WO2015057063A1 - Modified glycoprotein, protein-conjugate and process for the preparation thereof - Google Patents

Modified glycoprotein, protein-conjugate and process for the preparation thereof Download PDF

Info

Publication number
WO2015057063A1
WO2015057063A1 PCT/NL2014/050714 NL2014050714W WO2015057063A1 WO 2015057063 A1 WO2015057063 A1 WO 2015057063A1 NL 2014050714 W NL2014050714 W NL 2014050714W WO 2015057063 A1 WO2015057063 A1 WO 2015057063A1
Authority
WO
WIPO (PCT)
Prior art keywords
group
glycoprotein
antibody
conjugate
modified
Prior art date
Application number
PCT/NL2014/050714
Other languages
French (fr)
Inventor
Floris Louis Van Delft
Remon VAN GEEL
Maria Antonia WIJDEVEN
Jorge Merijn Mathieu Verkade
Ryan HEESBEEN
Original Assignee
Synaffix B.V.
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 Synaffix B.V. filed Critical Synaffix B.V.
Priority to CN201480068041.2A priority Critical patent/CN105814213B/en
Priority to EP14790774.5A priority patent/EP2935608A1/en
Priority to JP2016547822A priority patent/JP2016538877A/en
Priority to US15/029,123 priority patent/US10072096B2/en
Publication of WO2015057063A1 publication Critical patent/WO2015057063A1/en
Priority to US16/115,261 priority patent/US20190225706A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/40Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against enzymes
    • 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
    • A61K47/6871Medicinal 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 an enzyme
    • 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
    • C07K16/32Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against translation products of oncogenes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12PFERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
    • C12P21/00Preparation of peptides or proteins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12PFERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
    • C12P21/00Preparation of peptides or proteins
    • C12P21/005Glycopeptides, glycoproteins
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/40Immunoglobulins specific features characterized by post-translational modification
    • C07K2317/41Glycosylation, sialylation, or fucosylation

Definitions

  • the present invention relates to modified glycoproteins, in particular to glycoproteins comprising a modified sugar-moiety.
  • the invention also relates to a glycoprotein-conjugate wherein a glycoprotein according to the invention is conjugated to a molecule of interest.
  • Said molecule of interest may for example be a an active substance.
  • the invention further relates to a process for the preparation of a modified glycoprotein, and to a method for the preparation of a glycoprotein-conjugate.
  • the invention particularly relates to modified antibodies, antibody-conjugates, antibody- drug conjugates and methods for the preparation thereof.
  • Protein conjugates i.e. proteins conjugated to a molecule of interest via a linker
  • fluorescent labeling is a powerful technique for in vitro and in vivo visualisation
  • covalent immobilization of proteins is a useful strategy for industrial application and PEGylation of proteins leads to significantly enhanced circulation time.
  • the molecule of interest is a drug, for example a cytotoxic chemical.
  • Antibody-drug- conjugates are known in the art, and consist of a recombinant antibody covalently bound to a cytotoxic chemical via a synthetic linker.
  • Protein conjugates known from the prior art are commonly prepared by conjugation of a functional group to the side chain of amino acid lysine or cysteine, by acylation or alkylation, respectively.
  • conjugation takes place preferentially at lysine side chains with highest steric accessibility, the lowest pKa, or a combination thereof. Disadvantage of this method is that site-control of conjugation is low.
  • cysteines can be selectively liberated by a (partial) reductive step.
  • selective cysteine liberation by reduction is typically performed by treatment of a protein with a reducing agent ⁇ e.g. TCEP or DTT), leading to conversion of a disulfide bond into two free thiols.
  • the liberated thiols can then be alkylated with an electrophilic reagent, typically based on a maleimide chemistry, which generally proceeds fast and with high selectivity, or with haloacetamides, which also show strong preference for cysteine but side-reactions with lysine side-chains may be encountered.
  • an electrophilic reagent typically based on a maleimide chemistry, which generally proceeds fast and with high selectivity, or with haloacetamides, which also show strong preference for cysteine but side-reactions with lysine side-chains may be encountered.
  • T-DM1 is prepared by first (random) conjugation of lysines with succinimidyl 4-(N-maleimidomethyl)cyclohexane-l-carboxylate (SMCC), thereby effectively charging the antibody with maleimides.
  • SMCC succinimidyl 4-(N-maleimidomethyl)cyclohexane-l-carboxylate
  • the maleimide-functionalized antibody is treated with thiol-functionalized maytansinoid, leading to the conjugate.
  • An alternative strategy to prepare conjugates of a glycoprotein, a subclass of all proteins involves the generation of one or more aldehyde functions on the protein's glycan structure, either by chemical means (sodium periodate) or by enzymatic means (galactose oxidase).
  • the latter aldehyde function can subsequently be employed for a selective conjugation process, for example by condensation with a functionalized hydroxylamine or hydrazine molecule, thereby generating an oxime-linked or hydrazone-linked protein conjugate, respectively.
  • oximes and hydrazones in particular derived from aliphatic aldehydes, also show limited stability over time in water or at lower pH.
  • gemtuzumab ozogamicin is an oxime-linked antibody-drug conjugate and is known to suffer from premature deconjugation in vivo.
  • WO 2007/095506 and WO 2008/029281 disclose that the combination of GalT(Y289L) mutant with the C2- substituted azidoacetamido moiety 2-GalNAz-UDP leads to the incorporation of GalNAz at a terminal non-reducing GlcNAc of a glycan. Subsequent conjugation by Staudinger ligation or with copper-catalyzed click chemistry then provides the respective antibody conjugates wherein a fluorescent alkyne probe is conjugated to an antibody.
  • WO 2007/095506 and WO 2008/029281 further disclose that trimming of the glycan can take place with endo H, thereby hydrolyzing a GlcNAc-GlcNAc glycosidic bond and liberating a GlcNAc for enzymatic introduction of GalNAz.
  • galactose can be introduced to proteins featuring a terminal GlcNAc-moiety upon treatment with wild type Gal-Tl/UDP-Gal (leading to Gal-GlcN Ac-protein), while N-acetylgalactosamine can be introduced upon treatment with GalTl mutant Y289L (affording GalNAc-GlcNAc-protein). It has also been shown by Elling et al.
  • Conjugations of biomolecules based on thiols are well-known in the art.
  • reaction of thiols with maleimide is a fast and selective process, which typically rapidly leads to the desired conjugate.
  • Less popular but also regularly applied are halogenated acetamides that may also react with high selectivity with free thiols although chemoselectivity is compromised with respect to maleimide conjugation.
  • a particular advantage of conjugation with halogenated acetamides is the irreversible formation of a thioether, which compares favorably to maleimide conjugates with respect to stability. The latter stability also applied to conjugates formed by reaction of thiols with allenamide, as most recently reported by Abbas et al, Angew. Chem.
  • the invention relates to a process for the preparation of a modified glycoprotein, the process comprising contacting a glycoprotein comprising a glycan comprising a terminal GlcNAc-moiety with Su(A) x -P in the presence of a catalyst selected from the group consisting of /3(l,4)-galactosyltransf erases, /3(l,3)-N-galactosyltransferases, /3(l,4)-galactosyltransferases comprising a mutant catalytic domain and (1,3)- ⁇ - galactosyltransferases comprising a mutant catalytic domain; wherein Su(A) x is a sugar derivative Su comprising x functional groups A wherein x is 1, 2, 3 or 4 and wherein A is independently selected from the group consisting of a thiol group or a precursor thereof, a halogen, a sulfonyloxy group, a halogenated acetamido group,
  • b is 0 or 1;
  • d is O or l
  • G is a monosaccharide, or a linear or branched oligosaccharide comprising 2 to 20 sugar moieties.
  • the invention also relates to a glycoprotein comprising a glycan according to formula (105) or (106):
  • b is 0 or 1;
  • d is O or l
  • e is 0 or 1 ;
  • G is a monosaccharide, or a linear or branched oligosaccharide comprising 2 - 20 saccharide moieties;
  • Su(A) x is a sugar derivative Su comprising x functional groups A, wherein x is 1, 2, 3 or 4 and wherein A is independently selected from the group consisting a thiol group, a halogen, a sulfonyloxy group, a halogenated acetamido group, a mercaptoacetamido group and a sulfonated hydroxyacetamido group.
  • the invention also relates to the use of a glycoprotein according to the invention in the preparation of a glycoprotein-conjugate, wherein a glycoprotein-conjugate is defined as a glycoprotein that is conjugated to a molecule of interest D via a linker L; and to a process for the preparation of a glycoprotein-conjugate, said process comprising reacting a modified glycoprotein according to the invention with a linker- conjugate, wherein said linker-conjugate comprises a functional group B and one or more molecules of interest, wherein said functional group B is a functional group that is capable of reacting with a functional group A on a glycan of the modified glycoprotein, and wherein functional group A is as defined above.
  • the invention relates to modified antibodies, antibody-conjugates and antibody-drug conjugates. Description of the figures
  • Figure 1 shows examples of the different glycoforms of a protein that (A) can be obtained by regular expression followed by trimming with endo-glycosidase, or (B) by expression of a mAb in a mammalian system in the presence of swainsonine or by expression in an engineered host organism, e.g. Lecl CHO or Pichia, or (C) by trimming of the regular mixture of glycoforms (GO, Gl, G2, GOF, GIF and G2F) upon combined action of sialidase and galactosidase.
  • A can be obtained by regular expression followed by trimming with endo-glycosidase
  • B by expression of a mAb in a mammalian system in the presence of swainsonine or by expression in an engineered host organism, e.g. Lecl CHO or Pichia
  • C by trimming of the regular mixture of glycoforms (GO, Gl, G2, GOF, GIF and G2F) upon combined
  • Figure 2 shows a schematic representation of the enzymatic conversion of a GlcNAc-terminated protein with unnatural UDP-galactose derivative 1 (2- ketogalactose) or 2 (GalNAz), upon the action of galactosyl transferase mutant ⁇ (1,4)- Gal-T1(Y298L).
  • Figure 3 shows the structures of modified galactose derivatives (3-17) for transfer onto a GlcNAc-terminated sugar under the action of a galactosyl transferase (or a mutant thereof).
  • Figure 4 shows a range of thiol-derivatives or halogen derivatives of UDP- galactose (18-23) that may be transferred onto a GlcNAc-terminated sugar under the action of a galactosyl transferase (or a mutant thereof).
  • Figure 5 shows the scheme for the synthetic preparation of an acetyl-protected thiol variant (28a) or the deacetylated thiol variant (28b) or halogen variants (29+30) of UDP-GalNAc, substituted on the 2-NHAc moiety.
  • Figure 6 shows the scheme for the synthetic preparation of a biotin-substituted thiophenol (32), of azide- substituted thiophenol (41) and of azide-allenamide construct 43 from propionic amide precursos 42.
  • Figure 7 shows the structures of maleimide derivatives of biotin (33), MMAF (34), pyrene (44) and BCN (45).
  • Figure 8 shows a strategy for conversion of a trimmed monoclonal antibody 35 (comprising a core GlcNAc only).
  • Thiol-containing 36 can be reacted with maleimide derivatives 33 or 34, leading to conjugates 38 or 39, respectively.
  • halogenated derivative 36 can be conjugated by nucleophilic substitution of the halogen with a nucleophilic reagent such as DTT, MeOPhSH or N0 2 PhSH, or compound 32, leading to 40.
  • an azido-derivative of the antibody is formed (46).
  • the latter azide can be further conjugated with a BCN- toxin construct such as 47, leading to the antibody-drug conjugate 48.
  • Figure 11 shows a strategy for conversion of a thiolated monoclonal antibody 36 (comprising a core GlcNAc substituted with thiolated galactose derivative).
  • Thiol- containing 36 can be reacted with maleimide derivatives 43, 44 or 45 leading to conjugates 49, 50 or 51 respectively.
  • the latter conjugate 51, bearing the BCN-moiety can be further conjugated with an azide-biotin construct such as 52, leading to the antibody-biotin conjugate 53.
  • Figure 12 shows the synthetic route for the preparation of the acetyl-protected GalNAc derivatives UDP-GalNProSAc (57) and UDP-GalNBuSAc (58) and the thiol- containing GalNAc derivatives UDP-GalNProSH (59) and UDP-GalNBuSH (60). Also shown is the synthetic route for preparation of the maleimide conjugates of compounds 64-66, model compounds for conjugates of GalNAcSH, GalNProSH and GalNBuSH, respectively.
  • Figure 13 shows different glycoforms of a monoclonal antibody, e.g. IgG, which can be obtained by removing the native glycosylation site of a mAb and engineering a glycosylation site (based on sequence N-X-S/T, with X is any amino acid except proline) at another position.
  • a monoclonal antibody e.g. IgG
  • indefinite article “a” or “an” does not exclude the possibility that more than one of the elements is present, unless the context clearly requires that there is one and only one of the elements.
  • the indefinite article “a” or “an” thus usually means “at least one”.
  • the compounds disclosed in this description and in the claims may comprise one or more asymmetric centres, and different diastereomers and/or enantiomers may exist of the compounds.
  • the description of any compound in this description and in the claims is meant to include all diastereomers, and mixtures thereof, unless stated otherwise.
  • the description of any compound in this description and in the claims is meant to include both the individual enantiomers, as well as any mixture, racemic or otherwise, of the enantiomers, unless stated otherwise.
  • the structure of a compound is depicted as a specific enantiomer, it is to be understood that the invention of the present application is not limited to that specific enantiomer.
  • the compounds may occur in different tautomeric forms.
  • the compounds according to the invention are meant to include all tautomeric forms, unless stated otherwise.
  • the structure of a compound is depicted as a specific tautomer, it is to be understood that the invention of the present application is not limited to that specific tautomer.
  • Unsubstituted alkyl groups have the general formula C n H 2n+ i and may be linear or branched. Unsubstituted alkyl groups may also contain a cyclic moiety, and thus have the concomitant general formula C n H 2n -i. Optionally, the alkyl groups are substituted by one or more substituents further specified in this document. Examples of alkyl groups include methyl, ethyl, propyl, 2-propyl, t-butyl, 1-hexyl, 1-dodecyl, etc.
  • An aryl group comprises six to twelve carbon atoms and may include monocyclic and bicyclic structures.
  • the aryl group may be substituted by one or more substituents further specified in this document.
  • Examples of aryl groups are phenyl and naphthyl.
  • Arylalkyl groups and alkylaryl groups comprise at least seven carbon atoms and may include monocyclic and bicyclic structures.
  • the arylalkyl groups and alkylaryl may be substituted by one or more substituents further specified in this document.
  • An arylalkyl group is for example benzyl.
  • An alkylaryl group is for example 4-t-butylphenyl.
  • Heteroaryl groups comprise at least two carbon atoms (i.e. at least C 2 ) and one or more heteroatoms N, O, P or S.
  • a heteroaryl group may have a monocyclic or a bicyclic structure.
  • the heteroaryl group may be substituted by one or more substituents further specified in this document.
  • heteroaryl groups examples include pyridinyl, quinolinyl, pyrimidinyl, pyrazinyl, pyrazolyl, imidazolyl, thiazolyl, pyrrolyl, furanyl, triazolyl, benzofuranyl, indolyl, purinyl, benzoxazolyl, thienyl, phospholyl and oxazolyl.
  • Heteroarylalkyl groups and alkylheteroaryl groups comprise at least three carbon atoms (i.e. at least C 3 ) and may include monocyclic and bicyclic structures.
  • the heteroaryl groups may be substituted by one or more substituents further specified in this document.
  • an aryl group is denoted as a (hetero)aryl group, the notation is meant to include an aryl group and a heteroaryl group.
  • an alkyl(hetero)aryl group is meant to include an alkylaryl group and a alkylheteroaryl group
  • (hetero)arylalkyl is meant to include an arylalkyl group and a heteroarylalkyl group.
  • a C 2 - C 2 4 (hetero)aryl group is thus to be interpreted as including a C 2 - C 24 heteroaryl group and a C 6 - C 24 aryl group.
  • a C 3 - C 24 alkyl(hetero)aryl group is meant to include a C 7 - C 24 alkylaryl group and a C 3 - C 24 alkylheteroaryl group
  • a C 3 - C 24 (hetero)arylalkyl is meant to include a C 7 - C 24 arylalkyl group and a C 3 - C 24 heteroarylalkyl group.
  • alkyl groups, alkenyl groups, alkenes, alkynes, (hetero)aryl groups, (hetero)arylalkyl groups and alkyl(hetero)aryl groups may be substituted with one or more substituents selected from the group consisting of CI - C 12 alkyl groups, C 2 - C 12 alkenyl groups, C 2 - C 12 alkynyl groups, C 3 - C 12 cycloalkyl groups, C 5 - C 12 cycloalkenyl groups, C 8 - C 12 cycloalkynyl groups, Ci - C 12 alkoxy groups, C 2 - C 12 alkenyloxy groups, C 2 - C 12 alkynyloxy groups, C 3 - C 12 cycloalkyloxy groups, halogens, amino groups, oxo and silyl groups, wherein the silyl groups can be represented by the formula (R 10 ) 3 Si-, wherein R 10 is independently selected
  • An alkynyl group comprises a carbon-carbon triple bond.
  • An unsubstituted alkynyl group comprising one triple bond has the general formula C n H 2n -3 -
  • a terminal alkynyl is an alkynyl group wherein the triple bond is located at a terminal position of a carbon chain.
  • the alkynyl group is substituted by one or more substituents further specified in this document, and/or interrupted by heteroatoms selected from the group of oxygen, nitrogen and sulphur.
  • Examples of alkynyl groups include ethynyl, propynyl, butynyl, octynyl, etc.
  • a cycloalkynyl group is a cyclic alkynyl group.
  • An unsubstituted cycloalkynyl group comprising one triple bond has the general formula C n H 2n-5 .
  • a cycloalkynyl group is substituted by one or more substituents further specified in this document.
  • An example of a cycloalkynyl group is cyclooctynyl.
  • a heterocycloalkynyl group is a cycloalkynyl group interrupted by heteroatoms selected from the group of oxygen, nitrogen and sulphur.
  • a heterocycloalkynyl group is substituted by one or more substituents further specified in this document.
  • An example of a heterocycloalkynyl group is azacyclooctynyl.
  • a (hetero)aryl group comprises an aryl group and a heteroaryl group.
  • An alkyl(hetero)aryl group comprises an alkylaryl group and an alky 1 heteroaryl group.
  • a (hetero)arylalkyl group comprises a arylalkyl group and a heteroarylalkyl groups.
  • a (hetero)alkynyl group comprises an alkynyl group and a heteroalkynyl group.
  • a (hetero)cycloalkynyl group comprises an cycloalkynyl group and a heterocycloalkynyl group.
  • a (hetero)cycloalkyne compound is herein defined as a compound comprising a (hetero)cycloalkynyl group.
  • fused (hetero)cycloalkyne compounds i.e. (hetero)cycloalkyne compounds wherein a second ring structure is fused, i.e. annelated, to the (hetero)cycloalkynyl group.
  • a fused (hetero)cyclooctyne compound a cycloalkyl (e.g. a cyclopropyl) or an arene (e.g. benzene) may be annelated to the (hetero)cyclooctynyl group.
  • the triple bond of the (hetero)cyclooctynyl group in a fused (hetero)cyclooctyne compound may be located on either one of the three possible locations, i.e. on the 2, 3 or 4 position of the cyclooctyne moiety (numbering according to "IUPAC Nomenclature of Organic Chemistry", Rule A31.2).
  • the description of any fused (hetero)cyclooctyne compound in this description and in the claims is meant to include all three individual regioisomers of the cyclooctyne moiety.
  • alkyl group When an alkyl group, a (hetero)aryl group, alkyl(hetero)aryl group, a (hetero)arylalkyl group, a (hetero)cycloalkynyl group is optionally substituted, said groups are independently optionally substituted with one or more substituents independently selected from the group consisting of Ci - C 12 alkyl groups, C 2 - C 12 alkenyl groups, C 2 - C 12 alkynyl groups, C 3 - C 12 cycloalkyl groups, Ci - C 12 alkoxy groups, C 2 - C 12 alkenyloxy groups, C 2 - C 12 alkynyloxy groups, C 3 - C 12 cycloalkyloxy groups, halogens, amino groups, oxo groups and silyl groups, wherein the alkyl groups, alkenyl groups, alkynyl groups, cycloalkyl groups, alkoxy groups, alkenyloxy groups, al
  • sugar is herein used to indicate a monosaccharide, for example glucose (Glc), galactose (Gal), mannose (Man) and fucose (Fuc).
  • sugar derivative is herein used to indicate a derivative of a monosaccharide sugar, i.e. a monosaccharide sugar comprising substituents and/or functional groups. Examples of a sugar derivative include amino sugars and sugar acids, e.g.
  • glucosamine (Glc H 2 ), galactosamine (Gal H 2 ) N-acetylglucosamine (GlcNAc), N-acetylgalactosamine (GalNAc), sialic acid (Sia) which is also referred to as N-acetylneuraminic acid (NeuNAc), and N-acetylmuramic acid (MurNAc), glucuronic acid (GlcA) and iduronic acid (IdoA).
  • a sugar derivative also include compounds herein denoted Su(A)x, wherein Su is a sugar or a sugar derivative, and wherein Su comprises x functional groups A.
  • nucleotide is herein used in its normal scientific meaning.
  • nucleotide refers to a molecule that is composed of a nucleobase, a five-carbon sugar (either ribose or 2-deoxyribose), and one, two or three phosphate groups. Without the phosphate group, the nucleobase and sugar compose a nucleoside.
  • a nucleotide can thus also be called a nucleoside monophosphate, a nucleoside diphosphate or a nucleoside triphosphate.
  • the nucleobase may be adenine, guanine, cytosine, uracil or thymine.
  • nucleotide examples include uridine diphosphate (HDP), guanosine diphosphate (GDP), thymidine diphosphate (TDP), cytidine diphosphate (CDP) and cytidine monophosphate (CMP).
  • HDP uridine diphosphate
  • GDP guanosine diphosphate
  • TDP thymidine diphosphate
  • CDP cytidine diphosphate
  • CMP cytidine monophosphate
  • protein is herein used in its normal scientific meaning.
  • polypeptides comprising about 10 or more amino acids are considered proteins.
  • a protein may comprise natural, but also unnatural amino acids.
  • glycoprotein is herein used in its normal scientific meaning and refers to a protein comprising one or more monosaccharide or oligosaccharide chains (“glycans”) covalently bonded to the protein.
  • a glycan may be attached to a hydroxyl group on the protein (O-linked-glycan), e.g. to the hydroxyl group of serine, threonine, tyrosine, hydroxylysine or hydroxyproline, or to an amide function on the protein (N- glycoprotein), e.g. asparagine or arginine, or to a carbon on the protein (C- glycoprotein), e.g. tryptophan.
  • O-linked-glycan e.g. to the hydroxyl group of serine, threonine, tyrosine, hydroxylysine or hydroxyproline
  • N- glycoprotein e.g. asparagine or arginine
  • C- glycoprotein e.g. tryptophan
  • a glycoprotein may comprise more than one glycan, may comprise a combination of one or more monosaccharide and one or more oligosaccharide glycans, and may comprise a combination of N-linked, O-linked and C-linked glycans. It is estimated that more than 50% of all proteins have some form of glycosylation and therefore qualify as glycoprotein.
  • glycoproteins include PSMA (prostate-specific membrane antigen), CAL (candida antartica lipase), gp41, gpl20, EPO (erythropoietin), antifreeze protein and antibodies.
  • glycan is herein used in its normal scientific meaning and refers to a monosaccharide or oligosaccharide chain that is linked to a protein.
  • the term glycan thus refers to the carbohydrate-part of a glycoprotein.
  • the glycan is attached to a protein via the C-1 carbon of one sugar, which may be without further substitution (monosaccharide) or may be further substituted at one or more of its hydroxyl groups (oligosaccharide).
  • a naturally occurring glycan typically comprises 1 to about 10 saccharide moieties. However, when a longer saccharide chain is linked to a protein, said saccharide chain is herein also considered a glycan.
  • a glycan of a glycoprotein may be a monosaccharide.
  • a monosaccharide glycan of a glycoprotein consists of a single N-acetylglucosamine (GlcNAc), glucose (Glc), mannose (Man) or fucose (Fuc) covalently attached to the protein.
  • a glycan may also be an oligosaccharide.
  • An oligosaccharide chain of a glycoprotein may be linear or branched.
  • the sugar that is directly attached to the protein is called the core sugar.
  • a sugar that is not directly attached to the protein and is attached to at least two other sugars is called an internal sugar.
  • a sugar that is not directly attached to the protein but to a single other sugar, i.e. carrying no further sugar substituents at one or more of its other hydroxyl groups is called the terminal sugar.
  • a glycan may be an O-linked glycan, an N-linked glycan or a C-linked glycan.
  • O-linked glycan a monosaccharide or oligosaccharide glycan is bonded to an O- atom in an amino acid of the protein, typically via a hydroxyl group of serine (Ser) or threonine (Thr).
  • Thr threonine
  • a monosaccharide or oligosaccharide glycan is bonded to the protein via an N-atom in an amino acid of the protein, typically via an amide nitrogen in the side chain of asparagine (Asn) or arginine (Arg).
  • a monosaccharide or oligosaccharide glycan is bonded to a C-atom in an amino acid of the protein, typically to a C-atom of tryptophan (Trp).
  • the end of an oligosaccharide that is directly attached to the protein is called the reducing end of a glycan.
  • the other end of the oligosaccharide is called the non- reducing end of a glycan.
  • O-linked glycans For O-linked glycans, a wide diversity of chains exists. Naturally occurring O- linked glycans typically feature a serine or threonine-linked a-O-GalNAc moiety, further substituted with galactose, sialic acid and/or fucose. The hydroxylated amino acid that carries the glycan substitution may be part of any amino acid sequence in the protein.
  • N-linked glycans For N-linked glycans, a wide diversity of chains exists. Naturally occurring N- linked glycans typically feature an asparagine-linked ⁇ - ⁇ -GlcNAc moiety, in turn further substituted at its 4-OH with ⁇ -GlcNAc, in turn further substituted at its 4-OH with ⁇ -Man, in turn further substituted at its 3-OH and 6-OH with a-Man, leading to the glycan pentasaccharide Man 3 GlcNAc 2 .
  • the core GlcNAc moiety may be further substituted at its 6-OH by a-Fuc.
  • the pentasaccharide Man 3 GlcNAc 2 is the common oligosaccharide scaffold of nearly all N-linked glycoproteins and may carry a wide variety of other substituents, including but not limited to Man, GlcNAc, Gal and sialic acid.
  • the asparagine that is substituted with the glycan on its side-chain is typically part of the sequence Asn-X-Ser/Thr, with X being any amino acid but proline and Ser/Thr being either serine or threonine.
  • antibody is herein used in its normal scientific meaning.
  • An antibody is a protein generated by the immune system that is capable of recognizing and binding to a specific antigen.
  • An antibody is an example of a glycoprotein.
  • the term antibody herein is used in its broadest sense and specifically includes monoclonal antibodies, polyclonal antibodies, dimers, multimers, multispecific antibodies (e.g. bispecific antibodies), antibody fragments, and double and single chain antibodies.
  • antibody is herein also meant to include human antibodies, humanized antibodies, chimeric antibodies and antibodies specifically binding cancer antigen.
  • antibody is meant to include whole antibodies, but also fragments of an antibody, for example an antibody Fab fragment, (Fab') 2 , Fv fragment or Fc fragment from a cleaved antibody, a scFv-Fc fragment, a minibody, a diabody or a scFv.
  • antibody includes genetically engineered antibodies and derivatives of an antibody.
  • Antibodies, fragments of antibodies and genetically engineered antibodies may be obtained by methods that are known in the art.
  • Suitable marketed antibodies include, amongst others, abciximab, rituximab, basiliximab, palivizumab, infliximab, trastuzumab, alemtuzumab, adalimumab, tositumomab-1131, cetuximab, ibrituximab tiuxetan, omalizumab, bevacizumab, natalizumab, ranibizumab, panitumumab, eculizumab, certolizumab pegol, golimumab, canakinumab, catumaxomab, ustekinumab, tocilizumab, ofatumumab, denosumab, belimumab, ipilimumab and brentuximab.
  • treatment refers to obtaining a desired pharmacologic and/or physiologic effect.
  • the effect may be prophylactic in terms of completely or partially preventing a disease or symptom thereof and/or may be therapeutic in terms of a partial or complete cure for a disease and/or adverse affect attributable to the disease.
  • Treatment covers any treatment of a disease in a mammal, particularly in a human, and includes preventing the disease from occurring in a subject which may be predisposed to the disease but has not yet been diagnosed as having it; inhibiting the disease, i.e., arresting its development; relieving the disease, i.e., causing regression of the disease.
  • the present invention relates to a process for the preparation of a modified glycoprotein, the process comprising contacting a glycoprotein comprising a glycan comprising a terminal GlcNAc-moiety with Su(A) x -P in the presence of a suitable catalyst; wherein Su(A) x is a sugar derivative Su comprising x functional groups A wherein x is 1, 2, 3 or 4 and wherein A is independently selected from the group consisting of a thiol group or a precursor thereof, a halogen, a sulfonyloxy group, a halogenated acetamido group, a mercaptoacetamido group and a sulfonated hydroxyacetamido group; wherein P is a nucleotide; wherein a suitable catalyst is defined as a galactosyltransferase or a galactosyltransferase comprising a mutant catalytic domain, wherefore Su(A)
  • b is 0 or 1;
  • d is 0 or 1
  • G is a monosaccharide, or a linear or branched oligosaccharide comprising 2 to 20 sugar moieties.
  • the catalyst is selected from the group consisting of j3(l,4)- galactosyltransferases, /3(l,3)-N-galactosyltransferases, /3(l,4)-galactosyltransferases comprising a mutant catalytic domain and /3(l,3)-N-galactosyltransferases comprising a mutant catalytic domain.
  • the catalyst is described in more detail below.
  • the glycoprotein that is to be modified in the process according to the invention comprises a glycan, said glycan comprising a terminal GlcNAc-moiety, i.e. a Glc-NAc moiety that is present at the non-reducing end of the glycan.
  • Said glycan comprises one or more saccharide moieties, and may be linear or branched.
  • the GlcNAc- moiety of a glycan according to formula (101) wherein b is 1 i.e. a glycan consisting of a fucosylated GlcNAc
  • glycan (102) it is preferred that when d is 0 then e is 1, and when e is 0 then d is 1.
  • G represents a monosaccharide, or a linear or branched oligosaccharide comprising 2 to 20, preferably 2 to 12, more preferably 2 to 10, even more preferably 2 to 8 and most preferably 2 to 6 sugar moieties.
  • Sugar moieties that may be present in a glycan are known to a person skilled in the art, and include e.g. glucose (Glc), galactose (Gal), mannose (Man), fucose (Fuc), N-acetylglucosamine (GlcNAc), N- acetylgalactosamine (GalNAc), N-acetylneuraminic acid (NeuNAc) or sialic acid, xylose (Xyl).
  • the glycan comprising a terminal GlcNAc-moiety consists of one GlcNAc-moiety, and the glycan is a glycan according to formula (101), wherein b is 0.
  • said glycan consists of a fucosylated GlcNAc-moiety, and the glycan is a glycan according to formula (101), wherein b is 1.
  • said glycan is a glycan according to formula (102), wherein the core- GlcNAc, if present, is optionally fucosylated (b is 0 or 1).
  • a preferred glycan comprising a terminal GlcNAc-moiety is a glycan according to formula (101) or (102).
  • the glycoprotein that is to be modified in the process according to the invention is a glycoprotein according to formula (103) or (104):
  • Pr represents a protein
  • y is 1 to 20;
  • G is a linear or branched oligosaccharide
  • G comprises 2 to 12, more preferably 2 to 10, even more preferably 2 to 8 and most preferably 2 to 6 sugar moieties.
  • y is 1 to 12, more preferably y is 1, 2, 3, 4, 5, 6, 7 or 8, and even more preferably y is 1, 2, 3 or 4. Most preferably y is 1 or 2.
  • y is 2, 4, 6 or 8, preferably 2 or 4, most preferably 2. This embodiment is particularly preferred when the glycoprotein to be modified is an antibody, i.e. when Pr is Ab.
  • glycoprotein that is to be modified is a glycoprotein according to formula (103) or (104)
  • a glycoprotein mixture may be used as the starting glycoprotein, said mixture comprising glycoproteins comprising one or more fucosylated (b is 1) glycans (101) and/or (102) and/or one or more non-fucosylated (b is 0) glycans (101) and/or (101).
  • a glycoprotein comprising a glycan comprising a terminal GlcNAc-moiety is herein also referred to as a "terminal non-reducing GlcNAc-protein", and a glycan comprising a terminal GlcNAc-moiety is herein also referred to as a "terminal non- reducing GlcNAc-glycan”.
  • terminal non-reducing GlcNAc-protein includes a protein of formula (103) wherein b is 1
  • terminal non-reducing GlcNAc-glycan includes a glycan of formula (101) wherein b is 1.
  • the terminal non-reducing GlcNAc-protein may comprise a linear or a branched terminal non-reducing GlcNAc-glycan.
  • Said glycan is bonded via CI of the core-sugar- moiety to the protein, and said core-sugar-moiety preferably is a core-GlcNAc-moiety. Consequently, when the terminal non-reducing GlcNAc-glycan bonded to the protein is a glycan according to formula (102), it is preferred that d is 1.
  • CI of the core-sugar moiety of the terminal non- reducing GlcNAc-glycan is bonded to the protein via an N-glycosidic bond to a nitrogen atom in an amino acid residue in said protein, more preferably to an amide nitrogen atom in the side chain of an asparagine (Asn) or an arginine (Arg) amino acid.
  • CI of the core-sugar-moiety of the non-reducing GlcNAc-glycan may also be bonded to the protein via an O-glycosidic bond to an oxygen atom in an amino acid residue in said protein, more preferably to an oxygen atom in the side chain of a serine (Ser) or threonine (Thr) amino acid.
  • the core- sugar-moiety of said glycan is an O-GlcNAc-moiety or an O-GalNAc moiety, preferably an O-GlcNAc moiety.
  • CI of the core-sugar- moiety of the non-reducing GlcNAc-glycan may also be bonded to the protein via a C-glycosidic bond to a carbon atom on the protein, e.g. to tryptophan (Trp).
  • Trp tryptophan
  • a glycoprotein may comprise more than one glycan, and may comprise a combination of N-linked, O- linked and C-linked glycoproteins.
  • the terminal non-reducing GlcNAc-glycan may be present at a native glycosylation site of a protein, but may also be introduced on a different site on a protein.
  • the terminal non-reducing GlcNAc-protein is an antibody (Ab), the antibody comprising a glycan comprising a terminal GlcNAc- moiety.
  • Abs an antibody
  • Such an antibody is herein also referred to as a "terminal non-reducing GlcN Ac-antibody”.
  • a preferred terminal non-reducing GlcNAc-antibody is an antibody according to formula (103) or (104) as defined above, wherein Pr is Ab.
  • y is 1, 2, 3, 4, 5, 6, 7 or 8, and even more preferred that y is 2, 4, 6 or 8.
  • said antibody may be a whole antibody, but also an antibody fragment.
  • said antibody preferably comprises one or more, more preferably one, terminal non-reducing GlcNAc-glycan on each heavy chain.
  • Said whole antibody thus preferably comprises two or more, preferably two, four, six or eight of said glycans, more preferably two or four, and most preferably two glycans.
  • y is preferably 2, 4, 6 or 8, more preferably y is 2 or 4, and most preferably y is 2.
  • y is 1, 2, 3 or 4, and more preferably y is 1 or 2.
  • glycoprotein when said glycoprotein is an antibody, y is 1, 2 or 4.
  • said antibody is a monoclonal antibody (mAb).
  • mAb monoclonal antibody
  • said antibody is selected from the group consisting of IgA, IgD, IgE, IgG and IgM antibodies. More preferably, said antibody is an IgG antibody, and most preferably said antibody is an IgGl antibody.
  • the glycoprotein in an antibody is attached to the conserved N-glycosylation site in the Fc-fragment at asparagine in the region 290-305, typically N297.
  • the glycoprotein in an antibody is attached to a non-native glycosylation site in the antibody. It is further preferred that said non-native glycosylation site is a non-native N-glycosylation site.
  • a non-native glycosylation site may be introduced into the antibody via glycoengineering techniques.
  • Figure 1 shows a glycoprotein comprising a single, optionally fucosylated, GlcNAc-moiety; this GlcNAc-glycan may for example be linked to the protein via an N-glycosidic or an O- glycosidic bond.
  • Figure 1(b) shows a glycoprotein comprising a branched octasaccharide glycan wherein one of the branches comprises a terminal GlcNAc- moiety (this glycan is also referred to as GnM 5 )
  • the core-GlcNAc may optionally be fucosylated.
  • Figure 1(c) shows an antibody comprising a branched heptasaccharide glycan, wherein the core-GlcNAc moiety is optionally fucosylated and wherein all branches comprise a terminal GlcNAc-moiety.
  • a modified glycoprotein obtainable by the process for the modification of a glycoprotein according to the invention is defined as a glycoprotein comprising a modified glycan, said glycan comprising a GlcNAc-Su(A) x disaccharide-moiety at the non-reducing end, wherein Su(A) x is as defined above.
  • the modified glycan is according to the formula (105) or (106), wherein b, d and e, as well as their preferred embodiments, are as defined above, and wherein Su(A) x is a sugar derivative Su comprising x functional groups A, wherein x is 1, 2, 3 or 4 and wherein A is independently selected from the group consisting of a thiol group or a precursor thereof, a halogen, a sulfonyloxy group, a halogenated acetamido group, a mercaptoacetamido group and a sulfonated hydroxyacetamido group.
  • the present invention thus also relates to a process for the preparation of a modified glycoprotein, the process comprising contacting a glycoprotein comprising a glycan comprising a terminal GlcNAc-moiety with Su(A) x -P in the presence of a suitable catalyst; wherein Su(A) x is a sugar derivative Su comprising x functional groups A wherein x is 1, 2, 3 or 4 and wherein A is independently selected from the group consisting of a thiol group or a precursor thereof, a halogen, a sulfonyloxy group, a halogenated acetamido group, a mercaptoacetamido group and a sulfonated hydroxyacetamido group; wherein P is a nucleotide; wherein a suitable catalyst is defined as a galactosyltransferase or a galactosyltransferase comprising a mutant catalytic domain, wherefore Su(
  • the modified glycoprotein obtainable by the here described process is a glycoprotein according to formula (107) or (108), wherein Pr, b, d, e, y and Su(A) x , as well as their preferred embodiments, are as defined above.
  • a suitable catalyst is defined as a galactosyltransferase, or a galactosyltransferase comprising a mutant catalytic domain, wherefore Su(A) x -P is a substrate.
  • the catalyst is a galactosyltransferase
  • said galactosyltransferase preferably is a wild-type galactosyltransferase.
  • said mutant GalT domain may be present within a full-length GalT enzyme, but it may also be present in a recombinant molecule comprising a catalytic domain.
  • the catalyst is selected from the group consisting of ⁇ l,4)- galactosyltransferases, /3(l,3)-N-galactosyltransferases, ⁇ l,4)-galactosyltransferases comprising a mutant catalytic domain and /3(l,3)-N-galactosyltransferases comprising a mutant catalytic domain.
  • the catalyst is a wild-type galactosyltransferase, more preferably a wild-type ⁇ l,4)-galactosyltransf erase or a wild-type ⁇ l,3)-N- galactosyltransf erase, and even more preferably a wild-type ⁇ l,4)- galactosyltransf erase I.
  • ⁇ l,4)-Galactosyltransf erase I is herein further referred to as GalT.
  • the ⁇ l,4)-galactosyltransf erase I is selected from the group consisting of a bovine j8(l,4)-Gal-Tl, a human j8(l,4)-Gal-Tl, a human ⁇ (1,4)- Gal-T2, a human 3(1,4)-Gal-T3, a human 3(1,4)-Gal-T4 and a human ?(1,3)-Gal-T5.
  • This embodiment wherein the catalyst is a wild-type galactosyltransferase is particularly preferred when a functional group A in sugar derivative Su(A) x is present on C2 or C6, preferably C6, of said sugar derivative.
  • Su(A) x comprises not more than one functional group A, i.e. preferably x is 1.
  • Su(A) x and Su(A) x -P are described in more detail below.
  • the invention thus also relates to a process for the preparation of a modified glycoprotein, said process comprising a step of attaching, in the presence of a suitable catalyst, a modified sugar to a terminal GlcNAc-moiety on a glycoprotein;
  • a modified sugar is defined as a sugar comprising a functional group A wherein A is independently selected from the group consisting of a thiol group or a precursor thereof, a halogen, a sulfonyloxy group, a halogenated acetamido group, a mercaptoacetamido group and a sulfonated hydroxyacetamido group; and wherein a suitable catalyst is defined as a wild-type galactosyltransferase or a galactosyltransferase comprising a mutant catalytic domain, wherefore Su(A) x -P is a substrate; with the proviso that when the catalyst is a wild-type galacto
  • the invention relates to a process for the preparation of a modified glycoprotein, the process comprising contacting a glycoprotein comprising a glycan according to formula (101) or (102) as defined above with Su(A) x -P in the presence of a suitable catalyst; wherein Su(A) x is a sugar derivative Su comprising x functional groups A wherein x is 1, 2, 3 or 4 and A is independently selected from the group consisting of a thiol group or a precursor thereof, a halogen, a sulfonyloxy group, a halogenated acetamido group, a mercaptoacetamido group and a sulfonated hydroxyacetamido group; wherein P is a nucleotide; and wherein a suitable catalyst is defined as a wild-type galactosyltransferase or a galactosyltransferase comprising a mutant catalytic domain, wherefore Su
  • the wild-type galactosyltransferase is a ⁇ l,4)-galactosyltransf erase or a ⁇ l,3)-N-galactosyltransferase, more preferably a ⁇ l,4)-galactosyltransferase.
  • the catalyst is a galactosyltransferase comprising a mutant catalytic domain, preferably a ⁇ l,4)-galactosyltransf erase comprising a mutant catalytic domain or a ⁇ l,3)-N-galactosyltransferase comprising a mutant catalytic domain, more preferably a ⁇ l,4)-galactosyltransf erase comprising a mutant catalytic domain.
  • ⁇ l,4)-Galactosyltransferase I is herein further referred to as GalT.
  • the catalyst is selected from the group consisting of ⁇ l,4)-galactosyltransferases or ⁇ l,3)-N-galactosyltransferases, more preferably from the group of ⁇ l,4)-galactosyltransferases or ⁇ l,3)-N-galactosyltransferases, all comprising a mutant catalytic domain.
  • the catalyst is a ⁇ l,3)-N-galactosyltransferase comprising a mutant catalytic domain, and preferably said ⁇ l,3)-N- galactosyltransferase is a human ?(l-3)-Gal-T5.
  • the catalyst is a ⁇ l,4)-N-galactosyltransf erase comprising a mutant catalytic domain, more preferably, a ⁇ l,4)-galactosyltransferase I comprising a mutant catalytic domain, and even more preferably selected from the group consisting of a bovine ⁇ l,4)-Gal-Tl, a human /M-Gal-Tl, a human ?(1,4)-Gal-T2, a human?(1,4)-Gal-T3 and a human ?(1,4)-Gal-T4, all comprising a mutant catalytic domain.
  • the catalyst is a bovine ?(1,4)-Gal-Tl comprising a mutant catalytic domain.
  • a suitable catalyst for the process according to the invention is known in the art.
  • a suitable catalyst is for example a catalyst that comprises a mutant catalytic domain from a ⁇ l,4)-galactosyltransf erase I.
  • a catalytic domain herein refers to an amino acid segment that folds into a domain that is able to catalyze the linkage of the specific sugar derivative nucleotide Su(A) x -P to the terminal non-reducing GlcNAc- glycan in a specific process according to the invention.
