EP3568404A1 - Composés de type étoile de (guanidyl)x-oligosaccharides et conjugués ou complexes de ceux-ci - Google Patents

Composés de type étoile de (guanidyl)x-oligosaccharides et conjugués ou complexes de ceux-ci

Info

Publication number
EP3568404A1
EP3568404A1 EP17703926.0A EP17703926A EP3568404A1 EP 3568404 A1 EP3568404 A1 EP 3568404A1 EP 17703926 A EP17703926 A EP 17703926A EP 3568404 A1 EP3568404 A1 EP 3568404A1
Authority
EP
European Patent Office
Prior art keywords
formula
group
alkyl
branched
linear
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.)
Withdrawn
Application number
EP17703926.0A
Other languages
German (de)
English (en)
Inventor
Paolo Caliceti
Stefano Salmaso
Alessio MALFANTI
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.)
Universita degli Studi di Padova
Original Assignee
Universita degli Studi di Padova
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 Universita degli Studi di Padova filed Critical Universita degli Studi di Padova
Publication of EP3568404A1 publication Critical patent/EP3568404A1/fr
Withdrawn legal-status Critical Current

Links

Classifications

    • 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
    • C07H15/02Acyclic radicals, not substituted by cyclic structures
    • C07H15/04Acyclic radicals, not substituted by cyclic structures attached to an oxygen atom of the saccharide radical
    • 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
    • A61K47/56Medicinal 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
    • A61K47/61Medicinal 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 the organic macromolecular compound being a polysaccharide or a derivative thereof

