DBU, toluene, piperidine, DMF (5%, 5 %, 20%, 70%) (2 x 5 min) was applied to ensure the removal of the Fmoc group.
  • method 4 scale 100 pmols: The protected amino acid (3 eq 300 pmols)), DIG
  • amino acid to be deprotected was a proline
  • an additional treatment with DBU, toluene, piperidine, DMF (5%, 5 %, 20%, 70%) (2 x 5 min) was applied to ensure the removal of the Fmoc group.
  • Protocols used during the microwave assisted automated is: The compounds were synthesized at a 500 pmol scale using the following methods and protocols: The resin for the microwave assisted automated synthesis was selected depending on the group Y: If Y is a OH, the terminus will be COOH, CI-TCP(CI) ProTide resin will be chosen among others available. If Y is a NH2, the terminus will be CONH2, Rink amide ProTide resin will be chosen among others available.
  • Resin initial conditioning The resin was conditioned by washing with MeOH (5 x 30 s), DMF (5 x 30 s), DCM (5 x 30 s), 1% TFA in DCM (1 x 30 s and 2 x 10 min), DCM (5 x 30 s), DMF (5 x 30 s), DCM (5 x 30s), 5 % DIEA in DCM (1 x 30 s, 2 x 10 min), DCM (5 x 30 s), DMF (5 x 30 s).
  • Methods for the of the P disulfide or diselenide bond
  • the cyclization was performed in solution after the cleavage from the resin or on resin after the selective deprotection of the Cys, Sec or Pen residues.
  • the peptide was dissolved at a concentration of 100 pM in aqueous ammonium bicarbonate buffer 10 mM and pH 8.0.
  • the solution was intensely stirred for 24 h at room temperature. After that, the product was acidified with TFA to pH 2-3, frozen and lyophilized.
  • method 2 amide bond
  • the cyclization was performed on resin
  • the Fmoc group was removed with a 20% solution of piperidine in DMF (v/v) using a 30 s treatment and two treatments of 10 minutes.
  • the /V-terminal amine was protected with a Boc protecting group using BOC2O (3 eq, 1000 pmol, 56 mg) and DIEA (30 eq, 3000 pmol, 240 pL).
  • the OAI and Alloc groups were first deprotected by addition of tetrakis(triphenylphosphine)palladium(0) (0.1 eq, 10 pM, 12 mg), phenyl silane (10 eq, 1000 pmol, 123 mg) in DCM (3 x 15 min).
  • the resin was washed with 0.02 M sodium diethylcarbamate in DCM (3 x 5 min).
  • the /V-terminal amine was protected with a Boc protecting group using BOC2O (3 eq, 1000 pmol, 56 mg) and DIEA (30 eq, 3000 pmol, 240 pL).
  • the OAI and Alloc groups were first deprotected by addition of tetrakis(triphenylphosphine)palladium(0) (0.1 eq, 10 pM, 12 mg), phenyl silane (10 eq, 1000 pmol, 123 mg) in DCM (3 x 15 min).
  • the resin was washed with 0.02 M sodium diethyldithiocarbamate in DCM (3 x 5 min).
  • the /V-terminal amine was protected with a Boc protecting group using BOC2O (3 eq., 1000 pmols, 56 mg) and DIEA (30 eq., 3000 pmols, 240 pL).
  • the OAI and Alloc groups were first deprotected by addition of tetrakis(triphenylphosphine)palladium(0) (0.1 eq., 10 pM, 12 mg), phenyl silane (10 eq., 1000 pmols, 123 mg) in DCM (3 x 15 min).
  • the resin was washed with 0.02 M sodium diethyldithiocarbamate in DCM (3 x 5 min).
  • the disulfide bond can be accomplished by reaction of two thiols.
  • the thiols are dissolved at a concentration of 100 pM in aqueous ammonium bicarbonate buffer 10 mM and pH 8.0 and the solution is intensely stirred for 24 h at room temperature. After that, the solution is acidified with TFA to pH 2-3, frozen and lyophilized.
  • thioether bond is accomplished by reaction of an /V-terminal bromoacetyl group with a cysteine thiol as described in P.L.
  • ethers Ether formation can be accomplished by reaction of a hydroxyl group with a halo alkyl compound, preferably under basic conditions as described in Greene’s Protective Groups in Organic Synthesis, Fifth Edition. Peter G. M. Wuts. 2014 John Wiley & Sons, Inc. pp. 26-29.
  • esters can be accomplished by reaction of a hydroxyl group and a carboxylic acid using typical esterification conditions, such as Fischer esterification in the presence of acid catalysis, or alternatively with the reaction of the hydroxyl group with the corresponding acid chloride, as described in Greene’s Protective Groups in Organic Synthesis, Fifth Edition. Peter G. M. Wuts. 2014 John Wiley & Sons, Inc. pp. 271-279
  • thioesters The thioester bond is accomplished by reaction of a thiol with a carboxylic acid as described in M. Kazemi et al., Journal of Sulfur Chemistry, 2015, vol. 36:6, pp. 613-623. of Fmoc-TTDS-OH