  • ⁇ l,4)-galactosyltransferase I is herein further referred to as GalT.
  • Such mutant GalT catalytic domains are for example disclosed in J. Biol. Chem.
  • J. Biol. Chem. 2002, 277, 20833 and WO 2004/063344 disclose Tyr-289 mutants of bovine tf(l,4)-Gal-Tl, which are referred to as Y289L, Y289N and Y289I.
  • the method of preparation of said mutant catalytic domains Y289L, Y289N and Y289I is disclosed in detail in WO 2004/063344, p. 34, 1. 6 - p. 36, 1. 2, expressly incorporated by reference herein.
  • Mutant GalT domains that catalyze the formation of a glucose- ⁇ l,4)-N- acetylglucosamine bond are disclosed in WO 2004/063344 on p. 10, 1, 25 - p. 12, 1. 4 (expressly incorporated by reference herein).
  • Mutant GalT domains that catalyze the formation of an N-acetylgalactosamine-/3(l,4)-N-acetylglucosamine bond are disclosed in WO 2004/063344 on p. 12, 1, 6 - p. 13, 1. 2 (expressly incorporated by reference herein).
  • GalT domains that catalyze the formation of a N-acetylglucosamine- /3(l,4)-N-acetylglucosamine bond and a mannose-/3(l,4)-N-acetylglucosamine bond are disclosed in WO 2004/063344 on p. 12, 1, 19 - p. 14, 1. 6 (expressly incorporated by reference herein).
  • the disclosed mutant GalT domains may be included within full-length GalT enzymes, or in recombinant molecules containing the catalytic domains, as is disclosed in WO 2004/063344 on p. 14, 1, 31 - p. 16, 1. 28, expressly incorporated by reference herein.
  • GalT domain is for example Y284L, disclosed by Bojarova et al, Glycobiology 2009, 19, 509, expressly incorporated by reference herein, wherein Tyr284 is replaced by leucine.
  • GalT domain is for example R228K, disclosed by Qasba et al, Glycobiology 2002, 12, 691, expressly incorporated by reference herein, wherein Arg228 is replaced by lysine.
  • the catalyst may also comprise a mutant catalytic domain from a bovine ⁇ l,4)- galactosyltransferase, selected from the group consisting of GalT Y289N, GalT Y289F, GalT Y289M, GalT Y289V, GalT Y289G, GalT Y289I and GalT Y289A, preferably selected from the group consisting of GalT Y289F and GalT Y289M.
  • GalT Y289N, GalT Y289F, GalT Y289M, GalT Y289V, GalT Y289G, GalT Y289I and GalT Y289A may be provided via site-directed mutagenesis processes, in a similar manner as disclosed in WO 2004/063344, in Qasba et al, Prot. Expr. Pur. 2003, 30, 219 and in Qasba et al, J. Biol. Chem. 2002, 277, 20833 (all incorporated by reference) for Y289L, Y289N and Y289I.
  • GalT Y289N the tyrosine amino acid (Y) at position 289 is replaced by an asparagine (N) amino acid
  • GalT Y289F the tyrosine amino acid (Y) at position 289 is replaced by a phenyl alanine (F) amino acid
  • GalT Y289M said tyrosine is replaced by a methionine (M) amino acid
  • M methionine
  • GalT Y289V by a valine (V) amino acid
  • GalT Y289G by a glycine (G) amino acid
  • GalT Y289I by an isoleucine (I) amino acid
  • Y289A by an analine (A) amino acid.
  • said catalyst is a catalyst comprising a mutant catalytic domain from a /3(l,4)-galactosyltransferase, preferably from a bovine ⁇ (1,4)- Gal-Tl .
  • the catalyst is a catalyst comprising a mutant catalytic domain from a /3(l,4)-galactosyltransferase, preferably selected from the group consisting of bovine jff(l,4)-Gal-Tl GalT Y289L, GalT Y289N, GalT Y289I, GalT Y289F, GalT Y289M, GalT Y289V, GalT Y289G and GalT Y289A, more preferably selected from the group consisting of bovine ytf(l,4)-Gal-Tl GalT Y289L, GalT Y289N and GalT Y289I.
  • a mutant catalytic domain from a /3(l,4)-galactosyltransferase preferably selected from the group consisting of bovine jff(l,4)-Gal-Tl GalT Y289L, GalT Y289N, GalT Y289I
  • said catalyst is a catalyst comprising a GalT mutant catalytic domain selected from the group consisting of Y289L, Y289N, Y289I, Y284L, R228K, Y289F, Y289M, Y289V, Y289G and Y289A, preferably selected from the group consisting of Y289L, Y289N, Y289I, Y284L and R228K.
  • a GalT mutant catalytic domain selected from the group consisting of Y289L, Y289N, Y289I, Y284L, R228K, preferably selected from the group consisting of Y289L, Y289N, Y289I, Y284L and R228K.
  • said catalyst is a catalyst comprising a bovine ?(1,4)-Gal-Tl mutant catalytic domain selected from the group consisting of Y289F, Y289M, Y289V, Y289G and Y289A. More preferably said catalyst is a catalyst comprising a GalT mutant catalytic domain selected from the group consisting of Y289L, Y289N and Y289I, and most preferably said catalyst is a catalyst comprising a GalT mutant catalytic domain selected from the group consisting of Y289L.
  • a catalyst based on a(l,3)-N- galactosy transferase (further referred to as a3Gal-T), preferably a(l,3)-N- acetylgalactosaminyltransferase (further referred to as a3GalNAc-T), as disclosed in WO 2009/025646, incorporated by reference herein.
  • Mutation of a3Gal-T can broaden donor specificity of the enzyme, and make it an a3GalNAc-T.
  • Mutation of a3GalNAc- T can broaden donor specificity of the enzyme.
  • Polypeptide fragments and catalytic domains of a(l,3)-N-acetylgalactosaminyltransferases are disclosed in WO 2009/025646 on p. 26, 1. 18 - p. 47, 1. 15 and p. 77, 1. 21 - p. 82, 1. 4 (both expressly incorporated by reference herein).
  • the process for the preparation of a modified glycoprotein according to the invention is preferably performed in a suitable buffer solution, such as for example phosphate, buffered saline (e.g. phosphate-buffered saline, tris-buffered saline), citrate, HEPES, tris and glycine.
  • a suitable buffer solution such as for example phosphate, buffered saline (e.g. phosphate-buffered saline, tris-buffered saline), citrate, HEPES, tris and glycine.
  • Suitable buffers are known in the art.
  • the buffer solution is phosphate-buffered saline (PBS) or tris buffer.
  • the process is preferably performed at a temperature in the range of about 4 to about 50°C, more preferably in the range of about 10 to about 45°C, even more preferably in the range of about 20 to about 40°C, and most preferably in the range of about 30 to about 37°C.
  • the process is preferably performed a pH in the range of about 5 to about 9, preferably in the range of about 5.5 to about 8.5, more preferably in the range of about 6 to about 8. Most preferably, the process is performed at a pH in the range of about 7 to about 8.
  • Su(A) x is defined as a sugar derivative Su comprising x functional groups A, wherein A is independently selected from the group consisting of a thiol group or a precursor thereof, a halogen, a sulfonyloxy group, a halogenated acetamido group, a mercaptoacetamido group and a sulfonated hydroxyacetamido group, and wherein x is 1, 2, 3 or 4.
  • a Su(A) x -moiety may also be referred to as a "modified sugar".
  • a modified sugar is herein defined as a sugar or a sugar derivative, said sugar or sugar derivative comprising 1, 2, 3 or 4 functional groups A, wherein A is selected from the group consisting of a thiol group or a precursor thereof, a halogen, a sulfonyloxy group, a halogenated acetamido group, a mercaptoacetamido group and a sulfonated hydroxyacetamido group.
  • a modified sugar or sugar derivative comprises e.g. a thiol group
  • said sugar or sugar derivative may be referred to as a thiol-modified sugar or sugar derivative.
  • a modified sugar or sugar derivative comprises e.g. a thiol- precursor group
  • said sugar or sugar derivative may be referred to as a thiol-precursor- modified sugar or sugar derivative.
  • a modified sugar or sugar derivative comprises e.g. a halogen
  • said sugar or sugar derivative may be referred to as a halogen-modified sugar or sugar derivative.
  • a modified sugar or sugar derivative comprises e.g. a sulfonyloxy group
  • said sugar or sugar derivative may be referred to as a sulfonyloxy-modified sugar or sugar derivative.
  • a thiol group is herein defined as a -[C(R 7 )2] 0 SH group, wherein R 7 is independently selected from the group consisting of hydrogen, halogen and an (optionally substituted) Ci - C 24 alkyl group, and o is 0 - 24.
  • R 7 is hydrogen or a Ci, C 2 , C 3 or C 4 alkyl group, more preferably R 7 is hydrogen or -CH 3 .
  • o is 0 - 10, more preferably 0, 1, 2, 3, 4, 5 or 6. More preferably, R 7 is hydrogen, -CH 3 or a C 2 alkyl group and/or o is 0, 1, 2, 3 or 4.
  • R 7 is hydrogen and o is 0, 1, 2 or 3, more preferably o is 1 or 2, most preferably o is 0 or 1. Most preferably o is 0. Most preferably, said thiol group is -CH 2 CH 2 CH 2 SH, -CH 2 CH 2 SH, -CH 2 SH or -SH, preferably -SH.
  • a precursor of a thiol group is herein defined as a -[C(R 7 ) 2 ] 0 SC(0)CH 3 group, wherein R 7 and o, as well as their preferred embodiments, are as defined above for a thiol group.
  • said thiol-precursor is -CH 2 CH 2 CH 2 SC(0)CH 3 , -CH 2 CH 2 SC(0)CH 3 , -CH 2 SC(0)CH 3 or -SC(0)CH 3 , preferably -SC(0)CH 3 .
  • a sugar derivative Su(A) x wherein A is a precursor of a thiol group may be used.
  • the thiol-precursor is converted to a thiol group.
  • a halogen is herein defined as F, CI, Br or I.
  • said halogen is CI or Br, more preferably CI.
  • a sulfonyloxy group is herein defined as a -[C(R 7 ) 2 ] 0 OS(0) 2 R 8 group, wherein R 7 and o are as defined above for a thiol group, and R 8 is selected from the group consisting of Ci - C 24 alkyl groups, C 6 - C 24 aryl groups, C 7 - C 24 alkylaryl groups and C 7 - C 24 arylalkyl groups.
  • R 8 is preferably a Ci - C 4 alkyl group, a C 6 - Ci 2 aryl group, a C 7 - Ci 2 alkylaryl group or a C 7 - Ci 2 arylalkyl group, more preferably a Ci - C 4 alkyl group, a C 7 - Ci 2 alkylaryl group or a C 7 - Ci 2 arylalkyl group, even more preferably -CH 3 , -C 2 H 5 , a C 3 linear or branched alkyl group, a phenyl group or a C 7 alkylaryl group.
  • R 8 is most preferably a methyl group, an ethyl group, a phenyl group or a p-tolyl group.
  • R 7 is hydrogen or a Ci, C 2 , C 3 or C 4 alkyl group, more preferably R 7 is hydrogen or -CH 3 .
  • o is 0 - 10, more preferably 0, 1, 2, 3, 4, 5 or 6. More preferably, R 7 is hydrogen, -CH 3 or a C 2 alkyl group and/or o is 0, 1, 2, 3 or 4. Even more preferably R 7 is hydrogen and o is 1 or 2, most preferably o is 0.
  • R 8 is preferably a Ci - C 4 alkyl group, a C 7 - C 12 alkylaryl group or a C 7 - C 12 arylalkyl group, more preferably -CH 3 , -C 2 H 5 , a C 3 linear or branched alkyl group or a C 7 alkylaryl group.
  • the sulfonyloxy group is a mesylate group, -OS(0) 2 CH 3 , a benzenesulfonate group (-OS(0) 2 (C 6 H 5 ) or a tosylate group (-OS(0) 2 (C 6 H 4 CH 3 ).
  • a halogenated acetamido group is herein defined as an - HC(0)[C(R 7 ) 2 ] 0 X group, wherein R 7 is independently selected from the group consisting of hydrogen, halogen and an (optionally substituted) Ci - C 24 alkyl group, X is F, CI, Br or I, and o is 0 - 24.
  • R 7 is hydrogen or a Ci, C 2 , C 3 or C 4 alkyl group, more preferably R 7 is hydrogen or -CH 3 , most preferably hydrogen.
  • o is 0 to 10, more preferably 1, 2, 3, 4, 5 or 6, even more preferably 1, 2, 3 or 4 and most preferably o is 1.
  • R 7 is hydrogen, -CH 3 or a C 2 alkyl group and/or o is 1, 2, 3 or 4 and most preferably R 7 is hydrogen and o is 1.
  • X is CI or Br, more preferably X is CI.
  • R 7 is hydrogen, X is CI and o is 1.
  • a mercaptoacetamido group is herein defined as an - HC(0)[C(R 7 ) 2 ] o SH group, wherein R 7 is independently selected from the group consisting of hydrogen, halogen and an (optionally substituted) Ci - C 24 alkyl group and o is 0 - 24.
  • R 7 is hydrogen or a Ci, C 2 , C 3 or C 4 alkyl group, more preferably R 7 is hydrogen or -CH 3 , most preferably hydrogen.
  • o is 0 to 10, more preferably 1, 2, 3, 4, 5 or 6, even more preferably 1, 2, 3 or 4 and most preferably o is 2, 3 or 4.
  • R 7 is hydrogen, -CH 3 or a C 2 alkyl group and/or o is 1, 2, 3 or 4. More preferably, R 7 is hydrogen and o is 1, 2, 3 or 4. Most preferably, R 7 is hydrogen and o is 1, 2 or 3, preferably 1.
  • Preferred examples include a mercaptoethanoylamido group, a mercaptopropanoylamido group, a mercaptobutanoylamido group and a mercapto- pentanoylamido group, preferably a mercaptopropanoylamido group.
  • a sulfonated hydroxyacetamido group is herein defined as a - HC(0)[C(R 7 ) 2 ] 0 OS(0) 2 R 8 group, wherein R 7 is independently selected from the group consisting of hydrogen, halogen and an (optionally substituted) Ci - C 24 alkyl group, R 8 is selected from the group consisting of Ci - C 24 alkyl groups, C 6 - C 24 aryl groups, C 7 - C 24 alkylaryl groups and C 7 - C 24 arylalkyl groups, and o is 0 - 24.
  • R 8 is preferably a Ci - C 4 alkyl group, a C 6 - Ci 2 aryl group, a C 7 - Ci 2 alkylaryl group or a C 7 - Ci 2 arylalkyl group, more preferably -CH 3 , -C 2 H 5 , a C 3 linear or branched alkyl group, a C 6 - C9 aryl group or a C 7 alkylaryl group.
  • the sulfonyloxy group is a mesylate group -OS(0) 2 CH 3 , a benzenesulfonate group -OS(0) 2 (C 6 H 5 ) or a tosylate group -OS(0) 2 (C 6 H 4 CH 3 ).
  • R 7 is hydrogen or a Ci, C 2 , C 3 or C 4 alkyl group, more preferably R 7 is hydrogen or -CH 3 , most preferably hydrogen.
  • Preferably o is 0 to 10, more preferably 1, 2, 3, 4, 5 or 6, even more preferably 1, 2, 3 or 4 and most preferably o is 1. More preferably, R 7 is hydrogen, -CH 3 or a C 2 alkyl group and/or o is 1, 2, 3 or 4. Even more preferably R 7 is hydrogen and o is 1, 2 or 3. Yet even more preferably, R 7 is H, o is 1 and R 8 is a mesylate group, a benzenesulfonate group or a tosylate group. Most preferably, R 7 is hydrogen, R 8 is -CH 3 and o is 1.
  • the sugar derivative Su(A) x may comprise one or more functional groups A.
  • each functional group A is independently selected, i.e. one Su(A) x may comprise different functional groups A, e.g. a thiol group and a halogen, etc.
  • x is 1 or 2, more preferably x is 1.
  • functional group A is a thiol or a halogen, more preferably a halogen.
  • Sugar derivative Su(A) x is derived from a sugar or a sugar derivative Su, e.g. an amino sugar or an otherwise derivatized sugar.
  • sugars and sugar derivatives include galactose (Gal), mannose (Man), glucose (Glc), N-acetylneuraminic acid or sialic acid (Sial) and fucose (Fuc).
  • An aminosugar is a sugar wherein a hydroxyl (OH) group is replaced by an amine group and examples include glucosamine (Glc H 2 ) and galactosamine (Gal H 2 ).
  • examples of an otherwise derivatized sugar include N-acetylneuraminic acid (sialic acid, Sia or NeuNAc) or fucose (Fuc).
  • Sugar derivative Su(A) x is preferably derived from galactose (Gal), mannose
  • Man N-acetylglucosamine (GlcNAc), glucose (Glc), N-acetylgalactosamine (GalNAc), fucose (Fuc) and N-acetylneuraminic acid (sialic acid Sia or NeuNAc), preferably from the group consisting of GlcNAc, Glc, Gal and GalNAc. More preferably Su(A) x is derived from Gal or GalNAc, and most preferably Su(A) x is derived from GalNAc.
  • the one or more functional groups A in Su(A) x may be linked to the sugar or sugar derivative Su in several ways.
  • the one or more functional groups A may be bonded to C2, C3, C4 and/or C6 of the sugar or sugar derivative, instead of a hydroxyl (OH) group.
  • OH hydroxyl
  • fucose lacks an OH-group on C6, if A is bonded to C6 of Fuc, then A takes the place of an H-atom.
  • the one or more functional groups A in Su(A) x may be present on C2 and/or C6 of the sugar or sugar derivative Su.
  • A is preferably selected from the group consisting of a halogenated acetamido group, a mercaptoacetamido group and a sulfonated hydroxyacetamido group.
  • A is present on C2 of a 2-aminosugar derivative, e.g. GalNAc or GlcNAc, A is preferably selected from the group consisting of a halogen, a thiol group and a sulfonyloxy group.
  • A is a thiol group
  • it is preferred that said thiol group is present on the 6- position of a sugar derivative or on a 2-aminosugar derivative.
  • An example of Su(A)x wherein A is a thiol group is 2-(mercaptoacetamido)-2-deoxy-galactose (2- GalNAcSH).
  • Another example of Su(A)x wherein A is a thiol group is 6-mercapto-6- deoxy-galactose.
  • A is a halogen
  • it is preferred that said halogen is present on the 6-position of a sugar derivative or on a 2-amino sugar derivative.
  • An example of Su(A)x wherein A is a halogen is 2-(chloroacetamido)-2-deoxy-galactose (2-GalNAcCl).
  • Another example of Su(A)x wherein A is a halogen is 6-iodo-6-deoxy-galactose.
  • Another example of Su(A)x wherein A is a halogen is 6-(chloroacetamido)-6-deoxy-galactose.
  • A is a sulfonyloxy group
  • it is preferred that said sulfonyloxy group is present on the 6-position of a sugar derivative or on a 2-amino sugar derivative.
  • An example of Su(A)x wherein A is a sulfonyloxy group is 2- (methylsulfonyloxyacetamido)-2-deoxy-galactose (2-GalNAcOMs).
  • Another example of Su(A)x wherein A is a sulfonyloxy group is 2-(benzenesulfonyloxyacetamido)-2- deoxy-galactose (2-GalNAcOMs.
  • Another example of Su(A)x wherein A is a sulfonyloxy group is 6-(methylsulfonyl)-galactose.
  • A is a halogenated acetamido group, a mercaptoacetamido group or a sulfonated hydroxyacetamido group it is preferred that said groups are present on the 6- position of a sugar derivative.
  • Su(A) x -P include 6-A-6-deoxygalactose-UDP (6-A-Gal- UDP), such as 6-chloro-6-deoxygalactose-UDP (6-ClGal-UDP), 6-thio-6- deoxygalactose-UDP (6-HSGal-UDP) or 2-A-2-deoxygalactose-UDP (2-A-Gal-UDP), such as 2-chloro-2-deoxygalactose-UDP (2-ClGal-UDP), 2-thio-2-deoxygalactose- UDP (2-HSGal-UDP).
  • 6-A-6-deoxygalactose-UDP (6-A-Gal- UDP)
  • 6-chloro-6-deoxygalactose-UDP (6-ClGal-UDP
  • 6-thio-6- deoxygalactose-UDP 6-HSGal-UDP
  • A may be indirectly substituted to the sugar derivative as part of an acetamido group that in turn is substituting a hydroxyl group.
  • A may be indirectly substituted to the sugar derivative as part of another functional group that in turn is substituting a hydroxyl group or is attached to a hydroxyl group.
  • Examples of such other functional group include an (hetero)alkyl chain or a (hetero)aryl chain.
  • P is herein defined as a nucleotide.
  • P is preferably selected from the group consisting of a nucleoside monophosphate and a nucleoside diphosphate, more preferably from the group consisting of uridine diphosphate (UDP), guanosine diphosphate (GDP), thymidine diphosphate (TDP), cytidine diphosphate (CDP) and cytidine monophosphate (CMP), more preferably from the group consisting of uridine diphosphate (UDP), guanosine diphosphate (GDP), cytidine diphosphate and (CDP). Most preferably, P is UDP.
  • Su(A) x -UDP uridine diphosphates linked to thiol-, halo- and mesityloxy-substitued sugars and sugar derivatives
  • A a thiol precursor
  • o 1, 2, 3, 4, 5 or 6, preferably 1, 2, 3 or 4, more preferably 1, 2 or 3.
  • R is H or Ac.
  • sugar derivative nucleotides Su(A) x -P that may be employed in the process for the preparation of a modified glycoprotein according to the invention are a substrate for a wild type galactosyltransferase.
  • the process according to the invention may be performed in the presence of a wild type galactosyltransferase, preferably a wild type /3(l,4)-galactosyltransferase, more preferably a /3(l,4)-galactosyltransf erase I, as a catalyst.
  • Su(A) x -P is selected from the group consisting of Su(A) x -P wherein x is 1 and wherein A is present on C6 of the sugar or sugar derivative, and wherein A is as defined above.
  • A may be directly substituted to the sugar derivative instead of an hydroxyl group.
  • A may be indirectly substituted to the sugar derivative as part of an acetamido group that in turn is substituting a hydroxyl group.
  • A may be indirectly substituted to the sugar derivative as part of another functional group that in turn is substituting a hydroxyl group or is attached to a hydroxyl group.
  • Examples of such other functional group include an (hetero)alkyl chain or a (hetero)aryl chain.
  • Su(A) x comprises 1 or 2 functional groups A, i.e. preferably x is 1 or 2. More preferably, x is 1. In a preferred embodiment, Su is galactose (Gal). In a further preferred embodiment, x is 1 or 2 and Su is Gal, and most preferably, x is 1 and Su is Gal.
  • Su(A) x is selected from the group consisting of 6- GalNAcCl, 6-GalNAcSH, 2-GalNAcCl, 2-GalNAcSH, 6-ClGal, 2-ClGal, 2-HSGal and 6-HSGal, more preferably form the group consisting of 6-GalNAcCl, 6-GalNAcSH, 2- GalNAcCl, 2-GalNAcSH, 6-ClGal-and 2-ClGal.
  • x is 1 and Su(A) x is selected from the group consisting of 6-GalNAcCl, 6-GalNAcSH, 2-GalNAcCl, 2-GalNAcSH, 6-ClGal, 2- ClGal, 2-HSGal and 6-HSGal, more preferably from the group consisting of 6- GalNAcCl, 6-GalNAcSH, 2-GalNAcCl, 2-GalNAcSH, 6-ClGal-and 2-ClGal.
  • A is a thiol group or a halogen.
  • Su(A) x is selected from the group consisting of 2-GalNAcSH, 2-GalNAcX, 2-GalNAcOS(0) 2 R 8 , 6-GalNAcSH, 6- GalNAcX and 6-GalNAcOS(0) 2 R 8 .
  • x is 1 or 2 and Su(A)x is selected from the group consisting of 2-GalNAcSH, 2-GalNAcX, 2- GalNAcOS(0) 2 R 8 , 6-GalNAcSH, 6-GalNAcX and 6-GalNAcOS(0) 2 R 8 .
  • x is 1 and Su(A) x is selected from the group consisting of 2- GalNAcSH, 2-GalNAcX, 2-GalNAcOS(0) 2 R 8 , 6-GalNAcSH, 6-GalNAcX and 6- GalNAcOS(0) 2 R 8 , wherein X is CI or Br, preferably CI.
  • 2-GalNAcX-UDP is 2-GalNAcCl-UDP or 2- GalNAcBr-UDP, more preferably 2-GalNAcCl-UDP
  • 6-GalNAcX-UDP is 6- GalNAcCl-UDP or 6-GalNAcBr-UDP, more preferably 6-GalNAcCl-UDP
  • R 8 in 2-GalNAcOS(0) 2 R 8 -UDP is methyl, phenyl or p-tolyl, most preferably methyl
  • R 8 in 6-GalNAcOS(0) 2 R 8 -UDP is methyl, phenyl or p- tolyl, most preferably R 8 is methyl.
  • Su(A) x -P is selected from the group consisting of 6-HSGal-UDP, 6-XGal-UDP, 6-R 8 S(0) 2 OGal-UDP, and the catalyst is a wild-type human GalT; wherein X is CI, Br or I; and wherein R 8 is a methyl group, an ethyl group, a phenyl group or a p-tolyl group.
  • X is more preferably CI or Br, most preferably CI.
  • R 8 is more preferably methyl, phenyl or p-tolyl, most preferably methyl.
  • the human GalT is preferably a human /3 ⁇ 4-Gal-Tl, a human ?(1,4)-Gal-T2, a human ytf(l,4)-Gal-T3 and a human ytf(l,4)-Gal-T4.
  • the process for the preparation of a modified glycoprotein according to the invention may further comprise a step comprising providing a glycoprotein comprising a glycan comprising a terminal GlcNAc-moiety at the non-reducing end.
  • the invention thus also relates to a process for the preparation of a modified glycoprotein, the process comprising the steps of:
  • Su(A) x is a sugar derivative Su comprising x functional groups A wherein x is 1, 2, 3 or 4 and wherein A is independently selected from the group consisting of a thiol group or a precursor thereof, a halogen, a sulfonyloxy group, a halogenated acetamido group, a mercaptoacetamido group and a sulfonated hydroxy acetamido group; wherein P is a nucleotide; wherein a suitable catalyst is defined as a galactosyltransferase or a galactosyltransferase comprising a mutant catalytic domain, wherefore Su(A) x -P is a substrate; and wherein a glycan
  • the catalyst in step(ii) is selected from the group consisting of (1,4)- galactosyltransferases, /3(l,3)-N-galactosyltransferases, /3(l,4)-galactosyltransferases comprising a mutant catalytic domain and /3(l,3)-N-galactosyltransferases comprising a mutant catalytic domain.
  • the catalyst is described in more detail above.
  • a glycoprotein comprising a glycan comprising a terminal GlcNAc-moiety at the non-reducing end may be provided in several ways, for example by (a) trimming of N-glycoprotein with an endo-glycosidase as described in EMBO J.2001, 72, 3046 (incorporated by reference) or (b) expression of hybrid N-glycoprotein in the presence of swainsonine as for example described by Satoh et al. in Glycobiology 2006, 77, 104-1 18, incorporated by reference (followed by sialidase/galactosidase treatment).
  • the invention also relates to a process for the preparation of a modified glycoprotein, the process comprising the steps of:
  • Su(A) x is a sugar derivative Su comprising x functional groups A wherein x is 1, 2, 3 or 4 and wherein A is independently selected from the group consisting of a thiol group or a precursor thereof, a halogen, a sulfonyloxy group, a halogenated acetamido group, a mercaptoacetamido group and a sulfonated hydroxy acetamido group; wherein P is a nucleotide; wherein a suitable catalyst is defined as a galactosyltransferase or a galactosyltransferase comprising a mutant catalytic domain, wherefore Su(A) x -P is a substrate; and wherein a glycan
  • the catalyst is selected from the group consisting of (1,4)- galactosyltransferases, /3(l,3)-N-galactosyltransferases, /3(l,4)-galactosyltransferases comprising a mutant catalytic domain and /3(l,3)-N-galactosyltransferases comprising a mutant catalytic domain.
  • the catalyst is described in more detail above.
  • a glycan may be attached to glycoprotein via CI of a GlcNAc-residue, that is bonded to the amide side chain of an asparagine amino acid that is part of the glycoprotein.
  • the oligosaccharide may be attached via an N-glycosidic bond, in most cases to an asparagine (Asn) or arginine (Arg) amino acid side chain.
  • glycans Numerous different types exist. For example the Fc regions of Ig antibodies bear a highly conserved N-glycosylation site. The N-glycans attached to this site are predominantly core-fucosylated diantennary structures of the complex type.
  • Another type of glycan is the class of high mannose glycans. High-mannose typically comprises two N-acetylglucosamines and a varying number of mannose residues.
  • a glycan may be bonded to the glycoprotein via a GlcNAc- residue, and this GlcNAc residue may be fucosylated.
  • this is denoted by b: when b is 0, said GlcNAc-residue is non- fucosylated and when b is 1, said GlcNAc is fucosylated.
  • a second GlcNAc-residue is bonded to the GlcNac- residue that is directly bonded to the glycoprotein, as is also seen in Figure 1, (b) and (c).
  • a glycan wherein a second GlcNAc-residue is bonded to the GlcNac-residue that is directly bonded to the glycoprotein can be trimmed in order to obtain a glycoprotein comprising a glycan comprising a terminal GlcNAc-moiety according to formula (101) at the non-reducing end. Trimming occurs in between said two GlcNAc-residues.
  • Trimming of a glycan provides a glycan according to formula (101), as shown in Figure 1 (a).
  • a GlcNAc-residue that is covalently bonded to an N-glycosylation site, preferably via an N-glycosidic bond via CI of said GlcNAc and an amide present in an amino acid side chain of the antibody, is herein referred to as a "terminal GlcNAc-moiety", and also "core-GlcNAc-moiety”. Said terminal GlcNAc- moiety or core-GlcNAc-moiety is optionally fucosylated.
  • the trimming of a glycan occurs by the action of a suitable enzyme, and after trimming the oligosaccharide (also referred to as glycan) a terminal GlcNAc-moiety is obtained.
  • a glycan comprising a terminal GlcNAc-moiety according to formula (101) is obtained.
  • This terminal GlcNAc-moiety may be fucosylated (b in Figure 1(a) is 1) or non-fucosylated (b is 0).
  • a "suitable enzyme” is defined as an enzyme wherefore the oligosaccharide that is to be trimmed is a substrate.
  • the preferred type of enzyme that is to be used in step (i) of this particular embodiment of the process according to the invention depends on the specific glycan or glycans that is or are trimmed.
  • the enzyme in step (i) of this particular embodiment of the process is selected from the group of endo- glycosidases. Endo-glycosidases are capable of cleaving internal glycosidic linkages in glycan structures, which provides a benefit to remodeling and synthetic endeavors.
  • endo-glycosidases can be employed for facile homogenization of heterogeneous glycan populations, when they cleave at predictable sites within conserved glycan regions.
  • One of the most significant classes of endoglycosidases in this respect comprises the endo-P-N-acetylglucosaminidases (EC 3.2.1.96, commonly known as Endos and ENGases), a class of hydrolytic enzymes that remove N-glycans from glycoproteins by hydrolyzing the -l,4-glycosidic bond in the ⁇ , ⁇ '- diacetylchitobiose core (reviewed by Wong et al. Chem. Rev.
  • Endo-P-N-acetylglucosaminidases are found widely distributed through nature with common chemoenzymatic variants including Endo D, which is specific for pauci mannose; Endo A and Endo H, which are specific for high mannose; Endo F subtypes, which range from high mannose to biantennary complex; and Endo M, which can cleave most N-glycan structures (high mannose/compl ex-type/hybrid-type), except fucosylated glycans, and the hydrolytic activity for the high-mannose type oligosaccharides is significantly higher than that for the complex-and hybrid-type oligosaccharides.
  • These ENGases show specificity toward the distal N-glycan structure and not the protein displaying it, making them useful for cleaving most N-linked glycans from glycoproteins under native conditions.
  • Endoglycosidases Fl, F2, and F3 are most suitable for deglycosylation of native proteins.
  • the linkage specificities of endo Fl, F2, and F3 suggest a general strategy for deglycosylation of proteins that may remove all classes of N-linked oligosaccharides without denaturing the protein.
  • Biantennary and triantennary structures can be immediately removed by endoglycosidases F2 and F3, respectively.
  • Oligo- mannose and hybrid structures can be removed by Endo Fl .
  • Endo F3 is unique in that its cleavage is sensitive to the state of peptide linkage of the oligosaccharide, as well as the state of core fucosylation.
  • Endoglycosidase F3 cleaves asparagine-linked biantennary and triantennary complex oligosaccharides. It will cleave non-fucosylated biantennary and triantennary structures at a slow rate, but only if peptide-linked.
  • Core fucosylated biantennary structures are efficient substrates for Endo F3, which activity up to 400-fold. There is no activity on oligomannose and hybrid molecules. See for example Tarentino et al. Glycobiology 1995, 5, 599, incorporated by reference herein.
  • Endo S is a secreted endoglycosidase from Streptococcus pyogenes, and also belongs to the glycoside hydrolase family 18, as disclosed by Collin et al. (EMBO J. 2001, 20, 3046, incorporated by reference herein). In contrast to the ENGases mentioned above, however, the endo S has a more defined specificity and is specific for cleaving only the conserved N-glycan in the Fc domain of human IgGs (no other substrate has been identified to date), suggesting that a protein-protein interaction between the enzyme and IgG provides this specificity.
  • Endo S49 is described in WO 2013/037824 (Genovis AB), incorporated by reference herein.
  • Endo S49 is isolated from Streptococcus poyogenes NZ131 and is a homologue of Endo S.
  • Endo S49 has a specific endoglycosidase activity on native IgG and cleaves a larger variety of Fc glycans than Endo S.
  • the enzyme in step (i) of this embodiment is an endo- ⁇ - ⁇ -acetylglucosaminidase.
  • the endo- ⁇ - ⁇ - acetylglucosaminidase is selected from the group consisting of Endo S, Endo S 49, Endo Fl, Endo F2, Endo F3, Endo H, Endo M, Endo A, and any combination thereof.
  • the endo-P-N-acetylglucosaminidase is preferably selected from the group consisting of Endo S, Endo S49, Endo Fl, Endo F2, Endo F3, and a combination thereof.
  • the endo-P-N-acetylglucosaminidase is preferably selected from the group consisting of Endo S, Endo S49, Endo Fl, Endo F2, Endo F3, and a combination thereof, more preferably from the group consisting of Endo S, Endo S49, and a combination thereof.
  • the endo- ⁇ - ⁇ - acetylglucosaminidase is preferably selected from the group consisting of Endo Fl, Endo F2, Endo F3, and a combination thereof.
  • the endo- ⁇ - ⁇ - acetylglucosaminidase is preferably selected from the group consisting of Endo H, Endo M, Endo A and Endo F 1.
  • the present invention thus also relates to a process for the preparation of a modified glycoprotein, the process comprising the steps of:
  • Su(A) x is a sugar derivative Su comprising x functional groups A wherein x is 1, 2, 3 or 4 and wherein A is independently selected from the group consisting of a thiol group or a precursor thereof, a halogen, a sulfonyloxy group, a halogenated acetamido group, a mercaptoacetamido group and a sulfonated hydroxy acetamido group; wherein P is a nucleotide; wherein a suitable catalyst is defined as a galactosyltransferase or a galactosyltransferase comprising a mutant catalytic domain, wherefore Su(A) x -P is a substrate; and wherein a glycan comprising a terminal GlcNAc
  • the catalyst in step(ii) is selected from the group consisting of (1,4)- galactosyltransferases, /3(l,3)-N-galactosyltransferases, /3(l,4)-galactosyltransferases comprising a mutant catalytic domain and /3(l,3)-N-galactosyltransferases comprising a mutant catalytic domain.
  • (1,4)- galactosyltransferases /3(l,3)-N-galactosyltransferases
  • /3(l,4)-galactosyltransferases comprising a mutant catalytic domain
  • /3(l,3)-N-galactosyltransferases comprising a mutant catalytic domain.
  • Galactosyltransferases are described in more detail above.
  • the endo- -N-acetylglucosaminidase is selected from the group consisting of Endo S, Endo S 49, Endo Fl, Endo F2, Endo F3, Endo H, Endo M, Endo A, and any combination thereof, more preferably, the endo- ⁇ - N-acetylglucosaminidase is selected from the group consisting of Endo S, Endo S 49, Endo Fl, Endo F2, Endo F3 and any combination thereof. Most preferably, the endo- ⁇ - N-acetylglucosaminidase is Endo S or Endo S49.
  • the trimming step (2) of the process according to the invention is preferably performed in a suitable buffer solution, such as for example phosphate, buffered saline (e.g. phosphate-buffered saline, tris-buffered saline), citrate, HEPES, tris and glycine.
  • a suitable buffer solution such as for example phosphate, buffered saline (e.g. phosphate-buffered saline, tris-buffered saline), citrate, HEPES, tris and glycine.
  • Suitable buffers are known in the art.
  • the buffer solution is phosphate- buffered saline (PBS) or tris buffer.
  • the process is preferably performed at a temperature in the range of about 4 to about 50°C, more preferably in the range of about 10 to about 45°C, even more preferably in the range of about 20 to about 40°C, and most preferably in the range of about 30 to about 37°C.
  • the process is preferably performed a pH in the range of about 5 to about 9, preferably in the range of about 5.5 to about 8.5, more preferably in the range of about 6 to about 8. Most preferably, the process is performed at a pH in the range of about 7 to about 8.
  • the present invention also relates to a glycoprotein, obtainable by the process for the preparation of a modified glycoprotein according to the invention.
  • a modified glycoprotein is defined as a glycoprotein comprising a modified glycan, said glycan comprising a GlcNAc-Su(A) x disaccharide-moiety at the non- reducing end, wherein Su(A) x is as defined above.
  • the glycoprotein is an antibody.
  • the glycoprotein comprises a glycan according to formula (105) or (106):
  • b is 0 or 1;
  • d is 0 or 1
  • e is 0 or 1 ;
  • G is a monosaccharide, or a linear or branched oligosaccharide comprising 2 to 20 sugar moieties;
  • Su(A) x is a sugar derivative Su comprising x functional groups A wherein x is 1, 2, 3 or 4 and wherein A is independently selected from the group consisting of a thiol group, a halogen, a sulfonyloxy group, a halogenated acetamido group, a mercaptoacetamido group and a sulfonated hydroxyacetamido group.
  • functional group A is a precursor for a thiol group
  • said precursor is transformed into a thiol group during said process.
  • a sugar derivative Su(A) x comprising a precursor for a thiol group as a functional group A
  • a modified glycoprotein comprising a thiol group as a functional group A is obtained.
  • d when d is 0 then e is 1, and when e is 0 then d is 1.
  • G represents a monosaccharide, or a linear or branched oligosaccharide comprising 2 to 20, preferably 2 to 12, more preferably 2 to 10, even more preferably 2 to 8 and most preferably 2 to 6 sugar moieties.
  • Sugar moieties that may be present in a glycan are known to a person skilled in the art, and include e.g. glucose (Glc), galactose (Gal), mannose (Man), fucose (Fuc), N-acetylglucosamine (GlcNAc), N- acetylgalactosamine (GalNAc), N-acetylneuraminic acid (NeuNAc) or sialic acid, or xylose (Xyl).
  • the modified glycoprotein obtainable by the process described above is a glycoprotein according to formula (107) or (108): -
  • Pr represents a protein
  • y is 1 to 20;
  • Su(A) x , b, d, e and G are as defined above.
  • y is 1 to 12, more preferably y is 1, 2, 3, 4, 5, 6, 7 or 8, and even more preferably y is 1, 2, 3 or 4. Most preferably y is 1 or 2. In yet another preferred embodiment, y is 2, 4, 6 or 8. This embodiment is particularly preferred when the glycoprotein to be modified is an antibody, i.e. when Pr is Ab.
  • CI of the core-sugar moiety of the modified glycan is bonded to the protein via an N-glycosidic bond to a nitrogen atom in an amino acid residue in said protein, more preferably to an amide nitrogen atom in the side chain of an asparagine (Asn) or an arginine (Arg) amino acid.
  • CI of the core-sugar- moiety of said modified glycan may also be bonded to the protein via an O-glycosidic bond to an oxygen atom in an amino acid residue in said protein, more preferably to an oxygen atom in the side chain of a serine (Ser) or threonine (Thr) amino acid.
  • the core-sugar-moiety of said glycan is an O-GlcNAc- moiety or an O-GalNAc moiety, preferably an O-GlcNAc moiety.
  • CI of the core- sugar-moiety of said modified glycan may also be bonded to the protein via a C- glycosidic bond to a carbon atom on the protein, e.g. to tryptophan (Trp).
  • a glycoprotein may comprise more than one oligosaccharide chains, and may constitute of a combination of N-linked, O-linked and C-linked glycoproteins.
  • Said modified glycan may be present at a native glycosylation site of a protein, but may also be introduced on a different site on a protein.
  • the modified glycoprotein is a modified antibody.
  • Said antibody (Ab) comprises a glycan comprising a GlcNAc-Su(A) x disaccharide-moiety at the non-reducing end.
  • Such an antibody is herein also referred to as a modified antibody.
  • a preferred modified antibody is an antibody according to formula (107) or (108) as defined above, wherein Pr is Ab.
  • y is 1, 2, 3, 4, 5, 6, 7 or 8, and even more preferred that y is 1, 2, 3 or 4.
  • said antibody may be a whole antibody, but also an antibody fragment.
  • said antibody preferably comprises one or more, more preferably one, modified glycans on each heavy chain.
  • Said whole antibody thus preferably comprises two or more, preferably two, four, six or eight of said glycans, more preferably two or four, and most preferably two glycans.
  • y is preferably 2, 4, 6 or 8, more preferably y is 2 or 4, and most preferably y is 2.
  • y is 1, 2, 3 or 4, and more preferably y is 1 or 2.
  • said antibody is a monoclonal antibody (mAb).
  • mAb monoclonal antibody
  • said antibody is selected from the group consisting of IgA, IgD, IgE, IgG and IgM antibodies. More preferably, said antibody is an IgG antibody, and most preferably said antibody is an IgGl antibody.
  • the glycoprotein in the modified antibody is attached to the conserved N-glycosylation site in the Fc-fragment at asparagine in the region 290-305, typically N297.
  • said modified glycan may be present at a native glycosylation site of a protein, in this case an antibody, but may also be present on a different site.
  • Figure 13 shows different glycoforms of a monoclonal antibody, e.g. IgG.
  • the modified glycan may be on any position in the heavy or light chain.
  • sugar derivative Su(A) x and preferred embodiments thereof are described in detail above.
  • the definitions of the groups A and preferred embodiments thereof, and preferred embodiments of the process for the preparation of a modified glycoprotein also hold for the modified glycoprotein obtainable by said process.
  • x is 1 or 2, more preferably x is 1.
  • functional group A is a thiol group, a halogen, or a halogenated acetamido group, more preferably a halogen.
  • Su(A) x present in a modified glycoprotein and in a modified antibody according to the invention include 6-A-6-deoxygalactose (6-A-Gal), such as 6-chloro-6-deoxygalactose (6-ClGal), 6-thio-6-deoxygalactose (6-HSGal) or 2-A-2- deoxygalactose (2-A-Gal), such as 2-chloro-2-deoxygalactose (2-ClGal), 2-thio-2- deoxygalactose (2-HSGal).
  • A may be indirectly substituted to the sugar derivative as part of an acetamido group that in turn is substituting a hydroxyl group.