Definitions

  • the present invention origins in the biomedical and pharmaceutical field.
  • the invention relates to star-like (guanidyl)x-oligosaccharidic compounds as delivery carriers of diagnostic and therapeutic molecules.
  • the invention concerns with polycationic macromolecules having the capacity of carrying and delivering small-sized drugs or biological molecules such as oligonucleotides, polypetides and proteins across biological membranes.
  • oligo- and polynucleotides are good candidates for treatment of a variety of common genetic diseases (i.e. cancer [6], cystic fibrosis [7], muscular dystrophy [8], vascular disease [9], neurogenerative disorder [10], etc.).
  • Engineered viral vectors are also used to enhance the cell uptake of ONs.
  • Viruses have evolved efficient mechanism to bypass the cell membrane to reach the cytoplasm and nucleus.
  • Most used viral carrier for gene therapy are adenovirus, lentivirus, herpes simplex virus (HSV) or retrovirus.
  • HSV herpes simplex virus
  • the cost- effectiveness and availability of these carriers and their high capacity to activate the immune system have limited their use.
  • the use of viruses as drug carriers with few exceptions, entails the risks reported above [13, 14].
  • cationic materials including cationic polymers, lipids and cell penetration peptides, which have been used to complex the anionic ONs by coulombic interactions [15-18]. These nano-complexes are stable in the blood stream and represent a shield to degradation of ONs from nucleases. Compared to viral vectors, they have low antigenicity and risks of insertional oncogenesis.
  • PEI polyethyleneimine
  • Chitosan is a cationic polysaccharide with high content of amino group able to condense ONs [20].
  • Cationic lipids namely, 1 ,2-di-O-octadecenyl-3-trimethylammonium propane (DOTMA), 1 ,2-dioleoyl-3-trimethylammonium-propane (DOTAP), dimethyldioctadecylammonium (DDAB) and their colloidal formulations have been also used to complex ONs [21 ].
  • DOTMA 1,2-di-O-octadecenyl-3-trimethylammonium propane
  • DOTAP 1 ,2-dioleoyl-3-trimethylammonium-propane
  • DDAB dimethyldioctadecylammonium
  • Lipofectamine a liposomal formulation containing cationic lipids is regarded as a 'gold standard' for in vitro cell transfection.
  • cationic/ONs complexes are unstable, immunogenic and difficult to be manufactured.
  • Cationic cyclodextrins are cyclo-oligosaccharidic vehicles grafted with poly-cationic polymers such as PEI or poly-lysines that have been successfully used to deliver ONs or plasmids [22, 23].
  • poly-cationic polymers such as PEI or poly-lysines
  • a class of cyclodextrin-based gene delivery systems targeted to tumours by grafting transferrin to a PEG chain conjugated with adamantane that forms host-guest complexes with cationic cyclodextrins have been developed [24].
  • Covalent bioconjugates have been obtained by ON derivatization with several molecules including lipids, polymers and cell penetration enhancers.
  • HSA Human serum albumin
  • Bioconjugates have been obtained by attaching targeting agents to the ONs in order to yield specific cell targeting and cell up-take.
  • targeting agents used for this purpose there are: asialoglycoproteins for hepatocyte delivery [33], small peptides such as RGD that targets ⁇ / ⁇ 3 integrin receptor [34], bombesin that targets the BB2 G-protein overexpressed in prostate cancer cells [35], insulin growth factor-1 (IGF-1 ) peptide mimetic that targets the IGF-1 receptor of breast cancer cells [36], antibodies and antibody fragments, such as herceptin antibody, anti-Lewis-Y monoclonal antibody, antibody against a-CD19, a biomarker against acute lymphoblastic leukaemia [33].
  • One of the purposes of the present invention is to provide a class of molecules acting as intracellular vehicle of therapeutic or diagnostic molecules.
  • Another aim of the invention resides in the provisions of drug delivery systems for transporting therapeutic molecules in the intracellular target sites.
  • Another aim of the invention is the provision of a class of polycationic macromolecules (pOG-M-PEG), without interferring anionic residues, acting as cell permeation enhancers.
  • pOG-M-PEG polycationic macromolecules
  • the inventors have found that the insertion of guanidine moieties on a saccharide core provides a class of polycationic molecules with delivery features making them useful as intracellular delivery vectors of biological molecules such as oligo- and polynucleotides, aptamers and peptides for therapeutic or diagnostic purposes.
  • Ri , R2, R3, R4, are independently
  • R5 is a linker or spacer having formula -(Y)N- R"'-C, wherein
  • Y is an hydrogen or an acyl moiety having formula R*-CO-, wherein R* is a linear or branched C1 -C22 alkyl or C1 -C22 alkenyl chain or a steroidic scaffold,
  • C is a terminating group selected from C-i -Cs alkoxy-, halogen-, vinyl-, carboxyl-, amino-, aldehyde-, hydroxyl-, thiol-, maleimido-, biotin-, alkine- or azido- group or CO-W-R'", wherein W is N H , O, and R'" has the same meaning defined above; Re, if present, is a group selected from
  • n is an integer ranging from 1 -8 sugar units, and salts thereof.
  • the substituent R5 or Re if present is bound, typically by covalent bonds, to a functional moiety which is a compound having biological, physicochemical, biopharmaceutical and/or targeting activity or a fluorophore or a radionuclide or macrosurfaces.
  • a functional moiety which is a compound having biological, physicochemical, biopharmaceutical and/or targeting activity or a fluorophore or a radionuclide or macrosurfaces.
  • the compounds of formula (I) of the invention forms a conjugate with a functional moiety which make them useful as targeting or labelling compounds.