  • the coupling of the Fmoc-TTDS-OH (2 equivalents) was achieved by either 2 cycles of 30 min of 4 equivalents of oxyma and 4 of N,N’- Diisopropylcarbodiimide (DIC) in DMF or 4 equivalents of DIC and 4 of HOBt in DCM during 2 h.
  • DIC Diisopropylcarbodiimide
  • NanoMate Advanced BioSciences, Ithaca, NY, USA aspirated the samples from a 384-well plate (protein Lobind) with disposable, conductive pipette tips, and infused the samples through the nanoESI Chip (which consists of 400 nozzles in a 20 x 20 array) towards the mass spectrometer.
  • Spray voltage was 1.70 kV and delivery pressure were 0.50 psi; the ionization was NanoESI, positive ionization.
  • NMR experiments were carried out on a Bruker Avance III 600 MHz spectrometer equipped with a TCI cryoprobe. Samples were prepared by dissolving compounds in 90% H2O/10% D2O at 3-4 mM and pH was adjusted to 2-3. Chemical shifts were referenced to internal sodium-3-(trimethylsilyl)propane sulfonate (DSS). Suppression of the water signal was achieved by excitation sculpting. Residue specific assignments were obtained from 2D total correlated spectroscopy (TOCSY) and correlation spectroscopy (COSY) experiments, while 2D nuclear Overhauser effect spectroscopy (NOESY) permitted sequence specific assignments. 13C resonances were assigned from 2D 1 H13C HSQC spectra.
  • TOCSY total correlated spectroscopy
  • COSY correlation spectroscopy
  • NOESY nuclear Overhauser effect spectroscopy
  • Amino Acid Analysis was performed to assess the amino acids present and the amount obtained for each peptide. To this end, ion exchange chromatographic analysis after acid hydrolysis was performed. The samples were hydrolyzed with 6 M HCI at 110 °C for 16 h. They were then evaporated to dryness at reduced pressure and dissolved in 20 mM aqueous HCI. Finally, the amino acids were modified using the AccQ Tag protocol from Waters and analyzed by ion exchange HPLC. For amino acid analysis, 100 pL of peptide (1 mg/mL) was added to 100 pL HCI (12 M) and 20 pL aminoquinolyl-N-hydroxysuccinimidyl carbamate derivatization reagent. This mixture was left overnight at 110°C. The liquid was fully evaporated and 200 pL 20 mM HCI was added before the Waters AccQ-Tag protocol was performed.
  • BBS Borate buffered saline
  • EDTA ethylenediaminetetraacetic acid
  • PBS Phosphate-buffered saline
  • TCEP Tris(2- carboxyethyl)phosphine hydrochloride
  • HHL heavy-heavy-light
  • HH heavy-heavy
  • HL heavylight
  • He heavy chain
  • Lc light chain
  • the column outlet was directly introduced into the electrospray ionization (ESI) source of a Waters LCT-Premier XE mass spectrometer (TOF).
  • Capillary voltage and cone voltage were set to 3000 V and 100 V respectively.
  • Desolvation and source temperatures were set to 350°C and 120°C, respectively.
  • Cone and desolvation gas flow were set to 50 L/h and 600 L/h, respectively.
  • the mass spectrometer acquired full MS scans (400-4000 m/z) working in positive polarity mode.
  • Output parameters were as follows: mass range 5000-70000 and resolution 1 Da/channel. A uniform Gaussian model was used, with the corresponding peak widths at half height.
  • DBM ((3,4-dibromo-2,5-dioxo-2,5-dihydro-1 H-pyrrol-1-yl)acetic acid) was prepared as described in Mol. Pharm. 12, 3986-3998 (2015).
  • glycine 0.294 mg, 3.91 mmol
  • acetic acid 20 mL
  • the reaction mixture was heated to 100 °C overnight.
  • the solution was concentrated under vacuum and purified by silica gel chromatography (eluent DCM/MeOH 9:1).
  • Example 2 Preparation of NH2-TTDS-Dap-Lys-Ala-Pro-Glu-Thr-Ala-Leu-Asp-NH2 (NH2- TTDS-SEQ ID NO: 1) with an amide bond between Dap side-chain amino group and Asp side-chain carboxylic acid.
  • the cyclization was performed on resin following cyclization method 2:
  • the Fmoc group was removed with a 20% solution of piperidine in DMF (v/v) using a 30 s treatment and two treatments of 10 minutes.
  • the /V-terminal amine was protected with a Boc protecting group using BOC2O (3 eq., 1000 pmols, 56 mg) and DIEA (30 eq., 3000 pmols, 240 pL).
  • the OAI and Alloc groups were first deprotected by addition of tetrakis(triphenylphosphine)palladium(0) (0.1 eq., 10 pM, 12 mg), phenyl silane (10 eq., 1000 pmols, 123 mg) in DCM (3 x 15 min). The resin was washed with 0.02 M sodium diethylcarbamate in DCM (3 x 5 min).
  • Example 3 Preparation of DBM-TTDS-Dap-Lys-Ala-Pro-Glu-Thr-Ala-Leu-Asp-NH2 (DBM- TTDS-SEQ ID NO: 1) with an amide bond between Dap side-chain amino group and Asp side-chain carboxylic acid.