  • 6-A-acetamido-6-deoxygalactose such as 6- chloroacetamido-6-deoxygalactose (6-GalNAcCl), 6-thioacetamido-6-deoxygalactose (6-GalNAcSH) or 2-A-acetamido-2-deoxygalactose (2-GalNAcA), such as 2- chloroacetamido-2-deoxygalactose (2-GalNAcCl), 2-thioacetamido-2-deoxygalactose (2-GalNAcSH).
  • 6-GalNAcA such as 6- chloroacetamido-6-deoxygalactose (6-GalNAcCl), 6-thioacetamido-6-deoxygalactose (6-GalNAcSH)
  • 2-A-acetamido-2-deoxygalactose such as 2- chloroacetamido-2-deoxygalactose (2-GalNAc
  • A may be indirectly substituted to the sugar derivative as part of another functional group that in turn is substituting a hydroxyl group or is attached to a hydroxyl group.
  • Examples of such other functional group include an (hetero)alkyl chain or a (hetero)aryl chain.
  • Su(A) x is selected from the group consisting of 6- GalNAcCl, 6-GALNAcSH, 2-GalNAcCl and 2-GalNAcSH.
  • Su(A) x comprises 1 or 2 functional groups A, i.e. preferably x is 1 or 2. More preferably, x is 1. In another preferred embodiment, Su is galactose (Gal). In a further preferred embodiment, x is 1 or 2 and Su is Gal, and most preferably, x is 1 and Su is Gal.
  • Su(A) x is selected from the group consisting of 6- GalNAcCl, 6-GalNAcSH, 2-GalNAcCl, 2-GalNAcSH, 6-ClGal, 2-ClGal, 2-HSGal and 6-HSGal, more preferably form the group consisting of 6-GalNAcCl, 6-GalNAcSH, 2- GalNAcCl, 2-GalNAcSH, 6-ClGal-and 2-ClGal.
  • x is 1 and Su(A) x is selected from the group consisting of 6-GalNAcCl, 6-GalNAcSH, 2-GalNAcCl, 2-GalNAcSH, 6-ClGal, 2- ClGal, 2-HSGal and 6-HSGal, more preferably from the group consisting of 6- GalNAcCl, 6-GalNAcSH, 2-GalNAcCl, 2-GalNAcSH, 6-ClGal-and 2-ClGal.
  • A is a thiol group or a halogen.
  • the modified glycoprotein is preferably a thiol-modified glycoprotein or a halogen-modified glycoprotein.
  • the glycoprotein is a halogen-modified glycoprotein, it is preferably a chloride-modified glycoprotein, a bromide-modified glycoprotein or an iodide-modified glycoprotein, more preferably a chloride-modified glycoprotein or a bromide-modified glycoprotein, and most preferably a chloride-modified glycoprotein.
  • x is 1 and the modified glycoprotein is preferably a thiol-modified glycoprotein or a halogen-modified glycoprotein (preferably a chloride- or a bromide-modified glycoprotein, most preferably a chloride-modified glycoprotein).
  • the modified glycoprotein is preferably a thiol-modified glycoprotein or a halogen-modified glycoprotein (preferably a chloride- or a bromide-modified glycoprotein, most preferably a chloride-modified glycoprotein).
  • Su(A) x is selected from the group consisting of 2-GalNAcSH, 2- GalNAcX, 2-GalNAcOS(0) 2 R 8 , 6-GalNAcSH, 6-GalNAcX and 6-GalNAcOS(0) 2 R 8 .
  • x is 1 or 2 and Su(A) x is selected from the group consisting of 2-GalNAcSH, 2-GalNAcX, 2-GalNAcOS(0) 2 R 8 , 6-GalNAcSH, 6- GalNAcX and 6-GalNAcOS(0) 2 R 8 .
  • x is 1 and Su(A) x is selected from the group consisting of 2-GalNAcSH, 2-GalNAcX, 2- GalNAcOS(0) 2 R 8 , 6-GalNAcSH, 6-GalNAcX and 6-GalNAcOS(0) 2 R 8 .
  • R 8 and preferred embodiments of R 8 , are as defined above.
  • modified glycoprotein when the modified glycoprotein according to the invention comprises a thiol group, said modified glycoprotein may be referred to as a thiol-modified glycoprotein.
  • a modified glycoprotein comprises a halogen, said glycoprotein may be referred to as a halogen-modified glycoprotein.
  • a modified glycoprotein comprises a sulfonyloxy group, said glycoprotein may be referred to as a sulfonyloxy-modified glycoprotein.
  • a modified glycoprotein comprises a mercaptoacetamido group
  • said glycoprotein when a modified glycoprotein comprises a mercaptoacetamido-modified glycoprotein.
  • a modified glycoprotein comprises a halogenated acetamido group
  • said glycoprotein may be referred to as a halogenated acetamido-modified glycoprotein.
  • a modified glycoprotein comprises a sulfonated hydroxyacetamido group
  • said glycoprotein may be referred to as a sulfonated hydroxyacetamido-modified glycoprotein.
  • the invention also relates to the use of a modified glycoprotein according to the invention in the preparation of a glycoprotein-conjugate, wherein a glycoprotein- conjugate is defined as a glycoprotein that is conjugated to a molecule of interest D via a linker L.
  • the present invention also relates to the use of a modified glycoprotein according to the invention in the preparation of a protein-conjugate.
  • a protein-conjugate is herein defined as a protein that is conjugated to a molecule of interest (D) via a linker (L).
  • the protein-conjugate according to the invention may be conjugated to one or to more than one molecule of interest (D) via a linker (L).
  • the invention also relates to the use of a modified antibody according to the invention in the preparation of an antibody-conjugate, wherein an antibody-conjugate is defined as an antibody that is conjugated to a molecule of interest (D) via a linker (L).
  • an antibody-conjugate is defined as an antibody that is conjugated to a molecule of interest (D) via a linker (L).
  • a molecule of interest may for example be a reporter molecule, a diagnostic molecule, an active substance, an enzyme, an amino acid (including an unnatural amino acid), a (non-catalytic) protein, a peptide, a polypeptide, an oligonucleotide, a glycan, a (poly)ethylene glycol diamine (e.g.
  • the molecule of interest is selected from the group consisting of an amino acid (in particular lysine), an active substance, a reporter molecule, an azide and a (hetero)cycloalkynyl moiety.
  • An active substance is a pharmacological and/or biological substance, i.e. a substance that is biologically and/or pharmaceutically active, for example a drug or a prodrug, a diagnostic agent, an amino acid, a protein, a peptide, a polypeptide, a glycan, a lipid, a vitamin, a steroid, a nucleotide, a nucleoside, a polynucleotide, RNA or DNA.
  • suitable peptide tags include cell-penetrating peptides like human lactoferrin or polyarginine.
  • An example of a suitable glycan is oligomannose.
  • the active substance is selected from the group consisting of drugs and prodrugs. More preferably, the active substance is selected from the group consisting of pharmaceutically active compounds, in particular low to medium molecular weight compounds (e.g. about 200 to about 1500 Da, preferably about 300 to about 1000 Da), such as for example cytotoxins, antiviral agents, antibacterial agents, peptides and oligonucleotides.
  • cytotoxins include camptothecins, doxorubicin, daunorubicin, taxanes, calicheamycins, duocarmycins, maytansines, auristatins and pyrrolobenzodiazepines (PBDs).
  • the cytotoxin is selected from the group consisting of colchicine, vinca alkaloids, camptothecins, doxorubicin, daunorubicin, taxanes, calicheamycins, tubulysins, irinotecans, an inhibitory peptide, amanitin, deBouganin, duocarmycins, maytansines, auristatins and pyrrolobenzodiazepines (PBDs).
  • colchicine vinca alkaloids
  • camptothecins camptothecins
  • doxorubicin daunorubicin
  • taxanes calicheamycins
  • tubulysins tubulysins
  • irinotecans irinotecans
  • an inhibitory peptide an inhibitory peptide
  • amanitin amanitin
  • deBouganin duocarmycins
  • maytansines auristatins
  • the cytotoxin is selected from the group consisting of camptothecins, doxorubicin, daunorubicin, taxanes, calicheamycins, duocarmycins, maytansines, auristatins and pyrrolobenzodiazepines (PBDs).
  • the cytotoxin is selected from the group consisting of colchicine, vinca alkaloids, tubulysins, irinotecans, an inhibitory peptide, amanitin and deBouganin.
  • a reporter molecule is a molecule whose presence is readily detected, for example a diagnostic agent, a dye, a fluorophore, a radioactive isotope label, a contrast agent, a magnetic resonance imaging agent or a mass label.
  • a fluorophore include all kinds of Alexa Fluor (e.g. Alexa Fluor 555), cyanine dyes (e.g. Cy3 or Cy5), coumarin derivatives, fluorescein, rhodamine, allophycocyanin and chromomycin.
  • radioactive isotope label examples include 99m Tc, U 1 ln, 18 F, 14 C, 64 Cu, 13 I or 123 I, which may or may not be connected via a chelating moiety such as DTP A, DOT A, NOTA or HYNIC.
  • the molecule of interest (D) is conjugated to the antibody via a linker (L).
  • Linkers or linking units are well known in the art, and are described in more detail below.
  • a linker-conjugate is herein defined as a linker (L) that is conjugated to a molecule of interest (D).
  • the linker-conjugate according to the invention may comprise one or more than one molecule of interest (D).
  • the linker-conjugate preferably is of the formula B-L(D) r , wherein D is as defined above, and B and L are as defined below, and r is 1 to 20.
  • r is 1 to 10, more preferably r is 1 to 8, even more preferably r is 1, 2, 3, 4, 5 or 6, even more preferably r is 1, 2, 3 or 4, even more preferably r is 1 or 2 and most preferably r is 1.
  • the invention also relates to a process for the preparation of a glycoprotein- conjugate, said process comprising reacting a modified glycoprotein according to the invention with a linker-conjugate, wherein said linker-conjugate comprises a functional group B and one or more molecules of interest, wherein said functional group B is a functional group that is capable of reacting with a functional group A on a glycan of the modified glycoprotein, and wherein functional group A is as defined above.
  • the invention relates to a process for the preparation of a glycoprotein-conjugate, the process comprising the steps of:
  • Su(A) x is a sugar derivative Su comprising x functional groups A wherein x is 1, 2, 3 or 4 and wherein A is independently selected from the group consisting of a thiol group or a precursor thereof, a halogen, a sulfonyloxy group, a halogenated acetamido group, a mercaptoacetamido group and a sulfonated hydroxyacetamido group; wherein P is a nucleotide; wherein a suitable catalyst is defined as a galactosyltransferase or a galactosyltransf erase comprising a mutant catalytic domain, wherefore Su(A) x -P is a substrate; and wherein a glycan comprising a
  • b is 0 or 1;
  • d is 0 or 1
  • e is 0 or 1 ;
  • G is a monosaccharide, or a linear or branched oligosaccharide comprising 2 to 20 sugar moieties;
  • linker-conjugate comprises a functional group B and one or more molecules of interest (D), wherein said functional group B is a functional group that is capable of reacting with a functional group A on a glycan of the modified glycoprotein, and wherein functional group A is independently selected from the group consisting of a thiol group, a halogen, a sulfonyloxy group, a halogenated acetamido group, a mercaptoacetamido group and a sulfonated hydroxyacetamido group.
  • Su(A) x comprises 1 or 2 functional groups A, i.e. preferably x is 1 or 2. More preferably, x is 1. In another preferred embodiment, Su is galactose (Gal). In a further preferred embodiment, x is 1 or 2 and Su is Gal, and most preferably, x is 1 and Su is Gal. In these preferred embodiments it is further preferred that the linker- conjugate comprises 1 or 2, and most preferably 1, molecules of interest.
  • Su(A) x is selected from the group consisting of 6- GalNAcCl, 6-GalNAcSH, 2-GalNAcCl, 2-GalNAcSH, 6-ClGal, 2-ClGal, 2-HSGal and 6-HSGal, more preferably form the group consisting of 6-GalNAcCl, 6-GalNAcSH, 2- GalNAcCl, 2-GalNAcSH, 6-ClGal-and 2-ClGal.
  • the linker-conjugate comprises 1 or 2, and most preferably 1, molecules of interest.
  • x is 1 and Su(A) x is selected from the group consisting of 6-GalNAcCl, 6-GalNAcSH, 2-GalNAcCl, 2-GalNAcSH, 6-ClGal, 2- ClGal, 2-HSGal and 6-HSGal, more preferably from the group consisting of 6- GalNAcCl, 6-GalNAcSH, 2-GalNAcCl, 2-GalNAcSH, 6-ClGal-and 2-ClGal.
  • the linker-conjugate comprises 1 or 2, and most preferably 1, molecules of interest.
  • Su(A) x is selected from the group consisting of 2-GalNAcSH, 2-GalNAcX, 2-GalNAcOS(0) 2 R 8 , 6-GalNAcSH, 6-GalNAcX and 6-GalNAcOS(0) 2 R 8 .
  • x is 1 or 2 and Su(A) x is selected from the group consisting of 2-GalNAcSH, 2-GalNAcX, 2-GalNAcOS(0) 2 R 8 , 6-GalNAcSH, 6- GalNAcX and 6-GalNAcOS(0) 2 R 8 .
  • x is 1 and Su(A) x is selected from the group consisting of 2-GalNAcSH, 2-GalNAcX, 2- GalNAcOS(0) 2 R 8 , 6-GalNAcSH, 6-GalNAcX and 6-GalNAcOS(0) 2 R 8 .
  • the linker-conjugate comprises 1 or 2, and most preferably 1, molecules of interest.
  • X and R 8 are as defined above.
  • X is CI.
  • the invention further relates to a process for the preparation of a glycoprotein- conjugate, said process comprising reacting a modified glycoprotein according to the invention with a linker-conjugate, wherein said linker-conjugate comprises a functional group B and one or more molecules of interest, wherein said functional group B is a functional group that is capable of reacting with a functional group A on a glycan of the modified glycoprotein, and wherein functional group A is independently selected from the group consisting of a thiol group, a halogen, a sulfonyloxy group, a halogenated acetamido group, a mercaptoacetamido group and a sulfonated hydroxyacetamido group.
  • a suitable linker-conjugate for the preparation of a protein-conjugate according to the invention is a linker-conjugate comprising a functional group B and a molecule of interest.
  • Linkers (L) also referred to as linking units, are well known in the art.
  • L is linked to a molecule of interest (D) as well as to a functional group (B), as was described above. Numerous methods for linking said functional group (B) and said molecule of interest (D) to L are known in the art.
  • a linker may have the general structure F 1 -L(F 2 ) r , wherein F 1 represents either a functional group B or a functional group that is able to react with a functional group F on the functional group B as described above, e.g. a (hetero)cycloalkynyl group, a terminal alkynyl group, a primary amine, an aminooxy group, a hydrazyl group, an azido group, an N-maleimidyl group, an acetamido group or a thiol group.
  • F 2 represents a functional group that is able to react with a functional group F on the molecule of interest.
  • r is 1 to 20, preferably 1 to 10, more preferably 1 to 8, even more preferably 1, 2, 3, 4, 5 or 6, even more preferably 1, 2, 3 or 4 and most preferably, r is 1 or 2.
  • L may for example be selected from the group consisting of linear or branched
  • C1-C200 alkylene groups C2-C200 alkenylene groups, C2-C200 alkynylene groups, C 3 - C200 cycloalkylene groups, C5-C200 cycloalkenylene groups, C8-C200 cycloalkynylene groups, C7-C200 alkylarylene groups, C7-C200 arylalkylene groups, C8-C200 arylalkenylene groups, C9-C200 arylalkynylene groups.
  • alkylene groups, alkenylene groups, alkynylene groups, cycloalkylene groups, cycloalkenylene groups, cycloalkynylene groups, alkylarylene groups, arylalkylene groups, arylalkenylene groups and arylalkynylene groups may be substituted, and optionally said groups may be interrupted by one or more heteroatoms, preferably 1 to 100 heteroatoms, said heteroatoms preferably being selected from the group consisting of O, S and R 5 , wherein R 5 is independently selected from the group consisting of hydrogen, halogen, Ci - C24 alkyl groups, C 6 - C24 (hetero)aryl groups, C 7 - C24 alkyl(hetero)aryl groups and C 7 - C24 (hetero)arylalkyl groups. Most preferably, the heteroatom is O.
  • F, F 1 and F 2 may for example be independently selected from the group consisting of hydrogen, halogen, R 5 , C4 - C 10 (hetero)cycloalkyne groups,
  • linking units include (poly)ethylene glycol diamines (e.g. l,8-diamino-3,6-dioxaoctane or equivalents comprising longer ethylene glycol chains), polyethylene glycol or polyethylene oxide chains, polypropylene glycol or polypropylene oxide chains and l,x-diaminoalkanes wherein x is the number of carbon atoms in the alkane.
  • polyethylene glycol diamines e.g. l,8-diamino-3,6-dioxaoctane or equivalents comprising longer ethylene glycol chains
  • polyethylene glycol or polyethylene oxide chains e.g. l,8-diamino-3,6-dioxaoctane or equivalents comprising longer ethylene glycol chains
  • polyethylene glycol or polyethylene oxide chains e.g. l,8-diamino-3,6-dioxaoctane or equivalents compris
  • cleavable linkers comprises cleavable linkers.
  • Cleavable linkers are well known in the art. For example Shabat et al. , Soft Matter 2012, 6, 1073, incorporated by reference herein, discloses cleavable linkers comprising self- immolative moieties that are released upon a biological trigger, e.g. an enzymatic cleavage or an oxidation event.
  • a biological trigger e.g. an enzymatic cleavage or an oxidation event.
  • suitable cleavable linkers are peptide-linkers that are cleaved upon specific recognition by a protease, e.g. cathepsin, plasmin or metalloproteases, or glycoside-based linkers that are cleaved upon specific recognition by a glycosidase, e.g.
  • linking of the thiol-modified glycoprotein with the linker-conjugate preferably takes place via a Michael-type addition reaction or a nucleophilic substitution reaction or a thiol-ene reaction.
  • Functional group B is then preferably an N-maleimidyl group for the Michael-type addition, a halogenated acetamido group for the nucleophilic substitution reaction, or a terminal alkene for the thiol-ene reaction.
  • the linker-conjugate is then preferably X-CH 2 C(0) HL(D) r or X- CH 2 C(0)N[L(D) r ] 2 wherein X is F, CI, Br or I, or a maleimide-linker-conjugate (120) as illustrated below.
  • linking of the modified glycoprotein with the linker-conjugate preferably takes place via reaction with a thiol to form a thioether.
  • linking of the modified glycoprotein with the linker-conjugate preferably takes place via reaction with a thiol to form a thioether.
  • linking of the modified glycoprotein with the linker-conjugate preferably takes place via reaction with a thiol to form a thioether.
  • Functional group B comprises then preferably a thiol group, and a preferred linker-conjugate is HS-L(D) r .
  • functional group B may also comprise an alcohol group or an amine group.
  • the invention relates to a process for the preparation of a glycoprotein-conjugate, wherein: (a) when said modified glycoprotein is a halogen-modified glycoprotein or a halogenated acetamido-modified glycoprotein, functional group B comprises a thiol group, an alcohol group or an amine group; or
  • functional group B comprises an N-maleimide group or a halogenated acetamido group or an alkene
  • functional group B comprises thiol group, an alcohol group or an amine group.
  • said modified glycoprotein is a halogen-modified glycoprotein and functional group B comprises a thiol group
  • said thiol group may be an aliphatic or an aromatic thiol group.
  • said thiol group is an aromatic thiol group.
  • the modified glycoprotein is a thiol-modified glycoprotein and functional group B comprises an N-maleimide group or a halogenated acetamido group.
  • the modified glycoprotein is a thiol-modified glycoprotein and functional group B comprises an allenamide group.
  • the invention therefore also relates to a process for the preparation of a glycoprotein-conjugate, wherein said modified glycoprotein is a thiol-modified glycoprotein and functional group B comprises an allenamide group.
  • the present invention further relates to the use of the glycoprotein according to the invention in the preparation of a glycoprotein-conjugate, wherein a glycoprotein- conjugate is defined as a glycoprotein that is conjugated to a molecule of interest D via a linker L.
  • the invention also relates to a glycoprotein-conjugate obtainable by the process for the preparation of a glycoprotein-conjugate according to the invention.
  • the glycoprotein-conjugate according to the invention is an antibody-conjugate.
  • the molecule of interest D is selected from the group consisting of a reporter molecule, an active substance, an enzyme, an amino acid, a protein, a peptide, a polypeptide, an oligonucleotide, a glycan, a (poly)ethylene glycol diamine, a polyethylene glycol chain, a polyethylene oxide chain, a polypropylene glycol chain, a polypropylene oxide chain and l,x-diaminoalkane, wherein x is the number of carbon atoms in the alkane and x is 1 - 200, an azide or a (hetero)cycloalkynyl moiety, preferably a bivalent or bifunctional (hetero)cycloalkynyl moiety.
  • the invention relates to a glycoprotein-conjugate obtainable by a process comprising the steps of:
  • Su(A) x is a sugar derivative Su comprising x functional groups A wherein x is 1, 2, 3 or 4 and wherein A is independently selected from the group consisting of a thiol group or a precursor thereof, a halogen, a sulfonyloxy group, a halogenated acetamido group, a mercaptoacetamido group and a sulfonated hydroxy acetamido group; wherein P is a nucleotide; wherein a suitable catalyst is defined as a galactosyltransferase or a galactosyltransf erase comprising a mutant catalytic domain, wherefore Su(A) x -P is a substrate; and wherein a glycan comprising
  • b is O or l
  • d is 0 or 1
  • e is 0 or 1 ;
  • G is a monosaccharide, or a linear or branched oligosaccharide comprising 2 to 20 sugar moieties;
  • step (a) and (b) reacting the modified glycoprotein with a linker-conjugate, wherein said linker-conjugate comprises a functional group B and one or more molecules of interest (D), wherein said functional group B is a functional group that is capable of reacting with a functional group A on a glycan of the modified glycoprotein, and wherein functional group A is independently selected from the group consisting of a thiol group, a halogen, a sulfonyloxy group, a halogenated acetamido group, a mercaptoacetamido group and a sulfonated hydroxyacetamido group.
  • step (a) and (b) as well as their preferred embodiments are described in detail above.
  • glycoprotein-conjugate comprising a functional group B that comprises an N-maleimide group
  • the glycoprotein-conjugate according to the invention is according to formula (121) or (122):
  • Pr represents a protein
  • L is a Linker
  • D is a molecule of interest
  • r 1 to 20;
  • x is 1, 2, 3 or 4;
  • y is 1 to 20;
  • b is 0 or 1;
  • d is 0 or 1
  • e is 0 or 1 ;
  • p O or l
  • Q is -N(H)C(0)CH 2 - or CH 2 ;
  • Su is a sugar or sugar derivative
  • G is a monosaccharide, or a linear or branched oligosaccharide comprising 2 to 20 sugar moieties.
  • Su(A) x The value of p and the nature of Q depend on the nature of the modified sugar or sugar derivative Su(A) x that is used for the preparation of the glycoprotein-conjugate according to the process of the invention. If in Su(A) x a thiol group is present on the C2, C3, or C4 position of the sugar or the sugar derivative (instead of a sugar OH- group), then p is 0. If the Su(A) x is a 2-(mercaptoacetamido)-2-deoxy sugar derivative (i.e. a mercaptoacetamido group is present on C2 of said sugar or sugar derivative), Su(A) x is e.g.
  • p is 1 and Q is -N(H)C(0)CH 2 -. If the thiol in Su(A) x is present on the C6 position of the sugar or the sugar derivative, then p is 1 and Q is -CH 2 -.
  • glycoprotein-conjugate comprising a functional group B that comprises a halogenated acetamido group
  • the glycoprotein-conjugate according to the invention is according to formula (123) or (124):
  • R 9 is selected from the group consisting of L(D) r , hydrogen, Ci - C24 alkyl groups, C 6 - C24 aryl groups C 7 - C24 alkylaryl groups and C 7 - C24 arylalkyl groups, the Ci - C24 alkyl groups, C 6 - C24 aryl groups C 7 - C24 alkylaryl groups and C 7 - C24 arylalkyl groups optionally being substituted.
  • R 9 is selected from the group consisting of L(D) r , hydrogen, Ci - C 12 alkyl groups, C 6 - C 12 aryl groups C 7 - C 12 alkylaryl groups and C 7 - C122 arylalkyl groups, the Ci - C 12 alkyl groups, C 6 - C 12 aryl groups C 7 - C12 alkylaryl groups and C 7 - C 12 arylalkyl groups optionally being substituted.
  • R 9 is selected from the group consisting of L(D) r , hydrogen, Ci - C 6 alkyl groups, C 6 - C 12 aryl groups C 7 - C 12 alkylaryl groups and C 7 - C12 arylalkyl groups, the Ci - C 6 alkyl groups, C 6 - C 12 aryl groups C 7 - C 12 alkylaryl groups and C 7 - C 12 arylalkyl groups optionally being substituted. Even more preferably, R 9 is H, Ci, C 2 , C4 or C4 alkyl or C 6 - C 12 aryl. Most preferably, R 9 is H or methyl.
  • the value of p and the nature of Q depend on the nature of the modified sugar or sugar derivative Su(A) x that is used for the preparation of the glycoprotein-conjugate according to the process of the invention. If in Su(A) x a halogen is present on the C2, C3, or C4 position of the sugar or the sugar derivative (instead of a sugar OH-group), then p is 0. If the Su(A) x is a 2-(chloroacetamido)-2-deoxy sugar derivative (i.e. a halogenated acetamido group is present on C2 of said sugar or sugar derivative), Su(A) x is e.g.
  • p is 1 and Q is -N(H)C(0)CH 2 -. If the halogen in Su(A) x is present on the C6 position of the sugar or the sugar derivative, then p is 1 and Q is -CH 2 -.
  • glycoprotein- conjugate according to the invention is according to formula (125) or (126):
  • Pr, L, D, r, x, y, b, d, e, p, Q, Su, G and R are as defined above.
  • the value of p and the nature of Q depend on the nature of the modified sugar or sugar derivative Su(A) x that is used for the preparation of the glycoprotein-conjugate according to the process of the invention. If in Su(A) x a thiol group is present on the C2, C3, or C4 position of the sugar or the sugar derivative (instead of a sugar OH-group), then p is 0. If the Su(A) x is a 2-(mercaptoacetamido)-2- deoxy sugar derivative (i.e. a mercaptoacetamido group is present on C2 of said sugar or sugar derivative instead of an OH-group), Su(A) x is e.g.
  • p is 1 and Q is -N(H)C(0)CH 2 -. If the thiol in Su(A) x is present on the C6 position of the sugar or the sugar derivative, then p is 1 and Q is -CH 2 -.
  • G is a linear or branched oligosaccharide
  • G comprises 2 to 12, more preferably 2 to 10, even more preferably 2 to 8 and most preferably 2 to 6 sugar moieties.
  • y is 1 to 12, more preferably y is 1, 2, 3, 4, 5, 6, 7 or 8, and even more preferably y is 1, 2, 3 or 4. Most preferably y is 1 or 2.
  • y is 2, 4, 6 or 8. This embodiment is particularly preferred when the glycoprotein to be modified is an antibody, i.e. when Pr is Ab.
  • r is 1 to 10, more preferably 1, 2, 3, 4, 5, 6, 7, or 8, even more preferably 1, 2, 3 or 4, even more preferably 1 or 2, and most preferably r is 1.
  • y is 1 to 12, more preferably y is 1, 2, 3, 4, 5, 6, 7 or 8, and even more preferably y is 1, 2, 3 or 4. Most preferably y is 1 or 2.
  • y is 2, 4, 6 or 8. This embodiment is particularly preferred when the glycoprotein to be modified is an antibody, i.e. when Pr is Ab.
  • CI of the core-sugar moiety of the glycoprotein- glycan is bonded to the protein via an N-glycosidic bond to a nitrogen atom in an amino acid residue in said protein, more preferably to an amide nitrogen atom in the side chain of an asparagine (Asn) or an arginine (Arg) amino acid.
  • CI of the core-sugar-moiety of said glycan may also be bonded to the protein via an O-glycosidic bond to an oxygen atom in an amino acid residue in said protein, more preferably to an oxygen atom in the side chain of a serine (Ser) or threonine (Thr) amino acid.
  • the core-sugar-moiety of said glycan is an O-GlcNAc- moiety or an O-GalNAc moiety, preferably an O-GlcNAc moiety.
  • CI of the core- sugar-moiety of said glycan may also be bonded to the protein via a C-glycosidic bond to a carbon atom on the protein, e.g. to tryptophan (Trp).
  • a glycoprotein may comprise more than one oligosaccharide chains, and may constitute of a combination of N-linked, O-linked and C-linked glycoproteins.
  • Said modified glycan may be present at a native glycosylation site of a protein, but may also be introduced on a different site on a protein.
  • the glycoprotein-conjugate is an antibody-conjugate, said antibody comprising a glycan comprising a GlcNAc-Su(A) x disaccharide-moiety at the non-reducing end.
  • an antibody is herein also referred to as a modified antibody.
  • a preferred modified antibody is an antibody according to formula (107) or (108) as defined above, wherein Pr is Ab.
  • y is 1, 2, 3, 4, 5, 6, 7 or 8, and even more preferred that y is 1, 2, 3 or 4.
  • said antibody may be a whole antibody, but also an antibody fragment.
  • said antibody preferably comprises one or more, more preferably one, modified glycans on each heavy chain.
  • Said whole antibody thus preferably comprises two or more, preferably two, four, six or eight of said glycans, more preferably two or four, and most preferably two glycans.
  • y is preferably 2, 4, 6 or 8, more preferably y is 2 or 4, and most preferably y is 2.
  • y is 1, 2, 3 or 4, and more preferably y is 1 or 2.
  • said antibody is a monoclonal antibody (mAb).
  • mAb monoclonal antibody
  • said antibody is selected from the group consisting of IgA, IgD, IgE, IgG and IgM antibodies. More preferably, said antibody is an IgG antibody, and most preferably said antibody is an IgGl antibody.
  • the glycoprotein in the modified antibody is attached to the conserved N-glycosylation site in the Fc-fragment at asparagine in the region 290-305, typically N297.
  • the sugar derivative Su(A) x and preferred embodiments thereof are described in detail above.
  • the preferred embodiments of the process for the preparation of a modified glycoprotein also hold for the modified glycoprotein obtainable by said process.
  • x is 1 or 2, more preferably x is 1.
  • functional group A is a thiol group, a halogen, or a halogenated acetamido group, more preferably a halogen.
  • Su(A) x present in a modified antibody according to the invention include 6-A-6-deoxygalactose (6-A-Gal), such as 6-chloro-6-deoxygalactose (6-ClGal), 6-thio-6-deoxygalactose (6-HSGal) or 2-A-2-deoxygalactose (2-A-Gal), such as 2-chloro-2-deoxygalactose (2-ClGal), 2-thio-2-deoxygalactose (2-HSGal).
  • A may be indirectly substituted to the sugar derivative as part of an acetamido group that in turn is substituting a hydroxyl group.
  • 6-A- acetamido-6-deoxygalactose (6-GalNAcA), such as 6-chloroacetamido-6- deoxygalactose (6-GalNAcCl), 6-thioacetamido-6-deoxygalactose (6-GalNAcSH) or 2- A-acetamido-2-deoxygalactose (2-GalNAcA), such as 2-chloroacetamido-2- deoxygalactose (2-GalNAcCl), 2-thioacetamido-2-deoxygalactose (2-GalNAcSH).
  • A may be indirectly substituted to the sugar derivative as part of another functional group that in turn is substituting a hydroxyl group or is attached to a hydroxyl group.
  • Examples of such other functional group include an (hetero)alkyl chain or a (hetero)aryl chain.
  • y is 1, 2, 3 or 4 (preferably 2 or 4) and/or x is 1 or 2 (preferably 1).
  • y is 1 to 12, more preferably y is 1, 2, 3, 4, 5, 6, 7 or 8, and even more preferably y is 1, 2, 3 or 4. Most preferably y is 1 or 2. In yet another preferred embodiment, y is 2, 4, 6 or 8. This embodiment is particularly preferred when the glycoprotein to be modified is an antibody, i.e. when Pr is Ab.
  • y is 1, 2, 3 or 4 (preferably 2 or 4) and/or x is 1 or 2 (preferably 1), more preferably y is 1 or 2 and x is 1 or 2 (preferably 1), even more preferably y is 1 or 2 and x is 1.
  • CI of the core-sugar moiety of the modified glycan is bonded to the protein via an N-glycosidic bond to a nitrogen atom in an amino acid residue in said protein, more preferably to an amide nitrogen atom in the side chain of an asparagine (Asn) or an arginine (Arg) amino acid.
  • CI of the core-sugar- moiety of said modified glycan may also be bonded to the protein via an O-glycosidic bond to an oxygen atom in an amino acid residue in said protein, more preferably to an oxygen atom in the side chain of a serine (Ser) or threonine (Thr) amino acid.
  • the core-sugar-moiety of said glycan is an O-GlcNAc- moiety or an O-GalNAc moiety, preferably an O-GlcNAc moiety.
  • CI of the core- sugar-moiety of said modified glycan may also be bonded to the protein via a C- glycosidic bond to a carbon atom on the protein, e.g. to tryptophan (Trp).
  • a glycoprotein may comprise more than one oligosaccharide chains, and may constitute of a combination of N-linked, O-linked and C-linked glycoproteins.
  • Said modified glycan may be present at a native glycosylation site of a protein, but may also be introduced on a different site on a protein.
  • the modified glycoprotein is a modified antibody (Ab), said antibody comprising a glycan comprising a GlcNAc-Su(A) x disaccharide- moiety at the non-reducing end.
  • a modified antibody is an antibody according to formula (107) or (108) as defined above, wherein Pr is Ab.
  • y is 1, 2, 3, 4, 5, 6, 7 or 8, and even more preferred that y is 1, 2, 3 or 4.
  • said antibody may be a whole antibody, but also an antibody fragment.
  • said antibody preferably comprises one or more, more preferably one, modified glycans on each heavy chain.
  • Said whole antibody thus preferably comprises two or more, preferably two, four, six or eight of said glycans, more preferably two or four, and most preferably two glycans.
  • y is preferably 2, 4, 6 or 8, more preferably y is 2 or 4, and most preferably y is 2.
  • y is 1, 2, 3 or 4, and more preferably y is 1 or 2.
  • y is 1, 2, 3 or 4 and x is 1 or 2, and it is further preferred that y is 2 or 4 and x is 1.
  • said antibody is a monoclonal antibody (mAb).
  • mAb monoclonal antibody
  • said antibody is selected from the group consisting of IgA, IgD, IgE, IgG and IgM antibodies. More preferably, said antibody is an IgG antibody, and most preferably said antibody is an IgGl antibody.
  • the glycoprotein in the modified antibody is attached to the conserved N-glycosylation site in the Fc-fragment at asparagine in the region 290-305, typically N297.
  • sugar derivative Su(A) x and preferred embodiments thereof are described in detail above.
  • the preferred embodiments of the process for the preparation of a modified glycoprotein also hold for the modified glycoprotein obtainable by said process.
  • x is 1 or 2, more preferably x is 1.
  • functional group A is a thiol group, a halogen, or a halogenated acetamido group, more preferably a halogen.
  • the present invention particularly relates to a glycoprotein-conjugate obtainable by the process for the preparation of an glycoprotein, wherein the glycoprotein- conjugate is an antibody-conjugate.
  • An antibody-conjugate is herein defined as a an antibody that is conjugated to a molecule of interest (D) via a linker (L).
  • the antibody- conjugate according to the invention may be conjugated to one or to more than one molecule of interest (D) via a linker (L).
  • the preferred embodiments disclosed above for a glycoprotein-conjugate also apply to an antibody-conjugate.
  • the antibody-conjugate according to the invention is an antibody-drug conjugate (ADC).
  • An antibody-drug conjugate is herein defined as an antibody that is conjugated to a molecule of interest (D) via a linker (L), wherein D is selected from the group consisting of pharmaceutically active substances, and more preferably D is selected from the group consisting of drugs and prodrugs.
  • the active substance is selected from the group consisting of pharmaceutically active compounds, in particular low to medium molecular weight compounds (e.g. about 200 to about 1500 Da, preferably about 300 to about 1000 Da), such as for example cytotoxins, antiviral agents, antibacterial agents, peptides and oligonucleotides.
  • pharmaceutically active compounds in particular low to medium molecular weight compounds (e.g. about 200 to about 1500 Da, preferably about 300 to about 1000 Da), such as for example cytotoxins, antiviral agents, antibacterial agents, peptides and oligonucleotides.
  • cytotoxins examples include colchicine, vinca alkaloids, camptothecins, doxorubicin, daunorubicin, taxanes, calicheamycins, tubulysins, irinotecans, an inhibitory peptide, amanitin, deBouganin, duocarmycins, maytansines, auristatins and pyrrolobenzodiazepines (PBDs).
  • cytotoxins include colchicine, vinca alkaloids, camptothecins, doxorubicin, daunorubicin, taxanes, calicheamycins, tubulysins, irinotecans, an inhibitory peptide, amanitin, deBouganin, duocarmycins, maytansines, auristatins and pyrrolobenzodiazepines (PBDs).
  • PBDs pyrrolobenzodiazepines
  • the cytotoxin is selected from the group consisting of camptothecins, doxorubicin, daunorubicin, taxanes, calicheamycins, duocarmycins, maytansines, auristatins and pyrrolobenzodiazepines (PBDs).
  • the cytotoxin is selected from the group consisting of colchicine, vinca alkaloids, tubulysins, irinotecans, an inhibitory peptide, amanitin and deBouganin.
  • the invention thus also relates to an antibody-conjugate obtainable by the process for the preparation of an antibody-conjugate according to the invention.
  • the invention further relates to an antibody-conjugate according to the invention, wherein the molecule of interest D is an active substance, for use as a medicament.
  • the invention also relates to the use of an antibody-conjugate according to the invention, wherein the molecule of interest D is an active substance, for use in the treatment of cancer.
  • the invention further relates to an antibody-conjugate according to the invention, wherein the molecule of interest D is an active substance, for use in the treatment of breast cancer, more preferably for use in the treatment of HER2-positive breast cancer.
  • the invention also relates to a method for the treatment of cancer, comprising administering an antibody-drug conjugate according to the invention, in a therapeutically effective amount to a subject in need thereof.
  • the invention also relates to a method for the treatment of breast cancer, comprising administering an antibody-drug conjugate according to the invention, in a therapeutically effective amount to a subject in need thereof.
  • the invention also relates to a method for the treatment of HER2-positive breast cancer, comprising administering an antibody-drug conjugate according to the invention, in a therapeutically effective amount to a subject in need thereof.
  • modified antibody the antibody-conjugate and the processes for the preparation thereof according to the invention have several advantages over the processes, modified antibodies and antibody-conjugates known in the art.
  • DAR Drug to Antibody Ratio
  • modified antibodies and the antibody-conjugates according to the invention are homogeneous, both in site-specificity and stoichiometry. Said modified antibodies and antibody-conjugates are obtained with a DAR very near to the theoretical value, and with a very low standard deviation. This also means that the antibody-conjugates according to the invention result in a more consistent product for preclinical testing.
  • the homogeneity of the antibody-conjugate prepared by the process described herein becomes for example clear from the MS profile given in Figure 9.
  • Another advantage of the processes and antibodies according to the invention involves the reduction of waste in manufacturing, thereby enhancing companies' cost- of-goods.
  • a thiol-modified antibody according to the invention is coupled to a linker- conjugate comprising a maleimide
  • the process is well-known in the art, highly robust and validated.
  • Some examples of maleimide conjugates are provided in Figures 8 and 11.
  • Many maleimide-functionalized toxins have been described, because currently the preferred methodology for antibody-drug conjugation involves the combination of a cysteine mutant of a mAb (THIOmAb) and a maleimide derivative of a toxin. It is well known that such thiol-maleimide conjugates can be prepared with a highly beneficial stoichiometry of reagents (small excess of maleimide component).
  • the resulting thiol-maleimide conjugates may have limited stability, but in case the thiol is present on fucose, the stability is significantly enhanced.
  • a thiol-modified antibody according to the invention is coupled to a linker-conjugate comprising a halogenated acetamide derivative of a toxin, the efficiency of the process may be somewhat compromized with respect to maleimide conjugation and more undesired alternative conjugation may take place (e.g. on lysine side chains), but the desired product is an irreversibly formed (highly stable) thio-ether conjugate.
  • a thiol- modified antibody according to the invention When a thiol- modified antibody according to the invention is coupled to a linker-conjugate comprising a allenamide derivative of a toxin, the efficiency of the process may be somewhat compromized with respect to maleimide conjugation, and no undesired alternative conjugation is likely to take place (e.g. on lysine side chains), while the desired product is an irreversibly formed (highly stable) thio-ether conjugate.
  • An example of an allenamide structure is provided in Figure 6.
  • a halogen-modified antibody according to the invention is coupled to a linker-conjugate comprising a derivative of a toxin containing a nucleophilic group (thiol, alcohol, amine), such as the thiol-comprising structures represented in Figure 6, the resulting conjugate is a thio- ether, such as the structures represented in Figures 8 and 10, a regular ether or an amino-ether, all of which are formed irreversibly.
  • a linker-conjugate comprising a derivative of a toxin containing a nucleophilic group (thiol, alcohol, amine), such as the thiol-comprising structures represented in Figure 6, the resulting conjugate is a thio- ether, such as the structures represented in Figures 8 and 10, a regular ether or an amino-ether, all of which are formed irreversibly.
  • halogenated acetamides for conjugation to proteins containing free thiols (as in THIOmAbs or in a thiofucose-containing mAb)
  • the enzymatic incorporation of a halogenated sugar substrate is not compromised by competitive aspecific reaction with nucleophilic side chains of other amino acids (e.g. lysine).
  • nucleophilic side chains of other amino acids e.g. lysine
  • a halogenated antibody provides the opportunity to perform two-stage conjugation of a toxic payload.
  • reaction of the halogenated antibody with a thiophenol derivative bearing a pendant azide provides an intermediate that can serve as the starting point for introduction of the toxic payload by means of copper-free click chemistry.
  • the stoichiometry of the (cheap and non-toxic) thiophenol derivative can be high, while the stoichiometry of the (expensive and highly toxic) payload can be kept low.
  • a thiol-comprising antibody provides the opportunity to perform two-stage conjugation of a toxic payload.
  • reaction of the thiol-comprising antibody with an allenamide derivative bearing a pendant azide provides a stable intermediate that can serve as the starting point for introduction of the toxic payload by means of copper-free click chemistry (in the example biotin is used as a representative for a toxic payload).
  • the stoichiometry of the (cheap and non-toxic) allenamide derivative can be high, while the stoichiometry of the (expensive and highly toxic) payload can be kept low.
  • UDP-D-galactosamine (27) (45 mg, 0.0796 mmol) was dissolved in buffer pH 7 (0.5 M K 2 HP0 4 ) (2 mL). N-Succinimidyl-S-acetylthioacetate (37 mg, 0.159 mmol) and DMF (2 mL) were added and the reaction was stirred overnight at rt. Another 36 mg of N- succinimidyl-S-acetylthioacetate were added and after 3 h the reaction was concentrated in vacuo. Flash chromatography (7:2: 1-5:2: 1 EtOAc:MeOH:H 2 0) afforded UDP-GalNAcSAc (28a) (28 mg, 0.041 mmol, 52%).
  • UDP-D-galactosamine (27) (42 mg, 0.088 mmol) was dissolved in 0.1 M NaHC0 3 (3 mL) and N-(bromoacetoxy)succinimide 63 mg, 0.265 mmol) (prepared according to Hosztafi et al, Helv. Chim. Acta, 1996, 79, 133-136) and DMF (2 mL) were added. The reaction was stirred overnight at r.t. and concentrated in vacuo. The compound was purified by flash chromatography (7:2: 1-5:2: 1 EtOAc:MeOH:H 2 0) afforded UDP- GalNAcBr (30) (28 mg, 0.048 mmol, 65%).