  • the invention provides a conjugate comprising a compound of formula (I) bound by either the R5 or Re substituent group to a functional moiety selected from i) macromolecules or ii) a physicochemical and biopharmaceutical modifier iii) a fluorophore or iv) a radionuclide or v) macrosurfaces or a vi) targeting agent.
  • the compound of Formula (I) is bound, typically by a covalent bond, to the compound having biological activity or fluorophore or radionuclide or targeting agent via either R5 or Re substituent group.
  • the above conjugate is used to target said functional moiety in a in vivo or in vitro biological site where it exerts its action.
  • the cationic (guanidyl)x-oligosaccharidic compounds of formula (I) interact, especially by means of non-covalent bounds, with organic molecules bearing an anionic group (anionic macromolecules) and preferably having biological activity
  • the cationic (guanidyl)x- oligosaccharidic compounds of formula (I) may interact or bound anionic macromolecules such as ONs, polypeptides, proteins, anionic macromolecules having biological activity, especially biological drugs, or cell membranes, typically by interaction with one or more of the substituents groups Ri, R2, R3, R4.
  • the invention also provides a complex comprising a cationic (guanidyl)x-oligosaccharidic compound of formula (I) as defined above and biological anionic macromolecules, especially ONs, peptides, proteins, plasmids, poly-nucleic acids or drugs.
  • said conjugate is bound with non- covalent bonds with the compound of formula (I) via the substituents groups Ri,
  • the conjugate defined above is encapsulated or bond with a delivery system such as liposomes, micelles, polymeric or inorganic nanoparticles, micro- and nanocapsules or micro- or nanospheres or other matrices.
  • a delivery system such as liposomes, micelles, polymeric or inorganic nanoparticles, micro- and nanocapsules or micro- or nanospheres or other matrices.
  • the invention provides the use of the cationic (guanidyl)x-oligosaccharidic compounds of Formula (I) as carriers for delivering molecules having biological activities such as macromolecules, colloidal systems, peptides, proteins, drugs into a target cell, tissue or organ of a mammal, especially a human body or in vitro transfect agent.
  • molecules having biological activities such as macromolecules, colloidal systems, peptides, proteins, drugs into a target cell, tissue or organ of a mammal, especially a human body or in vitro transfect agent.
  • a compound of Formula I which is bound with non-covalent bonds by substituents group Ri, R2, R3, R4 to a molecule having biological activity especially ONs, peptides, proteins, plasmids or poly-nucleic acids or drugs, and typically with covalent bounds by either the substituent group R5 or R6 to a compound having biological activity for example a drug, an antibody, a peptide, protein or nucleic acids, a targeting agent, a fluorophore or a radionuclide.
  • Figure 1 illustrates the synthesis of maltotriosyl-N-acetyl-amino-hexanoic acid of the example 1 .
  • Figure 2 shows the synthesis of the star-like (2-bromoisobutyryl)6-maltotryosyl-N- acetyl-amino-hexanoic acid of the example 1 .
  • Figure 3 shows the 1 H NMR analysis of star-like (agmatinyl)6-maltotriosyl-N-acetyl- amino-hexanoic acid of the example 1 .
  • Figure 4 illustrates the synthesis of star-like (agmatinyl)6-maltotriosyl-N-acetyl- amino-hexanoic acid of the example 1 .
  • Figure 5 shows the molecular structure of star-like (agmatinyl)6-maltotriosyl-N- acetyl-amino-hexanoic acid of the example 1 .
  • Figure 6 illustrates the synthesis of star-like (agmatinyl)6-maltotriosyl-N-acetyl- amino-hexanoate-PEGekDa-NH2 of the example 2.
  • Figure 7 illustrates the synthesis of star-like (agmatinyl)6-maltotriosyl-N-acetyl- amino-hexanoate-oleylamide of the example 3.
  • Figure 8 shows the gel electrophoretic profiles of dsDNA/pOG-M-PEG complexes with N/P ratio between 0.5-5 at pH 7.4. The samples were run in polyacrylamide gel using TBE as running buffer.
  • Figure 10A shows the TEM images of dsDNA/pOG-M-PEG complexes with 3 N/P ratio.
  • Figure 10B shows the TEM images of dsDNA/pOG-M-PEG complexes with 5 N/P ratio.
  • Figure 13 shows the hemolytic profiles of dsDNA/pOG-M-PEG complexes with 3 and 5 N/P ratio at pH 7.4.
  • PBS pH 7.4 and ON solution in PBS, pH 7.4, were used as negative control whereas 1 % w/v of Triton X-100 was employed as a positive control.
  • Figure 14 shows the isothermal calorimetry profiles of pOG-M-PEG titrated with dsDNA in 10 mM TRIS, 150 mM NaCI, pH 7.4 and relative table.
  • Figure 15 C Viability of MC3T3-E1 cells incubated with dsDNA/pOG-M-PEG complexes with 3 and 5 N/P ratio.
  • Figure 16 B shows the Cell cytometric profiles of KB cells after 6 hour cell treatment with Cy3-dsDNA/pOG-M-PEG complexes with 3 and 5 N/P ratio. Untreated cells are used as control.
  • Figure 16 C shows the cell cytometric profiles of MC3T3-E1 cells after 6 hour cell treatment with Cy3-dsDNA/pOG-M-PEG complexes with 3 and 5 N/P ratio. Untreated cells are used as control.
  • Figure 17 A shows the confocal microscopy images of MCF-7 cells treated with Cy3-dsDNA/pOG-M-PEG complexes with 3 and 5 N/P ratio. Untreated cells are used as control.
  • Figure 17 B shows the confocal microscopy images of KB cells treated with Cy3- dsDNA/pOG-M-PEG with 3 and 5 N/P ratio. Untreated cells are used as control.
  • Figure 17 C shows the confocal microscopy images of MC3T3-E1 cells treated with Cy3-dsDNA/pOG-M-PEG complexes with 3 and 5 N/P ratio. Untreated cells are used as control.