  • NH2-TTDS-Dap-Lys-Ala-Pro-Glu-Thr-Ala-Leu-Asp-NH 2 (NH2-TTDS-SEQ ID NO: 1) prepared as in example 2, and using DBM prepared as in example 1, coupling method 2 was used to achieve compound DBM-TTDS-Dap-Lys-Ala-Pro-Glu-Thr-Ala-Leu- Asp-NH 2 (DBM-TTDS-SEQ ID NO: 1).
  • the coupling method 4 was applied using Fmoc-L-Tyr(tBu)-OH (137.7 mg).
  • the subsequent amino acids were coupled sequentially as follows using coupling method 4:
  • the cyclization was performed on resin following cyclization method 2:The Fmoc group was removed with a 20% solution of piperidine in DMF (v/v) using a 30 s treatment and two treatments of 10 minutes. The /V-terminal amine was protected with a Boc protecting group using BOC2O (3 eq., 1000 pmols, 56 mg) and DIEA (30 eq., 3000 pmols, 240 pL).
  • the OAI and Alloc groups were first deprotected by addition of tetrakis(triphenylphosphine)palladium(0) (0.1 eq., 10 pM, 12 mg), phenyl silane (10 eq., 1000 pmols, 123 mg) in DCM (3 x 15 min). The resin was washed with 0.02 M sodium diethylcarbamate in DCM (3 x 5 min).
  • Example 4 Preparation of Trastuzumab-DBM-TTDS-Dap-Lys-Ala-Pro-Glu-Thr-Ala-Leu- ASP-NH2 (Tz-DBM-TTDS-SEQ ID NO: 1) with an amide bond between Dap side-chain amino group and Asp side-chain carboxylic acid.
  • DBM-TTDS-Dap-Lys-Ala-Pro-Glu-Thr-Ala-Leu-Asp-NH2 (DBM-TTDS-SEQ ID NO: 1) prepared as in example 3 and Trastuzumab after general method for monoclonal antibody conditioning were conjugated using the following protocol to achieve the compound of formula Tz-DBM-TTDS-SEQ ID NO: 1.
  • Trastuzumab was obtained in its clinical form (Roche, lyophilized), resuspended in 7.2 mL sterile water and the buffer exchanged completely for BBS pH 8.5 with PD10 g25 columns (GE Healthcare), following General Methods for monoclonal antibody conditioning.
  • trastuzumab (111 pM, 4.9 mL, 544 nmol) was diluted with BBS (pH 8.5) to a final concentration of 22.9 pM.
  • BBS pH 8.5
  • a fresh solution of TCEP was added (10 mM, 332.2 pL, 3.26 pmol, 6 eq.) and the reaction was incubated at 37 °C for 2 h under mild agitation.
  • TCEP was removed by SEC using PD10 G25 columns with BBS as buffer, following manufacturer's instructions.
  • the DBM peptide (Example 3) in dry DMF (10 mM, 247 pL, 4.35 pmol, 8 eq.) was added to the reduced trastuzumab and the reaction was left at r.t. for 30 min. Afterwards, excess reagents were removed by SEC using PD10 G25 columns with PBS. The final conjugates were characterized by LC-MS confirming the integrity of the antibody after peptide conjugation.
  • DBM-TTDS- Thr-Phe-Phe-Tyr-Gly-Gly-Ser-Arg-Gly-Lys-Arg-Asn-Asn-Phe- Lys-Thr-Glu-Glu-Tyr-NH2 (DBM-TTDS-SEQ ID NO: 15) prepared as in Comparative Example 2 and Trastuzumab after general method for monoclonal antibody conditioning were conjugated using the following protocol to achieve the compound of formula Tz- DBM-TTDS-SEQ ID NO: 15.
  • Trastuzumab was obtained in its clinical form (Roche, lyophilized), resuspended in 7.2 mL sterile water and the buffer exchanged completely for BBS pH 8.5 with PD10 g25 columns (GE Healthcare), following General Methods for monoclonal antibody conditioning.
  • trastuzumab (111 pM, 4.9 mL, 544 nmol) was diluted with BBS (pH 8.5) to a final concentration of 22.9 pM.
  • BBS pH 8.5
  • a fresh solution of TCEP was added (10 mM, 332.2 pL, 3.26 pmol, 6 eq.) and the reaction was incubated at 37 °C for 2 h under mild agitation.
  • TCEP was removed by SEC using PD10 G25 columns with BBS as buffer, following manufacturer's instructions.
  • the DBM peptide (Comparative Example 2) in dry DMF (10 mM, 247 pL, 4.35 pmol, 8 eq.) was added to the reduced trastuzumab and the reaction was left at r.t. for 30 min. Afterwards, excess reagents were removed by SEC using PD10 G25 columns with PBS. The final conjugates were characterized by LC-MS confirming the integrity of the antibody after peptide conjugation.
  • Example 5 Preparation of Cetuximab-DBM-TTDS-Dap-Lys-Ala-Pro-Glu-Thr-Ala-Leu-Asp- NH2 (Cx-DBM-TTDS-SEQ ID NO: 1) with an amide bond between Dap side-chain amino and Asp side-chain carboxylic acid.
  • DBM-TTDS-Dap-Lys-Ala-Pro-Glu-Thr-Ala-Leu-Asp-NH2 (DBM-TTDS-SEQ ID NO: 1) prepared as in example 3 and Cetuximab after general method for monoclonal antibody conditioning were conjugated using the following protocol to achieve the compound of formula Cx-DBM-TTDS-SEQ ID NO: 1.