  • trastuzumab Specific mutants of trastuzumab were derived from literature (Qu et al, J. Immunol. Methods 1998, 213, 131), in particular L196N and G164S) or de novo designed, in particular V363T. In all three cases, asparagine 297 was mutated to glutamine (N297Q) to remove the native glycosylation site.
  • IgG glycans Trimming of IgG glycans was performed using endo S from Streptococcus pyogenes (commercially available from Genovis, Sweden). The IgG (10 mg/mL) was incubated with endo S (40 U/mL final concentration) in 25 mM Tris pH 8.0 for 16 hours at 37 °C.
  • trastuzumab with base MS peak 50591 Da was subjected to the trimming protocol above. After deconvolution of peaks, the mass spectrum showed one peak of the light chain and two peaks of the heavy chain. The two peaks of heavy chain belonged to one major product (49495 Da, 90% of total heavy chain), resulting from core GlcNAc(Fuc)-substituted trastuzumab, and a minor product (49351 Da, ⁇ 10% of total heavy chain), resulting from core GlcNAc-substituted trastuzumab.
  • Example 12 Trimming of trastuzumab-mutant N297Q, V363T
  • Endoglycosidase F3 EndoF3, 25 mU/mg IgG
  • Elizabethkingia meningosepticum commercially available from QA-Bio
  • Enzymatic introduction of modified sugar onto IgG was effected with a mutant of bovine pi,4-galactosyltransf erase (pi,4-Gal-Tl-Y289L).
  • the deglycosylated IgG (prepared as described above, 10 mg/mL) was incubated with a modified UDP-sugar derivative (0.4 mM) and p(l,4)-Gal-Tl-Y289L (1 mg/mL) in 10 mM MnCl 2 and 25 mM Tris-HCl pH 8.0 for 15 hours at 30 °C.
  • the modified IgG was incubated with protein A agarose (40 ⁇ . per mg IgG) for 2 hours at 4 °C.
  • the protein A agarose was washed three times with PBS and the IgG was eluted with 100 mM glycine-HCl pH 2.7.
  • the eluted IgG was neutralized with 1 M Tris-HCl pH 8.0 and concentrated and washed with PBS using an Amicon Ultra-0.5, Ultracel-10 Membrane (Millipore) to a concentration of 10 mg/mL.
  • Trimmed trastuzumab (250 ⁇ ., 10 mg/mL, 16.5 nmol) was incubated with UDP- GalNAcSAc (28a) (18.5 ⁇ , 10 mM) and p(l,4)-Gal-Tl(Y289L) (12.5 ⁇ , 2 mg/mL) in 10 mM MnCl 2 and 25 mM Tris-HCl pH 6.0 for 16 hours at 30 °C.
  • the crude mixture was purified with ProtA and in order to maintain the low pH the column was washed with 25 mM Tris-HCl pH 6.0 and after elution with glycine.HCl buffer (pH 2.7, 0.1 M), the elution buffer was neutralized with Tris-HCl (pH 7.2, 1 M).
  • Example 14 Glycosyltransfer of UDP-sugar 28a (GalNAcSAc) to deglycosylated trastuzumab (N297Q, V363T).
  • Trimmed trastuzumab(N297Q,V363T) (100 ⁇ L, 10 mg/mL, 6.6 nmol) was incubated with UDP-GalNAcSAc (28a) (14 ⁇ , 10 mM) and p(l,4)-Gal-Tl(Y289L) (10 ⁇ , 2 mg/mL) in 10 mM MnCl 2 and 25 mM Tris-HCl pH 6.0 for 16 hours at 30 °C.
  • Trimmed trastuzumab (100 ⁇ ., 10 mg/mL, 6.6 nmol) was incubated with UDP- GalNAcCl (29) (5 ⁇ , 10 mM) and p(l,4)-Gal-Tl(Y289L) (5 ⁇ , 2 mg/mL) in 10 mM MnCl 2 and 25 mM Tris-HCl pH 6.0 for 16 hours at 30 °C.
  • the crude mixture was purified with ProtA according to the protocol for GalNAcSAc to afford trast- (GalNAcCl) 2 (0.36 mg). AccuTOF analysis showed complete conversion to the desired product (mass 49731, expected mass 49732).
  • Example 16 Glycosyltransfer of UDP-sugar 29 (GalNAcCl) to deglycosylated trastuzumab (N297Q, V363T).
  • Trimmed trastuzumab N297Q V363T mutant (100 ⁇ ⁇ , 10 mg/mL, 6.6 nmol) was incubated with UDP-GalNAcCl (29) (5 ⁇ , 10 mM) and p(l,4)-Gal-Tl(Y289L) (5 ⁇ , 2 mg/mL) in 10 mM MnCl 2 and 25 mM Tris-HCl pH 6.0 for 16 hours at 30 °C.
  • the crude mixture was purified with ProtA according to the protocol for GalNAcSAc to afford trast-(GalNAcCl) 2 (0.35 mg). AccuTOF analysis showed complete conversion to the desired product (mass 49750, expected mass 49750).
  • Glycosyltransfer of UDP-sugar 30 (GalNAcBr) to deglycosylated trastuzumab.
  • the trimmed trastuzumab (100 ⁇ L, 10 mg/mL, 6.6 nmol) was incubated with UDP- GalNAcBr (RHX) (15 ⁇ , 10 mM) and pl,4-Gal-Tl-Y289L (5 ⁇ , 2 mg/mL) in 10 mM MnCl 2 and 25 mM Tris-HCl pH 6.0 for 16 hours at 30 °C.
  • the crude mixture was purified with ProtA according to the protocol for GalNaSAc to afford trast-(GalNacBr) 2 (0.34 mg).
  • AccuTOF analysis showed 50% conversion to the desired product (mass 49777, expected mass 49776 and mass + DTT (reaction during analysis sample) 49849.5).
  • the intramolecular substitution of the bromide was 30% (mass 49696, expected mass 49698) and 20% was remaining starting material.
  • the GalT mutant genes were amplified from a construct containing the sequence encoding the catalytic domain of GalT consisting of 130-402 aa residues, by the overlap extension PCR method.
  • the wild type enzyme is represented by SEQ ID NO: 17.
  • SEQ ID NO: 18 the first DNA fragment was amplified with a pair of primers: 01igo38_GalT_External_Fw (CAG CGA CAT ATG TCG CTG ACC GCA TGC CCT GAG GAG TCC represented by SEQ ID NO: 1) and 01igol9_GalT_Y289N_Rw (GAC ACC TCC AAA GTT CTG CAC GTA AGG TAG GCT AAA represented by SEQ ID NO: 2).
  • the Ndel restriction site is underlined, while the mutation site is highlighted in bold.
  • the second fragment was amplified with a pair of primers: 01igo29_GalT_External_Rw (CTG ATG GAT GGA TCC CTA GCT CGG CGT CCC GAT GTC CAC represented by SEQ ID NO: 3) and 01igol8_GalT_Y289N_Fw (CCT TAC GTG CAG AAC TTT GGA GGT GTC TCT GCT CTA represented by SEQ ID NO: 4).
  • the BamHI restriction site is underlined, while the mutation site is highlighted in bold.
  • the two fragments generated in the first round of PCR were fused in the second round using 01igo38_GalT_External_Fw and 01igo29_GalT_External_Rw primers. After digestion with Ndel and BamHI. This fragment was ligated into the pET16b vector cleaved with the same restriction enzymes.
  • the newly constructed expression vector contained the gene encoding Y289N mutant and the sequence encoding for the His-tag from pET16b vector, which was confirmed by DNA sequencing results.
  • Y289F represented by SEQ ID NO: 19
  • Y289M represented by SEQ ID NO: 20
  • Y289I represented by SEQ ID NO: 21
  • Y289V represented by SEQ ID NO: 22
  • Y289A represented by SEQ ID NO: 23
  • Y289G represented by SEQ ID NO: 24
  • the first DNA fragment was amplified with a pair of primers defined herein as SEQ ID NO: 1 and SEQ ID NO: 5 and the second fragment was amplified with a pair of primers defined herein as SEQ ID NO: 3 and SEQ ID NO: 6 (be referred to Table 1 for the related sequences).
  • the first DNA fragment was amplified with a pair of primers defined herein as SEQ ID NO: 1 and SEQ ID NO: 7 and the second fragment was amplified with a pair of primers defined herein as SEQ ID NO: 3 and SEQ ID NO: 8.
  • the first DNA fragment was amplified with a pair of primers defined herein as SEQ ID NO: 1 and SEQ ID NO: 9 and the second fragment was amplified with a pair of primers defined herein as SEQ ID NO: 3 and SEQ ID NO: 10.
  • the first DNA fragment was amplified with a pair of primers defined herein as SEQ ID NO: 1 and SEQ ID NO: 11 and the second fragment was amplified with a pair of primers defined herein as SEQ ID NO: 3 and SEQ ID NO: 12.
  • the first DNA fragment was amplified with a pair of primers defined herein as SEQ ID NO: 1 and SEQ ID NO: 13 and the second fragment was amplified with a pair of primers defined herein as SEQ ID NO: 3 and SEQ ID NO: 14.
  • the first DNA fragment was amplified with a pair of primers defined herein as SEQ ID NO: 1 and SEQ ID NO: 15 and the second fragment was amplified with a pair of primers defined herein as SEQ ID NO: 3 and SEQ ID NO: 16 (be referred to Table 1 for the related sequences).
  • GalT mutants were expressed, isolated and refolded from inclusion bodies according to the reported procedure by Qasba et al. ⁇ Prot. Expr. Pur. 2003, 30, 219- 229). After refolding, the precipitate was removed and the soluble and folded protein was isolated using a Ni-NTA column (HisTrap excel 1 mL column, GE Healthcare). After elution with 25 mM Tris-HCl pH 8.0, 300 mM NaCl and 200 mM imidazole, the protein was dialyzed against 25 mM Tris-HCl pH 8.0 and concentrated to 2 mg/mL using a spinfilter (Amicon Ultra- 15 Centrifugal Filter Unit with Ultracel-10 membrane, Merck Millipore).
  • Example 24 Synthesis of 4,4'-disulfanediyldibenzoic acid bishydroxysuccinimide ester
  • EDC.HC1 770 mg, 3.95 mmol
  • NHS 447 mg, 3.95 mmol
  • the mixture was stirred for 4 h and subsequent washed with water (2 x 20 mL), aqueous saturated NaHC0 3 (2 x 20 mL), dried over Na 2 S0 4 , filtrated and concentrated in vacuo.
  • 4,4'-Disulfanediyldibenzoic acid bishydroxysuccinimide ester was used crude in the next reaction.
  • Example 28 Synthesis of N-(2-(2-(2-azidoethoxy)ethoxy)ethyl)buta-2-allenamide (43) Butynamide 42 (9 mg, 0.04 mmol) was dissolved in dioxane (0.5 mL) followed by the addition K 2 C0 3 (18% in water, 0.5 mL). The reaction mixture was heated to 40 °C for 2h followed by the addition of citric acid (2 mL, 10% in water) and CH 2 C1 2 (2 mL). The water layer was extracted with CH 2 C1 2 (2 x 3 mL) and the organic layers were combined, dried over Na 2 S0 4 , filtrated and concentrated in vacuo to yield the product 43 (8 mg, 0.04 mmol, 89%).
  • Boc-protected pyrene derivative (460 mg, 0.97 mmol) was dissolved in methanol (10 mL). Acetyl chloride 1.9 mmol) was added and after 1 and 3 h additional acetyl chloride (2 x 140 mol) was added. After stirring for 4 h the mixture was concentrated under reduced pressure.
  • Trastuzumab-(Gal ProSH) 2 (1.0 mg, 10 mg/mL) was incubated with TCEP (6.6 ⁇ ., 10 mM in MiliQ), in PBS and 20 mM EDTA for 2 hours at rt. Subsequently, the reaction medium was exchanged to PBS using an Amicon Ultra-0.5, Ultracel-10 Membrane (Millipore). Dehydroascorbic acid (5.4 ⁇ ., 10 mM in DMSO) was added followed by incubation for 3 h. Maleimide-vc-PABA-MMAE 34 (1 ⁇ ,, 10 mM in DMA) was added and the mixture was incubated for 35 min.
  • Example 34 SPAAC of 46 with 47, leading to antibody-drug conjugate 48
  • Trastuzumab-(Gal ProSH) 2 (0.2 mg, 20 mg/mL) in 50 mM Tris-HCl pH 6.0 was incubated with N-(2"-(2'-(2-azidoethoxy)ethoxy)ethyl)buta-2-allenamide (43) (1 ⁇ , 40 mM in DMA) and 50 mM Tris-HCl pH 8.8 (10 ⁇ _, ) overnigth at rt.
  • Example 36 Conjugation of trast(GalNProSH) 2 with 44, leading to conjugate 50 Trastuzumab-(Gal ProSH) 2 (0.5 mg, 10 mg/mL) was incubated with TCEP (3.3 ⁇ ., 10 mM in MiliQ), in PBS and 20 mM EDTA for 2 hours at rt.
  • reaction medium was exchanged to PBS using an Amicon Ultra-0.5, Ultracel-10 Membrane (Millipore).
  • Dehydroascorbic acid (2.8 ⁇ ., 10 mM in DMSO) was added followed by incubation for 3h.
  • Pyrene maleimide 44 (1 ⁇ ., 10 mM in DMA) was added and the mixture was incubated for 35 min.
  • a sample (2 ⁇ ) was taken for digestion with Fabricator TM (50 U in 10 ⁇ _, PBS) and washed with MiliQ using an Amicon Ultra-0.5, Ultracel-10 Membrane (Millipore) and mass analysis showed the formation of the desired product 50 (24929, expected mass 24929) as main product (about 85%).
  • Trastuzumab-(Gal BuSH) 2 (1 mg, 10 mg/mL) was incubated with TCEP (8.3 ⁇ ., 10 mM in MiliQ), in PBS and 20 mM EDTA for 2 hours at rt. Subsequent the reaction medium was exchanged to PBS using an Amicon Ultra-0.5, Ultracel-10 Membrane (Millipore). Dehydroascorbic acid (6.5 ⁇ ., 10 mM in DMSO) was added followed by incubation for 3 h. Compound 45 (3 ⁇ _, premix, 40 mM in DMA) was added and the mixture was incubated for 35 min.
  • trastuzumab (49503, expected mass 49495).
  • Example 39 Synthesis of UDP-GalNProSH (59) and UDP-GalNBuSH (60) derivatives
  • Example 40 3, 4, 6-tri-O-acetyl-D-galactosamine-l -phosphate (54)
  • UDP-galactosamine derivative 57 (178 mg, 0.21 mmol) was dissolved in a mixture of water: MeOH :Et 3 N (3 :7:3, 5 mL) and stirred until full deprotection was achieved according to LCMS. The reaction was concentrated under reduced pressure and split in two portions. One portion was stored, while the other half was purified on anion exchange column (Q HITRAP, 2 x 5 mL columns). First binding on the column was achieved via loading with buffer A (10 mM NaHC0 3 ) and the column was rinsed with 100 mL buffer A. Next a gradient to 25% B (250 mM NaHC0 3 ) was performed to elute the impurities such as diUMP.
  • buffer A (10 mM NaHC0 3
  • B 250 mM NaHC0 3
  • Example 44 Synthesis of acetylmercapto-4-butanoic acid 4-bromobutanoic acid (1 g, 6 mmol) was dissolved in DMF (7 mL) followed by the addition of potassium thioacetate (1 g, 9 mmol). The reaction was stirred for 1 h and quenched by 1 M HC1 (20 mL) and Et 2 0 (20 mL). The organic layer was washed with 1 M HC1 (3 x 20 mL), dried over Na 2 S0 4 , filtrated and concentrated.
  • Example 45 Synthesis of acetylmercapto-4-butanoic acid hydroxysuccinimide ester
  • Acetylmercapto-4-butanoic acid (450 mg, 2.7 mmol) was dissolved in CH 2 C1 2 (30 mL) followed by the addition of EDC.HC1 (813 mg, 4.1 mmol) and N-hydroxysuccinimide (500 mg, 4.1 mmol).
  • the reaction was stirred overnight and quenched with saturated aqueous NaHC0 3 (20 mL).
  • the organic layer was washed with saturated aqueous NaHC0 3 (2 x 20 mL) and citric acid (1 x 10 mL).
  • the organic layer was dried over Na 2 SC"4, filtrated and concentrated, to give the title compound as a crude product (630 mg, 2.4 mmol, 90%).
  • Acetylmercaptoacetic acid pentafluorophenol ester (100 mg, 0.33 mmol) was dissolved in CH 2 C1 2 (3 mL) and benzylamine (69 ⁇ , 0.5 mmol) and Et 3 N (44 ⁇ , 0.40 mmol) were added. The mixture was stirred overnight and quenched by the addition of water (3 mL), followed by washing with water (3 x 3 mL). The organic layer was dried over Na 2 S0 4 , filtrated and concentrated in vacuo. Flash chromatography (EtOAc: heptane 1 :4 1 : 1) afforded 61, which was dissolved in degassed methanol (7 mL).
  • Acetylmercapto-3 -propanoic acid hydroxysuccinimide ester (100 mg, 0.41 mmol) was dissolved in CH 2 C1 2 (3 mL) and benzylamine (69 ⁇ ,, 0.5 mmol) and Et 3 N (44 ⁇ ,, 0.40 mmol) were added. The mixture was stirred overnight and quenched by the addition of water (3 mL), followed by washing with water (3 x 3 mL). The organic layer was dried over Na 2 SC"4, filtrated and concentrated in vacuo. Flash chromatography (EtOAc: heptane 1 :4 ⁇ 1 : 1) afforded product 62, which was dissolved in degassed methanol (7 mL).
  • Acetylmercapto-4-butanoic acid hydroxysuccinimide ester (100 mg, 0.38 mmol) was dissolved in CH 2 C1 2 (3 mL) and benzylamine (69 ⁇ , 0.5 mmol) and Et 3 N (44 ⁇ , 0.40 mmol) were added. The mixture was stirred overnight and quenched by the addition of water (3 mL), followed by washing with water (3 x 3 mL). The organic layer was dried over Na 2 S0 4 , filtrated and concentrated in vacuo. Flash chromatography (EtOAc: heptane 1 :4 - 1 : 1) afforded product 63, which was dissolved in degassed methanol (7 mL).
  • succinimide conjugates 67-69 (1 mg) was dissolved in DMF (400 ⁇ ) followed by the addition of buffer solution 1, 2 or 3 (800 ⁇ ).
  • Buffer solution 1 is PBS
  • buffer solution 2 is PBS + 1 mM glutathione (GSH, reduced)
  • buffer solution 3 is PBS + 1 mM glutathione (oxidized).
  • Final concentration of glutathione (through dilution with DMF) is 0.71 mM.
  • the samples were incubated at 37 °C and measured in time. Ratio of products was determined by LC-MS. Table 2. Relative ratio of starting succinimide (67-69) versus hydrolyzed succinimide versus glutathione adduct formed by reverse Michael, then GSH addition. Structures of compounds 67-69 are shown in Figure 12.
  • Example 56 Glycosyltransfer of UDP-sugar 59 (GalNProSH) to deglycosylated trastuzumab.
  • Deglycosylated trastuzumab (10 mg/mL) was incubated with 59 (1.3 mM) and ⁇ (1,4)- Gal-T1(Y289L,C342T) (0.2 mg/mL) in 10 mM MnCl 2 and 50 mM Tris-HCl pH 6.0 for 16 hours at 30 °C.
  • the functionalized trastuzumab was incubated with protein A agarose (40 ⁇ . per mg IgG) for 1 hours at rt.
  • the protein A agarose was washed three times with TBS (pH 6.0) and the IgG was eluted with 100 mM glycine-HCl pH 2.5.
  • the eluted IgG was neutralized with 1 M Tris-HCl pH 7.0 and concentrated and washed with 50 mM Tris- HC1 pH 6.0 using an Amicon Ultra-0.5, Ultracel-10 Membrane (Millipore) to a concentration of 15-20 mg/mL.
  • Example 57 Glycosyltransfer of UDP -sugar 60 (GalNBuSH) to deglycosylated trastuzumab.
  • trastuzumab (10 mg/mL) was incubated with 60 (1.3 mM) and ⁇ (1,4)- Gal-T1(Y289M) (2 mg/mL) in 10 mM MnCl 2 and 50 mM Tris-HCl pH 6.0 for 16 hours at 30 °C.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Biochemistry (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Medicinal Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Immunology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Biotechnology (AREA)
  • Microbiology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biophysics (AREA)
  • General Engineering & Computer Science (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Epidemiology (AREA)
  • Oncology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicinal Preparation (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Enzymes And Modification Thereof (AREA)

Abstract

The invention relates to a glycoprotein comprising an optionally fucosylated glycan according to formula (105) or (106), wherein Su(A)x is a modified sugar moiety comprising one or more functional groups A. Functional group A is independently selected from the group consisting of a thiol group, a halogen, a sulfonyloxy group, a halogenated acetamido group, a mercaptoacetamido group and a sulfonated hydroxyacetamido group. The invention also relates to a glycoprotein-conjugate wherein a glycoprotein according to the invention is conjugated to a molecule of interest. Said molecule of interest may for example be an active substance. The invention further relates to a process for the preparation of a modified glycoprotein, and to a method for the preparation of a glycoprotein-conjugate. The invention particularly relates to modified antibodies, antibody-conjugates, antibody-drug conjugates and methods for the preparation thereof.

Description

Modified glycoprotein, protein-conjugate and process for the preparation thereof
Technical field of the invention
The present invention relates to modified glycoproteins, in particular to glycoproteins comprising a modified sugar-moiety. The invention also relates to a glycoprotein-conjugate wherein a glycoprotein according to the invention is conjugated to a molecule of interest. Said molecule of interest may for example be a an active substance. The invention further relates to a process for the preparation of a modified glycoprotein, and to a method for the preparation of a glycoprotein-conjugate. The invention particularly relates to modified antibodies, antibody-conjugates, antibody- drug conjugates and methods for the preparation thereof.
Background of the invention
Protein conjugates, i.e. proteins conjugated to a molecule of interest via a linker, are known in the art. For example, fluorescent labeling is a powerful technique for in vitro and in vivo visualisation, covalent immobilization of proteins is a useful strategy for industrial application and PEGylation of proteins leads to significantly enhanced circulation time. In addition, there is great interest in antibody-conjugates wherein the molecule of interest is a drug, for example a cytotoxic chemical. Antibody-drug- conjugates are known in the art, and consist of a recombinant antibody covalently bound to a cytotoxic chemical via a synthetic linker. Protein conjugates known from the prior art are commonly prepared by conjugation of a functional group to the side chain of amino acid lysine or cysteine, by acylation or alkylation, respectively.
For lysines, conjugation takes place preferentially at lysine side chains with highest steric accessibility, the lowest pKa, or a combination thereof. Disadvantage of this method is that site-control of conjugation is low.
Better control of site-specificity is obtained by alkylation of cysteines, based on the fact that typically no or few free cysteines are present in a typical protein, thereby offering the option of alkylating only those cysteines that are already present in reduced form or selectively engineered into a protein. Alternatively, cysteines can be selectively liberated by a (partial) reductive step. For example, selective cysteine liberation by reduction is typically performed by treatment of a protein with a reducing agent {e.g. TCEP or DTT), leading to conversion of a disulfide bond into two free thiols. The liberated thiols can then be alkylated with an electrophilic reagent, typically based on a maleimide chemistry, which generally proceeds fast and with high selectivity, or with haloacetamides, which also show strong preference for cysteine but side-reactions with lysine side-chains may be encountered.
A recent report (N.M. Okeley et al., Bioconj. Chem. 2013, 24, 1650, incorporated by reference herein) describes the metabolic incorporation of 6-thiofucose into the glycan of a monoclonal antibody, followed by reduction-oxidation-maleimide conjugation. Interestingly, it was found that the 6-thiofucose maleimide conjugate described above was found to display enhanced stability with respect to cysteine maleimide conjugates. However, efficiency of incorporation of 6-thiofucose was found to be only 70%.
One alternative variant of maleimide conjugation, which was applied for the generation of an antibody-drug conjugate, involves a strategy where not the nucleophilic thiol is introduced in the monoclonal antibody, but rather the maleimide. For example, T-DM1 is prepared by first (random) conjugation of lysines with succinimidyl 4-(N-maleimidomethyl)cyclohexane-l-carboxylate (SMCC), thereby effectively charging the antibody with maleimides. In the next stage of the process, the maleimide-functionalized antibody is treated with thiol-functionalized maytansinoid, leading to the conjugate. Hence, this is a unique example where the antibody is effectively converted into an electrophilic reaction partner (instead of the common use of nucleophilic amino acid side chains for conjugation), upon treatment with SMCC. However, also in this case, by nature of the approach, only random conjugation of antibody is achieved.
Notwithstanding the versatility of the above technologies, a general disadvantage of protein conjugates obtained via alkylation with maleimides is that in general the resulting conjugates can be potentially unstable due to the reverse of alkylation, i.e. a retro-Michael reaction.
An alternative strategy to prepare conjugates of a glycoprotein, a subclass of all proteins, involves the generation of one or more aldehyde functions on the protein's glycan structure, either by chemical means (sodium periodate) or by enzymatic means (galactose oxidase). The latter aldehyde function can subsequently be employed for a selective conjugation process, for example by condensation with a functionalized hydroxylamine or hydrazine molecule, thereby generating an oxime-linked or hydrazone-linked protein conjugate, respectively. However, it is known that oximes and hydrazones, in particular derived from aliphatic aldehydes, also show limited stability over time in water or at lower pH. For example, gemtuzumab ozogamicin is an oxime-linked antibody-drug conjugate and is known to suffer from premature deconjugation in vivo.
Qasba et al. disclose in J. Biol. Chem. 2002, 277, 20833, incorporated by reference herein, that mutant galactosyltransferases GalT(Y289L), GalT(Y289I) and GalT(Y289N) can enzymatically attach GalNAc to a non-reducing GlcNAc sugar (β- benzyl-GlcNAc).
WO 2007/095506 and WO 2008/029281 (Invitrogen Corporation), incorporated by reference herein, disclose that the combination of GalT(Y289L) mutant with the C2- substituted azidoacetamido moiety 2-GalNAz-UDP leads to the incorporation of GalNAz at a terminal non-reducing GlcNAc of a glycan. Subsequent conjugation by Staudinger ligation or with copper-catalyzed click chemistry then provides the respective antibody conjugates wherein a fluorescent alkyne probe is conjugated to an antibody. WO 2007/095506 and WO 2008/029281 further disclose that trimming of the glycan can take place with endo H, thereby hydrolyzing a GlcNAc-GlcNAc glycosidic bond and liberating a GlcNAc for enzymatic introduction of GalNAz.
Qasba et al. disclose in Bioconjugate Chem. 2009, 20, 1228, incorporated by reference herein, that β-galactosidase-treated monoclonal antibodies (e.g. Rituxan, Remicade, Herceptin) having a GO glycoform (obtained by treatment of the crude mAbs with galactosidase) are fully regalactosylated to the G2 glycoform after transfer of a galactose moiety comprising a (GalNAz) to the terminal GlcNAc residues of the glycan, leading to tetraazido-substituted antibodies, i.e. two GalNAz moieties per heavy chain. The conjugation of said tetraazido-substituted antibodies to a molecule of interest, for example by Staudinger ligation or cycloaddition with an alkyne, is not disclosed. The transfer of a galactose moiety comprising a C2-substituted keto group (C2-keto-Gal) to the terminal GlcNAc residues of a GO glycoform glycan, as well as the linking of C2-keto-Gal to aminooxy biotin, is also disclosed. However, as mentioned above, the resulting oxime conjugates may display limited stability due to aqueous hydrolysis. A disadvantage of the methods disclosed in WO 2007/095506, WO 2004/063344 and Bioconjugate Chem. 2009, 20, 1228 is that the conjugates obtained by azide-alkyne click chemistry in all cases feature a triazole linkage, which may be disadvantageous with respect to immunogenicity of the ADC. Moreover, in case copper-catalyzed click chemistry is employed, protein damage resulting from undesired oxidative processes may occur, as is disclosed in e.g. Hong et al, Angew. Chem. Int. Ed. Engl. 2009, 48, 9879 (incorporated by reference).
Based on the above, it is clear that galactose can be introduced to proteins featuring a terminal GlcNAc-moiety upon treatment with wild type Gal-Tl/UDP-Gal (leading to Gal-GlcN Ac-protein), while N-acetylgalactosamine can be introduced upon treatment with GalTl mutant Y289L (affording GalNAc-GlcNAc-protein). It has also been shown by Elling et al. (ChemBioChem 2001, 2, 884, incorporated by reference herein) that a variety of human galactosyltransferases ( 4-Gal-Tl, 4-Gal-T4 and β3- Gal-T5), but not bovine 4-Gal-Tl, can accommodate a 6-biotinylated modification of galactose in UDP-Gal, in the absence of Mn2+, leading to effective transfer to model proteins BSA-(GlcNAc)n and ovalbumin. Similarly, Pannecoucke et al. (Tetrahedron Lett. 2008, 49, 2294, incorporated by reference herein) demonstrated that commercially available bovine 4-Gal-Tl under standard conditions is also able to transfer UDP-6- azidogalactose to a model GlcNAc-substrate, but the transfer to a GlcNAc-protein was not demonstrated.
Based on the above, it may be concluded that a strategy involving chemoselective or enzymatic modification of glycans on glycoproteins is a versatile strategy for the site-specific preparation of protein conjugates. However, the current technologies generate linkages of unpredictable stability (maleimides, oximes, hydrazones) or unnatural constitution (triazoles). Moreover, the mode of preparation may be slow and require large excesses of reagent (oxime or hydrazone ligation) or may lead to protein damage (copper-catalyzed click chemistry). Finally, the recently disclosed strategy for metabolic thiofucose incorporation has promise, but efficiency of thiofucose incorporation is low (±70%) due to competition with natural fucose.
Conjugations of biomolecules based on thiols are well-known in the art. In particular, reaction of thiols with maleimide is a fast and selective process, which typically rapidly leads to the desired conjugate. Less popular but also regularly applied are halogenated acetamides that may also react with high selectivity with free thiols although chemoselectivity is compromised with respect to maleimide conjugation. A particular advantage of conjugation with halogenated acetamides is the irreversible formation of a thioether, which compares favorably to maleimide conjugates with respect to stability. The latter stability also applied to conjugates formed by reaction of thiols with allenamide, as most recently reported by Abbas et al, Angew. Chem. Int. Ed. 2014, 53, 7491-7494, incorporated by reference. Other alternatives for conjugation to thiols are also known, for example vinylsulfone conjugation, but less frequently applied. Finally, light-induced thiol-ene reaction has also been shown to be suitable for protein conjugation, see for example Kunz et al. Angew. Chem. Int. Ed. 2007, 46, 5226- 5230), also in this case leading to highly stable thioethers.
Summary of the invention
The invention relates to a process for the preparation of a modified glycoprotein, the process comprising contacting a glycoprotein comprising a glycan comprising a terminal GlcNAc-moiety with Su(A)x-P in the presence of a catalyst selected from the group consisting of /3(l,4)-galactosyltransf erases, /3(l,3)-N-galactosyltransferases, /3(l,4)-galactosyltransferases comprising a mutant catalytic domain and (1,3)-Ν- galactosyltransferases comprising a mutant catalytic domain; wherein Su(A)x is a sugar derivative Su comprising x functional groups A wherein x is 1, 2, 3 or 4 and wherein A is independently selected from the group consisting of a thiol group or a precursor thereof, a halogen, a sulfonyloxy group, a halogenated acetamido group, a mercaptoacetamido group and a sulfonated hydroxyacetamido group; wherein P is a nucleotide; and wherein a glycan comprising a terminal GlcNAc-moiety is a glycan according to formula (101) or (102):
Figure imgf000007_0001
101 102
wherein:
b is 0 or 1;
d is O or l;
e is 0 or 1 ; and G is a monosaccharide, or a linear or branched oligosaccharide comprising 2 to 20 sugar moieties.
The invention also relates to a glycoprotein comprising a glycan according to formula (105) or (106):
-
Figure imgf000008_0001
105 106 wherein:
b is 0 or 1;
d is O or l;
e is 0 or 1 ;
G is a monosaccharide, or a linear or branched oligosaccharide comprising 2 - 20 saccharide moieties; and
Su(A)x is a sugar derivative Su comprising x functional groups A, wherein x is 1, 2, 3 or 4 and wherein A is independently selected from the group consisting a thiol group, a halogen, a sulfonyloxy group, a halogenated acetamido group, a mercaptoacetamido group and a sulfonated hydroxyacetamido group.
The invention also relates to the use of a glycoprotein according to the invention in the preparation of a glycoprotein-conjugate, wherein a glycoprotein-conjugate is defined as a glycoprotein that is conjugated to a molecule of interest D via a linker L; and to a process for the preparation of a glycoprotein-conjugate, said process comprising reacting a modified glycoprotein according to the invention with a linker- conjugate, wherein said linker-conjugate comprises a functional group B and one or more molecules of interest, wherein said functional group B is a functional group that is capable of reacting with a functional group A on a glycan of the modified glycoprotein, and wherein functional group A is as defined above.
In particular, the invention relates to modified antibodies, antibody-conjugates and antibody-drug conjugates. Description of the figures
Figure 1 shows examples of the different glycoforms of a protein that (A) can be obtained by regular expression followed by trimming with endo-glycosidase, or (B) by expression of a mAb in a mammalian system in the presence of swainsonine or by expression in an engineered host organism, e.g. Lecl CHO or Pichia, or (C) by trimming of the regular mixture of glycoforms (GO, Gl, G2, GOF, GIF and G2F) upon combined action of sialidase and galactosidase.
Figure 2 shows a schematic representation of the enzymatic conversion of a GlcNAc-terminated protein with unnatural UDP-galactose derivative 1 (2- ketogalactose) or 2 (GalNAz), upon the action of galactosyl transferase mutant β(1,4)- Gal-T1(Y298L).
Figure 3 shows the structures of modified galactose derivatives (3-17) for transfer onto a GlcNAc-terminated sugar under the action of a galactosyl transferase (or a mutant thereof).
Figure 4 shows a range of thiol-derivatives or halogen derivatives of UDP- galactose (18-23) that may be transferred onto a GlcNAc-terminated sugar under the action of a galactosyl transferase (or a mutant thereof).
Figure 5 shows the scheme for the synthetic preparation of an acetyl-protected thiol variant (28a) or the deacetylated thiol variant (28b) or halogen variants (29+30) of UDP-GalNAc, substituted on the 2-NHAc moiety.
Figure 6 shows the scheme for the synthetic preparation of a biotin-substituted thiophenol (32), of azide- substituted thiophenol (41) and of azide-allenamide construct 43 from propionic amide precursos 42.
Figure 7 shows the structures of maleimide derivatives of biotin (33), MMAF (34), pyrene (44) and BCN (45).
Figure 8 shows a strategy for conversion of a trimmed monoclonal antibody 35 (comprising a core GlcNAc only). Upon the action of Gal-T1(Y289L) on 35, UDP- GalNAc derivatives 28-30 can be selectively introduced onto the GlcNAc to give 36 (R=SH after deacetylation, or halogen). Thiol-containing 36 can be reacted with maleimide derivatives 33 or 34, leading to conjugates 38 or 39, respectively. Alternatively, halogenated derivative 36 can be conjugated by nucleophilic substitution of the halogen with a nucleophilic reagent such as DTT, MeOPhSH or N02PhSH, or compound 32, leading to 40. Figure 9 shows the MS profile of conjugate 40, crude after treatment of 37 (X = CI) with 32.
Figure 10 shows a strategy for conversion of chlorinated antibody 36 with R=C1 (comprising a core GlcNAc substituted with a GalNAcCl). Upon conjugation of chlorinated derivative 36 by substitution with azido-bearing 41, an azido-derivative of the antibody is formed (46). The latter azide can be further conjugated with a BCN- toxin construct such as 47, leading to the antibody-drug conjugate 48.
Figure 11 shows a strategy for conversion of a thiolated monoclonal antibody 36 (comprising a core GlcNAc substituted with thiolated galactose derivative). Thiol- containing 36 can be reacted with maleimide derivatives 43, 44 or 45 leading to conjugates 49, 50 or 51 respectively. The latter conjugate 51, bearing the BCN-moiety can be further conjugated with an azide-biotin construct such as 52, leading to the antibody-biotin conjugate 53.
Figure 12 shows the synthetic route for the preparation of the acetyl-protected GalNAc derivatives UDP-GalNProSAc (57) and UDP-GalNBuSAc (58) and the thiol- containing GalNAc derivatives UDP-GalNProSH (59) and UDP-GalNBuSH (60). Also shown is the synthetic route for preparation of the maleimide conjugates of compounds 64-66, model compounds for conjugates of GalNAcSH, GalNProSH and GalNBuSH, respectively.
Figure 13 shows different glycoforms of a monoclonal antibody, e.g. IgG, which can be obtained by removing the native glycosylation site of a mAb and engineering a glycosylation site (based on sequence N-X-S/T, with X is any amino acid except proline) at another position.
Detailed description of the invention
Definitions
The verb "to comprise" as is used in this description and in the claims and its conjugations is used in its non-limiting sense to mean that items following the word are included, but items not specifically mentioned are not excluded.
In addition, reference to an element by the indefinite article "a" or "an" does not exclude the possibility that more than one of the elements is present, unless the context clearly requires that there is one and only one of the elements. The indefinite article "a" or "an" thus usually means "at least one".
The compounds disclosed in this description and in the claims may comprise one or more asymmetric centres, and different diastereomers and/or enantiomers may exist of the compounds. The description of any compound in this description and in the claims is meant to include all diastereomers, and mixtures thereof, unless stated otherwise. In addition, the description of any compound in this description and in the claims is meant to include both the individual enantiomers, as well as any mixture, racemic or otherwise, of the enantiomers, unless stated otherwise. When the structure of a compound is depicted as a specific enantiomer, it is to be understood that the invention of the present application is not limited to that specific enantiomer.
The compounds may occur in different tautomeric forms. The compounds according to the invention are meant to include all tautomeric forms, unless stated otherwise. When the structure of a compound is depicted as a specific tautomer, it is to be understood that the invention of the present application is not limited to that specific tautomer.
Unsubstituted alkyl groups have the general formula CnH2n+i and may be linear or branched. Unsubstituted alkyl groups may also contain a cyclic moiety, and thus have the concomitant general formula CnH2n-i. Optionally, the alkyl groups are substituted by one or more substituents further specified in this document. Examples of alkyl groups include methyl, ethyl, propyl, 2-propyl, t-butyl, 1-hexyl, 1-dodecyl, etc.
An aryl group comprises six to twelve carbon atoms and may include monocyclic and bicyclic structures. Optionally, the aryl group may be substituted by one or more substituents further specified in this document. Examples of aryl groups are phenyl and naphthyl.
Arylalkyl groups and alkylaryl groups comprise at least seven carbon atoms and may include monocyclic and bicyclic structures. Optionally, the arylalkyl groups and alkylaryl may be substituted by one or more substituents further specified in this document. An arylalkyl group is for example benzyl. An alkylaryl group is for example 4-t-butylphenyl.
Heteroaryl groups comprise at least two carbon atoms (i.e. at least C2) and one or more heteroatoms N, O, P or S. A heteroaryl group may have a monocyclic or a bicyclic structure. Optionally, the heteroaryl group may be substituted by one or more substituents further specified in this document. Examples of suitable heteroaryl groups include pyridinyl, quinolinyl, pyrimidinyl, pyrazinyl, pyrazolyl, imidazolyl, thiazolyl, pyrrolyl, furanyl, triazolyl, benzofuranyl, indolyl, purinyl, benzoxazolyl, thienyl, phospholyl and oxazolyl.
Heteroarylalkyl groups and alkylheteroaryl groups comprise at least three carbon atoms (i.e. at least C3) and may include monocyclic and bicyclic structures. Optionally, the heteroaryl groups may be substituted by one or more substituents further specified in this document.
Where an aryl group is denoted as a (hetero)aryl group, the notation is meant to include an aryl group and a heteroaryl group. Similarly, an alkyl(hetero)aryl group is meant to include an alkylaryl group and a alkylheteroaryl group, and (hetero)arylalkyl is meant to include an arylalkyl group and a heteroarylalkyl group. A C2 - C24 (hetero)aryl group is thus to be interpreted as including a C2 - C24 heteroaryl group and a C6 - C24 aryl group. Similarly, a C3 - C24 alkyl(hetero)aryl group is meant to include a C7 - C24 alkylaryl group and a C3 - C24 alkylheteroaryl group, and a C3 - C24 (hetero)arylalkyl is meant to include a C7 - C24 arylalkyl group and a C3 - C24 heteroarylalkyl group.
Unless stated otherwise, alkyl groups, alkenyl groups, alkenes, alkynes, (hetero)aryl groups, (hetero)arylalkyl groups and alkyl(hetero)aryl groups may be substituted with one or more substituents selected from the group consisting of CI - C12 alkyl groups, C2 - C12 alkenyl groups, C2 - C12 alkynyl groups, C3 - C12 cycloalkyl groups, C5 - C12 cycloalkenyl groups, C8 - C12 cycloalkynyl groups, Ci - C12 alkoxy groups, C2 - C12 alkenyloxy groups, C2 - C12 alkynyloxy groups, C3 - C12 cycloalkyloxy groups, halogens, amino groups, oxo and silyl groups, wherein the silyl groups can be represented by the formula (R10)3Si-, wherein R10 is independently selected from the group consisting of Ci - C12 alkyl groups, C2 - C12 alkenyl groups, C2 - C12 alkynyl groups, C3 - C12 cycloalkyl groups, Ci - C12 alkoxy groups, C2 - C12 alkenyloxy groups, C2 - C12 alkynyloxy groups and C3 - C12 cycloalkyloxy groups, wherein the alkyl groups, alkenyl groups, alkynyl groups, cycloalkyl groups, alkoxy groups, alkenyloxy groups, alkynyloxy groups and cycloalkyloxy groups are optionally substituted, the alkyl groups, the alkoxy groups, the cycloalkyl groups and the cycloalkoxy groups being optionally interrupted by one of more hetero-atoms selected from the group consisting of O, N and S. An alkynyl group comprises a carbon-carbon triple bond. An unsubstituted alkynyl group comprising one triple bond has the general formula CnH2n-3 - A terminal alkynyl is an alkynyl group wherein the triple bond is located at a terminal position of a carbon chain. Optionally, the alkynyl group is substituted by one or more substituents further specified in this document, and/or interrupted by heteroatoms selected from the group of oxygen, nitrogen and sulphur. Examples of alkynyl groups include ethynyl, propynyl, butynyl, octynyl, etc.
A cycloalkynyl group is a cyclic alkynyl group. An unsubstituted cycloalkynyl group comprising one triple bond has the general formula CnH2n-5. Optionally, a cycloalkynyl group is substituted by one or more substituents further specified in this document. An example of a cycloalkynyl group is cyclooctynyl.
A heterocycloalkynyl group is a cycloalkynyl group interrupted by heteroatoms selected from the group of oxygen, nitrogen and sulphur. Optionally, a heterocycloalkynyl group is substituted by one or more substituents further specified in this document. An example of a heterocycloalkynyl group is azacyclooctynyl.
A (hetero)aryl group comprises an aryl group and a heteroaryl group. An alkyl(hetero)aryl group comprises an alkylaryl group and an alky 1 heteroaryl group. A (hetero)arylalkyl group comprises a arylalkyl group and a heteroarylalkyl groups. A (hetero)alkynyl group comprises an alkynyl group and a heteroalkynyl group. A (hetero)cycloalkynyl group comprises an cycloalkynyl group and a heterocycloalkynyl group.