  • cationic (guanidyl)x-oligosaccharidic compounds of Formula (I) are useful as carriers of therapeutic and diagnostic molecules, in particular oligonucleotides, such as asRNA (single stranded antisense RNA), mRNA (messanger RNA), snRNA (small nuclear RNA), siRNA (small interfering RNA), miRNA (micro RNA), pDNA (plasmidic DNA) or aptamers.
  • oligonucleotides such as asRNA (single stranded antisense RNA), mRNA (messanger RNA), snRNA (small nuclear RNA), siRNA (small interfering RNA), miRNA (micro RNA), pDNA (plasmidic DNA) or aptamers.
  • the compounds of Formula (I) are useful as agents to formulate and deliver the above mentioned biological molecules through physical or covalent conjugation and optional encapsulation into a delivery system such as liposomes, micelles, polymeric or inorganic nanoparticles, micro- and nanocapsules or micro- and nanospheres or other matrices.
  • the invention concerns with compounds of Formula (I)
  • Ri , R2, R3, R4, are independently:
  • R5 is a linker or spacer having formula -(Y)N-R"'-C, wherein
  • R' is a linear or branched C2-C22 alkyl or C2-C22 alkenyl
  • Y is an hydrogen or an acyl moiety having formula FT-CO-, wherein R* is a linear or branched C1-C22 alkyl or C1-C22 alkenyl chain or a steroidic scaffold,
  • C is a terminating group selected from C-i-Cs alkoxy-, halogen-, vinyl-, carboxyl-, amino-, aldehyde-, hydroxyl-, thiol-, maleimido-, biotin-, alkine- or azido- group or CO-W-R'", wherein W is NH, O, and R'" has the same meaning defined above; R6, if present, is a linker selected from
  • n is an integer ranging from 1 -8 sugar units, and salts thereof.
  • the substituents R5 or Re if present, is bond preferably by covalent bounds to a functional moiety such as a molecule having biological activity or a fluorophore or radionuclide.
  • a functional moiety such as a molecule having biological activity or a fluorophore or radionuclide.
  • the inventor have found that the (guanidyl)x-oligosaccharidic compounds of Formula (I) are useful to target molecules having biological activity and a fluorophore or a radionuclide or a targeting agent, optionally beared to the molecules, in the desired site.
  • the invention provides the use of the compounds of Formula (I) bound to a functional moiety, such as a molecule having biological activity, to target the functional moiety at the desired site of action.
  • said functional moiety is covalently bound to the substituent Rs or Re, if present, of the compounds of Formula (I) of the invention.
  • R6 is linked to Rs with a functional group selected from phenyl-, alkoxy-, halogen-, vinyl-, carboxyl-, amino-, amido-, aldehyde-, hydroxyl-, thiol-, maleimido-, biotin-, alkine- or azido-group.
  • a functional group selected from phenyl-, alkoxy-, halogen-, vinyl-, carboxyl-, amino-, amido-, aldehyde-, hydroxyl-, thiol-, maleimido-, biotin-, alkine- or azido-group.
  • the functional moieties include low and high molecular weight molecules having biological activity or physicochemical and biopharmaceutical modifiers such as phospholipids, natural or synthetic, linear or branched oligomers, polymers, copolymers or conjugates, a polyethylene glycol or a cyclic structure, a peptide, a protein, a drug, organic and/or inorganic surfaces.
  • biological activity or physicochemical and biopharmaceutical modifiers such as phospholipids, natural or synthetic, linear or branched oligomers, polymers, copolymers or conjugates, a polyethylene glycol or a cyclic structure, a peptide, a protein, a drug, organic and/or inorganic surfaces.
  • the functional moiety is a polymer or a compound having biological activity.
  • Exemplary polymers or molecules/compounds having biological activity or acting as physicochemical and biopharmaceutical modifiers bound to either Rs or Re substituent of the compounds of formula (I) include:
  • a phospholipid such as 1 ,2-distearoyl-sn-glycero-3- phosphoethanolamine
  • polypeptides and/or proteins for example transferrin, albumin, lysozyme, antibody or its fragments, or peptides;
  • Ri, R2, R3 and R 4 substituents are -CO-R°-X, wherein R° is a C2-C3 alkyl branched group and X is bromine or chlorine and preferably -CO- R°-X is a 2-bromoisobutyryl group.
  • substituents Ri, R2, R3 and R 4 is an isopropyl group and X is represented by an halogen preferably CI or Br.
  • R5 is bound to or Re is a polymer selected from dextran, pullulan, chitosan, PGA, POX and PEG. The latter being preferred.
  • the poly(ethylene glycol) derivative, linear or branched, of formula -[CH2CH2O] a -(CH2)b-C has an average molecular weight (Mw) from 132 to 20000 Da, preferably in the range of 4000 to 8000 MW.
  • a is 136 b2 and the molecular weight is 6000.
  • the (average) molecular weight of the above polymers may be determined with standard methods such as those disclosed in Ueno et al., 1988, Chem Pharm Bull. 36, 4971 -4975; Wyatt 1993, Anal Chim Acta 272: 1 -40; Watt Technologies 1999 "Light scattering University Dawn Course Manual and "Dawn Eos Manual” Wyatt Technology Corp. Santa Barbara CA (USA).
  • the alkyl chain is oleylamine.
  • R" is a C 4 alkyl chain bearing a -COOH group.
  • Rs is N-acetyl-12-aminododecanoyl acid.
  • the compounds of Formula (I) according to the invention have star-like (guanidyl)x-oligosaccharidic moieties and may physically form, typically by non- covalent bonds, complex with compounds having a negative charge, especially anionic compounds having a biological activity.
  • the compounds of Formula (I) may form complexes with anionic macromolecules especially those having a biological activity and protect them by enzymatic degradation and improve the delivery and cell uptake.