  • Cetuximab was obtained in its clinical form (SelleckChem, lyophilized), resuspended in sterile water and the buffer exchanged completely for BBS pH 8.5 with PD10 g25 columns (GE Healthcare), following General Methods for monoclonal antibody conditioning.
  • the conjugation protocol was adapted from Org. Biomol. Chem. 15, 2947-2952 (2017).
  • Cetuximab (1mg, 660 nmol) was diluted with BBS (pH 8.5) to a final concentration of 22.9 pM.
  • a fresh solution of DTT was added (10 mM, 4 pL, 2.64 pmol, 6 eq.) and the reaction was incubated at 37 °C for 2 h under mild agitation. DTT was removed by SEC using PD10 G25 columns with BBS as buffer, following manufacturer's instructions.
  • Example 6 Preparation of Bevacizumab-DBM-TTDS-Dap-Lys-Ala-Pro-Glu-Thr-Ala-Leu- ASP-NH2 (Bv-DBM-TTDS-SEQ ID NO: 1) with an amide bond between Dap side-chain amino group and Asp side-chain carboxylic acid.
  • DBM-TTDS-Dap-Lys-Ala-Pro-Glu-Thr-Ala-Leu-Asp-NH2 (DBM-TTDS-SEQ ID NO: 1) prepared as in example 3 and Bevacizumab after general method for monoclonal antibody conditioning were conjugated using the following protocol to achieve the compound of formula Bv-DBM-TTDS-SEQ ID NO: 1.
  • Bevacizumab was obtained in its clinical form (HSJD, lyophilized), resuspended in sterile water and the buffer exchanged completely for BBS pH 8.5 with PD10 g25 columns (GE Healthcare), following General Methods for monoclonal antibody conditioning.
  • the conjugation protocol was adapted from Org. Biomol. Chem. 15, 2947-2952 (2017).
  • Bevacizumab (1mg, 660 nmol) was diluted with BBS (pH 8.5) to a final concentration of 22.9 pM.
  • a fresh solution of DTT was added (10 mM, 4 pL, 2.64 pmol, 6 eq.) and the reaction was incubated at 37 °C for 2 h under mild agitation. DTT was removed by SEC using PD10 G25 columns with BBS as buffer, following manufacturer's instructions.
  • Example 7 Preparation of Pertuzumab-DBM-TTDS-Dap-Lys-Ala-Pro-Glu-Thr-Ala-Leu- ASP-NH2 (Pt-DBM-TTDS-SEQ ID NO: 1) with an amide bond between Dap side-chain amino group and Asp side-chain carboxylic acid.
  • the conjugation protocol was adapted from Org. Biomol. Chem. 15, 2947-2952 (2017).
  • Pertuzumab (1mg, 660 nmol) was diluted with BBS (pH 8.5) to a final concentration of 22.9 pM.
  • a fresh solution of DTT was added (10 mM, 4 pL, 2.64 pmol, 6 eq.) and the reaction was incubated at 37 °C for 2 h under mild agitation. DTT was removed by SEC using PD10 G25 columns with BBS as buffer, following manufacturer's instructions.
  • Example 8 Stability of the shuttle in mouse serum.
  • One of the major advantages of the shuttles of this invention is that unlike the vast majority of peptides composed exclusively of L-amino acids (which are rapidly metabolized by a series of enzymes present in the serum of the blood, thus limiting their therapeutic effects), these are made with D-amino acids, thus are not recognized by the metabolic enzymes present in the serum, thereby significantly increasing their half-life in serum.
  • Example 4 Regarding to the stability studies in mouse serum of the BBB-shuttles peptides present in Example 4 and Comparative Example 3, these were incubated at a concentration of 150 pM in buffer HBSS at 37°C, in the presence of 90% mouse serum. At a range of times, 50 pL aliquots were collected to which methanol was added in order to precipitate the serum proteins. The samples were centrifuged, filtered and analyzed by HPLC in order to determine the degree of degradation. FIG.
  • Example 7 shows stability studies in mouse serum of the BBB-shuttles peptides present in Example 4 and Comparative Example 3
  • Example 9 Binding of Tz, Tz-DBM-TTDS-SEQ ID NO: 1 (Example 4) and Tz-DBM-TTDS- SEQ ID NO: 15 (Comparative Example 3) to breast cancer cells.
  • Tz, Tz-DBM-TTDS-SEQ ID NO: 1 (Example 4) and Tz-DBM-TTDS-SEQ ID NO: 15 (Comparative Example 3) was performed with increasing concentrations in ice-cold PBS for 30 minutes at 4°C. Cells were then washed and incubated with an anti-human- Dylight 650 secondary antibody (Abeam pic) in ice-cold PBS for 30 minutes at 4°C. Cells were washed with ice-cold PBS and analyzed by flow cytometry (10,000 gated events per condition).
  • FIG. 8 shows binding of Tz, Tz-DBM-TTDS-SEQ ID NO: 1 (Example 4) and Tz- DBM-TTDS-SEQ ID NO: 15 (Comparative Example 3) to HER-2 overexpressing cells.
  • Example 10 Binding of Cx, Cx-DBM-TTDS-SEQ ID NO: 1 (Example 5) to breast cancer cells.
  • FIG. 9 shows binding of Cx, Cx-DBM-TTDS-SEQ ID NO: 1 (Example 5) to EGFR-positive MDA-MB-231 breast cancer cells.