A (hetero)cycloalkyne compound is herein defined as a compound comprising a (hetero)cycloalkynyl group.
Several of the compounds disclosed in this description and in the claims may be described as fused (hetero)cycloalkyne compounds, i.e. (hetero)cycloalkyne compounds wherein a second ring structure is fused, i.e. annelated, to the (hetero)cycloalkynyl group. For example in a fused (hetero)cyclooctyne compound, a cycloalkyl (e.g. a cyclopropyl) or an arene (e.g. benzene) may be annelated to the (hetero)cyclooctynyl group. The triple bond of the (hetero)cyclooctynyl group in a fused (hetero)cyclooctyne compound may be located on either one of the three possible locations, i.e. on the 2, 3 or 4 position of the cyclooctyne moiety (numbering according to "IUPAC Nomenclature of Organic Chemistry", Rule A31.2). The description of any fused (hetero)cyclooctyne compound in this description and in the claims is meant to include all three individual regioisomers of the cyclooctyne moiety.
When an alkyl group, a (hetero)aryl group, alkyl(hetero)aryl group, a (hetero)arylalkyl group, a (hetero)cycloalkynyl group is optionally substituted, said groups are independently optionally substituted with one or more substituents independently selected from the group consisting of Ci - C12 alkyl groups, C2 - C12 alkenyl groups, C2 - C12 alkynyl groups, C3 - C12 cycloalkyl groups, Ci - C12 alkoxy groups, C2 - C12 alkenyloxy groups, C2 - C12 alkynyloxy groups, C3 - C12 cycloalkyloxy groups, halogens, amino groups, oxo groups and silyl groups, wherein the alkyl groups, alkenyl groups, alkynyl groups, cycloalkyl groups, alkoxy groups, alkenyloxy groups, alkynyloxy groups and cycloalkyloxy groups are optionally substituted, the alkyl groups, the alkoxy groups, the cycloalkyl groups and the cycloalkoxy groups being optionally interrupted by one of more hetero-atoms selected from the group consisting of O, N and S, wherein the silyl groups are represented by the formula (R6)3Si-, wherein R6 is independently selected from the group consisting of Ci - C12 alkyl groups, C2 - C12 alkenyl groups, C2 - C12 alkynyl groups, C3 - C12 cycloalkyl groups, Ci - C12 alkoxy groups, C2 - C12 alkenyloxy groups, C2 - C12 alkynyloxy groups and C3 - C12 cycloalkyloxy groups, wherein the alkyl groups, alkenyl groups, alkynyl groups, cycloalkyl groups, alkoxy groups, alkenyloxy groups, alkynyloxy groups and cycloalkyloxy groups are optionally substituted, the alkyl groups, the alkoxy groups, the cycloalkyl groups and the cycloalkoxy groups being optionally interrupted by one of more hetero-atoms selected from the group consisting of O, N and S. The general term "sugar" is herein used to indicate a monosaccharide, for example glucose (Glc), galactose (Gal), mannose (Man) and fucose (Fuc). The term "sugar derivative" is herein used to indicate a derivative of a monosaccharide sugar, i.e. a monosaccharide sugar comprising substituents and/or functional groups. Examples of a sugar derivative include amino sugars and sugar acids, e.g. glucosamine (Glc H2), galactosamine (Gal H2) N-acetylglucosamine (GlcNAc), N-acetylgalactosamine (GalNAc), sialic acid (Sia) which is also referred to as N-acetylneuraminic acid (NeuNAc), and N-acetylmuramic acid (MurNAc), glucuronic acid (GlcA) and iduronic acid (IdoA). Examples of a sugar derivative also include compounds herein denoted Su(A)x, wherein Su is a sugar or a sugar derivative, and wherein Su comprises x functional groups A.
The term "nucleotide" is herein used in its normal scientific meaning. The term "nucleotide" refers to a molecule that is composed of a nucleobase, a five-carbon sugar (either ribose or 2-deoxyribose), and one, two or three phosphate groups. Without the phosphate group, the nucleobase and sugar compose a nucleoside. A nucleotide can thus also be called a nucleoside monophosphate, a nucleoside diphosphate or a nucleoside triphosphate. The nucleobase may be adenine, guanine, cytosine, uracil or thymine. Examples of a nucleotide include uridine diphosphate (HDP), guanosine diphosphate (GDP), thymidine diphosphate (TDP), cytidine diphosphate (CDP) and cytidine monophosphate (CMP).
The term "protein" is herein used in its normal scientific meaning. Herein, polypeptides comprising about 10 or more amino acids are considered proteins. A protein may comprise natural, but also unnatural amino acids.
The term "glycoprotein" is herein used in its normal scientific meaning and refers to a protein comprising one or more monosaccharide or oligosaccharide chains ("glycans") covalently bonded to the protein. A glycan may be attached to a hydroxyl group on the protein (O-linked-glycan), e.g. to the hydroxyl group of serine, threonine, tyrosine, hydroxylysine or hydroxyproline, or to an amide function on the protein (N- glycoprotein), e.g. asparagine or arginine, or to a carbon on the protein (C- glycoprotein), e.g. tryptophan. A glycoprotein may comprise more than one glycan, may comprise a combination of one or more monosaccharide and one or more oligosaccharide glycans, and may comprise a combination of N-linked, O-linked and C-linked glycans. It is estimated that more than 50% of all proteins have some form of glycosylation and therefore qualify as glycoprotein. Examples of glycoproteins include PSMA (prostate-specific membrane antigen), CAL (candida antartica lipase), gp41, gpl20, EPO (erythropoietin), antifreeze protein and antibodies.
The term "glycan" is herein used in its normal scientific meaning and refers to a monosaccharide or oligosaccharide chain that is linked to a protein. The term glycan thus refers to the carbohydrate-part of a glycoprotein. The glycan is attached to a protein via the C-1 carbon of one sugar, which may be without further substitution (monosaccharide) or may be further substituted at one or more of its hydroxyl groups (oligosaccharide). A naturally occurring glycan typically comprises 1 to about 10 saccharide moieties. However, when a longer saccharide chain is linked to a protein, said saccharide chain is herein also considered a glycan.
A glycan of a glycoprotein may be a monosaccharide. Typically, a monosaccharide glycan of a glycoprotein consists of a single N-acetylglucosamine (GlcNAc), glucose (Glc), mannose (Man) or fucose (Fuc) covalently attached to the protein.
A glycan may also be an oligosaccharide. An oligosaccharide chain of a glycoprotein may be linear or branched. In an oligosaccharide, the sugar that is directly attached to the protein is called the core sugar. In an oligosaccharide, a sugar that is not directly attached to the protein and is attached to at least two other sugars is called an internal sugar. In an oligosaccharide, a sugar that is not directly attached to the protein but to a single other sugar, i.e. carrying no further sugar substituents at one or more of its other hydroxyl groups, is called the terminal sugar. For the avoidance of doubt, there may exist multiple terminal sugars in an oligosaccharide of a glycoprotein, but only one core sugar.
A glycan may be an O-linked glycan, an N-linked glycan or a C-linked glycan. In an O-linked glycan a monosaccharide or oligosaccharide glycan is bonded to an O- atom in an amino acid of the protein, typically via a hydroxyl group of serine (Ser) or threonine (Thr). In an N-linked glycan a monosaccharide or oligosaccharide glycan is bonded to the protein via an N-atom in an amino acid of the protein, typically via an amide nitrogen in the side chain of asparagine (Asn) or arginine (Arg). In a C-linked glycan, a monosaccharide or oligosaccharide glycan is bonded to a C-atom in an amino acid of the protein, typically to a C-atom of tryptophan (Trp).
The end of an oligosaccharide that is directly attached to the protein is called the reducing end of a glycan. The other end of the oligosaccharide is called the non- reducing end of a glycan.
For O-linked glycans, a wide diversity of chains exists. Naturally occurring O- linked glycans typically feature a serine or threonine-linked a-O-GalNAc moiety, further substituted with galactose, sialic acid and/or fucose. The hydroxylated amino acid that carries the glycan substitution may be part of any amino acid sequence in the protein.
For N-linked glycans, a wide diversity of chains exists. Naturally occurring N- linked glycans typically feature an asparagine-linked β-Ν-GlcNAc moiety, in turn further substituted at its 4-OH with β-GlcNAc, in turn further substituted at its 4-OH with β-Man, in turn further substituted at its 3-OH and 6-OH with a-Man, leading to the glycan pentasaccharide Man3GlcNAc2. The core GlcNAc moiety may be further substituted at its 6-OH by a-Fuc. The pentasaccharide Man3GlcNAc2 is the common oligosaccharide scaffold of nearly all N-linked glycoproteins and may carry a wide variety of other substituents, including but not limited to Man, GlcNAc, Gal and sialic acid. The asparagine that is substituted with the glycan on its side-chain is typically part of the sequence Asn-X-Ser/Thr, with X being any amino acid but proline and Ser/Thr being either serine or threonine.
The term "antibody" is herein used in its normal scientific meaning. An antibody is a protein generated by the immune system that is capable of recognizing and binding to a specific antigen. An antibody is an example of a glycoprotein. The term antibody herein is used in its broadest sense and specifically includes monoclonal antibodies, polyclonal antibodies, dimers, multimers, multispecific antibodies (e.g. bispecific antibodies), antibody fragments, and double and single chain antibodies The term "antibody" is herein also meant to include human antibodies, humanized antibodies, chimeric antibodies and antibodies specifically binding cancer antigen. The term "antibody" is meant to include whole antibodies, but also fragments of an antibody, for example an antibody Fab fragment, (Fab')2, Fv fragment or Fc fragment from a cleaved antibody, a scFv-Fc fragment, a minibody, a diabody or a scFv. Furthermore, the term includes genetically engineered antibodies and derivatives of an antibody. Antibodies, fragments of antibodies and genetically engineered antibodies may be obtained by methods that are known in the art. Suitable marketed antibodies include, amongst others, abciximab, rituximab, basiliximab, palivizumab, infliximab, trastuzumab, alemtuzumab, adalimumab, tositumomab-1131, cetuximab, ibrituximab tiuxetan, omalizumab, bevacizumab, natalizumab, ranibizumab, panitumumab, eculizumab, certolizumab pegol, golimumab, canakinumab, catumaxomab, ustekinumab, tocilizumab, ofatumumab, denosumab, belimumab, ipilimumab and brentuximab.
The terms "treatment," "treating," and the like refer to obtaining a desired pharmacologic and/or physiologic effect. The effect may be prophylactic in terms of completely or partially preventing a disease or symptom thereof and/or may be therapeutic in terms of a partial or complete cure for a disease and/or adverse affect attributable to the disease. "Treatment," as used herein, covers any treatment of a disease in a mammal, particularly in a human, and includes preventing the disease from occurring in a subject which may be predisposed to the disease but has not yet been diagnosed as having it; inhibiting the disease, i.e., arresting its development; relieving the disease, i.e., causing regression of the disease.
Process for the preparation of a modified glycoprotein
The present invention relates to a process for the preparation of a modified glycoprotein, the process comprising contacting a glycoprotein comprising a glycan comprising a terminal GlcNAc-moiety with Su(A)x-P in the presence of a suitable catalyst; wherein Su(A)x is a sugar derivative Su comprising x functional groups A wherein x is 1, 2, 3 or 4 and wherein A is independently selected from the group consisting of a thiol group or a precursor thereof, a halogen, a sulfonyloxy group, a halogenated acetamido group, a mercaptoacetamido group and a sulfonated hydroxyacetamido group; wherein P is a nucleotide; wherein a suitable catalyst is defined as a galactosyltransferase or a galactosyltransferase comprising a mutant catalytic domain, wherefore Su(A)x-P is a substrate; and wherein a glycan comprising a terminal GlcNAc-moiety is a glycan according to formula (101) or (102):
Figure imgf000018_0001
101 102
wherein:
b is 0 or 1;
d is 0 or 1;
e is 0 or 1 ; and G is a monosaccharide, or a linear or branched oligosaccharide comprising 2 to 20 sugar moieties.
Preferably, the catalyst is selected from the group consisting of j3(l,4)- galactosyltransferases, /3(l,3)-N-galactosyltransferases, /3(l,4)-galactosyltransferases comprising a mutant catalytic domain and /3(l,3)-N-galactosyltransferases comprising a mutant catalytic domain. The catalyst is described in more detail below.
The glycoprotein that is to be modified in the process according to the invention comprises a glycan, said glycan comprising a terminal GlcNAc-moiety, i.e. a Glc-NAc moiety that is present at the non-reducing end of the glycan. Said glycan comprises one or more saccharide moieties, and may be linear or branched. Herein, the GlcNAc- moiety of a glycan according to formula (101) wherein b is 1 (i.e. a glycan consisting of a fucosylated GlcNAc), is also considered a terminal GlcNAc-moiety. In glycan (102) it is preferred that when d is 0 then e is 1, and when e is 0 then d is 1.
G represents a monosaccharide, or a linear or branched oligosaccharide comprising 2 to 20, preferably 2 to 12, more preferably 2 to 10, even more preferably 2 to 8 and most preferably 2 to 6 sugar moieties. Sugar moieties that may be present in a glycan are known to a person skilled in the art, and include e.g. glucose (Glc), galactose (Gal), mannose (Man), fucose (Fuc), N-acetylglucosamine (GlcNAc), N- acetylgalactosamine (GalNAc), N-acetylneuraminic acid (NeuNAc) or sialic acid, xylose (Xyl). In one embodiment, the glycan comprising a terminal GlcNAc-moiety consists of one GlcNAc-moiety, and the glycan is a glycan according to formula (101), wherein b is 0. In another embodiment, said glycan consists of a fucosylated GlcNAc-moiety, and the glycan is a glycan according to formula (101), wherein b is 1. In yet another embodiment, said glycan is a glycan according to formula (102), wherein the core- GlcNAc, if present, is optionally fucosylated (b is 0 or 1).
When a core-GlcNAc-moiety is fucosylated, fucose is most commonly linked a- 1,6 to C6 of the core-GlcNAc substituent. As described above, the GlcNAc-moiety of a glycan according to formula (101) wherein b is 1, is herein also considered a terminal GlcNAc-moiety.
As described above, a preferred glycan comprising a terminal GlcNAc-moiety is a glycan according to formula (101) or (102). In a further preferred embodiment the glycoprotein that is to be modified in the process according to the invention is a glycoprotein according to formula (103) or (104):
Figure imgf000020_0001
103 104
wherein:
Pr represents a protein;
y is 1 to 20; and
b, d, e and G are as defined above.
Preferably, when d is 0 then e is 1, and when e is 0 then d is 1. When G is a linear or branched oligosaccharide, it is preferred that G comprises 2 to 12, more preferably 2 to 10, even more preferably 2 to 8 and most preferably 2 to 6 sugar moieties. In another preferred embodiment, y is 1 to 12, more preferably y is 1, 2, 3, 4, 5, 6, 7 or 8, and even more preferably y is 1, 2, 3 or 4. Most preferably y is 1 or 2. In yet another preferred embodiment, y is 2, 4, 6 or 8, preferably 2 or 4, most preferably 2. This embodiment is particularly preferred when the glycoprotein to be modified is an antibody, i.e. when Pr is Ab.
In the process according to the invention, when the glycoprotein that is to be modified is a glycoprotein according to formula (103) or (104), a glycoprotein mixture may be used as the starting glycoprotein, said mixture comprising glycoproteins comprising one or more fucosylated (b is 1) glycans (101) and/or (102) and/or one or more non-fucosylated (b is 0) glycans (101) and/or (101).
A glycoprotein comprising a glycan comprising a terminal GlcNAc-moiety is herein also referred to as a "terminal non-reducing GlcNAc-protein", and a glycan comprising a terminal GlcNAc-moiety is herein also referred to as a "terminal non- reducing GlcNAc-glycan". It should be noted that the term "terminal non-reducing GlcNAc-protein" includes a protein of formula (103) wherein b is 1, and that the term "terminal non-reducing GlcNAc-glycan" includes a glycan of formula (101) wherein b is 1.
The terminal non-reducing GlcNAc-protein may comprise a linear or a branched terminal non-reducing GlcNAc-glycan. Said glycan is bonded via CI of the core-sugar- moiety to the protein, and said core-sugar-moiety preferably is a core-GlcNAc-moiety. Consequently, when the terminal non-reducing GlcNAc-glycan bonded to the protein is a glycan according to formula (102), it is preferred that d is 1.
In a preferred embodiment, CI of the core-sugar moiety of the terminal non- reducing GlcNAc-glycan is bonded to the protein via an N-glycosidic bond to a nitrogen atom in an amino acid residue in said protein, more preferably to an amide nitrogen atom in the side chain of an asparagine (Asn) or an arginine (Arg) amino acid. However, CI of the core-sugar-moiety of the non-reducing GlcNAc-glycan may also be bonded to the protein via an O-glycosidic bond to an oxygen atom in an amino acid residue in said protein, more preferably to an oxygen atom in the side chain of a serine (Ser) or threonine (Thr) amino acid. In this embodiment, it is preferred that the core- sugar-moiety of said glycan is an O-GlcNAc-moiety or an O-GalNAc moiety, preferably an O-GlcNAc moiety. CI of the core-sugar- moiety of the non-reducing GlcNAc-glycan may also be bonded to the protein via a C-glycosidic bond to a carbon atom on the protein, e.g. to tryptophan (Trp). As described above, a glycoprotein may comprise more than one glycan, and may comprise a combination of N-linked, O- linked and C-linked glycoproteins.
The terminal non-reducing GlcNAc-glycan may be present at a native glycosylation site of a protein, but may also be introduced on a different site on a protein.
In a preferred embodiment the terminal non-reducing GlcNAc-protein is an antibody (Ab), the antibody comprising a glycan comprising a terminal GlcNAc- moiety. Such an antibody is herein also referred to as a "terminal non-reducing GlcN Ac-antibody". A preferred terminal non-reducing GlcNAc-antibody is an antibody according to formula (103) or (104) as defined above, wherein Pr is Ab. In this embodiment, it is further preferred that y is 1, 2, 3, 4, 5, 6, 7 or 8, and even more preferred that y is 2, 4, 6 or 8.
As was defined above, said antibody may be a whole antibody, but also an antibody fragment. When the antibody is a whole antibody, said antibody preferably comprises one or more, more preferably one, terminal non-reducing GlcNAc-glycan on each heavy chain. Said whole antibody thus preferably comprises two or more, preferably two, four, six or eight of said glycans, more preferably two or four, and most preferably two glycans. In other words, when said antibody is a whole antibody, y is preferably 2, 4, 6 or 8, more preferably y is 2 or 4, and most preferably y is 2. When the antibody is an antibody fragment, it is preferred that y is 1, 2, 3 or 4, and more preferably y is 1 or 2.
In a particular preferred embodiment, when said glycoprotein is an antibody, y is 1, 2 or 4.
In a preferred embodiment, said antibody is a monoclonal antibody (mAb). Preferably, said antibody is selected from the group consisting of IgA, IgD, IgE, IgG and IgM antibodies. More preferably, said antibody is an IgG antibody, and most preferably said antibody is an IgGl antibody.
In a preferred embodiment, the glycoprotein in an antibody is attached to the conserved N-glycosylation site in the Fc-fragment at asparagine in the region 290-305, typically N297. In another preferred embodiment, the glycoprotein in an antibody is attached to a non-native glycosylation site in the antibody. It is further preferred that said non-native glycosylation site is a non-native N-glycosylation site. A non-native glycosylation site may be introduced into the antibody via glycoengineering techniques.
Several examples of the position of a glycosylation site in an antibody are shown in Figure 13.
Several examples of a terminal non-reducing GlcNAc-protein that may be modified in the process according to the invention are shown in Figure 1. Figure 1 (a) shows a glycoprotein comprising a single, optionally fucosylated, GlcNAc-moiety; this GlcNAc-glycan may for example be linked to the protein via an N-glycosidic or an O- glycosidic bond. Figure 1(b) shows a glycoprotein comprising a branched octasaccharide glycan wherein one of the branches comprises a terminal GlcNAc- moiety (this glycan is also referred to as GnM5) The core-GlcNAc may optionally be fucosylated. Figure 1(c) shows an antibody comprising a branched heptasaccharide glycan, wherein the core-GlcNAc moiety is optionally fucosylated and wherein all branches comprise a terminal GlcNAc-moiety.
A process for the attachment of an unnatural galactose derivative to a terminal non-reducing GlcNAc-protein under the action of galactosyltransferase mutant Y289L is shown in Figure 2.
A modified glycoprotein obtainable by the process for the modification of a glycoprotein according to the invention is defined as a glycoprotein comprising a modified glycan, said glycan comprising a GlcNAc-Su(A)x disaccharide-moiety at the non-reducing end, wherein Su(A)x is as defined above.
Preferably, the modified glycan is according to the formula (105) or (106), wherein b, d and e, as well as their preferred embodiments, are as defined above, and wherein Su(A)x is a sugar derivative Su comprising x functional groups A, wherein x is 1, 2, 3 or 4 and wherein A is independently selected from the group consisting of a thiol group or a precursor thereof, a halogen, a sulfonyloxy group, a halogenated acetamido group, a mercaptoacetamido group and a sulfonated hydroxyacetamido group.
Figure imgf000023_0001
105 106
The present invention thus also relates to a process for the preparation of a modified glycoprotein, the process comprising contacting a glycoprotein comprising a glycan comprising a terminal GlcNAc-moiety with Su(A)x-P in the presence of a suitable catalyst; wherein Su(A)x is a sugar derivative Su comprising x functional groups A wherein x is 1, 2, 3 or 4 and wherein A is independently selected from the group consisting of a thiol group or a precursor thereof, a halogen, a sulfonyloxy group, a halogenated acetamido group, a mercaptoacetamido group and a sulfonated hydroxyacetamido group; wherein P is a nucleotide; wherein a suitable catalyst is defined as a galactosyltransferase or a galactosyltransferase comprising a mutant catalytic domain, wherefore Su(A)x-P is a substrate; wherein a glycan comprising a terminal GlcNAc-moiety is a glycan according to formula (101) or (102), as defined above; and wherein a modified glycoprotein is defined as a glycoprotein comprising a modified glycan according to formula (105) or (106), wherein (105) and (106) are as defined above.
More preferably, the modified glycoprotein obtainable by the here described process is a glycoprotein according to formula (107) or (108), wherein Pr, b, d, e, y and Su(A) x, as well as their preferred embodiments, are as defined above.
-
Figure imgf000024_0001
107 108
Su(A)x and preferred embodiments thereof are described in more detail below.
The process for the preparation of a modified glycoprotein according to the invention is performed in the presence of a suitable catalyst. A suitable catalyst is defined as a galactosyltransferase, or a galactosyltransferase comprising a mutant catalytic domain, wherefore Su(A)x-P is a substrate. When the catalyst is a galactosyltransferase, said galactosyltransferase preferably is a wild-type galactosyltransferase. When the catalyst is a galactosyltransferase comprising a mutant catalytic domain, said mutant GalT domain may be present within a full-length GalT enzyme, but it may also be present in a recombinant molecule comprising a catalytic domain.
Preferably, the catalyst is selected from the group consisting of \l,4)- galactosyltransferases, /3(l,3)-N-galactosyltransferases, \l,4)-galactosyltransferases comprising a mutant catalytic domain and /3(l,3)-N-galactosyltransferases comprising a mutant catalytic domain.
In one embodiment, the catalyst is a wild-type galactosyltransferase, more preferably a wild-type \l,4)-galactosyltransf erase or a wild-type \l,3)-N- galactosyltransf erase, and even more preferably a wild-type \l,4)- galactosyltransf erase I. \l,4)-Galactosyltransf erase I is herein further referred to as GalT. Even more preferably, the \l,4)-galactosyltransf erase I is selected from the group consisting of a bovine j8(l,4)-Gal-Tl, a human j8(l,4)-Gal-Tl, a human β(1,4)- Gal-T2, a human 3(1,4)-Gal-T3, a human 3(1,4)-Gal-T4 and a human ?(1,3)-Gal-T5.
This embodiment wherein the catalyst is a wild-type galactosyltransferase is particularly preferred when a functional group A in sugar derivative Su(A)x is present on C2 or C6, preferably C6, of said sugar derivative. In this embodiment, it is further preferred that Su(A)x comprises not more than one functional group A, i.e. preferably x is 1. Su(A)x and Su(A)x-P are described in more detail below.
The invention thus also relates to a process for the preparation of a modified glycoprotein, said process comprising a step of attaching, in the presence of a suitable catalyst, a modified sugar to a terminal GlcNAc-moiety on a glycoprotein; wherein a modified sugar is defined as a sugar comprising a functional group A wherein A is independently selected from the group consisting of a thiol group or a precursor thereof, a halogen, a sulfonyloxy group, a halogenated acetamido group, a mercaptoacetamido group and a sulfonated hydroxyacetamido group; and wherein a suitable catalyst is defined as a wild-type galactosyltransferase or a galactosyltransferase comprising a mutant catalytic domain, wherefore Su(A)x-P is a substrate; with the proviso that when the catalyst is a wild-type galactosyltransferase, said modified sugar comprises not more than one functional group A and said functional group A is present on C6 of said modified sugar.
In a further embodiment, the invention relates to a process for the preparation of a modified glycoprotein, the process comprising contacting a glycoprotein comprising a glycan according to formula (101) or (102) as defined above with Su(A)x-P in the presence of a suitable catalyst; wherein Su(A)x is a sugar derivative Su comprising x functional groups A wherein x is 1, 2, 3 or 4 and A is independently selected from the group consisting of a thiol group or a precursor thereof, a halogen, a sulfonyloxy group, a halogenated acetamido group, a mercaptoacetamido group and a sulfonated hydroxyacetamido group; wherein P is a nucleotide; and wherein a suitable catalyst is defined as a wild-type galactosyltransferase or a galactosyltransferase comprising a mutant catalytic domain, wherefore Su(A)x-P is a substrate; with the proviso that when the catalyst is a wild-type galactosyltransferase, Su(A)x-P comprises one functional group A (x is 1), and said functional group A is present on C2 or C6 of Su(A)x, preferably C6. Preferably, the wild-type galactosyltransferase is a \l,4)-galactosyltransf erase or a \l,3)-N-galactosyltransferase, more preferably a \l,4)-galactosyltransferase.
In another embodiment the catalyst is a galactosyltransferase comprising a mutant catalytic domain, preferably a \l,4)-galactosyltransf erase comprising a mutant catalytic domain or a \l,3)-N-galactosyltransferase comprising a mutant catalytic domain, more preferably a \l,4)-galactosyltransf erase comprising a mutant catalytic domain. \l,4)-Galactosyltransferase I is herein further referred to as GalT.
In another embodiment the catalyst is selected from the group consisting of \l,4)-galactosyltransferases or \l,3)-N-galactosyltransferases, more preferably from the group of \l,4)-galactosyltransferases or \l,3)-N-galactosyltransferases, all comprising a mutant catalytic domain.
In a preferred embodiment the catalyst is a \l,3)-N-galactosyltransferase comprising a mutant catalytic domain, and preferably said \l,3)-N- galactosyltransferase is a human ?(l-3)-Gal-T5.
More preferably, the catalyst is a \l,4)-N-galactosyltransf erase comprising a mutant catalytic domain, more preferably, a \l,4)-galactosyltransferase I comprising a mutant catalytic domain, and even more preferably selected from the group consisting of a bovine \l,4)-Gal-Tl, a human /M-Gal-Tl, a human ?(1,4)-Gal-T2, a human?(1,4)-Gal-T3 and a human ?(1,4)-Gal-T4, all comprising a mutant catalytic domain.
Most preferably the catalyst is a bovine ?(1,4)-Gal-Tl comprising a mutant catalytic domain.
Several suitable catalysts for the process according to the invention are known in the art. A suitable catalyst is for example a catalyst that comprises a mutant catalytic domain from a \l,4)-galactosyltransf erase I. A catalytic domain herein refers to an amino acid segment that folds into a domain that is able to catalyze the linkage of the specific sugar derivative nucleotide Su(A)x-P to the terminal non-reducing GlcNAc- glycan in a specific process according to the invention. \l,4)-galactosyltransferase I is herein further referred to as GalT. Such mutant GalT catalytic domains are for example disclosed in J. Biol. Chem. 2002, 277, 20833 and WO 2004/063344 (National Institutes of Health), incorporated by reference herein. J. Biol. Chem. 2002, 277, 20833 and WO 2004/063344 disclose Tyr-289 mutants of bovine tf(l,4)-Gal-Tl, which are referred to as Y289L, Y289N and Y289I. The method of preparation of said mutant catalytic domains Y289L, Y289N and Y289I is disclosed in detail in WO 2004/063344, p. 34, 1. 6 - p. 36, 1. 2, expressly incorporated by reference herein.
Mutant GalT domains that catalyze the formation of a glucose- \l,4)-N- acetylglucosamine bond are disclosed in WO 2004/063344 on p. 10, 1, 25 - p. 12, 1. 4 (expressly incorporated by reference herein). Mutant GalT domains that catalyze the formation of an N-acetylgalactosamine-/3(l,4)-N-acetylglucosamine bond are disclosed in WO 2004/063344 on p. 12, 1, 6 - p. 13, 1. 2 (expressly incorporated by reference herein). Mutant GalT domains that catalyze the formation of a N-acetylglucosamine- /3(l,4)-N-acetylglucosamine bond and a mannose-/3(l,4)-N-acetylglucosamine bond are disclosed in WO 2004/063344 on p. 12, 1, 19 - p. 14, 1. 6 (expressly incorporated by reference herein).
The disclosed mutant GalT domains may be included within full-length GalT enzymes, or in recombinant molecules containing the catalytic domains, as is disclosed in WO 2004/063344 on p. 14, 1, 31 - p. 16, 1. 28, expressly incorporated by reference herein.
Another mutant GalT domain is for example Y284L, disclosed by Bojarova et al, Glycobiology 2009, 19, 509, expressly incorporated by reference herein, wherein Tyr284 is replaced by leucine.
Another mutant GalT domain is for example R228K, disclosed by Qasba et al, Glycobiology 2002, 12, 691, expressly incorporated by reference herein, wherein Arg228 is replaced by lysine. The catalyst may also comprise a mutant catalytic domain from a bovine \l,4)- galactosyltransferase, selected from the group consisting of GalT Y289N, GalT Y289F, GalT Y289M, GalT Y289V, GalT Y289G, GalT Y289I and GalT Y289A, preferably selected from the group consisting of GalT Y289F and GalT Y289M. GalT Y289N, GalT Y289F, GalT Y289M, GalT Y289V, GalT Y289G, GalT Y289I and GalT Y289A may be provided via site-directed mutagenesis processes, in a similar manner as disclosed in WO 2004/063344, in Qasba et al, Prot. Expr. Pur. 2003, 30, 219 and in Qasba et al, J. Biol. Chem. 2002, 277, 20833 (all incorporated by reference) for Y289L, Y289N and Y289I. In GalT Y289N the tyrosine amino acid (Y) at position 289 is replaced by an asparagine (N) amino acid, in GalT Y289F the tyrosine amino acid (Y) at position 289 is replaced by a phenyl alanine (F) amino acid, in GalT Y289M said tyrosine is replaced by a methionine (M) amino acid, in GalT Y289V by a valine (V) amino acid, in GalT Y289G by a glycine (G) amino acid, in GalT Y289I by an isoleucine (I) amino acid and in Y289A by an analine (A) amino acid.
In a preferred embodiment of the process for the preparation of a modified glycoprotein according to the invention, said catalyst is a catalyst comprising a mutant catalytic domain from a /3(l,4)-galactosyltransferase, preferably from a bovine β(1,4)- Gal-Tl .
Preferably, the catalyst is a catalyst comprising a mutant catalytic domain from a /3(l,4)-galactosyltransferase, preferably selected from the group consisting of bovine jff(l,4)-Gal-Tl GalT Y289L, GalT Y289N, GalT Y289I, GalT Y289F, GalT Y289M, GalT Y289V, GalT Y289G and GalT Y289A, more preferably selected from the group consisting of bovine ytf(l,4)-Gal-Tl GalT Y289L, GalT Y289N and GalT Y289I.
In a further preferred embodiment, said catalyst is a catalyst comprising a GalT mutant catalytic domain selected from the group consisting of Y289L, Y289N, Y289I, Y284L, R228K, Y289F, Y289M, Y289V, Y289G and Y289A, preferably selected from the group consisting of Y289L, Y289N, Y289I, Y284L and R228K. In another preferred embodiment, said catalyst is a catalyst comprising a bovine ?(1,4)-Gal-Tl mutant catalytic domain selected from the group consisting of Y289F, Y289M, Y289V, Y289G and Y289A. More preferably said catalyst is a catalyst comprising a GalT mutant catalytic domain selected from the group consisting of Y289L, Y289N and Y289I, and most preferably said catalyst is a catalyst comprising a GalT mutant catalytic domain selected from the group consisting of Y289L.
Another type of suitable catalysts is a catalyst based on a(l,3)-N- galactosy transferase (further referred to as a3Gal-T), preferably a(l,3)-N- acetylgalactosaminyltransferase (further referred to as a3GalNAc-T), as disclosed in WO 2009/025646, incorporated by reference herein. Mutation of a3Gal-T can broaden donor specificity of the enzyme, and make it an a3GalNAc-T. Mutation of a3GalNAc- T can broaden donor specificity of the enzyme. Polypeptide fragments and catalytic domains of a(l,3)-N-acetylgalactosaminyltransferases are disclosed in WO 2009/025646 on p. 26, 1. 18 - p. 47, 1. 15 and p. 77, 1. 21 - p. 82, 1. 4 (both expressly incorporated by reference herein).
The process for the preparation of a modified glycoprotein according to the invention is preferably performed in a suitable buffer solution, such as for example phosphate, buffered saline (e.g. phosphate-buffered saline, tris-buffered saline), citrate, HEPES, tris and glycine. Suitable buffers are known in the art. Preferably, the buffer solution is phosphate-buffered saline (PBS) or tris buffer.
The process is preferably performed at a temperature in the range of about 4 to about 50°C, more preferably in the range of about 10 to about 45°C, even more preferably in the range of about 20 to about 40°C, and most preferably in the range of about 30 to about 37°C.
The process is preferably performed a pH in the range of about 5 to about 9, preferably in the range of about 5.5 to about 8.5, more preferably in the range of about 6 to about 8. Most preferably, the process is performed at a pH in the range of about 7 to about 8.
Su(A)x is defined as a sugar derivative Su comprising x functional groups A, wherein A is independently selected from the group consisting of a thiol group or a precursor thereof, a halogen, a sulfonyloxy group, a halogenated acetamido group, a mercaptoacetamido group and a sulfonated hydroxyacetamido group, and wherein x is 1, 2, 3 or 4.
A Su(A)x-moiety may also be referred to as a "modified sugar". A modified sugar is herein defined as a sugar or a sugar derivative, said sugar or sugar derivative comprising 1, 2, 3 or 4 functional groups A, wherein A is selected from the group consisting of a thiol group or a precursor thereof, a halogen, a sulfonyloxy group, a halogenated acetamido group, a mercaptoacetamido group and a sulfonated hydroxyacetamido group. When a modified sugar or sugar derivative comprises e.g. a thiol group, said sugar or sugar derivative may be referred to as a thiol-modified sugar or sugar derivative. When a modified sugar or sugar derivative comprises e.g. a thiol- precursor group, said sugar or sugar derivative may be referred to as a thiol-precursor- modified sugar or sugar derivative. When a modified sugar or sugar derivative comprises e.g. a halogen, said sugar or sugar derivative may be referred to as a halogen-modified sugar or sugar derivative. When a modified sugar or sugar derivative comprises e.g. a sulfonyloxy group, said sugar or sugar derivative may be referred to as a sulfonyloxy-modified sugar or sugar derivative. A thiol group is herein defined as a -[C(R7)2]0SH group, wherein R7 is independently selected from the group consisting of hydrogen, halogen and an (optionally substituted) Ci - C24 alkyl group, and o is 0 - 24. Preferably R7 is hydrogen or a Ci, C2, C3 or C4 alkyl group, more preferably R7 is hydrogen or -CH3. Preferably o is 0 - 10, more preferably 0, 1, 2, 3, 4, 5 or 6. More preferably, R7 is hydrogen, -CH3 or a C2 alkyl group and/or o is 0, 1, 2, 3 or 4. Even more preferably R7 is hydrogen and o is 0, 1, 2 or 3, more preferably o is 1 or 2, most preferably o is 0 or 1. Most preferably o is 0. Most preferably, said thiol group is -CH2CH2CH2SH, -CH2CH2SH, -CH2SH or -SH, preferably -SH.
A precursor of a thiol group is herein defined as a -[C(R7)2]0SC(0)CH3 group, wherein R7 and o, as well as their preferred embodiments, are as defined above for a thiol group. Most preferably, said thiol-precursor is -CH2CH2CH2SC(0)CH3, -CH2CH2SC(0)CH3, -CH2SC(0)CH3 or -SC(0)CH3, preferably -SC(0)CH3. In the process for the preparation of a modified glycoprotein according to the invention, a sugar derivative Su(A)x wherein A is a precursor of a thiol group may be used. During said process, the thiol-precursor is converted to a thiol group.
A halogen is herein defined as F, CI, Br or I. Preferably, said halogen is CI or Br, more preferably CI.
A sulfonyloxy group is herein defined as a -[C(R7)2]0OS(0)2R8 group, wherein R7 and o are as defined above for a thiol group, and R8 is selected from the group consisting of Ci - C24 alkyl groups, C6 - C24 aryl groups, C7 - C24 alkylaryl groups and C7 - C24 arylalkyl groups. R8 is preferably a Ci - C4 alkyl group, a C6 - Ci2 aryl group, a C7 - Ci2 alkylaryl group or a C7 - Ci2 arylalkyl group, more preferably a Ci - C4 alkyl group, a C7 - Ci2 alkylaryl group or a C7 - Ci2 arylalkyl group, even more preferably -CH3, -C2H5, a C3 linear or branched alkyl group, a phenyl group or a C7 alkylaryl group. R8 is most preferably a methyl group, an ethyl group, a phenyl group or a p-tolyl group. Preferably R7 is hydrogen or a Ci, C2, C3 or C4 alkyl group, more preferably R7 is hydrogen or -CH3. Preferably o is 0 - 10, more preferably 0, 1, 2, 3, 4, 5 or 6. More preferably, R7 is hydrogen, -CH3 or a C2 alkyl group and/or o is 0, 1, 2, 3 or 4. Even more preferably R7 is hydrogen and o is 1 or 2, most preferably o is 0. R8 is preferably a Ci - C4 alkyl group, a C7 - C12 alkylaryl group or a C7 - C12 arylalkyl group, more preferably -CH3, -C2H5, a C3 linear or branched alkyl group or a C7 alkylaryl group. Most preferably the sulfonyloxy group is a mesylate group, -OS(0)2CH3, a benzenesulfonate group (-OS(0)2(C6H5) or a tosylate group (-OS(0)2(C6H4CH3).
A halogenated acetamido group is herein defined as an - HC(0)[C(R7)2]0X group, wherein R7 is independently selected from the group consisting of hydrogen, halogen and an (optionally substituted) Ci - C24 alkyl group, X is F, CI, Br or I, and o is 0 - 24. Preferably R7 is hydrogen or a Ci, C2, C3 or C4 alkyl group, more preferably R7 is hydrogen or -CH3, most preferably hydrogen. Preferably o is 0 to 10, more preferably 1, 2, 3, 4, 5 or 6, even more preferably 1, 2, 3 or 4 and most preferably o is 1. More preferably, R7 is hydrogen, -CH3 or a C2 alkyl group and/or o is 1, 2, 3 or 4 and most preferably R7 is hydrogen and o is 1. Preferably, X is CI or Br, more preferably X is CI. Most preferably, R7 is hydrogen, X is CI and o is 1.
A mercaptoacetamido group is herein defined as an - HC(0)[C(R7)2]oSH group, wherein R7 is independently selected from the group consisting of hydrogen, halogen and an (optionally substituted) Ci - C24 alkyl group and o is 0 - 24. Preferably R7 is hydrogen or a Ci, C2, C3 or C4 alkyl group, more preferably R7 is hydrogen or -CH3, most preferably hydrogen. Preferably o is 0 to 10, more preferably 1, 2, 3, 4, 5 or 6, even more preferably 1, 2, 3 or 4 and most preferably o is 2, 3 or 4. More preferably, R7 is hydrogen, -CH3 or a C2 alkyl group and/or o is 1, 2, 3 or 4. More preferably, R7 is hydrogen and o is 1, 2, 3 or 4. Most preferably, R7 is hydrogen and o is 1, 2 or 3, preferably 1. Preferred examples include a mercaptoethanoylamido group, a mercaptopropanoylamido group, a mercaptobutanoylamido group and a mercapto- pentanoylamido group, preferably a mercaptopropanoylamido group.
A sulfonated hydroxyacetamido group is herein defined as a - HC(0)[C(R7)2]0OS(0)2R8 group, wherein R7 is independently selected from the group consisting of hydrogen, halogen and an (optionally substituted) Ci - C24 alkyl group, R8 is selected from the group consisting of Ci - C24 alkyl groups, C6 - C24 aryl groups, C7 - C24 alkylaryl groups and C7 - C24 arylalkyl groups, and o is 0 - 24. R8 is preferably a Ci - C4 alkyl group, a C6 - Ci2 aryl group, a C7 - Ci2 alkylaryl group or a C7 - Ci2 arylalkyl group, more preferably -CH3, -C2H5, a C3 linear or branched alkyl group, a C6 - C9 aryl group or a C7 alkylaryl group. Most preferably the sulfonyloxy group is a mesylate group -OS(0)2CH3, a benzenesulfonate group -OS(0)2(C6H5) or a tosylate group -OS(0)2(C6H4CH3). Preferably R7 is hydrogen or a Ci, C2, C3 or C4 alkyl group, more preferably R7 is hydrogen or -CH3, most preferably hydrogen. Preferably o is 0 to 10, more preferably 1, 2, 3, 4, 5 or 6, even more preferably 1, 2, 3 or 4 and most preferably o is 1. More preferably, R7 is hydrogen, -CH3 or a C2 alkyl group and/or o is 1, 2, 3 or 4. Even more preferably R7 is hydrogen and o is 1, 2 or 3. Yet even more preferably, R7 is H, o is 1 and R8 is a mesylate group, a benzenesulfonate group or a tosylate group. Most preferably, R7 is hydrogen, R8 is -CH3 and o is 1.
The sugar derivative Su(A)x may comprise one or more functional groups A. When Su(A)x comprises two or more functional groups A, each functional group A is independently selected, i.e. one Su(A)x may comprise different functional groups A, e.g. a thiol group and a halogen, etc. In a preferred embodiment, x is 1 or 2, more preferably x is 1. In another preferred embodiment, functional group A is a thiol or a halogen, more preferably a halogen.
Sugar derivative Su(A)x is derived from a sugar or a sugar derivative Su, e.g. an amino sugar or an otherwise derivatized sugar. Examples of sugars and sugar derivatives include galactose (Gal), mannose (Man), glucose (Glc), N-acetylneuraminic acid or sialic acid (Sial) and fucose (Fuc).
An aminosugar is a sugar wherein a hydroxyl (OH) group is replaced by an amine group and examples include glucosamine (Glc H2) and galactosamine (Gal H2). Examples of an otherwise derivatized sugar include N-acetylneuraminic acid (sialic acid, Sia or NeuNAc) or fucose (Fuc).