  • the (guanidyl)x-oligosaccharidic molecules of the invention may form complexes with anionic therapeutic macromolecules such as ONs, especially asRNA (single stranded antisense RNA), mRNA (messenger RNA), snRNA (small nuclear RNA), siRNA (small interfering RNA), miRNA (micro RNA), pDNA (plasmidic DNA) or aptamers or with peptides, proteins, drugs, polymer therapeutics such as linear or branched bioconjugates, liposomes, micelles or nanoparticles.
  • anionic therapeutic macromolecules such as ONs, especially asRNA (single stranded antisense RNA), mRNA (messenger RNA), snRNA (small nuclear RNA), siRNA (small interfering RNA), miRNA (micro RNA), pDNA (plasmidic DNA) or aptamers or with peptides, proteins, drugs, polymer therapeutics such as linear or branched bioconjugates, lip
  • the complexation may be exploited by anchoring the (guanidyl)x- oligosaccharidic compound of Formula (I) to hydrophilic polymers, lipids, proteins, surfaces to obtain colloidal systems suitable to promote the cell entry of therapeutic macromolecules or their use in in vitro tools.
  • the scaffold of the compounds of Formula (I) contains oligosaccharides moieties, especially from 1 to 8 saccharidic units, that provide the star-like shape, and guanidinium groups to provide positive charges.
  • the saccharidic units are maltotriose, glucose, lactose, trealose, gentiobiose, mannose, cellobiose, maltose, isomaltose, maltotetraose, maltopentaose, maltohexaose, maltoheptaose, maltooctaose and preferably are lactose, maltose or maltotriose.
  • the star-like (guanidyl)x-oligosaccharidic compounds of Formula (I) have a pKa in the range of from 12 to 13 which is fully protonated in physiological environment.
  • This feature enables the compounds of Formula (I) to complex negative macromolecules such as ONs.
  • the invention concerns compounds of Formula (I) for use as carriers for intracellular delivery of macromolecules or supramolecular colloidal structures physically or covalently conjugated thereto.
  • the compounds of Formula (I) are conjugated with hydrophilic polymer, such as a poly(ethylene glycol) to give stealth properties at the system, improving pharmacokinetic parameters such as half-life and degradation.
  • the star-like (guanidyl)x-oligosaccharidic compounds of Formula (I) may be complexed with ONs taking advantage of the natural attraction between opposite charges or may covalently bind functional moieties such as molecules or colloidal systems or anchoring to surfaces of molecules or products having biological activity.
  • the present invention provides a method for the production of the (guanidyl)x-oligosaccharidic derivatives of formula (I) as defined above, said method comprising the steps of:
  • (CH2)b-C where a is an integer selected in the range from 2 to 454, b is an integer selected in the range from 2 to 6;
  • Y is an hydrogen or an acyl moiety having general formula R*-CO-, where R* is a linear or branched with or without multiple bond when is possible C1 -C22 alkyl chain or a molecule derivative with a steroidic scaffold;
  • C have the same meanings for both and is an terminating group which alkoxy-, halogen-, vinyl-, carboxyl-, amino-, aldehyde-, hydroxyl-, thiol-, maleimido- , biotin-, alkine- or azido- group;
  • the stabilization of the amino-oligosaccharide derivative obtained is carried out trough N-acetylation; 2.
  • a poly(ethylene glycol) chain is used having formula -NH-[CH2CH20] a -(CH2)b-C where a, b and C have the same meaning described above.
  • the oligosaccharide was functionalized with a spacer through the reaction of the amino group of the selected spacer R5, preferably ⁇ - aminohexanoic acid with the anomeric carbon of the selected oligosaccharides, preferably maltotriose.
  • the reaction was performed in methanol supplemented with of acetic acid (1 % v/v) at 50 °C.
  • the bond between the selected sugar and the spacer R5 was stabilized preferably through the addition of acetic anhydride in order to prevent the separation of the spacer from the oligosaccharide bulk.
  • the reaction of acetylation was stopped after 24 hours.
  • the product of conjugation was purified by precipitation in a cold ethyl ether.
  • the hydroxyl groups of the N-acetylamino spacer oligosaccharide derivative were then functionalized by the use of an appropriate linker, preferably 2- bromoisobutyryl bromide. This functionalization was carried out in order to obtain a star shape oligosaccharide derivative.
  • the N-acetylamino spacer oligosaccharide was dispersed in cold chloroform with trietylamine and the suspension was added of 2-bromoisobutyryl bromide. The reaction was stirred for 72 hours and the product was isolated by sequential extractions.
  • the (2-bromoisobutyryl) star-like oligosaccharide was then modified with the guanidine moiety.
  • the conjugation was performed through the use of copper salts (preferably CuBr), TPMA (tris[(2-pyridyl)methyl amine], a ligand of Cu (I)) and ascorbic acid as reducing agent.
  • the monomer employed in this synthesis was an acryloyl guanidyl derivative (preferably acryloyl agmatine, synthesized according with the literature).
  • the reaction was carried out under nitrogen conditions for 3 days at 65 °C and the final product was precipitated in a solution of ethyl ethenacetone (1 :1 ) with the addition of 1 % v/v of acetic acid.
  • the last step of synthesis concerns the PEGylation of the star-like (guanidyl)x- oligosaccharidic derivative.
  • the attachment of the polymeric side chain was performed on the lead derivative star-like (agmatinyl)6-maltotriosyl-N-acetyl-amino- hexanoic acid.
  • MES mM morpholino-ethan- sulphonic acid buffer
  • the final product was purified by dialysis and recovered by lyophilization.