  • Example 11 Binding of Pt, Pt-DBM-TTDS-SEQ ID NO: 1 (Example 7) to breast cancer cells.
  • FIG. 10 shows binding of Pt, Pt-DBM- TTDS-SEQ ID NO: 1 (Example 7) to HER-2 overexpressing cells.
  • Cells were grown in 12-well plates in the monolayer up to 50% of confluence and serum starved overnight. Then cells were treated with PG (100 nM) and/or Tz (10 pg/ml), 24 h after stimulation, cells were trypsinized, washed twice with ice-cold PBS, fixed in 70% ethanol at -20°C for 15 min, resuspended in RNaseA 1 mg/ml (ELIRX Ltd. Gdansk, Tru) and stained with propidium iodide (2,5 pg/ml). Cell cycle was analyzed with BD LSR II flow cytometer (BD Biosciences).
  • Tz-DBM-TTDS-SEQ ID NO: 1 Example 4
  • Tz-DBM- TTDS-SEQ ID NO: 15 Comparative Example 3 treated cells.
  • SKBR3, BT-474 or MDA- MB-231 cells were serum starved and stimulated with Tz, Tz-DBM-TTDS-SEQ ID NO: 1 (Example 4) and Tz-DBM-TTDS-SEQ ID NO: 15 (Comparative Example 3) (100 nM) for 5 days.
  • Cells were stained with propidium iodide and cell cycle was analyzed by flow cytometry. Results are shown in FiG. 11.
  • the reaction was stopped by removing the solution from the reaction vessel and adding it to a PD MiniTrap G-25 column (GE Healtcare) previously equilibrated with PBS.
  • the iodinated protein was dialyzed (Slide-A- Lyzer® minidialysis devices, 20 KDa, 0.5 mL) overnight against PBS to further remove the unincorporated 125 l.
  • the radioactivity of 10pL fractions was measured for 2 min using a Packard Cobra II Gamma Counter, and the protein concentration was determined using BCA analysis (Thermo Scientific).
  • the samples were diluted with Ringer Hepes to a final concentration of 100 nM.
  • Example 14 AlexaFluor 488-NHS labelling and quantification of Cx, Cx-DBM-TTDS-SEQ ID NO: 1 (Example 5); Bv, Bv-DBM-TTDS-SEQ ID NO: 1 (Example 6), Pt, Pt-DBM-TTDS- SEQ ID NO: 1 (Example 7)
  • Example 15 Permeability assays in the in vitro human BBB cellular model
  • the amount of protein was quantified using a gamma counter and the apparent permeability using the following formula: where P app is obtained in cm/s, Q A (t) is the amount of compound at the time t in the acceptor well, VD is the volume in the donor well, t is time in seconds, A is the area of the membrane in cm and Qo(to) is the amount of compound in the donor compartment at the beginning of the experiment.
  • MS analysis proteins from the acceptor compartment were purified by immunoprecipitation with Protein A magnetic beads following manufacturer’s instructions.
  • 25 pL of beads were placed into a 1.5 microcentrifuge tube, diluted with PBST and gently mixed. The tube was placed into a magnetic stand to facilitate the supernatant removal.
  • 500 pL were added of PBS solution were added to the tube to wash the beads. After mixing the solution was removed after collecting the beads with the magnetic stand. This operation was repeated 3 times. 1 mL of acceptor solution was added and left mixing with the beads o/n at 4°C. Then the supernatant was discarded, and the beads were washed (3 x 500 pL PBST and 3 x 500 pL PBS).
  • FIG. 12 shows permeability results in the human in vitro BBB cellular model for Tz, Tz-DBM-TTDS-SEQ ID NO: 1 (Example 4) and Tz-DBM-TTDS-SEQ ID NO: 15 (Comparative Example 3) and FIG 13, FIG 14, FIG 15 show permeability results in the human in vitro BBB cellular model for Cx, Cx-DBM-TTDS-SEQ ID NO: 1 (Example 5); Bv, Bv-DBM-TTDS-SEQ ID NO: 1 (Example 6), Pt, Pt-DBM-TTDS-SEQ ID NO: 1 (Example 7), respectively.
  • FIG 16 shows comparison of permeability of the conjugates.
  • Example 16 Biodistribution studies of Tz, Tz-DBM-TTDS-SEQ ID NO: 1 (Example 4) and Tz-DBM-TTDS-SEQ ID NO: 15 (Comparative Example 3) in mice.
  • mice Biodistribution studies in mice were performed by ChemPartner animal facility according to protocols approved by ChemPartner Institutional Animal Care and Use Committee (IACUC) following Assessment and Accreditation of Laboratory Animal Care (AAALAC) guidelines.
  • CD-1 male mice (6-8 weeks) were injected with Tz, Tz-DBM-TTDS-SEQ ID NO: 1 (Example 4) and Tz-DBM-TTDS-SEQ ID NO: 15 (Comparative Example 3). (10 mg/kg) via tail vein injection. 8h after injection blood was collected for serum generation and brains were terminally collected.
  • FIG. 17 shows biodistribution studies in mice.
  • Example 17 Conjugation of DBM peptides to lgG1 mAb at pH 8.5
  • the conjugation protocol was adapted from Org. Biomol. Chem. 15, 2947-2952 (2017).