Sugar derivative Su(A)x is preferably derived from galactose (Gal), mannose
(Man), N-acetylglucosamine (GlcNAc), glucose (Glc), N-acetylgalactosamine (GalNAc), fucose (Fuc) and N-acetylneuraminic acid (sialic acid Sia or NeuNAc), preferably from the group consisting of GlcNAc, Glc, Gal and GalNAc. More preferably Su(A)x is derived from Gal or GalNAc, and most preferably Su(A)x is derived from GalNAc.
The one or more functional groups A in Su(A)x may be linked to the sugar or sugar derivative Su in several ways. The one or more functional groups A may be bonded to C2, C3, C4 and/or C6 of the sugar or sugar derivative, instead of a hydroxyl (OH) group. It should be noted that, since fucose lacks an OH-group on C6, if A is bonded to C6 of Fuc, then A takes the place of an H-atom. Preferably, the one or more functional groups A in Su(A)x may be present on C2 and/or C6 of the sugar or sugar derivative Su. When a functional group A is present instead of an OH-group on C2 of a sugar or sugar derivative, A is preferably selected from the group consisting of a halogenated acetamido group, a mercaptoacetamido group and a sulfonated hydroxyacetamido group. However, when A is present on C2 of a 2-aminosugar derivative, e.g. GalNAc or GlcNAc, A is preferably selected from the group consisting of a halogen, a thiol group and a sulfonyloxy group.
When A is a thiol group, it is preferred that said thiol group is present on the 6- position of a sugar derivative or on a 2-aminosugar derivative. An example of Su(A)x wherein A is a thiol group is 2-(mercaptoacetamido)-2-deoxy-galactose (2- GalNAcSH). Another example of Su(A)x wherein A is a thiol group is 6-mercapto-6- deoxy-galactose.
When A is a halogen, it is preferred that said halogen is present on the 6-position of a sugar derivative or on a 2-amino sugar derivative. An example of Su(A)x wherein A is a halogen is 2-(chloroacetamido)-2-deoxy-galactose (2-GalNAcCl). Another example of Su(A)x wherein A is a halogen is 6-iodo-6-deoxy-galactose. Another example of Su(A)x wherein A is a halogen is 6-(chloroacetamido)-6-deoxy-galactose.
When A is a sulfonyloxy group, it is preferred that said sulfonyloxy group is present on the 6-position of a sugar derivative or on a 2-amino sugar derivative. An example of Su(A)x wherein A is a sulfonyloxy group is 2- (methylsulfonyloxyacetamido)-2-deoxy-galactose (2-GalNAcOMs). Another example of Su(A)x wherein A is a sulfonyloxy group is 2-(benzenesulfonyloxyacetamido)-2- deoxy-galactose (2-GalNAcOMs. Another example of Su(A)x wherein A is a sulfonyloxy group is 6-(methylsulfonyl)-galactose.
When A is a halogenated acetamido group, a mercaptoacetamido group or a sulfonated hydroxyacetamido group it is preferred that said groups are present on the 6- position of a sugar derivative.
Further examples of Su(A)x-P include 6-A-6-deoxygalactose-UDP (6-A-Gal- UDP), such as 6-chloro-6-deoxygalactose-UDP (6-ClGal-UDP), 6-thio-6- deoxygalactose-UDP (6-HSGal-UDP) or 2-A-2-deoxygalactose-UDP (2-A-Gal-UDP), such as 2-chloro-2-deoxygalactose-UDP (2-ClGal-UDP), 2-thio-2-deoxygalactose- UDP (2-HSGal-UDP). Alternatively, A may be indirectly substituted to the sugar derivative as part of an acetamido group that in turn is substituting a hydroxyl group. Examples include 6-A-acetamido-6-deoxygalactose-UDP (6-GalNAcA-UDP), such as 6-chloroacetamido-6-deoxygalactose-UDP (6-GalNAcCl-UDP, (23) with X = CI), 6- thioacetamido-6-deoxygalactose-UDP (6-GalNAcSH-UDP, (20) with 1 CH2 and R = H) or 2-A-acetamido-2-deoxygalactose-UDP (2-GalNAcA-UDP), such as 2- chloroacetamido-2-deoxygalactose-UDP (2-GalNAcCl-UDP, (21) with X = CI), 2- thioacetamido-2-deoxygalactose-UDP (2-GalNAcSH-UDP, (18) with 1 CH2 and R = H). Alternatively, A may be indirectly substituted to the sugar derivative as part of another functional group that in turn is substituting a hydroxyl group or is attached to a hydroxyl group. Examples of such other functional group include an (hetero)alkyl chain or a (hetero)aryl chain.
P is herein defined as a nucleotide. P is preferably selected from the group consisting of a nucleoside monophosphate and a nucleoside diphosphate, more preferably from the group consisting of uridine diphosphate (UDP), guanosine diphosphate (GDP), thymidine diphosphate (TDP), cytidine diphosphate (CDP) and cytidine monophosphate (CMP), more preferably from the group consisting of uridine diphosphate (UDP), guanosine diphosphate (GDP), cytidine diphosphate and (CDP). Most preferably, P is UDP.
Several compounds of formula Su(A)x-P, wherein a nucleoside monophosphate or a nucleoside diphosphate P is linked to a sugar derivative Su(A)x, are known in the art. For example Wang et al, Chem. Eur. J. 2010, 16, 13343-13345, Piller et al, ACS Chem. Biol. 2012, 7, 753, Piller et al, Bioorg. Med. Chem. Lett. 2005, 15, 5459-5462 and WO 2009/102820 (Qasba et al), all incorporated by reference herein, disclose a number of compounds Su(A)x-P and their syntheses.
Several examples (18 - 23) of uridine diphosphates linked to thiol-, halo- and mesityloxy-substitued sugars and sugar derivatives, Su(A)x-UDP, are shown below. Also an example of Su(A)x-P wherein A is a thiol precursor (18 - 20, with R = Ac), is shown. As described above, in 18 - 20 it is preferred that o is 1, 2, 3, 4, 5 or 6, preferably 1, 2, 3 or 4, more preferably 1, 2 or 3. Furthermore, R is H or Ac.
Figure imgf000035_0001
Additional examples (3 - 17) of keto-, alkynyl-, halogen, thiol, thiolated acetamido- and halogenated acetamido-substitued sugars and sugar derivatives are shown in Figure 3, all of which may be converted into their corresponding UDP sugars Su(A)x-UDP, some of which are depicted in Figure 4. The synthesis of some of the thiol derivatives or halogenated acetamide derivatives, all of which can be readily prepared from the common precursor 27, is depicted in Figure 5.
As was described above, several of the sugar derivative nucleotides Su(A)x-P that may be employed in the process for the preparation of a modified glycoprotein according to the invention are a substrate for a wild type galactosyltransferase. For these sugar derivative nucleotides Su(A)x-P, the process according to the invention may be performed in the presence of a wild type galactosyltransferase, preferably a wild type /3(l,4)-galactosyltransferase, more preferably a /3(l,4)-galactosyltransf erase I, as a catalyst. When a wild type galactosyltransferase is used as a catalyst, it is preferred that Su(A)x-P is selected from the group consisting of Su(A)x-P wherein x is 1 and wherein A is present on C6 of the sugar or sugar derivative, and wherein A is as defined above. A may be directly substituted to the sugar derivative instead of an hydroxyl group. Examples include 6-A-6-deoxygalactose-UDP (6-A-Gal-UDP), 6-chloro-6- deoxygalactose-UDP (6-ClGal-UDP, (22) with 1 CH2 and X = CI), 6-thio-6- deoxygalactose-UDP (6-HSGal-UDP, (19) with 1 CH2 and R = H)
Alternatively, A may be indirectly substituted to the sugar derivative as part of an acetamido group that in turn is substituting a hydroxyl group. Examples include 6-A- acetamido-6-deoxygalactose-UDP (6-GalNAcA-UDP), such as 6-chloroacetamido-6- deoxygalactose-UDP (6-GalNAcCl-UDP, (23) with X = CI), 6-thioacetamido-6- deoxygalactose-UDP (6-GalNAcSH-UDP, (20) with 1 CH2 and R = H). Alternatively, A may be indirectly substituted to the sugar derivative as part of another functional group that in turn is substituting a hydroxyl group or is attached to a hydroxyl group. Examples of such other functional group include an (hetero)alkyl chain or a (hetero)aryl chain.
In a preferred embodiment of the process for the preparation of a modified glycoprotein, Su(A)x comprises 1 or 2 functional groups A, i.e. preferably x is 1 or 2. More preferably, x is 1. In a preferred embodiment, Su is galactose (Gal). In a further preferred embodiment, x is 1 or 2 and Su is Gal, and most preferably, x is 1 and Su is Gal.
In a preferred embodiment, Su(A)x is selected from the group consisting of 6- GalNAcCl, 6-GalNAcSH, 2-GalNAcCl, 2-GalNAcSH, 6-ClGal, 2-ClGal, 2-HSGal and 6-HSGal, more preferably form the group consisting of 6-GalNAcCl, 6-GalNAcSH, 2- GalNAcCl, 2-GalNAcSH, 6-ClGal-and 2-ClGal.
In a further preferred embodiment, x is 1 and Su(A)x is selected from the group consisting of 6-GalNAcCl, 6-GalNAcSH, 2-GalNAcCl, 2-GalNAcSH, 6-ClGal, 2- ClGal, 2-HSGal and 6-HSGal, more preferably from the group consisting of 6- GalNAcCl, 6-GalNAcSH, 2-GalNAcCl, 2-GalNAcSH, 6-ClGal-and 2-ClGal.
In a preferred embodiment, A is a thiol group or a halogen.
In a particularly preferred embodiment of the process for the modification of a modified glycoprotein according to the invention, Su(A)x is selected from the group consisting of 2-GalNAcSH, 2-GalNAcX, 2-GalNAcOS(0)2R8, 6-GalNAcSH, 6- GalNAcX and 6-GalNAcOS(0)2R8. In an even more preferred embodiment, x is 1 or 2 and Su(A)x is selected from the group consisting of 2-GalNAcSH, 2-GalNAcX, 2- GalNAcOS(0)2R8, 6-GalNAcSH, 6-GalNAcX and 6-GalNAcOS(0)2R8. In a most preferred embodiment, x is 1 and Su(A)x is selected from the group consisting of 2- GalNAcSH, 2-GalNAcX, 2-GalNAcOS(0)2R8, 6-GalNAcSH, 6-GalNAcX and 6- GalNAcOS(0)2R8, wherein X is CI or Br, preferably CI.
In a particularly preferred embodiment of the process for the preparation of a modified glycoprotein according to the invention, Su(A)x-P is selected from the group consisting of 2-GalNAcSH-UDP ((18) with 1 CH2 and R = H), 2-GalNAcX-UDP (21), 2-GalNAcOS(0)2R8-UDP, 6-GalNAcSH-UDP ((20) with 1 CH2 and R = H), 6- GalNAcX-UDP (23) and 6-GalNAcOS(0)2R8-UDP, and the catalyst is bovine β(1,4)- Gal-Tl comprising a mutant catalytic domain GalT (Y289L); wherein X is CI, Br or I; and wherein R8 is a methyl group, an ethyl group, a phenyl group or a p-tolyl group.
In a further preferred embodiment 2-GalNAcX-UDP is 2-GalNAcCl-UDP or 2- GalNAcBr-UDP, more preferably 2-GalNAcCl-UDP, and 6-GalNAcX-UDP is 6- GalNAcCl-UDP or 6-GalNAcBr-UDP, more preferably 6-GalNAcCl-UDP. In another preferred embodiment, R8 in 2-GalNAcOS(0)2R8-UDP is methyl, phenyl or p-tolyl, most preferably methyl, and R8 in 6-GalNAcOS(0)2R8-UDP is methyl, phenyl or p- tolyl, most preferably R8 is methyl.
In another particularly preferred embodiment of the process for the preparation of a modified glycoprotein according to the invention, Su(A)x-P is selected from the group consisting of 6-HSGal-UDP, 6-XGal-UDP, 6-R8S(0)2OGal-UDP, and the catalyst is a wild-type human GalT; wherein X is CI, Br or I; and wherein R8 is a methyl group, an ethyl group, a phenyl group or a p-tolyl group. X is more preferably CI or Br, most preferably CI. R8 is more preferably methyl, phenyl or p-tolyl, most preferably methyl. The human GalT is preferably a human /¾-Gal-Tl, a human ?(1,4)-Gal-T2, a human ytf(l,4)-Gal-T3 and a human ytf(l,4)-Gal-T4.
The process for the preparation of a modified glycoprotein according to the invention may further comprise a step comprising providing a glycoprotein comprising a glycan comprising a terminal GlcNAc-moiety at the non-reducing end. The invention thus also relates to a process for the preparation of a modified glycoprotein, the process comprising the steps of:
(i) providing a glycoprotein comprising a glycan comprising a terminal GlcNAc-moiety at the non-reducing end; and
(ii) contacting a glycoprotein comprising a glycan comprising a terminal GlcNAc-moiety with Su(A)x-P in the presence of a suitable catalyst; wherein Su(A)x is a sugar derivative Su comprising x functional groups A wherein x is 1, 2, 3 or 4 and wherein A is independently selected from the group consisting of a thiol group or a precursor thereof, a halogen, a sulfonyloxy group, a halogenated acetamido group, a mercaptoacetamido group and a sulfonated hydroxy acetamido group; wherein P is a nucleotide; wherein a suitable catalyst is defined as a galactosyltransferase or a galactosyltransferase comprising a mutant catalytic domain, wherefore Su(A)x-P is a substrate; and wherein a glycan comprising a terminal GlcNAc-moiety is a glycan according to formula (101) or (102):
Figure imgf000038_0001
101 102
wherein b, d, e and G are as defined above. Preferably, the catalyst in step(ii) is selected from the group consisting of (1,4)- galactosyltransferases, /3(l,3)-N-galactosyltransferases, /3(l,4)-galactosyltransferases comprising a mutant catalytic domain and /3(l,3)-N-galactosyltransferases comprising a mutant catalytic domain. The catalyst is described in more detail above.
A glycoprotein comprising a glycan comprising a terminal GlcNAc-moiety at the non-reducing end, i.e. a terminal non-reducing GlcNAc protein, may be provided in several ways, for example by (a) trimming of N-glycoprotein with an endo-glycosidase as described in EMBO J.2001, 72, 3046 (incorporated by reference) or (b) expression of hybrid N-glycoprotein in the presence of swainsonine as for example described by Satoh et al. in Glycobiology 2006, 77, 104-1 18, incorporated by reference (followed by sialidase/galactosidase treatment). In a particular embodiment, the invention also relates to a process for the preparation of a modified glycoprotein, the process comprising the steps of:
(i) providing a glycoprotein comprising a glycan comprising a terminal GlcNAc-moiety at the non-reducing end by trimming a glycoprotein comprising an oligosaccharide glycan by the action of an endo-glycosidase; and
(ii) contacting a glycoprotein comprising a glycan comprising a terminal GlcNAc-moiety with Su(A)x-P in the presence of a suitable catalyst; wherein Su(A)x is a sugar derivative Su comprising x functional groups A wherein x is 1, 2, 3 or 4 and wherein A is independently selected from the group consisting of a thiol group or a precursor thereof, a halogen, a sulfonyloxy group, a halogenated acetamido group, a mercaptoacetamido group and a sulfonated hydroxy acetamido group; wherein P is a nucleotide; wherein a suitable catalyst is defined as a galactosyltransferase or a galactosyltransferase comprising a mutant catalytic domain, wherefore Su(A)x-P is a substrate; and wherein a glycan comprising a terminal GlcNAc-moiety is a glycan according to formula (101) or (102):
Figure imgf000039_0001
101 102
wherein b, d, e and G are as defined above.
Preferably, the catalyst is selected from the group consisting of (1,4)- galactosyltransferases, /3(l,3)-N-galactosyltransferases, /3(l,4)-galactosyltransferases comprising a mutant catalytic domain and /3(l,3)-N-galactosyltransferases comprising a mutant catalytic domain. The catalyst is described in more detail above.
A glycan may be attached to glycoprotein via CI of a GlcNAc-residue, that is bonded to the amide side chain of an asparagine amino acid that is part of the glycoprotein. The oligosaccharide may be attached via an N-glycosidic bond, in most cases to an asparagine (Asn) or arginine (Arg) amino acid side chain.
Numerous different types of glycans exist. For example the Fc regions of Ig antibodies bear a highly conserved N-glycosylation site. The N-glycans attached to this site are predominantly core-fucosylated diantennary structures of the complex type. Another type of glycan is the class of high mannose glycans. High-mannose typically comprises two N-acetylglucosamines and a varying number of mannose residues.
As described above, a glycan may be bonded to the glycoprotein via a GlcNAc- residue, and this GlcNAc residue may be fucosylated. In Figure 1, this is denoted by b: when b is 0, said GlcNAc-residue is non- fucosylated and when b is 1, said GlcNAc is fucosylated.
In a large number of glycans, a second GlcNAc-residue is bonded to the GlcNac- residue that is directly bonded to the glycoprotein, as is also seen in Figure 1, (b) and (c). A glycan wherein a second GlcNAc-residue is bonded to the GlcNac-residue that is directly bonded to the glycoprotein (e.g. Figure 1(b) and (c)), can be trimmed in order to obtain a glycoprotein comprising a glycan comprising a terminal GlcNAc-moiety according to formula (101) at the non-reducing end. Trimming occurs in between said two GlcNAc-residues. Trimming of a glycan provides a glycan according to formula (101), as shown in Figure 1 (a). Such a GlcNAc-residue that is covalently bonded to an N-glycosylation site, preferably via an N-glycosidic bond via CI of said GlcNAc and an amide present in an amino acid side chain of the antibody, is herein referred to as a "terminal GlcNAc-moiety", and also "core-GlcNAc-moiety". Said terminal GlcNAc- moiety or core-GlcNAc-moiety is optionally fucosylated.
The trimming of a glycan occurs by the action of a suitable enzyme, and after trimming the oligosaccharide (also referred to as glycan) a terminal GlcNAc-moiety is obtained. By trimming, a glycan comprising a terminal GlcNAc-moiety according to formula (101) is obtained. This terminal GlcNAc-moiety may be fucosylated (b in Figure 1(a) is 1) or non-fucosylated (b is 0).
A "suitable enzyme" is defined as an enzyme wherefore the oligosaccharide that is to be trimmed is a substrate. The preferred type of enzyme that is to be used in step (i) of this particular embodiment of the process according to the invention depends on the specific glycan or glycans that is or are trimmed. In a preferred embodiment of this particular embodiment of the process according to the invention, the enzyme in step (i) of this particular embodiment of the process is selected from the group of endo- glycosidases. Endo-glycosidases are capable of cleaving internal glycosidic linkages in glycan structures, which provides a benefit to remodeling and synthetic endeavors. For example, endo-glycosidases can be employed for facile homogenization of heterogeneous glycan populations, when they cleave at predictable sites within conserved glycan regions. One of the most significant classes of endoglycosidases in this respect comprises the endo-P-N-acetylglucosaminidases (EC 3.2.1.96, commonly known as Endos and ENGases), a class of hydrolytic enzymes that remove N-glycans from glycoproteins by hydrolyzing the -l,4-glycosidic bond in the Ν,Ν'- diacetylchitobiose core (reviewed by Wong et al. Chem. Rev. 2011, 111, 4259, incorporated by reference herein), leaving a single core N-linked GlcNAc residue. Endo-P-N-acetylglucosaminidases are found widely distributed through nature with common chemoenzymatic variants including Endo D, which is specific for pauci mannose; Endo A and Endo H, which are specific for high mannose; Endo F subtypes, which range from high mannose to biantennary complex; and Endo M, which can cleave most N-glycan structures (high mannose/compl ex-type/hybrid-type), except fucosylated glycans, and the hydrolytic activity for the high-mannose type oligosaccharides is significantly higher than that for the complex-and hybrid-type oligosaccharides. These ENGases show specificity toward the distal N-glycan structure and not the protein displaying it, making them useful for cleaving most N-linked glycans from glycoproteins under native conditions.
Endoglycosidases Fl, F2, and F3 are most suitable for deglycosylation of native proteins. The linkage specificities of endo Fl, F2, and F3 suggest a general strategy for deglycosylation of proteins that may remove all classes of N-linked oligosaccharides without denaturing the protein. Biantennary and triantennary structures can be immediately removed by endoglycosidases F2 and F3, respectively. Oligo- mannose and hybrid structures can be removed by Endo Fl .
Endo F3 is unique in that its cleavage is sensitive to the state of peptide linkage of the oligosaccharide, as well as the state of core fucosylation. Endoglycosidase F3 cleaves asparagine-linked biantennary and triantennary complex oligosaccharides. It will cleave non-fucosylated biantennary and triantennary structures at a slow rate, but only if peptide-linked. Core fucosylated biantennary structures are efficient substrates for Endo F3, which activity up to 400-fold. There is no activity on oligomannose and hybrid molecules. See for example Tarentino et al. Glycobiology 1995, 5, 599, incorporated by reference herein.
Endo S is a secreted endoglycosidase from Streptococcus pyogenes, and also belongs to the glycoside hydrolase family 18, as disclosed by Collin et al. (EMBO J. 2001, 20, 3046, incorporated by reference herein). In contrast to the ENGases mentioned above, however, the endo S has a more defined specificity and is specific for cleaving only the conserved N-glycan in the Fc domain of human IgGs (no other substrate has been identified to date), suggesting that a protein-protein interaction between the enzyme and IgG provides this specificity.
Endo S49 is described in WO 2013/037824 (Genovis AB), incorporated by reference herein. Endo S49 is isolated from Streptococcus poyogenes NZ131 and is a homologue of Endo S. Endo S49 has a specific endoglycosidase activity on native IgG and cleaves a larger variety of Fc glycans than Endo S. In a preferred embodiment, the enzyme in step (i) of this embodiment is an endo- β-Ν-acetylglucosaminidase. In a further preferred embodiment, the endo-β-Ν- acetylglucosaminidase is selected from the group consisting of Endo S, Endo S 49, Endo Fl, Endo F2, Endo F3, Endo H, Endo M, Endo A, and any combination thereof.
When the glycan to be trimmed is a diantennary structure of the complex type, the endo-P-N-acetylglucosaminidase is preferably selected from the group consisting of Endo S, Endo S49, Endo Fl, Endo F2, Endo F3, and a combination thereof.
When the oligosaccharide to be trimmed is a diantennary structure of the complex type (i.e. according to Figure 1(c), and it is present at the IgG conserved N- glycosylation site at N297, the endo-P-N-acetylglucosaminidase is preferably selected from the group consisting of Endo S, Endo S49, Endo Fl, Endo F2, Endo F3, and a combination thereof, more preferably from the group consisting of Endo S, Endo S49, and a combination thereof.
When the glycan to be trimmed is a diantennary structure of the complex type, and it is not present at the IgG conserved N-glycosylation site at N297, the endo-β-Ν- acetylglucosaminidase is preferably selected from the group consisting of Endo Fl, Endo F2, Endo F3, and a combination thereof. When the glycan to be trimmed is a high mannose, the endo-β-Ν- acetylglucosaminidase is preferably selected from the group consisting of Endo H, Endo M, Endo A and Endo F 1.
The present invention thus also relates to a process for the preparation of a modified glycoprotein, the process comprising the steps of:
(i) trimming a glycan of a glycoprotein comprising an oligosaccharide glycan by the action of an endo- -N-acetylglucosaminidase, in order to provide a glycoprotein comprising a glycan comprising a terminal GlcNAc-moiety according to formula (101); and
(ii) contacting said glycoprotein comprising a glycan according to formula (101) with Su(A)x-P in the presence of a suitable catalyst; wherein Su(A)x is a sugar derivative Su comprising x functional groups A wherein x is 1, 2, 3 or 4 and wherein A is independently selected from the group consisting of a thiol group or a precursor thereof, a halogen, a sulfonyloxy group, a halogenated acetamido group, a mercaptoacetamido group and a sulfonated hydroxy acetamido group; wherein P is a nucleotide; wherein a suitable catalyst is defined as a galactosyltransferase or a galactosyltransferase comprising a mutant catalytic domain, wherefore Su(A)x-P is a substrate; and wherein a glycan comprising a terminal GlcNAc-moiety is a glycan according to formula (101) or (102):
-—
Figure imgf000043_0001
101 102
wherein b, d, e and G are as defined above.
Preferably, the catalyst in step(ii) is selected from the group consisting of (1,4)- galactosyltransferases, /3(l,3)-N-galactosyltransferases, /3(l,4)-galactosyltransferases comprising a mutant catalytic domain and /3(l,3)-N-galactosyltransferases comprising a mutant catalytic domain. Galactosyltransferases are described in more detail above. In a further preferred embodiment, the endo- -N-acetylglucosaminidase is selected from the group consisting of Endo S, Endo S 49, Endo Fl, Endo F2, Endo F3, Endo H, Endo M, Endo A, and any combination thereof, more preferably, the endo-β- N-acetylglucosaminidase is selected from the group consisting of Endo S, Endo S 49, Endo Fl, Endo F2, Endo F3 and any combination thereof. Most preferably, the endo-β- N-acetylglucosaminidase is Endo S or Endo S49.
The trimming step (2) of the process according to the invention is preferably performed in a suitable buffer solution, such as for example phosphate, buffered saline (e.g. phosphate-buffered saline, tris-buffered saline), citrate, HEPES, tris and glycine. Suitable buffers are known in the art. Preferably, the buffer solution is phosphate- buffered saline (PBS) or tris buffer.
The process is preferably performed at a temperature in the range of about 4 to about 50°C, more preferably in the range of about 10 to about 45°C, even more preferably in the range of about 20 to about 40°C, and most preferably in the range of about 30 to about 37°C.
The process is preferably performed a pH in the range of about 5 to about 9, preferably in the range of about 5.5 to about 8.5, more preferably in the range of about 6 to about 8. Most preferably, the process is performed at a pH in the range of about 7 to about 8.
Modified glycoprotein
The present invention also relates to a glycoprotein, obtainable by the process for the preparation of a modified glycoprotein according to the invention.
A modified glycoprotein is defined as a glycoprotein comprising a modified glycan, said glycan comprising a GlcNAc-Su(A)x disaccharide-moiety at the non- reducing end, wherein Su(A)x is as defined above. In a preferred embodiment, the glycoprotein is an antibody.
In a preferred embodiment, the glycoprotein comprises a glycan according to formula (105) or (106):
Figure imgf000045_0001
105 106
wherein:
b is 0 or 1;
d is 0 or 1;
e is 0 or 1 ;
G is a monosaccharide, or a linear or branched oligosaccharide comprising 2 to 20 sugar moieties; and
Su(A)x is a sugar derivative Su comprising x functional groups A wherein x is 1, 2, 3 or 4 and wherein A is independently selected from the group consisting of a thiol group, a halogen, a sulfonyloxy group, a halogenated acetamido group, a mercaptoacetamido group and a sulfonated hydroxyacetamido group.
As described above, when in the process for the preparation of a modified glycoprotein according the invention, functional group A is a precursor for a thiol group, said precursor is transformed into a thiol group during said process. As a consequence, when the process is performed with a sugar derivative Su(A)x comprising a precursor for a thiol group as a functional group A, a modified glycoprotein comprising a thiol group as a functional group A is obtained.
Preferably, when d is 0 then e is 1, and when e is 0 then d is 1.
G represents a monosaccharide, or a linear or branched oligosaccharide comprising 2 to 20, preferably 2 to 12, more preferably 2 to 10, even more preferably 2 to 8 and most preferably 2 to 6 sugar moieties. Sugar moieties that may be present in a glycan are known to a person skilled in the art, and include e.g. glucose (Glc), galactose (Gal), mannose (Man), fucose (Fuc), N-acetylglucosamine (GlcNAc), N- acetylgalactosamine (GalNAc), N-acetylneuraminic acid (NeuNAc) or sialic acid, or xylose (Xyl).
More preferably, the modified glycoprotein obtainable by the process described above is a glycoprotein according to formula (107) or (108): -
Figure imgf000046_0001
107
wherein:
Pr represents a protein;
y is 1 to 20; and
Su(A)x, b, d, e and G are as defined above.
In a preferred embodiment y is 1 to 12, more preferably y is 1, 2, 3, 4, 5, 6, 7 or 8, and even more preferably y is 1, 2, 3 or 4. Most preferably y is 1 or 2. In yet another preferred embodiment, y is 2, 4, 6 or 8. This embodiment is particularly preferred when the glycoprotein to be modified is an antibody, i.e. when Pr is Ab.
In a preferred embodiment, CI of the core-sugar moiety of the modified glycan, said glycan comprising a GlcNAc-Su(A)x disaccharide-moiety at the non-reducing end, is bonded to the protein via an N-glycosidic bond to a nitrogen atom in an amino acid residue in said protein, more preferably to an amide nitrogen atom in the side chain of an asparagine (Asn) or an arginine (Arg) amino acid. However, CI of the core-sugar- moiety of said modified glycan may also be bonded to the protein via an O-glycosidic bond to an oxygen atom in an amino acid residue in said protein, more preferably to an oxygen atom in the side chain of a serine (Ser) or threonine (Thr) amino acid. In this embodiment, it is preferred that the core-sugar-moiety of said glycan is an O-GlcNAc- moiety or an O-GalNAc moiety, preferably an O-GlcNAc moiety. CI of the core- sugar-moiety of said modified glycan may also be bonded to the protein via a C- glycosidic bond to a carbon atom on the protein, e.g. to tryptophan (Trp). As described above, a glycoprotein may comprise more than one oligosaccharide chains, and may constitute of a combination of N-linked, O-linked and C-linked glycoproteins.
Said modified glycan may be present at a native glycosylation site of a protein, but may also be introduced on a different site on a protein.
In a preferred embodiment the modified glycoprotein is a modified antibody. Said antibody (Ab) comprises a glycan comprising a GlcNAc-Su(A)x disaccharide-moiety at the non-reducing end. Such an antibody is herein also referred to as a modified antibody. A preferred modified antibody is an antibody according to formula (107) or (108) as defined above, wherein Pr is Ab. In this embodiment, it is further preferred that y is 1, 2, 3, 4, 5, 6, 7 or 8, and even more preferred that y is 1, 2, 3 or 4.
As was defined above, said antibody may be a whole antibody, but also an antibody fragment. When the antibody is a whole antibody, said antibody preferably comprises one or more, more preferably one, modified glycans on each heavy chain. Said whole antibody thus preferably comprises two or more, preferably two, four, six or eight of said glycans, more preferably two or four, and most preferably two glycans. In other words, when said antibody is a whole antibody, y is preferably 2, 4, 6 or 8, more preferably y is 2 or 4, and most preferably y is 2. When the antibody is an antibody fragment, it is preferred that y is 1, 2, 3 or 4, and more preferably y is 1 or 2.
In a preferred embodiment, said antibody is a monoclonal antibody (mAb). Preferably, said antibody is selected from the group consisting of IgA, IgD, IgE, IgG and IgM antibodies. More preferably, said antibody is an IgG antibody, and most preferably said antibody is an IgGl antibody.
In a preferred embodiment, the glycoprotein in the modified antibody is attached to the conserved N-glycosylation site in the Fc-fragment at asparagine in the region 290-305, typically N297. As described above, said modified glycan may be present at a native glycosylation site of a protein, in this case an antibody, but may also be present on a different site. Figure 13 shows different glycoforms of a monoclonal antibody, e.g. IgG. The modified glycan may be on any position in the heavy or light chain.
The sugar derivative Su(A)x and preferred embodiments thereof are described in detail above. The definitions of the groups A and preferred embodiments thereof, and preferred embodiments of the process for the preparation of a modified glycoprotein also hold for the modified glycoprotein obtainable by said process. Thus, in a preferred embodiment, x is 1 or 2, more preferably x is 1. In another preferred embodiment, functional group A is a thiol group, a halogen, or a halogenated acetamido group, more preferably a halogen. Further examples of Su(A)x present in a modified glycoprotein and in a modified antibody according to the invention include 6-A-6-deoxygalactose (6-A-Gal), such as 6-chloro-6-deoxygalactose (6-ClGal), 6-thio-6-deoxygalactose (6-HSGal) or 2-A-2- deoxygalactose (2-A-Gal), such as 2-chloro-2-deoxygalactose (2-ClGal), 2-thio-2- deoxygalactose (2-HSGal). Alternatively, A may be indirectly substituted to the sugar derivative as part of an acetamido group that in turn is substituting a hydroxyl group. Examples include 6-A-acetamido-6-deoxygalactose (6-GalNAcA), such as 6- chloroacetamido-6-deoxygalactose (6-GalNAcCl), 6-thioacetamido-6-deoxygalactose (6-GalNAcSH) or 2-A-acetamido-2-deoxygalactose (2-GalNAcA), such as 2- chloroacetamido-2-deoxygalactose (2-GalNAcCl), 2-thioacetamido-2-deoxygalactose (2-GalNAcSH). Alternatively, A may be indirectly substituted to the sugar derivative as part of another functional group that in turn is substituting a hydroxyl group or is attached to a hydroxyl group. Examples of such other functional group include an (hetero)alkyl chain or a (hetero)aryl chain.
In a preferred embodiment, Su(A)x is selected from the group consisting of 6- GalNAcCl, 6-GALNAcSH, 2-GalNAcCl and 2-GalNAcSH.
In a preferred embodiment, Su(A)x comprises 1 or 2 functional groups A, i.e. preferably x is 1 or 2. More preferably, x is 1. In another preferred embodiment, Su is galactose (Gal). In a further preferred embodiment, x is 1 or 2 and Su is Gal, and most preferably, x is 1 and Su is Gal.
In a preferred embodiment, Su(A)x is selected from the group consisting of 6- GalNAcCl, 6-GalNAcSH, 2-GalNAcCl, 2-GalNAcSH, 6-ClGal, 2-ClGal, 2-HSGal and 6-HSGal, more preferably form the group consisting of 6-GalNAcCl, 6-GalNAcSH, 2- GalNAcCl, 2-GalNAcSH, 6-ClGal-and 2-ClGal.
In a further preferred embodiment, x is 1 and Su(A)x is selected from the group consisting of 6-GalNAcCl, 6-GalNAcSH, 2-GalNAcCl, 2-GalNAcSH, 6-ClGal, 2- ClGal, 2-HSGal and 6-HSGal, more preferably from the group consisting of 6- GalNAcCl, 6-GalNAcSH, 2-GalNAcCl, 2-GalNAcSH, 6-ClGal-and 2-ClGal.
In a preferred embodiment of the modified glycoprotein according to the invention, A is a thiol group or a halogen. The modified glycoprotein is preferably a thiol-modified glycoprotein or a halogen-modified glycoprotein. When the glycoprotein is a halogen-modified glycoprotein, it is preferably a chloride-modified glycoprotein, a bromide-modified glycoprotein or an iodide-modified glycoprotein, more preferably a chloride-modified glycoprotein or a bromide-modified glycoprotein, and most preferably a chloride-modified glycoprotein. More preferably, x is 1 and the modified glycoprotein is preferably a thiol-modified glycoprotein or a halogen-modified glycoprotein (preferably a chloride- or a bromide-modified glycoprotein, most preferably a chloride-modified glycoprotein).
In a particularly preferred embodiment of the modified glycoprotein according to the invention, Su(A)x is selected from the group consisting of 2-GalNAcSH, 2- GalNAcX, 2-GalNAcOS(0)2R8, 6-GalNAcSH, 6-GalNAcX and 6-GalNAcOS(0)2R8. In an even more preferred embodiment, x is 1 or 2 and Su(A)x is selected from the group consisting of 2-GalNAcSH, 2-GalNAcX, 2-GalNAcOS(0)2R8, 6-GalNAcSH, 6- GalNAcX and 6-GalNAcOS(0)2R8. In a most preferred embodiment, x is 1 and Su(A)x is selected from the group consisting of 2-GalNAcSH, 2-GalNAcX, 2- GalNAcOS(0)2R8, 6-GalNAcSH, 6-GalNAcX and 6-GalNAcOS(0)2R8. R8, and preferred embodiments of R8, are as defined above.
When the modified glycoprotein according to the invention comprises a thiol group, said modified glycoprotein may be referred to as a thiol-modified glycoprotein. When a modified glycoprotein comprises a halogen, said glycoprotein may be referred to as a halogen-modified glycoprotein. When a modified glycoprotein comprises a sulfonyloxy group, said glycoprotein may be referred to as a sulfonyloxy-modified glycoprotein. When a modified glycoprotein comprises a mercaptoacetamido group, said glycoprotein may be referred to as a mercaptoacetamido-modified glycoprotein. When a modified glycoprotein comprises a halogenated acetamido group, said glycoprotein may be referred to as a halogenated acetamido-modified glycoprotein. When a modified glycoprotein comprises a sulfonated hydroxyacetamido group, said glycoprotein may be referred to as a sulfonated hydroxyacetamido-modified glycoprotein.
The invention also relates to the use of a modified glycoprotein according to the invention in the preparation of a glycoprotein-conjugate, wherein a glycoprotein- conjugate is defined as a glycoprotein that is conjugated to a molecule of interest D via a linker L.
Process for the preparation of a glycoprotein-conjugate
The present invention also relates to the use of a modified glycoprotein according to the invention in the preparation of a protein-conjugate. A protein-conjugate is herein defined as a protein that is conjugated to a molecule of interest (D) via a linker (L). The protein-conjugate according to the invention may be conjugated to one or to more than one molecule of interest (D) via a linker (L).
The invention also relates to the use of a modified antibody according to the invention in the preparation of an antibody-conjugate, wherein an antibody-conjugate is defined as an antibody that is conjugated to a molecule of interest (D) via a linker (L).
A molecule of interest may for example be a reporter molecule, a diagnostic molecule, an active substance, an enzyme, an amino acid (including an unnatural amino acid), a (non-catalytic) protein, a peptide, a polypeptide, an oligonucleotide, a glycan, a (poly)ethylene glycol diamine (e.g. l,8-diamino-3,6-dioxaoctane or equivalents comprising longer ethylene glycol chains), a polyethylene glycol chain, a polyethylene oxide chain, a polypropylene glycol chain, a polypropylene oxide chain, l,x- diaminoalkane (wherein x is the number of carbon atoms in the alkane), an azide or a (hetero)cycloalkynyl moiety, preferably a bivalent or bifunctional (hetero)cycloalkynyl moiety. In a preferred embodiment, the molecule of interest is selected from the group consisting of an amino acid (in particular lysine), an active substance, a reporter molecule, an azide and a (hetero)cycloalkynyl moiety.
An active substance is a pharmacological and/or biological substance, i.e. a substance that is biologically and/or pharmaceutically active, for example a drug or a prodrug, a diagnostic agent, an amino acid, a protein, a peptide, a polypeptide, a glycan, a lipid, a vitamin, a steroid, a nucleotide, a nucleoside, a polynucleotide, RNA or DNA. Examples of suitable peptide tags include cell-penetrating peptides like human lactoferrin or polyarginine. An example of a suitable glycan is oligomannose.
In a preferred embodiment, the active substance is selected from the group consisting of drugs and prodrugs. More preferably, the active substance is selected from the group consisting of pharmaceutically active compounds, in particular low to medium molecular weight compounds (e.g. about 200 to about 1500 Da, preferably about 300 to about 1000 Da), such as for example cytotoxins, antiviral agents, antibacterial agents, peptides and oligonucleotides. Examples of cytotoxins include camptothecins, doxorubicin, daunorubicin, taxanes, calicheamycins, duocarmycins, maytansines, auristatins and pyrrolobenzodiazepines (PBDs). Preferably, the cytotoxin is selected from the group consisting of colchicine, vinca alkaloids, camptothecins, doxorubicin, daunorubicin, taxanes, calicheamycins, tubulysins, irinotecans, an inhibitory peptide, amanitin, deBouganin, duocarmycins, maytansines, auristatins and pyrrolobenzodiazepines (PBDs). In a preferred embodiment, the cytotoxin is selected from the group consisting of camptothecins, doxorubicin, daunorubicin, taxanes, calicheamycins, duocarmycins, maytansines, auristatins and pyrrolobenzodiazepines (PBDs). In another preferred embodiment, the cytotoxin is selected from the group consisting of colchicine, vinca alkaloids, tubulysins, irinotecans, an inhibitory peptide, amanitin and deBouganin.
A reporter molecule is a molecule whose presence is readily detected, for example a diagnostic agent, a dye, a fluorophore, a radioactive isotope label, a contrast agent, a magnetic resonance imaging agent or a mass label. Examples of a fluorophore include all kinds of Alexa Fluor (e.g. Alexa Fluor 555), cyanine dyes (e.g. Cy3 or Cy5), coumarin derivatives, fluorescein, rhodamine, allophycocyanin and chromomycin.
Examples of radioactive isotope label include 99mTc, U 1ln, 18F, 14C, 64 Cu, 13 I or 123I, which may or may not be connected via a chelating moiety such as DTP A, DOT A, NOTA or HYNIC.
In the protein-conjugate according to the invention, the molecule of interest (D) is conjugated to the antibody via a linker (L). Linkers or linking units are well known in the art, and are described in more detail below. A linker-conjugate is herein defined as a linker (L) that is conjugated to a molecule of interest (D). The linker-conjugate according to the invention may comprise one or more than one molecule of interest (D). The linker-conjugate preferably is of the formula B-L(D)r, wherein D is as defined above, and B and L are as defined below, and r is 1 to 20. Preferably r is 1 to 10, more preferably r is 1 to 8, even more preferably r is 1, 2, 3, 4, 5 or 6, even more preferably r is 1, 2, 3 or 4, even more preferably r is 1 or 2 and most preferably r is 1.
The invention also relates to a process for the preparation of a glycoprotein- conjugate, said process comprising reacting a modified glycoprotein according to the invention with a linker-conjugate, wherein said linker-conjugate comprises a functional group B and one or more molecules of interest, wherein said functional group B is a functional group that is capable of reacting with a functional group A on a glycan of the modified glycoprotein, and wherein functional group A is as defined above. The invention relates to a process for the preparation of a glycoprotein-conjugate, the process comprising the steps of:
(a) contacting a glycoprotein comprising a glycan comprising a terminal GlcNAc-moiety with Su(A)x-P in the presence of a suitable catalyst; wherein Su(A)x is a sugar derivative Su comprising x functional groups A wherein x is 1, 2, 3 or 4 and wherein A is independently selected from the group consisting of a thiol group or a precursor thereof, a halogen, a sulfonyloxy group, a halogenated acetamido group, a mercaptoacetamido group and a sulfonated hydroxyacetamido group; wherein P is a nucleotide; wherein a suitable catalyst is defined as a galactosyltransferase or a galactosyltransf erase comprising a mutant catalytic domain, wherefore Su(A)x-P is a substrate; and wherein a glycan comprising a terminal GlcNAc-moiety is a glycan according to formula (101) or (102):
Figure imgf000052_0001
101 102
wherein:
b is 0 or 1;
d is 0 or 1;
e is 0 or 1 ; and
G is a monosaccharide, or a linear or branched oligosaccharide comprising 2 to 20 sugar moieties;
to obtain a modified glycoprotein; and
(b) reacting the modified glycoprotein with a linker-conjugate, wherein said linker-conjugate comprises a functional group B and one or more molecules of interest (D), wherein said functional group B is a functional group that is capable of reacting with a functional group A on a glycan of the modified glycoprotein, and wherein functional group A is independently selected from the group consisting of a thiol group, a halogen, a sulfonyloxy group, a halogenated acetamido group, a mercaptoacetamido group and a sulfonated hydroxyacetamido group. In a preferred embodiment of the process for the preparation of an glycoprotein- conjugate, Su(A)x comprises 1 or 2 functional groups A, i.e. preferably x is 1 or 2. More preferably, x is 1. In another preferred embodiment, Su is galactose (Gal). In a further preferred embodiment, x is 1 or 2 and Su is Gal, and most preferably, x is 1 and Su is Gal. In these preferred embodiments it is further preferred that the linker- conjugate comprises 1 or 2, and most preferably 1, molecules of interest.