  • the star-like (guanidyl)x-oligosaccharidic compound of formula (I) may be covalently or non-covalently linked to anionic molecules, preferably ONs.
  • Dynamic light scattering was used to determine the hydrodynamic radii of complexes at various N/P ratio. Complexes were approximately of 70-100 nm in diameter. The zeta potential were also evaluated and the results showed that the charge is approximately neutral ( ⁇ 4-8 mV). TEM images showed a rod like shape of the complexes having size in agreement with the DLS analysis. The formulation were stable in PBS, pH 7.4, and in cell culture medium in 12 hours. These features are very important for a gene delivery systems.
  • the hemolytic effect of the complexes were evaluated and the analysis showed a complete compatibility with the biomembranes of the red blood cells under physiological conditions.
  • the assay confirms that the star-like (guanydil)x-oligosacharidic compounds of the invention are suitable for the intracellular delivery of macromolecules as described above.
  • halogen indicates fluorine (F), chlorine (CI), bromine (Br) or iodine (I).
  • alkyl indicates a saturated aliphatic hydrocarbon radical, including straight chain and branched chain radicals of 1 to 6 carbon atoms referred to as C-i -e alkyl.
  • alkyl are methyl, ethyl, propyl, isopropyl, n-butyl, iso-butyl, tert-butyl, n-amyl, iso-amyl, n-hexyl, and the like.
  • the alkyl group of the compounds described herein may be designated as "C1-C22, C-i-Cs or C1-C4 alkyl" or similar designations.
  • Ci-C 4 alkyl indicates that there are one to four carbon atoms in the alkyl chain, i.e., the alkyl chain is selected from among methyl, ethyl, propyl, iso-propyl, n-butyl, iso-butyl, sec-butyl, and t-butyl.
  • saturated hydrocarbon radicals include, but are not limited to, groups such as methyl, ethyl, n-propyl, isopropyl, n-butyl, t-butyl, isobutyl, sec- butyl, cyclohexyl, cyclohexylmethyl, cyclopropylmethyl, homologs and isomers of, for example, n-pentyl, n-hexyl, n-heptyl, n-octyl, and the like.
  • An unsaturated alkyl group is one having one or more double bonds.
  • unsaturated alkyl groups include, but are not limited to, vinyl, 2-propenyl, crotyl, 2- isopentenyl, 2-butadienyl, 2,4-pentadienyl, 3-(1 ,4-pentadienyl), ethynyl, 1 - and 3- propynyl, 3-butynyl, and the higher homologs and isomers.
  • Alkyl groups which are limited to hydrocarbon groups are termed "homoalkyl".
  • Peptides means products derived from condensation of two or more amino carboxylic acid molecules (the same or different) by formation of a covalent bond from the a-carbonyl carbon of one to the a-nitrogen atom of another with formal loss of water.
  • the term is usually applied to structures formed from a-amino acids, but it includes those derived from any amino carboxylic acid.
  • Proteins means naturally occurring and synthetic polypeptides having molecular weights greater than about 5000 daltons.
  • alkoxy means an unsubstituted or substituted alkyl chain linked to the remainder of the molecule through an oxygen atom.
  • alkoxy include, but are not limited to, methoxy, ethoxy, propyloxy, isopropyloxy, benzyloxy and the like.
  • the term "optionally substituted” or “substituted” means that the referenced group may be substituted with one or more additional group(s) individually and independently selected for example from C1-22 alkyl, cycloalkyl, halo, carbonyl, thiocarbonyl, isocyanato, thiocyanato, isothiocyanato, and amino, including mono- and di-substituted amino groups, and the protected derivatives thereof.
  • the term steroidic scaffold means naturally occurring compounds and synthetic analogues, based on the cyclopenta[a]phenanthrene carbon skeleton, partially or completely hydrogenated; there are usually methyl groups at C-10 and C-13, and often an alkyl group at C-17. By extension, one or more bond scissions, ring expansions and/or ring contractions of the skeleton may have occurred.
  • Typical compounds having steroidic scaffold comprise cholestanes such as cholesterol and cholanes such as cholic acid.
  • phospholipid means lipids containing phosphoric acid as mono- or di- esters, including phosphatidic acids and phosphoglycerides.
  • Useful phospholipid include phosphatidylcholine, phosphatic acid, phosphatidylglycerol, phosphatidylethanolamine, phosphatidylglycerol, phosphatidylserine, phosphatidylserine, lysophosphatidylcholine.
  • molecule having biological activity means a compound exerting a biological activity, such as a drug, a protein, a polypeptide, or a oligonucleotide.
  • biological site includes biological matter such as cells, or tissues or organs of the human body.
  • physicochemical and biopharmaceutical modifiers mean agents improving the pharmacokinetic features or the biopharmaceutical features such as solubility, carrier of an active ingredient in the target site.
  • Bis amino terminating Polyethylen glicole was purchased from Iris Biotech GmbH (Marktredwiz, Germany).
  • dsDNA double strand DNA
  • C3-dsDNA cyanin-3 labeled dsDNA
  • the dsDNA intercalating agent GelRed was purchased from SICHIM ( Rome, Italy).
  • MCF-7 human breast carcinoma
  • KB human cervical carcinoma
  • KB murine embryonic fibroblast
  • maltotriose (1 .98 mmol) was dissolved in 30 mL of methanol acidified with 1 % v/v of acetic acid. The solution was heated to 60 °C until complete dissolution of maltoriose. Then, the solution was added of 1040.1 mg of 6- aminohexanoic acid (7.93 mmol) and maintained under stirring for 14 h at 40 °C. The volume was reduced to half volume under vacuum and the solution was added of 22.6 mL of acetic anhydride (237.6 mmol) over 60 min. The solution was stirred at room temperature for 24 h and then the volume was reduced to « 5 mL.