  • the selected monoclonal antibody (mAb) (1 mg, 660 nmol) was diluted with BBS (pH 8.5) to a final concentration of 22.9 pM.
  • a fresh solution of DTT was added (10 mM, 4 pL,
  • DBM-TTDS-MiniAp4 [M+H+] theorical: 1509,269 Da; [M+H+] experimental: 1508,517 Da
  • DBM-TDS-Ang2 [M+H+] theorical: 2898,765 Da; [M+2H+/2] experimental: 1449,571 Da
  • AlexaFluor ® 488 (Aex:485/Aem:535nm) to conduct transport assay, where antibodies were assayed at 500 nm and quantification was carried out using a fluorometer.
  • Example 19 Permeability assays in the in vitro human BBB cellular model
  • the amount of protein was quantified using a gamma counter and the apparent permeability using the following formula: where P app is obtained in cm/s, QA(Q is the amount of compound at the time t in the acceptor well, VD is the volume in the donor well, t is time in seconds, A is the area of the membrane in cm and Qo(to) is the amount of compound in the donor compartment at the beginning of the experiment.
  • the results are shown in FIG. 18.
  • the results show that the permeability of the modified antibody according to the present invention (Cx-DBM_MiniAp4) is higher than the permeability of an antibody modified with a maleimide MiniAp4 as in Example 23 of WO2015/001015A1 (Cx-mal-MiniAp4).
  • FIG 19 shows the ratio of Papp of Cx modified with a SN38 conjugated peptide shuttle (MiniAp4 or Ang2) and Cx modified with the naked shuttle (MiniAp4 and Ang2) assayed at 1 pM in the human in vitro BBB cellular model.
  • the results show that the MiniAp4 conjugate increases the transport of SN38 more efficiently than the Ang2 conjugates.
  • a antibody shuttle conjugate of formula (I) or a pharmaceutically acceptable salt thereof which comprises from 1-6 peptides of formula P incorporated inserted in the disulfide bonds of the antibody in the form of a -P-(W)s-Y and joined to the sulfides through a linker -[(Ll)-(L 2 )-(L3)m-; wherein:
  • Z represents the structure of a monoclonal antibody or a monoclonal antibody fragment thereof;
  • the disulfide bonds are any disulfide bond that is capable of structurally retaining its structure upon reduction conditions and is selected from the group consisting of a naturally present interchain disulfide bond of the antibody, a naturally present intrachain disulfide bond of the antibody, and a disulfide bond introduced in the antibody by genetic engineering;
  • Li is a linker selected from the group consisting of Li a and Lit>;
  • L 2 is a biradical composed from 2 to 8 biradicals selected from the group consisting of LA, LB, LC and has the formula -LA-(LB) U -LC-
  • D is a radical of a substance selected form a biologically active substance, a substance for use in a diagnostic method; and a radioligand for radiotherapy;
  • P is a biradical of a peptide, equal or different, selected from the group consisting of:
  • peptide which comprises the amino acid sequence XI KAPETALX 2 wherein Xi is selected from the group consisting of Dap and Dab; and X 2 is selected from the group consisting of D (aspartic acid) and E (glutamic acid) (SEQ ID NO:7) being a linear peptide;
  • CKAPETALCAAA having at least an intrapeptide disulfide bond between cysteines 1 and 9, that is SEQ ID NO:9: CKAPETALCAAA
  • an anticancer active pharmaceutical ingredient selected from the group consisting of auristatins, duocarmycins, PBD dimers, maytansinoids, calicheamicins, anthracyclines, camptothecines, alpha-amanitin, tubulysins, MMAE, T-
  • Clause 9 A process for preparing an antibody shuttle conjugate as defined in any of the clauses 1-8, comprising: a) reducing disulfide bridges of the antibody; b) rebridging the disulfide bridges by reacting the -SH groups of the antibody with a dibromomaleimide-peptide of formula (II), c) optionally, carrying out a hydrolysis; wherein q; L2, L3, D, P, m, W, s, and Y are as defined in the antibody shuttle conjugate of formula (I).
  • a pharmaceutical composition comprising a therapeutically effective amount of the antibody shuttle conjugate as defined in any of the clauses 1-8, together with appropriate amounts of pharmaceutically acceptable carriers or excipients.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Organic Chemistry (AREA)
  • Immunology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Microbiology (AREA)
  • Oncology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Cell Biology (AREA)
  • Mycology (AREA)
  • Peptides Or Proteins (AREA)