In a preferred embodiment, Su(A)x is selected from the group consisting of 6- GalNAcCl, 6-GalNAcSH, 2-GalNAcCl, 2-GalNAcSH, 6-ClGal, 2-ClGal, 2-HSGal and 6-HSGal, more preferably form the group consisting of 6-GalNAcCl, 6-GalNAcSH, 2- GalNAcCl, 2-GalNAcSH, 6-ClGal-and 2-ClGal. In these preferred embodiments it is further preferred that the linker-conjugate comprises 1 or 2, and most preferably 1, molecules of interest.
In a further preferred embodiment, x is 1 and Su(A)x is selected from the group consisting of 6-GalNAcCl, 6-GalNAcSH, 2-GalNAcCl, 2-GalNAcSH, 6-ClGal, 2- ClGal, 2-HSGal and 6-HSGal, more preferably from the group consisting of 6- GalNAcCl, 6-GalNAcSH, 2-GalNAcCl, 2-GalNAcSH, 6-ClGal-and 2-ClGal. In these preferred embodiments it is further preferred that the linker-conjugate comprises 1 or 2, and most preferably 1, molecules of interest.
In a particularly preferred embodiment of the modified antibody according to the invention, Su(A)x is selected from the group consisting of 2-GalNAcSH, 2-GalNAcX, 2-GalNAcOS(0)2R8, 6-GalNAcSH, 6-GalNAcX and 6-GalNAcOS(0)2R8. In an even more preferred embodiment, x is 1 or 2 and Su(A)x is selected from the group consisting of 2-GalNAcSH, 2-GalNAcX, 2-GalNAcOS(0)2R8, 6-GalNAcSH, 6- GalNAcX and 6-GalNAcOS(0)2R8. In a most preferred embodiment, x is 1 and Su(A)x is selected from the group consisting of 2-GalNAcSH, 2-GalNAcX, 2- GalNAcOS(0)2R8, 6-GalNAcSH, 6-GalNAcX and 6-GalNAcOS(0)2R8. In these preferred embodiments it is further preferred that the linker-conjugate comprises 1 or 2, and most preferably 1, molecules of interest. X and R8, and preferred embodiments thereof, are as defined above. Preferably X is CI.
The invention further relates to a process for the preparation of a glycoprotein- conjugate, said process comprising reacting a modified glycoprotein according to the invention with a linker-conjugate, wherein said linker-conjugate comprises a functional group B and one or more molecules of interest, wherein said functional group B is a functional group that is capable of reacting with a functional group A on a glycan of the modified glycoprotein, and wherein functional group A is independently selected from the group consisting of a thiol group, a halogen, a sulfonyloxy group, a halogenated acetamido group, a mercaptoacetamido group and a sulfonated hydroxyacetamido group.
A suitable linker-conjugate for the preparation of a protein-conjugate according to the invention is a linker-conjugate comprising a functional group B and a molecule of interest. Linkers (L), also referred to as linking units, are well known in the art. In a linker-conjugate as described herein, L is linked to a molecule of interest (D) as well as to a functional group (B), as was described above. Numerous methods for linking said functional group (B) and said molecule of interest (D) to L are known in the art. As will be clear to a person skilled in the art, the choice of a suitable method for linking a functional group (B) to one end and a molecule of interest (D) to another end of a linker depends on the exact nature of the functional group (B), the linker (L) and the molecule of interest (D).
A linker may have the general structure F1-L(F2)r, wherein F1 represents either a functional group B or a functional group that is able to react with a functional group F on the functional group B as described above, e.g. a (hetero)cycloalkynyl group, a terminal alkynyl group, a primary amine, an aminooxy group, a hydrazyl group, an azido group, an N-maleimidyl group, an acetamido group or a thiol group. F2 represents a functional group that is able to react with a functional group F on the molecule of interest.
Since more than one molecule of interest may be bonded to a linker, more than one functional group F2 may be present on L. As was described above, r is 1 to 20, preferably 1 to 10, more preferably 1 to 8, even more preferably 1, 2, 3, 4, 5 or 6, even more preferably 1, 2, 3 or 4 and most preferably, r is 1 or 2.
L may for example be selected from the group consisting of linear or branched
C1-C200 alkylene groups, C2-C200 alkenylene groups, C2-C200 alkynylene groups, C3- C200 cycloalkylene groups, C5-C200 cycloalkenylene groups, C8-C200 cycloalkynylene groups, C7-C200 alkylarylene groups, C7-C200 arylalkylene groups, C8-C200 arylalkenylene groups, C9-C200 arylalkynylene groups. Optionally the alkylene groups, alkenylene groups, alkynylene groups, cycloalkylene groups, cycloalkenylene groups, cycloalkynylene groups, alkylarylene groups, arylalkylene groups, arylalkenylene groups and arylalkynylene groups may be substituted, and optionally said groups may be interrupted by one or more heteroatoms, preferably 1 to 100 heteroatoms, said heteroatoms preferably being selected from the group consisting of O, S and R5, wherein R5 is independently selected from the group consisting of hydrogen, halogen, Ci - C24 alkyl groups, C6 - C24 (hetero)aryl groups, C7 - C24 alkyl(hetero)aryl groups and C7 - C24 (hetero)arylalkyl groups. Most preferably, the heteroatom is O.
F, F1 and F2 may for example be independently selected from the group consisting of hydrogen, halogen, R5, C4 - C10 (hetero)cycloalkyne groups,
Figure imgf000055_0001
-C≡CR5, -[C(R5)2C(R5)20]q-R5, wherein q is in the range of 1 to 200, -CN, -N3, -NCX, -XCN, -XR5, -N(R5)2, -+N(R5)3, -C(X)N(R5)2, -C(R5)2XR5, -C(X)R5, -C(X)XR5, -S(0)R5, -S(0)2R5, -S(0)OR5, -S(0)2OR5, -S(0)N(R5)2, -S(0)2N(R5)2, -OS(0)R5, -OS(0)2R5, -OS(0)OR5, -OS(0)2OR5, -P(0)(R5)(OR5), -P(0)(OR5)2, -OP(0)(OR5)2, -Si(R5)3, -XC(X)R5, -XC(X)XR5, -XC(X)N(R5)2, -N(R5)C(X)R5, -N(R5)C(X)XR5 and -N(R5)C(X)N(R5)2, wherein X is oxygen or sulphur and wherein R5 is as defined above.
Examples of suitable linking units include (poly)ethylene glycol diamines (e.g. l,8-diamino-3,6-dioxaoctane or equivalents comprising longer ethylene glycol chains), polyethylene glycol or polyethylene oxide chains, polypropylene glycol or polypropylene oxide chains and l,x-diaminoalkanes wherein x is the number of carbon atoms in the alkane.
Another class of suitable linkers comprises cleavable linkers. Cleavable linkers are well known in the art. For example Shabat et al. , Soft Matter 2012, 6, 1073, incorporated by reference herein, discloses cleavable linkers comprising self- immolative moieties that are released upon a biological trigger, e.g. an enzymatic cleavage or an oxidation event. Some examples of suitable cleavable linkers are peptide-linkers that are cleaved upon specific recognition by a protease, e.g. cathepsin, plasmin or metalloproteases, or glycoside-based linkers that are cleaved upon specific recognition by a glycosidase, e.g. glucoronidase, or nitroaromatics that are reduced in oxygen-poor, hypoxic areas. When A is a thiol group, linking of the thiol-modified glycoprotein with the linker-conjugate preferably takes place via a Michael-type addition reaction or a nucleophilic substitution reaction or a thiol-ene reaction. Functional group B is then preferably an N-maleimidyl group for the Michael-type addition, a halogenated acetamido group for the nucleophilic substitution reaction, or a terminal alkene for the thiol-ene reaction. The linker-conjugate is then preferably X-CH2C(0) HL(D)r or X- CH2C(0)N[L(D)r]2 wherein X is F, CI, Br or I, or a maleimide-linker-conjugate (120) as illustrated below.
Figure imgf000056_0001
When A is a halogen-modified glycoprotein, a halogenated acetamide-modified glycoprotein, a sulfonyloxy-modified glycoprotein or a sulfonated hydroxy acetamide- modified glycoprotein, linking of the modified glycoprotein with the linker-conjugate preferably takes place via reaction with a thiol to form a thioether. When A is a halogen, a halogenated acetamido group, a sulfonyloxy group or a sulfonated hydroxy acetamido group, linking of the modified glycoprotein with the linker-conjugate preferably takes place via reaction with a thiol to form a thioether. In other words, when the modified glycoprotein is a halogen-modified glycoprotein, a halogenated acetamide-modified glycoprotein, a sulfonyloxy-modified glycoprotein or a sulfonated hydroxy acetamide-modified glycoprotein, linking of the modified glycoprotein with the linker-conjugate preferably takes place via reaction with a thiol to form a thioether.
Functional group B comprises then preferably a thiol group, and a preferred linker-conjugate is HS-L(D)r. However, functional group B may also comprise an alcohol group or an amine group.
In particular, the invention relates to a process for the preparation of a glycoprotein-conjugate, wherein: (a) when said modified glycoprotein is a halogen-modified glycoprotein or a halogenated acetamido-modified glycoprotein, functional group B comprises a thiol group, an alcohol group or an amine group; or
(b) when said modified glycoprotein is a thiol-modified glycoprotein or a mercaptoacetamido-modified glycoprotein, functional group B comprises an N-maleimide group or a halogenated acetamido group or an alkene; or
(c) when said modified glycoprotein is a sulfonyloxy-modified glycoprotein or a sulfonated hydroxyacetamido-modified glycoprotein, functional group B comprises thiol group, an alcohol group or an amine group.
When said modified glycoprotein is a halogen-modified glycoprotein and functional group B comprises a thiol group, said thiol group may be an aliphatic or an aromatic thiol group. In a preferred embodiment said thiol group is an aromatic thiol group.
In a preferred embodiment, the modified glycoprotein is a thiol-modified glycoprotein and functional group B comprises an N-maleimide group or a halogenated acetamido group.
In another preferred embodiment, the modified glycoprotein is a thiol-modified glycoprotein and functional group B comprises an allenamide group. The invention therefore also relates to a process for the preparation of a glycoprotein-conjugate, wherein said modified glycoprotein is a thiol-modified glycoprotein and functional group B comprises an allenamide group.
The present invention further relates to the use of the glycoprotein according to the invention in the preparation of a glycoprotein-conjugate, wherein a glycoprotein- conjugate is defined as a glycoprotein that is conjugated to a molecule of interest D via a linker L.
Glycoprotein-conjugate
The invention also relates to a glycoprotein-conjugate obtainable by the process for the preparation of a glycoprotein-conjugate according to the invention.
In a preferred embodiment, the glycoprotein-conjugate according to the invention is an antibody-conjugate. In another preferred embodiment, the molecule of interest D is selected from the group consisting of a reporter molecule, an active substance, an enzyme, an amino acid, a protein, a peptide, a polypeptide, an oligonucleotide, a glycan, a (poly)ethylene glycol diamine, a polyethylene glycol chain, a polyethylene oxide chain, a polypropylene glycol chain, a polypropylene oxide chain and l,x-diaminoalkane, wherein x is the number of carbon atoms in the alkane and x is 1 - 200, an azide or a (hetero)cycloalkynyl moiety, preferably a bivalent or bifunctional (hetero)cycloalkynyl moiety.
In particular, the invention relates to a glycoprotein-conjugate obtainable by a process comprising the steps of:
(a) contacting a glycoprotein comprising a glycan comprising a terminal GlcNAc-moiety with Su(A)x-P in the presence of a suitable catalyst; wherein Su(A)x is a sugar derivative Su comprising x functional groups A wherein x is 1, 2, 3 or 4 and wherein A is independently selected from the group consisting of a thiol group or a precursor thereof, a halogen, a sulfonyloxy group, a halogenated acetamido group, a mercaptoacetamido group and a sulfonated hydroxy acetamido group; wherein P is a nucleotide; wherein a suitable catalyst is defined as a galactosyltransferase or a galactosyltransf erase comprising a mutant catalytic domain, wherefore Su(A)x-P is a substrate; and wherein a glycan comprising a terminal GlcNAc-moiety is a glycan according to formula (101) or (102):
Figure imgf000058_0001
101 102
wherein:
b is O or l;
d is 0 or 1;
e is 0 or 1 ; and
G is a monosaccharide, or a linear or branched oligosaccharide comprising 2 to 20 sugar moieties;
to obtain a modified glycoprotein; and (b) reacting the modified glycoprotein with a linker-conjugate, wherein said linker-conjugate comprises a functional group B and one or more molecules of interest (D), wherein said functional group B is a functional group that is capable of reacting with a functional group A on a glycan of the modified glycoprotein, and wherein functional group A is independently selected from the group consisting of a thiol group, a halogen, a sulfonyloxy group, a halogenated acetamido group, a mercaptoacetamido group and a sulfonated hydroxyacetamido group. The processes of step (a) and (b) as well as their preferred embodiments are described in detail above.
When a thiol-modified glycoprotein is reacted with a linker-conjugate comprising a functional group B that comprises an N-maleimide group, preferably the glycoprotein-conjugate according to the invention is according to formula (121) or (122):
Figure imgf000059_0001
121
Figure imgf000059_0002
wherein: Pr represents a protein;
L is a Linker;
D is a molecule of interest;
r is 1 to 20;
x is 1, 2, 3 or 4;
y is 1 to 20; and
b is 0 or 1;
d is 0 or 1;
e is 0 or 1 ;
p is O or l;
Q is -N(H)C(0)CH2- or CH2;
Su is a sugar or sugar derivative; and
G is a monosaccharide, or a linear or branched oligosaccharide comprising 2 to 20 sugar moieties.
The value of p and the nature of Q depend on the nature of the modified sugar or sugar derivative Su(A)x that is used for the preparation of the glycoprotein-conjugate according to the process of the invention. If in Su(A)x a thiol group is present on the C2, C3, or C4 position of the sugar or the sugar derivative (instead of a sugar OH- group), then p is 0. If the Su(A)x is a 2-(mercaptoacetamido)-2-deoxy sugar derivative (i.e. a mercaptoacetamido group is present on C2 of said sugar or sugar derivative), Su(A)x is e.g. GalNAcSH or GlcNAcSH, then p is 1 and Q is -N(H)C(0)CH2-. If the thiol in Su(A)x is present on the C6 position of the sugar or the sugar derivative, then p is 1 and Q is -CH2-.
When a thiol-modified glycoprotein is reacted with a linker-conjugate comprising a functional group B that comprises a halogenated acetamido group, preferably the glycoprotein-conjugate according to the invention is according to formula (123) or (124):
Figure imgf000061_0001
123
Figure imgf000061_0002
124 wherein Pr, L, D, r, x, y, b, d, e, p, Q, Su and G are as defined above for (121) and (122), and R9 is selected from the group consisting of L(D)r, hydrogen, Ci - C24 alkyl groups, C6 - C24 aryl groups C7 - C24 alkylaryl groups and C7 - C24 arylalkyl groups, the Ci - C24 alkyl groups, C6 - C24 aryl groups C7 - C24 alkylaryl groups and C7 - C24 arylalkyl groups optionally being substituted. Preferably, R9 is selected from the group consisting of L(D)r, hydrogen, Ci - C12 alkyl groups, C6 - C12 aryl groups C7 - C12 alkylaryl groups and C7 - C122 arylalkyl groups, the Ci - C12 alkyl groups, C6 - C12 aryl groups C7 - C12 alkylaryl groups and C7 - C12 arylalkyl groups optionally being substituted. More preferably, R9 is selected from the group consisting of L(D)r, hydrogen, Ci - C6 alkyl groups, C6 - C12 aryl groups C7 - C12 alkylaryl groups and C7 - C12 arylalkyl groups, the Ci - C6 alkyl groups, C6 - C12 aryl groups C7 - C12 alkylaryl groups and C7 - C12 arylalkyl groups optionally being substituted. Even more preferably, R9 is H, Ci, C2, C4 or C4 alkyl or C6 - C12 aryl. Most preferably, R9 is H or methyl.
Also in this embodiment the value of p and the nature of Q depend on the nature of the modified sugar or sugar derivative Su(A)x that is used for the preparation of the glycoprotein-conjugate according to the process of the invention. If in Su(A)x a halogen is present on the C2, C3, or C4 position of the sugar or the sugar derivative (instead of a sugar OH-group), then p is 0. If the Su(A)x is a 2-(chloroacetamido)-2-deoxy sugar derivative (i.e. a halogenated acetamido group is present on C2 of said sugar or sugar derivative), Su(A)x is e.g. GalNAcCl or GlcNAcCl, then p is 1 and Q is -N(H)C(0)CH2-. If the halogen in Su(A)x is present on the C6 position of the sugar or the sugar derivative, then p is 1 and Q is -CH2-.
When a thiol-modified glycoprotein is reacted with a linker-conjugate comprising a functional group B that comprises an allenamide group, preferably the glycoprotein- conjugate according to the invention is according to formula (125) or (126):
(iuc )b
Figure imgf000062_0001
125
Figure imgf000062_0002
126 wherein Pr, L, D, r, x, y, b, d, e, p, Q, Su, G and R are as defined above.
Also in (125) and (126), the value of p and the nature of Q depend on the nature of the modified sugar or sugar derivative Su(A)x that is used for the preparation of the glycoprotein-conjugate according to the process of the invention. If in Su(A)x a thiol group is present on the C2, C3, or C4 position of the sugar or the sugar derivative (instead of a sugar OH-group), then p is 0. If the Su(A)x is a 2-(mercaptoacetamido)-2- deoxy sugar derivative (i.e. a mercaptoacetamido group is present on C2 of said sugar or sugar derivative instead of an OH-group), Su(A)x is e.g. GalNAcSH or GlcNAcSH, then p is 1 and Q is -N(H)C(0)CH2-. If the thiol in Su(A)x is present on the C6 position of the sugar or the sugar derivative, then p is 1 and Q is -CH2-.
When G is a linear or branched oligosaccharide, it is preferred that G comprises 2 to 12, more preferably 2 to 10, even more preferably 2 to 8 and most preferably 2 to 6 sugar moieties. In another preferred embodiment, y is 1 to 12, more preferably y is 1, 2, 3, 4, 5, 6, 7 or 8, and even more preferably y is 1, 2, 3 or 4. Most preferably y is 1 or 2. In yet another preferred embodiment, y is 2, 4, 6 or 8. This embodiment is particularly preferred when the glycoprotein to be modified is an antibody, i.e. when Pr is Ab.
Preferably, r is 1 to 10, more preferably 1, 2, 3, 4, 5, 6, 7, or 8, even more preferably 1, 2, 3 or 4, even more preferably 1 or 2, and most preferably r is 1. In a preferred embodiment y is 1 to 12, more preferably y is 1, 2, 3, 4, 5, 6, 7 or 8, and even more preferably y is 1, 2, 3 or 4. Most preferably y is 1 or 2. In yet another preferred embodiment, y is 2, 4, 6 or 8. This embodiment is particularly preferred when the glycoprotein to be modified is an antibody, i.e. when Pr is Ab. In a preferred embodiment, CI of the core-sugar moiety of the glycoprotein- glycan, is bonded to the protein via an N-glycosidic bond to a nitrogen atom in an amino acid residue in said protein, more preferably to an amide nitrogen atom in the side chain of an asparagine (Asn) or an arginine (Arg) amino acid. However, CI of the core-sugar-moiety of said glycan may also be bonded to the protein via an O-glycosidic bond to an oxygen atom in an amino acid residue in said protein, more preferably to an oxygen atom in the side chain of a serine (Ser) or threonine (Thr) amino acid. In this embodiment, it is preferred that the core-sugar-moiety of said glycan is an O-GlcNAc- moiety or an O-GalNAc moiety, preferably an O-GlcNAc moiety. CI of the core- sugar-moiety of said glycan may also be bonded to the protein via a C-glycosidic bond to a carbon atom on the protein, e.g. to tryptophan (Trp). As described above, a glycoprotein may comprise more than one oligosaccharide chains, and may constitute of a combination of N-linked, O-linked and C-linked glycoproteins. Said modified glycan may be present at a native glycosylation site of a protein, but may also be introduced on a different site on a protein.
In a preferred embodiment the glycoprotein-conjugate is an antibody-conjugate, said antibody comprising a glycan comprising a GlcNAc-Su(A)x disaccharide-moiety at the non-reducing end. Such an antibody is herein also referred to as a modified antibody. A preferred modified antibody is an antibody according to formula (107) or (108) as defined above, wherein Pr is Ab. In this embodiment, it is further preferred that y is 1, 2, 3, 4, 5, 6, 7 or 8, and even more preferred that y is 1, 2, 3 or 4.
As was defined above, said antibody may be a whole antibody, but also an antibody fragment. When the antibody is a whole antibody, said antibody preferably comprises one or more, more preferably one, modified glycans on each heavy chain. Said whole antibody thus preferably comprises two or more, preferably two, four, six or eight of said glycans, more preferably two or four, and most preferably two glycans. In other words, when said antibody is a whole antibody, y is preferably 2, 4, 6 or 8, more preferably y is 2 or 4, and most preferably y is 2. When the antibody is an antibody fragment, it is preferred that y is 1, 2, 3 or 4, and more preferably y is 1 or 2.
In a preferred embodiment, said antibody is a monoclonal antibody (mAb). Preferably, said antibody is selected from the group consisting of IgA, IgD, IgE, IgG and IgM antibodies. More preferably, said antibody is an IgG antibody, and most preferably said antibody is an IgGl antibody.
In a preferred embodiment, the glycoprotein in the modified antibody is attached to the conserved N-glycosylation site in the Fc-fragment at asparagine in the region 290-305, typically N297. The sugar derivative Su(A)x and preferred embodiments thereof are described in detail above. The preferred embodiments of the process for the preparation of a modified glycoprotein also hold for the modified glycoprotein obtainable by said process. Thus, in a preferred embodiment, x is 1 or 2, more preferably x is 1. In another preferred embodiment, functional group A is a thiol group, a halogen, or a halogenated acetamido group, more preferably a halogen.
Further examples of Su(A)x present in a modified antibody according to the invention include 6-A-6-deoxygalactose (6-A-Gal), such as 6-chloro-6-deoxygalactose (6-ClGal), 6-thio-6-deoxygalactose (6-HSGal) or 2-A-2-deoxygalactose (2-A-Gal), such as 2-chloro-2-deoxygalactose (2-ClGal), 2-thio-2-deoxygalactose (2-HSGal). Alternatively, A may be indirectly substituted to the sugar derivative as part of an acetamido group that in turn is substituting a hydroxyl group. Examples include 6-A- acetamido-6-deoxygalactose (6-GalNAcA), such as 6-chloroacetamido-6- deoxygalactose (6-GalNAcCl), 6-thioacetamido-6-deoxygalactose (6-GalNAcSH) or 2- A-acetamido-2-deoxygalactose (2-GalNAcA), such as 2-chloroacetamido-2- deoxygalactose (2-GalNAcCl), 2-thioacetamido-2-deoxygalactose (2-GalNAcSH). Alternatively, A may be indirectly substituted to the sugar derivative as part of another functional group that in turn is substituting a hydroxyl group or is attached to a hydroxyl group. Examples of such other functional group include an (hetero)alkyl chain or a (hetero)aryl chain.
In a preferred embodiment of the glycoprotein-conjugate according to the invention, y is 1, 2, 3 or 4 (preferably 2 or 4) and/or x is 1 or 2 (preferably 1).
In a preferred embodiment y is 1 to 12, more preferably y is 1, 2, 3, 4, 5, 6, 7 or 8, and even more preferably y is 1, 2, 3 or 4. Most preferably y is 1 or 2. In yet another preferred embodiment, y is 2, 4, 6 or 8. This embodiment is particularly preferred when the glycoprotein to be modified is an antibody, i.e. when Pr is Ab.
In a further preferred embodiment y is 1, 2, 3 or 4 (preferably 2 or 4) and/or x is 1 or 2 (preferably 1), more preferably y is 1 or 2 and x is 1 or 2 (preferably 1), even more preferably y is 1 or 2 and x is 1.
In a preferred embodiment, CI of the core-sugar moiety of the modified glycan, said glycan comprising a GlcNAc-Su(A)x disaccharide-moiety at the non-reducing end, is bonded to the protein via an N-glycosidic bond to a nitrogen atom in an amino acid residue in said protein, more preferably to an amide nitrogen atom in the side chain of an asparagine (Asn) or an arginine (Arg) amino acid. However, CI of the core-sugar- moiety of said modified glycan may also be bonded to the protein via an O-glycosidic bond to an oxygen atom in an amino acid residue in said protein, more preferably to an oxygen atom in the side chain of a serine (Ser) or threonine (Thr) amino acid. In this embodiment, it is preferred that the core-sugar-moiety of said glycan is an O-GlcNAc- moiety or an O-GalNAc moiety, preferably an O-GlcNAc moiety. CI of the core- sugar-moiety of said modified glycan may also be bonded to the protein via a C- glycosidic bond to a carbon atom on the protein, e.g. to tryptophan (Trp). As described above, a glycoprotein may comprise more than one oligosaccharide chains, and may constitute of a combination of N-linked, O-linked and C-linked glycoproteins.
Said modified glycan may be present at a native glycosylation site of a protein, but may also be introduced on a different site on a protein.
In a preferred embodiment the modified glycoprotein is a modified antibody (Ab), said antibody comprising a glycan comprising a GlcNAc-Su(A)x disaccharide- moiety at the non-reducing end. Such an antibody is herein also referred to as a modified antibody. A preferred modified antibody is an antibody according to formula (107) or (108) as defined above, wherein Pr is Ab. In this embodiment, it is further preferred that y is 1, 2, 3, 4, 5, 6, 7 or 8, and even more preferred that y is 1, 2, 3 or 4.
As was defined above, said antibody may be a whole antibody, but also an antibody fragment. When the antibody is a whole antibody, said antibody preferably comprises one or more, more preferably one, modified glycans on each heavy chain. Said whole antibody thus preferably comprises two or more, preferably two, four, six or eight of said glycans, more preferably two or four, and most preferably two glycans. In other words, when said antibody is a whole antibody, y is preferably 2, 4, 6 or 8, more preferably y is 2 or 4, and most preferably y is 2. When the antibody is an antibody fragment, it is preferred that y is 1, 2, 3 or 4, and more preferably y is 1 or 2.
It is further preferred that y is 1, 2, 3 or 4 and x is 1 or 2, and it is further preferred that y is 2 or 4 and x is 1.
In a preferred embodiment, said antibody is a monoclonal antibody (mAb). Preferably, said antibody is selected from the group consisting of IgA, IgD, IgE, IgG and IgM antibodies. More preferably, said antibody is an IgG antibody, and most preferably said antibody is an IgGl antibody.
In a preferred embodiment, the glycoprotein in the modified antibody is attached to the conserved N-glycosylation site in the Fc-fragment at asparagine in the region 290-305, typically N297.
The sugar derivative Su(A)x and preferred embodiments thereof are described in detail above. The preferred embodiments of the process for the preparation of a modified glycoprotein also hold for the modified glycoprotein obtainable by said process. Thus, in a preferred embodiment, x is 1 or 2, more preferably x is 1. In another preferred embodiment, functional group A is a thiol group, a halogen, or a halogenated acetamido group, more preferably a halogen.
Antibody-drug conjugate
The present invention particularly relates to a glycoprotein-conjugate obtainable by the process for the preparation of an glycoprotein, wherein the glycoprotein- conjugate is an antibody-conjugate. An antibody-conjugate is herein defined as a an antibody that is conjugated to a molecule of interest (D) via a linker (L). The antibody- conjugate according to the invention may be conjugated to one or to more than one molecule of interest (D) via a linker (L). The preferred embodiments disclosed above for a glycoprotein-conjugate also apply to an antibody-conjugate. In a further preferred embodiment, the antibody-conjugate according to the invention is an antibody-drug conjugate (ADC). An antibody-drug conjugate is herein defined as an antibody that is conjugated to a molecule of interest (D) via a linker (L), wherein D is selected from the group consisting of pharmaceutically active substances, and more preferably D is selected from the group consisting of drugs and prodrugs.
More preferably, the active substance is selected from the group consisting of pharmaceutically active compounds, in particular low to medium molecular weight compounds (e.g. about 200 to about 1500 Da, preferably about 300 to about 1000 Da), such as for example cytotoxins, antiviral agents, antibacterial agents, peptides and oligonucleotides. Examples of cytotoxins include colchicine, vinca alkaloids, camptothecins, doxorubicin, daunorubicin, taxanes, calicheamycins, tubulysins, irinotecans, an inhibitory peptide, amanitin, deBouganin, duocarmycins, maytansines, auristatins and pyrrolobenzodiazepines (PBDs). In a preferred embodiment, the cytotoxin is selected from the group consisting of camptothecins, doxorubicin, daunorubicin, taxanes, calicheamycins, duocarmycins, maytansines, auristatins and pyrrolobenzodiazepines (PBDs). In another preferred embodiment, the cytotoxin is selected from the group consisting of colchicine, vinca alkaloids, tubulysins, irinotecans, an inhibitory peptide, amanitin and deBouganin. The invention thus also relates to an antibody-conjugate obtainable by the process for the preparation of an antibody-conjugate according to the invention.
The invention further relates to an antibody-conjugate according to the invention, wherein the molecule of interest D is an active substance, for use as a medicament.
The invention also relates to the use of an antibody-conjugate according to the invention, wherein the molecule of interest D is an active substance, for use in the treatment of cancer.
The invention further relates to an antibody-conjugate according to the invention, wherein the molecule of interest D is an active substance, for use in the treatment of breast cancer, more preferably for use in the treatment of HER2-positive breast cancer.
The invention also relates to a method for the treatment of cancer, comprising administering an antibody-drug conjugate according to the invention, in a therapeutically effective amount to a subject in need thereof.
The invention also relates to a method for the treatment of breast cancer, comprising administering an antibody-drug conjugate according to the invention, in a therapeutically effective amount to a subject in need thereof.
The invention also relates to a method for the treatment of HER2-positive breast cancer, comprising administering an antibody-drug conjugate according to the invention, in a therapeutically effective amount to a subject in need thereof.
The modified antibody, the antibody-conjugate and the processes for the preparation thereof according to the invention have several advantages over the processes, modified antibodies and antibody-conjugates known in the art.
As was described above, the processes known in the art for conjugation of a linker-toxin to antibodies still need to be improved, in terms of control of both site- specificity and stoichiometry. Despite the ability of ADCs to home in on their targets, the amount of drug estimated to get inside tumor cells is typically <2% of an administered dose. This problem is amplified by the unpredictable conjugation results of ADCs known in the art. It is important to avoid underconjugated antibodies, which decrease the potency, as well as highly conjugated species, which may have markedly decreased circulating half-lives, impaired binding to the target protein, and increased toxicity. For antibody-drug conjugates, a measure for the loading of molecules of interest (e.g. drugs, active substances) onto the antibody is the so-called Drug to Antibody Ratio (DAR), which gives the average number of active substance molecules per antibody, calculated from a statistical distribution. The theoretical maximum value of DAR for a certain type of ADC is equal to the number of anchoring sites. As was described above, processes for the preparation of ADCs known from the prior art generally result in a product comprising a mixture of antibody-conjugates with a varying number of molecules of interest present in each antibody-conjugate, and in a DAR with a high standard deviation.
One of the advantages of the modified antibodies and the antibody-conjugates according to the invention is that these antibodies and antibody-conjugates are homogeneous, both in site-specificity and stoichiometry. Said modified antibodies and antibody-conjugates are obtained with a DAR very near to the theoretical value, and with a very low standard deviation. This also means that the antibody-conjugates according to the invention result in a more consistent product for preclinical testing. The homogeneity of the antibody-conjugate prepared by the process described herein becomes for example clear from the MS profile given in Figure 9.
Another advantage of the processes and antibodies according to the invention involves the reduction of waste in manufacturing, thereby enhancing companies' cost- of-goods.
When a thiol-modified antibody according to the invention is coupled to a linker- conjugate comprising a maleimide, the process is well-known in the art, highly robust and validated. Some examples of maleimide conjugates are provided in Figures 8 and 11. Many maleimide-functionalized toxins have been described, because currently the preferred methodology for antibody-drug conjugation involves the combination of a cysteine mutant of a mAb (THIOmAb) and a maleimide derivative of a toxin. It is well known that such thiol-maleimide conjugates can be prepared with a highly beneficial stoichiometry of reagents (small excess of maleimide component). It is also well known that the resulting thiol-maleimide conjugates may have limited stability, but in case the thiol is present on fucose, the stability is significantly enhanced. When a thiol-modified antibody according to the invention is coupled to a linker-conjugate comprising a halogenated acetamide derivative of a toxin, the efficiency of the process may be somewhat compromized with respect to maleimide conjugation and more undesired alternative conjugation may take place (e.g. on lysine side chains), but the desired product is an irreversibly formed (highly stable) thio-ether conjugate. When a thiol- modified antibody according to the invention is coupled to a linker-conjugate comprising a allenamide derivative of a toxin, the efficiency of the process may be somewhat compromized with respect to maleimide conjugation, and no undesired alternative conjugation is likely to take place (e.g. on lysine side chains), while the desired product is an irreversibly formed (highly stable) thio-ether conjugate. An example of an allenamide structure is provided in Figure 6. When a halogen-modified antibody according to the invention is coupled to a linker-conjugate comprising a derivative of a toxin containing a nucleophilic group (thiol, alcohol, amine), such as the thiol-comprising structures represented in Figure 6, the resulting conjugate is a thio- ether, such as the structures represented in Figures 8 and 10, a regular ether or an amino-ether, all of which are formed irreversibly. In contrast to the use of halogenated acetamides for conjugation to proteins containing free thiols (as in THIOmAbs or in a thiofucose-containing mAb), the enzymatic incorporation of a halogenated sugar substrate is not compromised by competitive aspecific reaction with nucleophilic side chains of other amino acids (e.g. lysine). The lack of aspecific reactions also pertains to the subsequent conjugation step where in this case excess of a nucleophilic derivative of a functional group is applied to the halogenated mAb.
One particular advantage of the use of a halogenated antibody is that it provides the opportunity to perform two-stage conjugation of a toxic payload. For example as illustrated in Figure 10, reaction of the halogenated antibody with a thiophenol derivative bearing a pendant azide, provides an intermediate that can serve as the starting point for introduction of the toxic payload by means of copper-free click chemistry. By applying such a two-stage process, the stoichiometry of the (cheap and non-toxic) thiophenol derivative can be high, while the stoichiometry of the (expensive and highly toxic) payload can be kept low.
One particular advantage of the use of a thiol-comprising antibody is that it provides the opportunity to perform two-stage conjugation of a toxic payload. For example as illustrated in Figure 1 1, reaction of the thiol-comprising antibody with an allenamide derivative bearing a pendant azide, provides a stable intermediate that can serve as the starting point for introduction of the toxic payload by means of copper-free click chemistry (in the example biotin is used as a representative for a toxic payload). By applying such a two-stage process, the stoichiometry of the (cheap and non-toxic) allenamide derivative can be high, while the stoichiometry of the (expensive and highly toxic) payload can be kept low.
Additional advantages are thus the stability of antibody-conjugates according to the invention, as well as the straightforward and generally applicable process for the introduction of a thiol group, a halogen, a sulfonyloxy group, a halogenated acetamido group, a mercaptoacetamido group and a sulfonated hydroxyacetamido group into an antibody.
One additional advantage for the preparation of thiol-comprising antibodies by the process described herein is that the starting UDP-sugars can be readily synthesized, as exemplified in Figure 12.
Finally, an advantage preparing antibody conjugates by connecting via the glycan chain provides the opportunity to prepare a large number of isomers by means of engineering of glycomutants of the native antibody. Examples of different glycoforms (with R = glycan) of the native antibody are depicted in Figure 13.
Examples
Synthesis
Examples 1 - 5: Synthesis of UDP-GalNAc derivatives 28-31
The reaction scheme of the synthesis of UDP-GalNAc derivatives 28-31, starting from 24, as performed in Examples 1 - 4, is shown in Figure Y.
Example 1. Synthesis of UDP-GalNH2
Compound 24 was prepared from D-galactosamine according to the procedure described for D-glucosamine in Linhardt et al, J. Org. Chem. 2012, 77, 1449-1456. 1H-NMR (300 MHz, CD3OD): δ 5.69 (dd, J = 6.84, 6.84 Hz, 1H), 5.43-5.41 (m, 1H), 5.35 (dd, J= 10.9, 3.4 Hz, 1H), 4.54 (t, J= 6.48 Hz, 1H), 4.23-4.12 (m, 1H), 4.04 (dd, J = 10.9, 6.1 Hz, 1H), 3.82 (dt, J = 11.1, 2.7 Hz, 1H), 2.12 (s, 3H), 2.00 (s, 3H), 1.99 (s, 3H). LRMS (ESI-) calcd for Ci2Hi8N30n ( -H+) 410.06, found 410.1. Next, compound 24 was coupled to UMP according to Baisch et al. Bioorg. Med. Chem., 1997, 5, 383-391).
Thus, a solution of D-uridine-5'-monophosphate disodium salt (1.49 g, 4.05 mmol) in H20 (15 mL) was treated with DOWEX 50Wx8 (H+ form) for 30 minutes and filtered. The filtrate was stirred vigorously at rt while tributylamine (0.966 mL, 4.05 mmol) was added dropwise. After 30 minutes of further stirring, the reaction mixture was lyophilized and further dried over P205 under vacuum for 5 h.
The resulting tributylammonium uridine-5 '-monophosphate was dissolved in dry DMF (25 mL) in an argon atmosphere. Carbonyldiimidazole (1.38 g, 8.51 mmol) was added and the reaction mixture was stirred at r.t. for 30 min. Next, dry MeOH (180 was added and stirred for 15 min to remove the excess CDI. The leftover MeOH was removed under high vacuum for 15 min. Subsequently, compound 24 (2.0 g, 4.86 mmol) was dissolved in dry DMF (25 mL) and added dropwise to the reaction mixture. The reaction was allowed to stir at rt for 2 d before concentration in vacuo. The consumption of the imidazole-UMP intermediate was monitored by MS. Flash chromatography (7:2: 1-5:2: 1 EtOAc:MeOH:H20) afforded product 25 (1.08 g, 1.51 mmol, 37%).
1H-NMR (300 MHz, D20): δ 7.96 (d, J= 8.0 Hz, 1H), 5.98-5.94 (m, 2H), 5.81-5.79 (m, 1H), 5.70 (dd, J= 7.1, 3.3 Hz, 1H), 5.49 (dd, J= 15.2, 2.6 Hz, 1H), 5.30 (ddd, J= 18.5, 11.0, 3.2 Hz, 2H), 4.57 (q, J= 6.0 Hz, 2H), 4.35-4.16 (m, 9H), 4.07-3.95 (m, 2H), 2.17 (s, 3H), 2.08 (s, 3H), 2.07 (s, 3H).
LRMS (ESI-) calcd for C21H29N5O19P2 ( -H+) 716.09, found 716.3. Compound 25 was deacetylated according to Kiso et al, Glycoconj. J, 2006, 23, 565- 573).
Thus, compound 25 (222 mg, 0.309 mmol) was dissolved in H20 (2.5 mL) and triethylamine (2.5 mL) and MeOH (6 mL) were added. The reaction mixture was stirred for 3 h and then concentrated in vacuo to afford crude UDP-2-azido-2-deoxy-D- galactose (26). 1H- MR (300 MHz, D20): δ 7.99 (d, J = 8.2 Hz, 1H), 6.02-5.98 (m, 2H), 5.73 (dd, J = 7.4, 3.4 Hz, 1H), 4.42-4.37 (m, 2H), 4.30-4.18 (m, 4H), 4.14-4.04 (m, 2H), 3.80-3.70 (m, 2H), 3.65-3.58 (m, 1H).
LRMS (ESI-) calcd for Ci5H23N5Oi6P2 ( -H+) 590.05, found 590.2. Finally, to a solution of compound 26 in H20:MeOH 1 : 1 (4mL) was added Lindlar's catalyst ( mg). The reaction was stirred under a hydrogen atmosphere for 5 h and filtered over celite. The filter was rinsed with H20 (10 ml) and the filtrate was concentrated in vacuo to afford the UDP-D-galactosamine (UDP-Gal H2, 27) (169 mg, 0.286 mmol, 92% yield). 1H- MR (300 MHz, D20): δ 7.93 (d, J = 8.1 Hz, 1H), 5.99- 5.90 (m, 2H), 5.76-5.69 (m, 1H), 4.39-4.34 (m, 2H), 4.31-4.17 (m, 5H), 4.05-4.01 (m, 1H), 3.94-3.86 (m, 1H), 3.82-3.70 (m, 3H), 3.30-3.16 (m, 1H). LRMS (ESI-) calcd for Ci5H25N3Oi6P2 ( -H+) 564.06, found 564.1.
Example 2. Synthesis of UDP-GalNAcSAc (28a)
UDP-D-galactosamine (27) (45 mg, 0.0796 mmol) was dissolved in buffer pH 7 (0.5 M K2HP04) (2 mL). N-Succinimidyl-S-acetylthioacetate (37 mg, 0.159 mmol) and DMF (2 mL) were added and the reaction was stirred overnight at rt. Another 36 mg of N- succinimidyl-S-acetylthioacetate were added and after 3 h the reaction was concentrated in vacuo. Flash chromatography (7:2: 1-5:2: 1 EtOAc:MeOH:H20) afforded UDP-GalNAcSAc (28a) (28 mg, 0.041 mmol, 52%).
1H- MR (300 MHz, D20): δ 7.84 (d, J= 8.1 Hz, 1H), 5.90-5.82 (m, 2H), 5.48-5.41 (m, 1H), 4.29-4.22 (m, 2H), 4.20-4.00 (m, 5H), 3.98-3.82 (m, 2H), 3.79-3.59 (m, 4H), 2.30 (s, 3H). LRMS (ESI-) calcd for Ci9H29N3Oi8P2S ( -H+) 680.06, found 680.1. Example 3. Synthesis of UDP-GalNAcCl (29)
UDP-D-galactosamine (27) (42 mg, 0.074 mmol) was dissolved in 0.1 M NaHC03 (1 mL) and N-(Chloroacetoxy)succinimide (29 mg, 0.149 mmol) (prepared according to Hosztafi et al, Helv. Chim. Acta, 1996, 79, 133-136) and DMF (1 mL) were added. The reaction was stirred overnight at r. , another 10 mg of N- (Chloroacetoxy)succinimide was added and stirring was continued overnight. The reaction was concentrated in vacuo and purified by flash chromatography (7:2: 1-5:2: 1 EtOAc:MeOH:H20) afforded UDP-GalNAcCl (29) (25 mg, 0.039 mmol, 53%).
1H- MR (300 MHz, D20): δ 7.84 (d, J= 8.1 Hz, 1H), 5.89-5.84 (m, 2H), 5.53-5.46 (m, 1H), 4.33-4.00 (m, 9H), 3.99-3.88 (m, 2H), 3.77-3.59 (m, 2H), 1.83 (s, 1H).
LRMS (ESI-) calcd for Ci7H26ClN3Oi7P2 ( -H+) 640.03 (100%), 642.03 (32%), found 640.1 (100%), 642.2 (35%). Example 4. Synthesis of UDP-GalNAcBr (30)
UDP-D-galactosamine (27) (42 mg, 0.088 mmol) was dissolved in 0.1 M NaHC03 (3 mL) and N-(bromoacetoxy)succinimide 63 mg, 0.265 mmol) (prepared according to Hosztafi et al, Helv. Chim. Acta, 1996, 79, 133-136) and DMF (2 mL) were added. The reaction was stirred overnight at r.t. and concentrated in vacuo. The compound was purified by flash chromatography (7:2: 1-5:2: 1 EtOAc:MeOH:H20) afforded UDP- GalNAcBr (30) (28 mg, 0.048 mmol, 65%).