  • FT-I R 3472 (-OH), 2978 (-CH), 1 744 (isobutyryl -CO-O), 1 650 (acetyl -CO-N). Elemental analysis: found C, 40.02%; H, 4.89%; Br, 31 .1 2%; N, 0.84%; (O, 23, 1 3%). [calcd for (2-bromoisobutyryl)6-maltotryosyl-N-acetyl-amino-hexanoic acid (CsoHysBreNO; ), C, 38.66%; H, 4.87%; Br, 30.86%; N, 0.90%; O, 24.72%.].
  • Arcyloyl-agmatine was synthesized according to the protocol reported in the literature [patent US 6.703.468][38J. Briefly, 2.0 grams of agmatine sulfate (8.76 mmol) was dissolved in NaHC03 saturated water solution at 0 °C. Acryloyl chloride (749.18 ⁇ _, 9.64 mmol) was added over 30 min under vigorous stirring. After 1 h, the pH of the solution was adjusted to 1 .0 using 1 .0 N HCI and the mixture was saturated using sodium chloride.
  • the mixture was stirred in these conditions for 72 hours and then was exposed to the air. After 30 min the solution was extensively poured in 1 :1 v/v diethyl ether/acetone mixture added of 1 % v/v of acetic acid. The precipitate was collected and desiccated under vacuum. The product yield was 542 mg, corresponding to 65%.
  • Figure 4 shows the synthesis of (agmatinyl)6-maltotriosyl-N-acetyl-amino- hexanoic acid.
  • Figure 5 shows the general structure of the oligoguanidine derivative.
  • ON/pOG-M-PEG complexes were prepared by using a scrambled 1 1 kDa dsDNA (19 pb) as ON model.
  • the dsDNA and pOG-M-PEG concentrations were selected in order to yield fixed N/P ratios required for the specific analysis.
  • the dsDNA/pOG-M-PEG mixtures were maintained at room temperature for 30 min under gentle agitation to equilibrate before analysis.
  • Electrophoretic analyses were performed to evaluate the complexation capacity of (Oligoguanidyl)6-maltotriosyl-N-acetyl-amino-hexanoate-poly(ethylenglycol)-NH2 with oligonucleotides.
  • N/P nitrogen/phosphorous
  • At the complex were added 3 ⁇ _ of loading buffer containing 30% glycerol and 0.25% xylene cyanol in water.
  • the dsDNA/pOG-M-PEG samples were analyzed by gel-electrophoresis at 100 V for 1 hours on a 12% polyacrylamide gel in Tris-Borate-EDTA (TBE) 1 X buffer.
  • the migrated dsDNA was visualized by immersion of the gel in a Gel Red solution previously prepared by dilution of 15 L of staining marker in 30 ml_ of milliQ water for 30 min.
  • Figure 8 shows the gel image obtained using a UV-Transilluminator.
  • DLS Dynamic Light Scattering
  • dsDNA/pOG-M-PEG samples containing at 100 pg/mL of pOG-M-PEG and 3 and 5 N/P ratio were analyzed by Transmission Electron Microscopy (TEM).
  • the samples were deposited on a small copper grid (400 mesh), covered by "holey film” carbon layer and analyzed in negative staining mode.
  • the contrast agent used was uranyl acetate 1 % w/v.
  • Figure 10A and Figure 10B show the TEM images of complexes prepared with 3 and 5 N/P ratio, respectively.
  • Figure 1 1 A-B and Figure 12A-B show the size of the samples at the two NP ratios and in buffer or DMEM medium, respectively.
  • dsDNA (100 ⁇ ) and pOG-M-PEG (20 ⁇ ) solutions were prepared by dissolving dsDNA and pOG-M-PEG in 10 mM TRIS, 150 mM NaCI, pH 7.4. Both the solutions were degassed and thermostated at 25 °C before the analysis. At 5 minutes intervals, 10 ⁇ _ of dsDNA solution was injected in 10 sec into the calorimeter cell containing 1 .5 ml_ pOG-M-PEG.
  • Figure 14 shows the isothermal calorimetry profiles of pOG-M-PEG titrated with dsDNA in 10 mM TRIS, 150 mM NaCI, pH 7.4 and relative table.
  • the human MCF-7 breast adenocarcinoma, human KB cervical carcinoma and murine MC3T3-E1 embryonic fibroblast cell lines were grown at 37°C, in 5% CO2 atmosphere, using DMEM medium supplemented with 15% FBS (Fetal Bovine Serum), 2 mM L-glutamine, 100 lU/mL penicillin, 100 pg/mL streptomycin and 0.25 g/mL of amphotericin B.
  • FBS Fetal Bovine Serum
  • MCF-7, KB, MC3T3-E1 cells were seeded in 96 well plate (5 x 10 3 cell/well). After 24 hours the medium was removed and the cells were incubated at 37°C with increasing concentrations of pOG-M-PEG and increasing dsDNA concentrations of dsDNA/pOG-M-PEG samples at 3 and 5 N/P ratio.
  • Figure 15 shows the viability of MCF-7 ( Figure 15A), KB ( Figure 15B) and MC3T3-E1 ( Figure 15C) cells incubated with dsDNA/pOG-M-PEG complexes with 3 and 5 N/P ratio for 24 and 48 hours.
  • Cy3-dsDNA labeled Cyanine 3-dsDNA
  • Cells were seeded in 6 well plate at a density of 1 .5 x 1 0 6 cells/well. The DMEM medium was removed and the cells were washed twice with PBS, pH 7.4, and then treated with 500 ⁇ _ of Cy3-dsDNA/pOG-M-PEG complex solutions at fixed concentrations of 125 nM labelled ONs.
  • MCF-7, KB and MC3T3-E1 cells were seeded at density of 5 x 10 4 cells/cm 2 and grown for 24 h at 37°C and 5% C02.
  • the wells were washed three times and incubated with 500 ⁇ _ of 5.0 pg/mL of DAPI solution for nuclei staining for 15 min at room temperature.
  • the fixed cells were treated with 4.5 ug/mL solution of wheat germ agglutinin 488 AlexaFluor.
  • the wells were washed three times with PBS solution and the three times with milliQ water.
  • the samples images were acquired using a Zeiss LSM 800 confocal microscopy equipped of an immersion lens with 63 X magnification.
  • the lasers were fixed at 405 nm, 488 nm and 561 nm to detect DAPI, 488 AlexaFluor and Cy3-dsDNA.
  • Thapa, B., et al. Asialoglycoprotein receptor-mediated gene delivery to hepatocytes using galactosylated polymers. Biomacromolecules, 2015. 16(9): p. 3008-3020.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Organic Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Crystallography & Structural Chemistry (AREA)
  • Biotechnology (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicinal Preparation (AREA)