Abstract

L'invention concerne des conjugués d'anticorps et des navettes peptidiques sélectionnées qui traversent la barrière hémato-encéphalique, éventuellement, également conjugués avec un principe pharmaceutique actif, un agent radiothérapeutique ou un agent diagnostique, des compositions pharmaceutiques les comprenant, ainsi que les conjugués destinés à être utilisés en médecine et en tant qu'agent diagnostique.
PCT/EP2023/071199 2022-08-01 2023-07-31 Modification spécifique d'un site par une navette bhe d'entités à base d'anticorps pour traverser la barrière hémato-encéphalique WO2024028282A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP22382746 2022-08-01
EP22382746.0 2022-08-01

Publications (1)

Publication Number Publication Date
WO2024028282A1 true WO2024028282A1 (fr) 2024-02-08

Family

ID=82786472

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2023/071199 WO2024028282A1 (fr) 2022-08-01 2023-07-31 Modification spécifique d'un site par une navette bhe d'entités à base d'anticorps pour traverser la barrière hémato-encéphalique

Country Status (1)

Country Link
WO (1) WO2024028282A1 (fr)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015001015A1 (fr) 2013-07-04 2015-01-08 Universitat De Barcelona Peptides transportés activement et résistant aux protéases en tant que navettes bbb et produits de construction navette-cargaison
CA2957354A1 (fr) 2014-09-12 2016-03-17 Genentech, Inc. Anticorps et conjugues modifies genetiquement avec de la cysteine
WO2016064749A2 (fr) * 2014-10-20 2016-04-28 Igenica Biotherapeutics, Inc. Nouveaux conjugués anticorps-médicament et composés, compositions et procédés d'utilisation s'y rapportant
WO2016074093A1 (fr) * 2014-11-14 2016-05-19 Angiochem Inc. Conjugués comprenant une partie anticorps, un polypeptide capable de traverser la barrière hémato-encéphalique, et une cytokine
WO2022064052A1 (fr) * 2020-09-28 2022-03-31 Fundació Institut De Recerca Biomèdica (Irb Barcelona) Conjugués peptidiques de sn38 utiles dans le traitement du cancer

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015001015A1 (fr) 2013-07-04 2015-01-08 Universitat De Barcelona Peptides transportés activement et résistant aux protéases en tant que navettes bbb et produits de construction navette-cargaison
CA2957354A1 (fr) 2014-09-12 2016-03-17 Genentech, Inc. Anticorps et conjugues modifies genetiquement avec de la cysteine
WO2016064749A2 (fr) * 2014-10-20 2016-04-28 Igenica Biotherapeutics, Inc. Nouveaux conjugués anticorps-médicament et composés, compositions et procédés d'utilisation s'y rapportant
WO2016074093A1 (fr) * 2014-11-14 2016-05-19 Angiochem Inc. Conjugués comprenant une partie anticorps, un polypeptide capable de traverser la barrière hémato-encéphalique, et une cytokine
WO2022064052A1 (fr) * 2020-09-28 2022-03-31 Fundació Institut De Recerca Biomèdica (Irb Barcelona) Conjugués peptidiques de sn38 utiles dans le traitement du cancer

Non-Patent Citations (14)

* Cited by examiner, † Cited by third party
Title
A. MADDER ET AL., EUR. J. ORG. CHEM., pages 2787 - 2791
E. KAISERR. L. COLESCOTTC. D. BOSSINGERP. I. COOK: "Color test for detection of free terminal amino groups in the solid-phase synthesis of peptides", ANAL. BIOCHEM., vol. 34, 1970, pages 595 - 598, XP024828956, DOI: 10.1016/0003-2697(70)90146-6
EUR J PHARM BIOPHARM, vol. 115, 2017, pages 149 - 158
F. DAVID ET AL., ORG. PROCESS RES. DEV., vol. 14, no. 4, 2010, pages 999 - 1007
J. MED. CHEM., vol. 51, no. 21, 2008, pages 6916 - 6926
M. AMBLARD ET AL.: "Methods and protocols of modern solid-phase peptide synthesis", MOLECULAR BIOTECHNOLOGY, vol. 33, 2006, pages 239 - 254, XP009116689, DOI: 10.1385/MB:33:3:239
M. KAZEMI ET AL., JOURNAL OF SULFUR CHEMISTRY, vol. 36, no. 6, 2015, pages 613 - 623
M. T. NGUYEN ET AL., J. ORG. CHEM., vol. 20, no. 63, 1998, pages 6878 - 6885
MOL. PHARM., vol. 12, 2015, pages 3986 - 3998
MORAIS MAURÍCIO ET AL: "Optimisation of the dibromomaleimide (DBM) platform for native antibody conjugation by accelerated post-conjugation hydrolysis", ORGANIC & BIOMOLECULAR CHEMISTRY, vol. 15, no. 14, 1 January 2017 (2017-01-01), pages 2947 - 2952, XP093011814, ISSN: 1477-0520, DOI: 10.1039/C7OB00220C *
ORG. BIOMOL. CHEM., vol. 15, 2017, pages 2947 - 2952
P.L. BARKER ET AL., J. MED. CHEM., vol. 35, 1992, pages 2040 - 2048
PETER G. M. WUTS.: "Greene's Protective Groups in Organic Synthesis", 2014, JOHN WILEY & SONS, INC., pages: 456 - 463
S.F.M. VAN DONGEN ET AL., BIOCONJUGATE CHEM., vol. 20, 2009, pages 20 - 23

Similar Documents

Publication Publication Date Title
KR102647074B1 (ko) 친수성 항체-약물 컨쥬게이트
JP6817492B2 (ja) 高いインビボ忍容性を有するアントラサイクリン系の抗体薬物複合体
CN106856656B (zh) 一种稳定型抗体药物耦联物及其制备方法和用途
JP7402807B2 (ja) グリピカン3抗体およびそのコンジュゲート
JP3373849B2 (ja) 3価および4価の単一特異性抗原結合性タンパク質
JP7073487B2 (ja) 操作された抗体化合物およびこれらの抱合体
JP2024081670A (ja) トランスグルタミナーゼコンジュゲーション方法およびリンカー
EP2654803B1 (fr) Conjugués peptidiques radiomarqués se liant à her2
JP2015510877A (ja) 抗体の化学修飾
ES2855169T3 (es) Conjugados de polipéptido-anticuerpo derivados de aprotinina
TW202034961A (zh) 含有標識部-抗人類抗體Fab片段複合體之醫藥組成物
KR20190078573A (ko) 신규 항인간 MUC1 항체 Fab 프래그먼트
AU2020234394A1 (en) Site-specific antibody conjugation and antibody-drug conjugate as specific example thereof
BR112020023420A2 (pt) Complexo tendo fragmento fab de anticorpo anti-muc1 humano, peptídeo ligador e/ou ligante
US20230046947A1 (en) Reactive conjugates
JP2023529640A (ja) 治療用コンジュゲート
WO2024028282A1 (fr) Modification spécifique d'un site par une navette bhe d'entités à base d'anticorps pour traverser la barrière hémato-encéphalique
CA2572825A1 (fr) Compositions pour l'administration en plusieurs etapes de rayonnement hot-spots a des cellules cancereuses
WO2022078566A1 (fr) Conjugués réactifs
WO2024083166A1 (fr) Conjugué anticorps-médicament, son procédé de préparation et son utilisation dans la résistance tumorale
WO2023241621A1 (fr) Anticorps anti-liv-1 et conjugué anticorps-médicament
TW202421202A (zh) 抗cd33抗體和抗cd33抗體-藥物偶聯物及其用途
CN117338949A (zh) 一种带有高稳定性亲水连接单元的奥瑞他汀类药物及其偶联物
NZ761642B2 (en) Hydrophilic antibody-drug conjugates
NZ722252B2 (en) Hydrophilic antibody-drug conjugates

Legal Events

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

Ref document number: 23750999

Country of ref document: EP

Kind code of ref document: A1