Abstract

La présente invention concerne des composés de (guanidyl)x-oligosaccharides ayant une structure de type étoile possédant la capacité de transporter et d'administrer des médicaments ou des molécules biologiques tels que des oligonucléotides, des polypeptides et des protéines à travers des membranes biologiques. Les composés de (guanidyl)x-oligosaccharides de la présente invention peuvent former des conjugués avec des fractions fonctionnelles telles que des composés ayant une activité biologique, des fluorophores, des radionucléides, des macrosurfaces, des agents de ciblage et/ou des complexes de formes avec une macromolécule anionique telle que des ON, des peptides, des protéines, des acides nucléiques, des médicaments, des membranes cellulaires.
EP17703926.0A 2017-01-12 2017-01-12 Composés de type étoile de (guanidyl)x-oligosaccharides et conjugués ou complexes de ceux-ci Withdrawn EP3568404A1 (fr)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/EP2017/050534 WO2018130282A1 (fr) 2017-01-12 2017-01-12 Composés de type étoile de (guanidyl)x-oligosaccharides et conjugués ou complexes de ceux-ci

Publications (1)

Publication Number Publication Date
EP3568404A1 true EP3568404A1 (fr) 2019-11-20

Family

ID=57995172

Family Applications (1)

Application Number Title Priority Date Filing Date
EP17703926.0A Withdrawn EP3568404A1 (fr) 2017-01-12 2017-01-12 Composés de type étoile de (guanidyl)x-oligosaccharides et conjugués ou complexes de ceux-ci

Country Status (2)

Country Link
EP (1) EP3568404A1 (fr)
WO (1) WO2018130282A1 (fr)

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11729549B2 (en) 2019-12-30 2023-08-15 Harman International Industries, Incorporated Voice ducking with spatial speech separation for vehicle audio system
EP4285933A1 (fr) * 2022-05-30 2023-12-06 BioNTech SE Complexes oligosaccharides et utilisations
EP4285932A1 (fr) * 2022-05-30 2023-12-06 BioNTech SE Complexes oligosaccharides et utilisations
WO2023067123A1 (fr) * 2021-10-22 2023-04-27 BioNTech SE Complexes d'oligosaccharides et leurs utilisations

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR100756591B1 (ko) 1998-12-03 2007-09-10 포라 가세이 고교 가부시키가이샤 신규 화합물, 당해 화합물로부터 제조된 중합체 및 당해 중합체를 포함하는 조성물
WO2012097876A1 (fr) * 2011-01-21 2012-07-26 Universita' Degli Studi Di Padova Dérivés (guanidyl)-oligosaccharidiques de type couronne en tant qu'améliorateurs de la pénétration cellulaire pour l'administration intracellulaire de systèmes thérapeutiques colloïdaux

Also Published As

Publication number Publication date
WO2018130282A1 (fr) 2018-07-19

Similar Documents

Publication Publication Date Title
Muddineti et al. Cholesterol-grafted chitosan micelles as a nanocarrier system for drug-siRNA co-delivery to the lung cancer cells
Cavallaro et al. Polymeric nanoparticles for siRNA delivery: Production and applications
Ganesh et al. Hyaluronic acid based self-assembling nanosystems for CD44 target mediated siRNA delivery to solid tumors
Biswas et al. Lipid modified triblock PAMAM-based nanocarriers for siRNA drug co-delivery
US7001891B1 (en) Biodegradable polycation composition for delivery of an anionic macromolecule
AU2011353233B2 (en) Carrier for negatively charged drugs comprising a cationic lipid and a preparation method thereof
Lai et al. Nucleic acid delivery with chitosan and its derivatives
US6958325B2 (en) Cationic polysaccharide compositions
Tripathi et al. Linear polyethylenimine-graft-chitosan copolymers as efficient DNA/siRNA delivery vectors in vitro and in vivo
Layek et al. Hexanoic acid and polyethylene glycol double grafted amphiphilic chitosan for enhanced gene delivery: influence of hydrophobic and hydrophilic substitution degree
KR101912978B1 (ko) 페닐보론산기가 도입된 블록 공중합체 및 그의 용도
Huo et al. Polyion complex micelles composed of pegylated polyasparthydrazide derivatives for siRNA delivery to the brain
WO2018130282A1 (fr) Composés de type étoile de (guanidyl)x-oligosaccharides et conjugués ou complexes de ceux-ci
Gaspar et al. Biofunctionalized nanoparticles with pH-responsive and cell penetrating blocks for gene delivery
Yang et al. Protein corona precoating on redox-responsive chitosan-based nano-carriers for improving the therapeutic effect of nucleic acid drugs
JP2007145761A (ja) 細胞膜透過性ペプチド修飾多糖−コレステロールまたは多糖−脂質非ウイルス性ベクターおよびその製造方法
CN111214461B (zh) 糖靶向修饰siRNA纳米粒子的制备及应用
Kozlu et al. An aquaporin 4 antisense oligonucleotide loaded, brain targeted nanoparticulate system design
Hussein et al. Peptide-based targeted polymeric nanoparticles for siRNA delivery
US20160228567A1 (en) Hypoxia-Targeted Delivery System for Pharmaceutical Agents
Altangerel et al. PEGylation of 6-amino-6-deoxy-curdlan for efficient in vivo siRNA delivery
Kong et al. Benzylguanidine and galactose double-conjugated chitosan nanoparticles with reduction responsiveness for targeted delivery of doxorubicin to CXCR 4 positive tumors
Choi et al. Multi-layered nanogels with MMP-sheddable PEG masks: Preparation and promotion of tumor cell uptake by controlling surface characteristics
Malfanti et al. Oligo-guanidyl targeted bioconjugates forming rod shaped polyplexes as a new nanoplatform for oligonucleotide delivery
CN115151278A (zh) 用于核酸疗法的靶向肿瘤的多肽纳米颗粒递送系统

Legal Events

Date Code Title Description
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: UNKNOWN

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE

PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20190806

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

DAV Request for validation of the european patent (deleted)
DAX Request for extension of the european patent (deleted)
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20210803