1H- MR (300 MHz, D20): δ 7.86 (d, J= 3.2 Hz, 1H), 5.97-5.84 (m, 2H), 5.54-5.46 (m, 1H), 4.33-4.04 (m, 6H), 3.99-3.85 (m, 2H), 3.79-3.60 (m, 2H), 2.75-2.68 (m, 3H). LRMS (ESI-) calcd for Ci7H26BrN3Oi7P2 ( -H+) 683.98 (100%), 685.98 (98%), found 687.1 (100%), 688.0 (92%), 686.0 (85%), 689.0 (72%).
Example 6. Synthesis of 4,4'-disulfanediyldibenzoic acid (31)
To a solution of 4-mercaptobenzoic acid (0.20 g, 1.30 mmol) in EtOH (10 mL) was added I2 (0.20 g, 0.65 mmol) and Et3N (0.54 mL, 395 mg, 3.90 mmol). The resulting mixture was stirred for 16 h. After addition of 10% aqueous Na2S203 and 0.01 M aqueous HCl (25 mL), the mixture was partially concentrated and 1M aqueous HCl was added (1 mL). EtOAc (20 mL), DCM (50 mL) and water (10 mL) were added and the product was filtered off and dried in vacuo. The desired product (4,4 - disulfanediyldibenzoic acid) was obtained as white solid (180 mg, 0.59 mmol, 90%) and used without further purification (see below). 1H NMR (300 MHz, DMSO) δ (ppm) 7.96-7.88 (m, 4H), 7.68-7.59 (m, 4H).
Example 7. Synthesis of mercapto-D-biotin conjugate (32)
A solution of tert-butyl (2-(2-(2-aminoethoxy)ethoxy)ethyl)carbamate (451 mg, 1.82 mmol) in anhydrous DMF (5 mL) was placed under an argon atmosphere. D-Biotin (445 mg, 1.82 mmol), EDCI (418 mg, 2.18 mmol), HOBt (343 mg, 2.54 mmol), diisopropylethylamine (317 μΐ^, 235 mg, 1.82 mmol) and anhydrous DMF (2 mL) were added. The resulting mixture was stirred for 24 h and concentrated. The residue was purified via column chromatography (2%→ 10% MeOH in DCM). After concentration of the product containing fractions, the residue was dissolved in a mixture of EtOAc/DCM/MeOH (100 mL/20 mL/ 20 mL) and washed with aqueous saturated H4CI (2 x 100 mL). The organic mixture was dried (Na2S04) and concentrated to 20 mL. The desired product (biotin-PEG2- HBoc) precipitated as a colorless glass (228 mg, 0.48 mmol, 26%) and used in the next step. 1H NMR (300 MHz, CD3OD) δ (ppm) 4.49 (dd, J = 7.4, 4.5 Hz, 1H), 4.30 (dd, J = 7.9, 4.4 Hz, 1H), 3.62 (s, 4H), 3.55 (t, J = 5.5 Hz, 2H), 3.52 (t, J = 5.6 Hz, 2H) 3.40-3.15 (m, 5H), 2.93 (dd, J = 12.7, 5.0 Hz, lH)f, 2.71 (d, J = 12.8 Hz, 1H), 2.22 (t, J = 7.4 Hz, 2H), 1.81-1.40 (m, 6H), 1.44 (s, 9H).
A solution of biotin-PEG2-NHBoc (208 mg, 0.44 mmol) was dissolved in MeOH (5 mL). After addition of acetyl chloride (250 μΕ, 275 mg, 3.5 mmol), the mixture was stirred for 17 h and concentrated. The residue was co-evaporated with EtOAc (5 mL), which quantitatively yielded the product as a yellow solid. The product (biotin-PEG2- NH2) was used in the next step without further purification.
To a solution of 4,4'-disulfanediyldibenzoic acid (31, 20 mg, 0.065 mmol) in DMF (1 mL) were added HATU (55 mg, 0.144 mmol) and diisopropylamine (70 μΕ, 50 mg, 0.39 mmol). Biotin-PEG2-NH2 (53 mg, 0.130 mmol) was added as a solution in DMF. The resulting mixture was stirred for 19 h and poured out in a mixture of DCM (20 mL) and saturated aqueous NH4C1 (20 mL). After separation, the organic phase was dried (Na2SC"4). The residue was purified via column chromatography (5%→20% MeOH in DCM). The desired product (biotin-PEG2-SS-PEG2-Biotin) was obtained as a white solid (21 mg, 0.021 mmol, 32%). LRMS (ESI+) calcd for C46H66N8OioS4 ( +H+) 1019.39, found 1020.1.
To a cooled (0 °C) solution of biotin-PEG2-SS-PEG2-biotin (3.5 mg; 3.4 μιηοΐ) in a mixture of THF (0.5 mL) and EtOH (0.5 mL) was added NaBH4 (0.5 mg, 12 μιηοΐ). While warming up to rt, the mixture was stirred for 2 h and concentrated. The residue was taken up in water (1 mL) and 10 μΐ 1 M aqueous HC1 was added. The desired product was extracted with DCM (2 x 2 mL). The combined organic layers were dried (Na2S04) and concentrated to yield the title compound (32) (2.7 mg, 78%). LRMS (ESI+) calcd for C23H35N405S2 ( +H+) 511.20, found 511.2.
Example 8. Synthesis of maleimide-biotin conjugate 33
To a solution of N-succinimidyl 6-maleimidocaproate (10 mg, 0.032 mmol) in 1 mL DMF was added a solution of biotin-PEG2- H2 (13 mg, 0.032 mmol) and Et3N (9 μί, 6.5 mg, 0.064 mmol) in DMF (1 mL). The mixture was stirred for 1 h and poured out in a mixture of DCM (10 mL) and aqueous saturated NaHC03 (10 mL). After separation, the organic layer was dried (Na2S04) and concentrated, Column chromatography (2→20% MeOH in DCM) afforded the desired product 33 according to LRMS analysis. LRMS (ESI+) calcd for C26H42N507S ( +H+) 568.28, found 568.2.
Conjugations
Antibody glycosylation mutant
Specific mutants of trastuzumab were derived from literature (Qu et al, J. Immunol. Methods 1998, 213, 131), in particular L196N and G164S) or de novo designed, in particular V363T. In all three cases, asparagine 297 was mutated to glutamine (N297Q) to remove the native glycosylation site.
Both native trastuzumab and mutant antibodies were transiently expressed by Evitria (Zurich, Switzerland). General protocol for mass spectral analysis oflgG
A solution of 50 μg (modified) IgG, 1 M Tris-HCl pH 8.0, 1 mM EDTA and 30 mM DTT with a total volume of approximately 70 μΕ was incubated for 20 minutes at 37 °C to reduce the disulfide bridges allowing to analyze both light and heavy chain. If present, azide-functionalities are reduced to amines under these conditions. Reduced samples were washed trice with milliQ using an Amicon Ultra-0.5, Ultracel-10 Membrane (Millipore) and concentrated to 10 μΜ (modified) IgG. The reduced IgG was analyzed by electrospray ionization time-of-flight (ESI-TOF) on a JEOL AccuTOF. Deconvoluted spectra were obtained using Magtran software.
Examples 9 - 12: General Protocol for trimming of IgG glycans
Trimming of IgG glycans was performed using endo S from Streptococcus pyogenes (commercially available from Genovis, Sweden). The IgG (10 mg/mL) was incubated with endo S (40 U/mL final concentration) in 25 mM Tris pH 8.0 for 16 hours at 37 °C.
Example 9. Trimming of native trastuzumab
Trastuzumab with base MS peak 50591 Da was subjected to the trimming protocol above. After deconvolution of peaks, the mass spectrum showed one peak of the light chain and two peaks of the heavy chain. The two peaks of heavy chain belonged to one major product (49495 Da, 90% of total heavy chain), resulting from core GlcNAc(Fuc)-substituted trastuzumab, and a minor product (49351 Da, ±10% of total heavy chain), resulting from core GlcNAc-substituted trastuzumab. Example 12. Trimming of trastuzumab-mutant N297Q, V363T
Trastuzumab-(N297Q,V363T) mutant (10 mg/mL) with major base MS peaks at 50934, 51227 and 51517 Da, corresponding to G2F, G2FS1 and G2FS2 glycosylation isoforms, was incubated with Endoglycosidase F3 (EndoF3, 25 mU/mg IgG) from Elizabethkingia meningosepticum (commercially available from QA-Bio) in 100 mM sodium citrate pH 4.5 for 16 hrs, which led to complete deglycosylation (major heavy chain product of 49515 Da resulting from N297Q,V363T heavy chain with core GlNAc(Fuc) attached).
General protocol for glycosyltransfer of modified sugar to IgG
Enzymatic introduction of modified sugar onto IgG was effected with a mutant of bovine pi,4-galactosyltransf erase (pi,4-Gal-Tl-Y289L). The deglycosylated IgG (prepared as described above, 10 mg/mL) was incubated with a modified UDP-sugar derivative (0.4 mM) and p(l,4)-Gal-Tl-Y289L (1 mg/mL) in 10 mM MnCl2 and 25 mM Tris-HCl pH 8.0 for 15 hours at 30 °C.
The modified IgG was incubated with protein A agarose (40 μΐ. per mg IgG) for 2 hours at 4 °C. The protein A agarose was washed three times with PBS and the IgG was eluted with 100 mM glycine-HCl pH 2.7. The eluted IgG was neutralized with 1 M Tris-HCl pH 8.0 and concentrated and washed with PBS using an Amicon Ultra-0.5, Ultracel-10 Membrane (Millipore) to a concentration of 10 mg/mL.
Example 13. Glycosyltransfer of UDP-sugar 28a (UDF '-GalNAcSAc) to deglycosylated trastuzumab.
Trimmed trastuzumab (250 μΐ., 10 mg/mL, 16.5 nmol) was incubated with UDP- GalNAcSAc (28a) (18.5 μί, 10 mM) and p(l,4)-Gal-Tl(Y289L) (12.5 μί, 2 mg/mL) in 10 mM MnCl2 and 25 mM Tris-HCl pH 6.0 for 16 hours at 30 °C. The crude mixture was purified with ProtA and in order to maintain the low pH the column was washed with 25 mM Tris-HCl pH 6.0 and after elution with glycine.HCl buffer (pH 2.7, 0.1 M), the elution buffer was neutralized with Tris-HCl (pH 7.2, 1 M).
Via this protocol 1.1 mg of an IgG was obtained, AccuTOF analysis of which showed the 95% conversion of trast-(GalNAcSH)2 into a single desired product (GalNAcSAc minus the acetate group, probably due to in situ deprotection with DTT (mass 49729, expected mass 49730). The other 5% was remaining starting material (mass 49494).
Example 14. Glycosyltransfer of UDP-sugar 28a (GalNAcSAc) to deglycosylated trastuzumab (N297Q, V363T).
Trimmed trastuzumab(N297Q,V363T) (100 μL, 10 mg/mL, 6.6 nmol) was incubated with UDP-GalNAcSAc (28a) (14 μί, 10 mM) and p(l,4)-Gal-Tl(Y289L) (10 μί, 2 mg/mL) in 10 mM MnCl2 and 25 mM Tris-HCl pH 6.0 for 16 hours at 30 °C. The crude mixture was purified with ProtA and in order to maintain the low pH the column was washed with 25 mM Tris-HCl pH 6.0 and after elution with glycine.HCl buffer (pH 2.7, 0.1 M), the elution buffer was neutralized with Tris-HCl (pH 7.2, 1 M). Via this protocol 0.25 mg of an IgG was obtained, AccuTOF analysis of which showed the presence of trast(N297Q,V363T)-(GalNAcSH)2 as a single desired product (GalNAcSAc minus the acetate group, probably due to in situ deprotection with DTT (mass 49744, expected mass 49748). Example 15. Glycosyltransfer of UDP-sugar 29 (GalNAcCl) to deglycosylated trastuzumab.
Trimmed trastuzumab (100 μΐ., 10 mg/mL, 6.6 nmol) was incubated with UDP- GalNAcCl (29) (5 μί, 10 mM) and p(l,4)-Gal-Tl(Y289L) (5 μί, 2 mg/mL) in 10 mM MnCl2 and 25 mM Tris-HCl pH 6.0 for 16 hours at 30 °C. The crude mixture was purified with ProtA according to the protocol for GalNAcSAc to afford trast- (GalNAcCl)2 (0.36 mg). AccuTOF analysis showed complete conversion to the desired product (mass 49731, expected mass 49732).
Example 16. Glycosyltransfer of UDP-sugar 29 (GalNAcCl) to deglycosylated trastuzumab (N297Q, V363T).
Trimmed trastuzumab N297Q V363T mutant (100 μΐ^, 10 mg/mL, 6.6 nmol) was incubated with UDP-GalNAcCl (29) (5 μί, 10 mM) and p(l,4)-Gal-Tl(Y289L) (5 μί, 2 mg/mL) in 10 mM MnCl2 and 25 mM Tris-HCl pH 6.0 for 16 hours at 30 °C. The crude mixture was purified with ProtA according to the protocol for GalNAcSAc to afford trast-(GalNAcCl)2 (0.35 mg). AccuTOF analysis showed complete conversion to the desired product (mass 49750, expected mass 49750). Example 17. Glycosyltransfer of UDP-sugar 30 (GalNAcBr) to deglycosylated trastuzumab.
The trimmed trastuzumab (100 μL, 10 mg/mL, 6.6 nmol) was incubated with UDP- GalNAcBr (RHX) (15 μί, 10 mM) and pl,4-Gal-Tl-Y289L (5 μί, 2 mg/mL) in 10 mM MnCl2 and 25 mM Tris-HCl pH 6.0 for 16 hours at 30 °C. The crude mixture was purified with ProtA according to the protocol for GalNaSAc to afford trast-(GalNacBr)2 (0.34 mg). AccuTOF analysis showed 50% conversion to the desired product (mass 49777, expected mass 49776 and mass + DTT (reaction during analysis sample) 49849.5). The intramolecular substitution of the bromide was 30% (mass 49696, expected mass 49698) and 20% was remaining starting material.
Example 18: Conjugation of trast-GalNAcCl with DTT
Crude trast-(GalNAcCl)2 (2 μί, 10 mg/ml in 25 mM Tris-HCl pH 8.0) was incubated with DTT (2 μί, 0.2 M) for 10 minutes at 37 °C. Subsequent analysis with AccuTOF showed 70% incooperation of DTT (mass 49849, expected mass 49850) and 30% intramolecular substitution of the chloride (mass 49697, expected mass 49698).
Example 19: Conjugation of trast(GalNAcCl) 2 with ?-N02PhSH
To a solution of trast-(GalNAcCl)2 (7.5 iL, 26.6 mg/ml, 1.3 nmol) in 25 mM Tris-HCl pH 6.0 was added degassed Tris-HCl (7.5 μΐ., 100 mM pH 7.2) and a solution of degassed /?-N02PhSH (7.5 μί, 0.10 mM, 750 nmol in MiliQ + 10 mM EDTA) under argon atmosphere. Analysis after 4 h by AccuTOF showed 95% conversion to the desired product (mass 49849, expected mass 49851).
Example 20: Conjugation of trast(GalNAcCl) 2 with ?-MeOPhSH
To a solution of trast-(GalNAcCl)2 (7.5 iL, 26.6 mg/ml, 1.3 nmol) in 25 mM Tris-HCl pH 6.0 was added degassed Tris-HCl (7.5 μΐ., 100 mM pH 7.2) and a solution of degassed /?-MeOPhSH (1 μί, 0.10 mM, 100 nmol in MiliQ + 10 mM EDTA) under argon atmosphere. Analysis after 4 h by AccuTOF showed 95% conversion to the desired product (mass 49835, expected mass 49836). SDS-PAGE analysis indicated that no reduction of disulfide bonds of the antibody had taken place.
Example 21: Conjugation of trast(GalNAcCl) 2 with 32
To a solution of trast-(GalNAcCl)2 (7.5 iL, 26.6 mg/ml, 1.3 nmol) in 25 mM Tris-HCl pH 6.0 was added degassed Tris-HCl (7.5 μΐ., 100 mM pH 7.2) and a solution of degassed 32 (1 μί, 0.10 mM, 100 nmol in 10 mM EDTA in MiliQ/DMF=l : l) under argon atmosphere. After overnight incubation the excess of reagent was removed by spinfilter purification after which analysis by AccuTOF showed complete conversion to the desired product 40 (mass 50208, expected mass 50206).
Example 22: Conjugation of trast(GalNAcSH) 2 with mc-vc-PABA-MMAE (34)
Protein A purified trast-(GalNAcSH)2 (5 μί, 20 mg/ml in 25 mM Tris-HCl pH 8.0), which was obtained using UDP-sugar 29 as described above, was incubated with mc- vc-PABA-MMAE (5 μΐ,, 1 mM, commercially available from Concortis) in PBS for 16 hrs at room temperature. The excess of reagent was removed by spin-filter purification. Subsequent analysis with AccuTOF showed the presence of the desired product (mass 51051, expected mass+Na+ 51050). Example 23: Cloning and expression of GalT mutants Y289N, Y289F, Y289M, Y289V, Y289A, Y289G and Y289I.
The GalT mutant genes were amplified from a construct containing the sequence encoding the catalytic domain of GalT consisting of 130-402 aa residues, by the overlap extension PCR method. The wild type enzyme is represented by SEQ ID NO: 17. For Y289N mutant (represented by SEQ ID NO: 18), the first DNA fragment was amplified with a pair of primers: 01igo38_GalT_External_Fw (CAG CGA CAT ATG TCG CTG ACC GCA TGC CCT GAG GAG TCC represented by SEQ ID NO: 1) and 01igol9_GalT_Y289N_Rw (GAC ACC TCC AAA GTT CTG CAC GTA AGG TAG GCT AAA represented by SEQ ID NO: 2). The Ndel restriction site is underlined, while the mutation site is highlighted in bold. The second fragment was amplified with a pair of primers: 01igo29_GalT_External_Rw (CTG ATG GAT GGA TCC CTA GCT CGG CGT CCC GAT GTC CAC represented by SEQ ID NO: 3) and 01igol8_GalT_Y289N_Fw (CCT TAC GTG CAG AAC TTT GGA GGT GTC TCT GCT CTA represented by SEQ ID NO: 4). The BamHI restriction site is underlined, while the mutation site is highlighted in bold. The two fragments generated in the first round of PCR were fused in the second round using 01igo38_GalT_External_Fw and 01igo29_GalT_External_Rw primers. After digestion with Ndel and BamHI. This fragment was ligated into the pET16b vector cleaved with the same restriction enzymes. The newly constructed expression vector contained the gene encoding Y289N mutant and the sequence encoding for the His-tag from pET16b vector, which was confirmed by DNA sequencing results. For the construction of Y289F (represented by SEQ ID NO: 19), Y289M (represented by SEQ ID NO: 20), Y289I (represented by SEQ ID NO: 21), Y289V (represented by SEQ ID NO: 22), Y289A (represented by SEQ ID NO: 23) and Y289G (represented by SEQ ID NO: 24) mutants the same procedure was used, with the mutation sites changed to TTT, ATG, ATT, GTG, GCG or GGC triplets encoding for phenylalanine, methionine, isoleucine, valine, alanine or glycine, respectively. More specifically, for the construction of Y289F the first DNA fragment was amplified with a pair of primers defined herein as SEQ ID NO: 1 and SEQ ID NO: 5 and the second fragment was amplified with a pair of primers defined herein as SEQ ID NO: 3 and SEQ ID NO: 6 (be referred to Table 1 for the related sequences). Furthermore, for the construction of Y289M the first DNA fragment was amplified with a pair of primers defined herein as SEQ ID NO: 1 and SEQ ID NO: 7 and the second fragment was amplified with a pair of primers defined herein as SEQ ID NO: 3 and SEQ ID NO: 8. For the construction of Y289I the first DNA fragment was amplified with a pair of primers defined herein as SEQ ID NO: 1 and SEQ ID NO: 9 and the second fragment was amplified with a pair of primers defined herein as SEQ ID NO: 3 and SEQ ID NO: 10. For the construction of Y289V the first DNA fragment was amplified with a pair of primers defined herein as SEQ ID NO: 1 and SEQ ID NO: 11 and the second fragment was amplified with a pair of primers defined herein as SEQ ID NO: 3 and SEQ ID NO: 12. for the construction of Y289A the first DNA fragment was amplified with a pair of primers defined herein as SEQ ID NO: 1 and SEQ ID NO: 13 and the second fragment was amplified with a pair of primers defined herein as SEQ ID NO: 3 and SEQ ID NO: 14. For the construction of Y289G the first DNA fragment was amplified with a pair of primers defined herein as SEQ ID NO: 1 and SEQ ID NO: 15 and the second fragment was amplified with a pair of primers defined herein as SEQ ID NO: 3 and SEQ ID NO: 16 (be referred to Table 1 for the related sequences).
GalT mutants were expressed, isolated and refolded from inclusion bodies according to the reported procedure by Qasba et al. {Prot. Expr. Pur. 2003, 30, 219- 229). After refolding, the precipitate was removed and the soluble and folded protein was isolated using a Ni-NTA column (HisTrap excel 1 mL column, GE Healthcare). After elution with 25 mM Tris-HCl pH 8.0, 300 mM NaCl and 200 mM imidazole, the protein was dialyzed against 25 mM Tris-HCl pH 8.0 and concentrated to 2 mg/mL using a spinfilter (Amicon Ultra- 15 Centrifugal Filter Unit with Ultracel-10 membrane, Merck Millipore).
Table 1 Sequence identification of the primers used
SEQ ID NO Nucleotide sequence
SEQ ID NO: 1 CAG CGA CAT ATG TCG CTG ACC GCA TGC CCT GAG GAG TCC
SEQ ID NO: 2 GAC ACC TCC AAA GTT CTG CAC GTA AGG TAG GCT AAA
SEQ ID NO: 3 CTG ATG GAT GGA TCC CTA GCT CGG CGT CCC GAT GTC CAC
SEQ ID NO: 4 CCT TAC GTG CAG AAC TTT GGA GGT GTC TCT GCT CTA
SEQ ID NO: 5 GAC ACC TCC AAA AAA CTG CAC GTA AGG TAG GCT AAA
SEQ ID NO: 6 CCT TAC GTG CAG TTT TTT GGA GGT GTC TCT GCT CTA
SEQ ID NO: 7 GAC ACC TCC AAA CAT CTG CAC GTA AGG TAG GCT AAA
SEQ ID NO: 8 CCT TAC GTG CAG ATG TTT GGA GGT GTC TCT GCT CTA
SEQ ID NO: 9 GAC ACC TCC AAA AAT CTG CAC GTA AGG TAG GCT AAA SEQ ID NO: 10 CCT TAC GTG CAG ATT TTT GGA GGT GTC TCT GCT CTA
SEQ ID NO: 11 GAC ACC TCC AAA CAC CTG CAC GTA AGG TAG GCT AAA
SEQ ID NO: 12 CCT TAC GTG CAG GTG TTT GGA GGT GTC TCT GCT CTA
SEQ ID NO: 13 GAC ACC TCC AAA CGC CTG CAC GTA AGG TAG GCT AAA
SEQ ID NO: 14 CCT TAC GTG CAG GCG TTT GGA GGT GTC TCT GCT CTA
SEQ ID NO: 15 GAC ACC TCC AAA GCC CTG CAC GTA AGG TAG GCT AAA
SEQ ID NO: 16 CCT TAC GTG CAG GGC TTT GGA GGT GTC TCT GCT CTA
Example 24: Synthesis of 4,4'-disulfanediyldibenzoic acid bishydroxysuccinimide ester To a suspension of 4,4'-disulfanediyldibenzoic acid 31 (550 mg, 1.79 mmol) in DCM (20 mL) was added EDC.HC1 (770 mg, 3.95 mmol) and NHS (447 mg, 3.95 mmol). The mixture was stirred for 4 h and subsequent washed with water (2 x 20 mL), aqueous saturated NaHC03 (2 x 20 mL), dried over Na2S04, filtrated and concentrated in vacuo. 4,4'-Disulfanediyldibenzoic acid bishydroxysuccinimide ester was used crude in the next reaction.
Example 25: Synthesis of 4,4'-disulfanediylbis(N-(2"-(2'-(2-azidoethoxy)- ethoxy) ethyl) benzamide)
To a solution of 2"-(2'-(2-azidoethoxy)ethoxy)ethan-l -amine (210 mg, 1 mmol) in DCM (10 mL) was added Et3N (240 μΐ^, 1.7 mmol) and the mixture was stirred for 5 min followed by the addition of the 4,4'-dithiodibenzoic acid bishydroxysuccinimide ester (240 mg, 0.5 mmol). After stirring overnight, the reaction mixture was concentrated under reduced pressure and the product was purified on flash column chromatography (DCM - DCM:MeOH, 96:4). The fractions containing the product were washed with water (3 x 20 mL), subsequent dried over Na2S04, filtrated and concentrated in vacuo to yield 4,4'-disulfanediylbis(N-(2"-(2'-(2- azidoethoxy)ethoxy)ethyl)benzamide in 54% yield (168 mg, 0.27 mmol). 1H-NMR (400 MHz, CDC13) δ (ppm) 7.74-7.72 (m, 4H), 7.54-7.52 (m, 4H), 3.68-3.58 (m, 20H), 3.35 (t, 4H, J= 4.8 Hz). Example 26: Synthesis of mercaptobenzoic acid derivative 41
To a solution of 4,4'-disulfanediyl-bis(N-(2"-(2'-(2-azidoethoxy)ethoxy)ethyl)- benzamide (22 mg, 0.036 mmol) in THF at 0 °C (1 mL) was added NaBH4 (2.7 mg, 0.07 mmol). The slurry was stirred and after 3 and 6h extra NaBH4 (2 times 2.7 mg, 0.07 mmol) was added. The reaction was stirred for another hour followed by the addition of 0.1 M HC1 (1 mL) and DCM (2 mL). The mixture was extracted with DCM (2 x 5 mL) and the combined organic layers were dried over Na2S04, filtrated and concentrated in vacuo to yield 41 in 69% yield (16 mg, 0.05 mmol). 1H- MR (400 MHz, CDC13) δ (ppm) 7.65-7.63 (m, 2H), 7.29-7.26 (m, 2H), 3.68-3.56 (m, 10H), 3.35 (t, 2H, J= 6.4 Hz).
Example 27: Synthesis ofN-(2-(2-(2-azidoethoxy)ethoxy)ethyl)but-3-ynamide (42)
To a solution of but-3-ynoic acid (15 mg, 0.18 mmol) in CH2C12 (2 mL) was added Mukayama reagens (66 mg, 0.27 mmol) and the suspension was stirred for lh followed by the dropwise addition of a premixed solution of 2-(2-(2-azidoethoxy)ethoxy)ethan- 1-amine (17 mg, 0.20 mmol) and Et3N (60 μί, 0.45 mmol) in CH2C12 (2 mL). After 30 min the mixture was concentrated under reduced pressure and subsequent water (5 mL) and EtOAc (5 mL) were added. The organic layer was washed with water (2 x 5 mL), dried over Na2S04 filtrated and concentrated in vacuo. Flash chromatography (EtOAc:pentane 1 : 1→ 6: 1) afforded product 42 (9 mg, 0.04 mmol, 21%). 1H- MR (400 MHz, CDCI3): δ 3.64-3.58 (m, 6H), 3.55-3.52 (m, 2H), 3.45-3.42 (m, 2H), 3.33 (t, J= 4.8 Hz, 2H), 3.15 (d, J= 2.4 Hz, 2H), 2.29 (t, J= 2.8 Hz, 1H).
Example 28: Synthesis of N-(2-(2-(2-azidoethoxy)ethoxy)ethyl)buta-2-allenamide (43) Butynamide 42 (9 mg, 0.04 mmol) was dissolved in dioxane (0.5 mL) followed by the addition K2C03 (18% in water, 0.5 mL). The reaction mixture was heated to 40 °C for 2h followed by the addition of citric acid (2 mL, 10% in water) and CH2C12 (2 mL). The water layer was extracted with CH2C12 (2 x 3 mL) and the organic layers were combined, dried over Na2S04, filtrated and concentrated in vacuo to yield the product 43 (8 mg, 0.04 mmol, 89%). 1H- MR (400 MHz, CDC13): δ 5.63 (t, J = 6.4 Hz, 1H), 5.21 (d, J= 6.7 Hz, 2H), 3.70-3.60 (m, 6H), 3.60-3.58 (m, 2H), 3.53-3.50 (m, 2H), 3.39 (t, J= 4.8 Hz, 2H).
Example 29: Synthesis of 1-pyrenecarboxylic acid OSu ester
To a solution of 1-pyrenecarboxylic acid (65 mg, 0.24 mmol) in DCM/DMF (2 mL each) was added N-hydroxysuccinimide (34 mg, 0.29 mmol) and EDC.HC1 (70 mg, 0.36 mmol). The reaction was stirred for 2 h and subsequent diluted with DCM (10 mL), washed with aqueous citric acid (10%, 5 mL) and saturated NaHC03 (3 x 5 mL), dried over Na2S04, filtrated and concentrated in vacuo to give crude 1- pyrenecarboxylic acid hydroxysuccinimide ester.
Example 30: Synthesis of 44
1-Pyrenecarboxylic acid hydroxysuccinimide ester (480 mg, 1.38 mmol) was dissolved in DCM (15 mL) and tert-butyl (2-(2-(2-aminoethoxy)ethoxy)ethyl)carbamate (348 mg, 1.4 mmol and Et3N (286
Figure imgf000085_0001
2.1 mmol) were added. The reaction mixture was stirred overnight and quenched with water (15 mL), the organic layer was washed with water (1 x 15 mL) and saturated aqueous NaHC03 (2 x 15 mL), dried over Na2S04, filtrated and concentrated in vacuo. Purification via gradient flash column chromatography (DCM → DCM:MeOH 95:5) yielded the Boc-protected pyrene derivative (460 mg, 0.97 mmol, 70%).
Next, the Boc-protected pyrene derivative (460 mg, 0.97 mmol) was dissolved in methanol (10 mL). Acetyl chloride 1.9 mmol) was added and after 1 and 3 h additional acetyl chloride (2 x 140
Figure imgf000085_0002
mol) was added. After stirring for 4 h the mixture was concentrated under reduced pressure. Next, the crude product (100 mg, 0.24 mmol) was dissolved in DCM (3 mL) and 2,5-dioxopyrrolidin-l-yl 4-(2,5-dioxo- 2,5-dihydro-lH-pyrrol-l-yl)butanoate (50 mg, 0.17 mmol) and Et3N (73 μί, 0.52 mmol) were added. After stirring overnight the solution was quenched with water (3 mL), washed with water (2 x 3 mL), dried over Na2S04, filtrated and concentrated. Purification via gradient flash column chromatography (DCM→ DCM:MeOH 95 :5) yielded 44 (69 mg, 0.13 mmol, 75%). 1H-NMR (400 MHz, CDC13) δ (ppm) 8.48 (d, 1H, J = 9.2 Hz), 8.12 (d, 2H, J = 7.6 Hz), 8.04-7.93 (m, 6H), 6.59 (bs, 1H), 6.38 (s, 2H), 5.99 (bs, 1H), 3.76-3.71 (m, 4H), 3.62-3.60 (m, 2H), 3.54-3.52 (m, 2H), 3.37 (t, 2H, J = 5.2 Hz), 3.23-3.17 (m, 4H), 1.82 (t, 2H, J= 6.8 Hz), 1.63 (q, 2H, J= 7.2 Hz).
Example 31: Formation of maleimide BCN conjugate 45
To a solution of BCN-(POE)3NH2 (13 mg, 0.04 mmol) in DMA (1 mL) was added 4- maleimido butanoic acid hydroxysuccinimide ester (7 mg, 0.03 mmol), followed by incubation for 1 h. The solution was used as such in the conjugation experiments. Example 32: Conjugation of trast(GalNProSH) 2 with 34 to give 39
Trastuzumab-(Gal ProSH)2 (1.0 mg, 10 mg/mL) was incubated with TCEP (6.6 μΐ., 10 mM in MiliQ), in PBS and 20 mM EDTA for 2 hours at rt. Subsequently, the reaction medium was exchanged to PBS using an Amicon Ultra-0.5, Ultracel-10 Membrane (Millipore). Dehydroascorbic acid (5.4 μΐ., 10 mM in DMSO) was added followed by incubation for 3 h. Maleimide-vc-PABA-MMAE 34 (1 μΐ,, 10 mM in DMA) was added and the mixture was incubated for 35 min. Subsequently, a sample (2 μΕ) was taken for digestion with Fabricator™ (50 U in 10 μΙ_, PBS) and washed with MiliQ using an Amicon Ultra-0.5, Ultracel-10 Membrane (Millipore) and mass analysis showed the formation of 39 (25706, expected mass 25701).
Example 33: Formation of trastuzumab-conjugate 46.
To a solution of 36 (R=C1) (3 μί, 63 mg/ml, 1.3 nmol) in 25 mM Tris-HCl pH 6.0 was added Tris-HCl (10 μί, 100 mM pH 7.2) and a solution of 41 (10 μί, 10 mM, 100 nmol in MiliQ + 10 mM EDTA + 10% DMF). After incubation overnight, AccuTOF analysis showed >95% conversion to the desired product 46 (mass 50010, expected mass 50006).
Example 34: SPAAC of 46 with 47, leading to antibody-drug conjugate 48
The remaining reagent of example 33 was removed via spin-filtration (3 x 0.5 mL) to Tris-HCl (25 mM, pH 7.5) and BCN-vc-PABA-MMAF 47 (10 μί, 2 mM, 20 nmol in MiliQ + 5% DMF) was added. After incubation overnight, AccuTOF analysis showed >95% conversion to the desired product 48 (mass 51566, expected mass 51564). Example 35: Conjugation of trast(GalNProSH) 2 with allenamide 43, leading to conjugate 49
Trastuzumab-(Gal ProSH)2 (0.2 mg, 20 mg/mL) in 50 mM Tris-HCl pH 6.0 was incubated with N-(2"-(2'-(2-azidoethoxy)ethoxy)ethyl)buta-2-allenamide (43) (1 μί, 40 mM in DMA) and 50 mM Tris-HCl pH 8.8 (10 μΙ_, ) overnigth at rt. Subsequent a sample (2 μΕ) was taken for digestion with Fabricator TM (50 U in 10 μΙ_, PBS) and washed with MiliQ using an Amicon Ultra-0.5, Ultracel-10 Membrane (Millipore) and mass analysis showed the formation of the desired product 49 (24627, expected mass 24627) as main product (about 70%). Example 36: Conjugation of trast(GalNProSH) 2 with 44, leading to conjugate 50 Trastuzumab-(Gal ProSH)2 (0.5 mg, 10 mg/mL) was incubated with TCEP (3.3 μΐ., 10 mM in MiliQ), in PBS and 20 mM EDTA for 2 hours at rt. Subsequent the reaction medium was exchanged to PBS using an Amicon Ultra-0.5, Ultracel-10 Membrane (Millipore). Dehydroascorbic acid (2.8 μΐ., 10 mM in DMSO) was added followed by incubation for 3h. Pyrene maleimide 44 (1 μΐ., 10 mM in DMA) was added and the mixture was incubated for 35 min. Subsequent a sample (2 μΕ) was taken for digestion with Fabricator TM (50 U in 10 μΙ_, PBS) and washed with MiliQ using an Amicon Ultra-0.5, Ultracel-10 Membrane (Millipore) and mass analysis showed the formation of the desired product 50 (24929, expected mass 24929) as main product (about 85%).
Example 37: Conjugation of trast(GalNProSH) 2 with 45 followed by addition of biotine-Ni (52)
Trastuzumab-(Gal BuSH)2 (1 mg, 10 mg/mL) was incubated with TCEP (8.3 μΐ., 10 mM in MiliQ), in PBS and 20 mM EDTA for 2 hours at rt. Subsequent the reaction medium was exchanged to PBS using an Amicon Ultra-0.5, Ultracel-10 Membrane (Millipore). Dehydroascorbic acid (6.5 μΐ., 10 mM in DMSO) was added followed by incubation for 3 h. Compound 45 (3 μΙ_, premix, 40 mM in DMA) was added and the mixture was incubated for 35 min. Subsequently, a sample (2 μΐ.) was taken for digestion with Fabricator™ (50 U in 10 μΙ_, PBS) and washed with MiliQ using an Amicon Ultra-0.5, Ultracel-10 Membrane (Millipore) and mass analysis showed the formation of the desired product 51 (24891, expected mass 24891) as main product (about 85%). The reaction mixture was spinfiltered to 25 mM Tris-HCl pH 7.5, two times, followed by the addition of 52 (3 μΐ^, 40 mM in DMF). After incubation a sample was subjected to DTT as described above and mass analysis showed the formation of the desired product 53 (50658, expected mass 50650) as main product (about 85%)), one of the remaining species is trimmed trastuzumab (49503, expected mass 49495).
Example 38: Synthesis of 52
2"-(2'-(2-azidoethoxy)ethoxy)ethan-l -amine (50 mg, 0.17 mmol) was dissolved in CH2C12 (2 mL) and biotine hydroxysuccinimide derivative (60 mg, 0.18 mmol) and Et3N (52 μΐ^, 0.37 mmol) were added. The reaction mixture was stirred overnight and quenched with water (5 mL) and CH2CI2 (5 mL). The organic layer was washed with water (2 x 5 mL), dried over NaS04, filtrated and concentrated under reduced pressure. Flash chromatography (DCM→ DCM:MeOH 9: 1) afforded product 52 (13 mg, 0.03 mmol, 20%). LRMS (ESI+) calcd for Ci6H29N604S ( +H+) 401.19, found 401.34.
Example 39: Synthesis of UDP-GalNProSH (59) and UDP-GalNBuSH (60) derivatives The reaction scheme of the synthesis of UDP-GalNProSH and UDP-GalNBuSH derivatives 59 + 60, starting from 24, as performed in Examples xxx, is shown in Figure 12.
Example 40: 3, 4, 6-tri-O-acetyl-D-galactosamine-l -phosphate (54)
To a solution of azide 24 (105 mg, 0.255 mmol) in MeOH (3 mL) was added Pd/C (20 mg). The reaction was stirred under a H2 atmosphere for 2 h and filtered over celite. The filter was rinsed with MeOH (10 mL) and the filtrate was concentrated in vacuo to afford 54 (94 mg, 0.244 mmol, 96%).
1H- MR (300 MHz, D20): δ 5.87-5.76 (m, 1H), 5.44 (br s, 1H), 5.30-5.20 (m, 1H), 4.55 (t, J= 6.3 Hz, 1H), 4.28-4.00 (m, 3H), 2.11 (s, 3H), 2.03 (s, 3H), 2.00 (s, 3H). LRMS (EST) calcd for Ci2Hi9NOnP (M") 384.07, found 384.1.
Example 41: Synthesis of 55
To a solution of 54 (162 mg, 0.42 mmol) in DCM/DMF (2 mL each), acetylmercapto- 3-propanoic acid hydroxysuccinimide ester (175 mg, 0.69 mmol) and Et3N (88 μΕ, 0.63 mmol) were added. After stirring overnight, the reaction mixture was concentrated under reduced pressure. Purification via flash column chromatography (EtOAc → EtOAc:MeOH 30:70) yielded 55 (85 mg, 0.17 mmol, 39%). LRMS (EST) calcd for Ci7H25NOi3PS (M") 514.09, found 514.06.
Example 42: Synthesis of 57
Next, compound 55 was coupled to UMP according to Baisch et al. Bioorg. Med. Chem., 1997, 5, 383-391.
Thus, a solution of D-uridine-5'-monophosphate disodium salt (1.49 g, 4.05 mmol) in H20 (15 mL) was treated with DOWEX 50Wx8 (H+ form) for 30 minutes and filtered. The filtrate was stirred vigorously at rt while tributylamine (0.966 mL, 4.05 mmol) was added dropwise. After 30 minutes of further stirring, the reaction mixture was lyophilized.
The resulting tributylammonium uridine-5'-monophosphate (103 mg, 0.20 mmol) was dissolved in dry DMF (2 mL) in an argon atmosphere. Carbonyldiimidazole (57 mg, 0.36 mmol) was added and the reaction mixture was stirred at r.t. for 30 min. Next, dry MeOH (7 μL) was added and stirred for 15 min to remove the excess CDI. The leftover MeOH was removed under high vacuum for 15 min. Subsequently, compound x (87 mg, 0.17 mmol) and N-methylimidizole HC1 (118 mg, 0.85 mmol) were dissolved in dry DMF (2 mL) and added dropwise to the reaction mixture. The reaction was allowed to stir overnight before concentration in vacuo. Flash chromatography (9:2: 1-5:2: 1 EtOAc:MeOH:H20) afforded 57 (91 mg, 0.11 mmol, 65%). LRMS (EST) calcd for C26H36N3O21P2S (M") 820.11, found 820.24. Example 43: Synthesis of UDP-GalNProSH (59)
UDP-galactosamine derivative 57 (178 mg, 0.21 mmol) was dissolved in a mixture of water: MeOH :Et3N (3 :7:3, 5 mL) and stirred until full deprotection was achieved according to LCMS. The reaction was concentrated under reduced pressure and split in two portions. One portion was stored, while the other half was purified on anion exchange column (Q HITRAP, 2 x 5 mL columns). First binding on the column was achieved via loading with buffer A (10 mM NaHC03) and the column was rinsed with 100 mL buffer A. Next a gradient to 25% B (250 mM NaHC03) was performed to elute the impurities such as diUMP. Increasing the gradient to 100%> B eluted the product as dimer. The fractions were freeze-dried and subsequent dissolved in water (5 mL) followed by the addition of DTT (15 mg, 0.1 mmol) and Et3N (few drops). After lh the reduction was to completion and the solvents were removed under reduced pressure. Flash chromatography (6:2: 1-3 :2: 1 EtOAc:MeOH:H20) afforded product 59 (17 mg, 0.03 mmol, 26%). LRMS (EST) calcd for Ci8H28N3Oi7P2S (M") 652.07, found 652.03. 1H-NMR (400 MHz, D20): δ 7.86 (d, J = 8.0 Hz, 1H), 5.88-5.86 (m, 2H), 5.48-5.45 (m, 1H), 4.28-4.05 (m, 8H), 3.95-3.85(m, 1H), 3.68-3.65 (m, 2H), 2.69-2.67 (m, 2H), 2.60-2.55 (m, 2H).
Example 44: Synthesis of acetylmercapto-4-butanoic acid 4-bromobutanoic acid (1 g, 6 mmol) was dissolved in DMF (7 mL) followed by the addition of potassium thioacetate (1 g, 9 mmol). The reaction was stirred for 1 h and quenched by 1 M HC1 (20 mL) and Et20 (20 mL). The organic layer was washed with 1 M HC1 (3 x 20 mL), dried over Na2S04, filtrated and concentrated. Flash chromatography (1 :0-7: 1 heptane:EtOAc + 1% Ac20) afforded acetylmercapto-4- butanoic acid (450 mg, 2.7 mmol, 46%). 1H- MR (400 MHz, CDC13): δ 2.94 (t, J= 7.2 Hz, 2H), 2.45 (t, J= 7.2 Hz, 2H), 2.34 (s, 3H), 1.92 (q, J = 6.8 Hz, 2H).
Example 45: Synthesis of acetylmercapto-4-butanoic acid hydroxysuccinimide ester Acetylmercapto-4-butanoic acid (450 mg, 2.7 mmol) was dissolved in CH2C12 (30 mL) followed by the addition of EDC.HC1 (813 mg, 4.1 mmol) and N-hydroxysuccinimide (500 mg, 4.1 mmol). The reaction was stirred overnight and quenched with saturated aqueous NaHC03 (20 mL). The organic layer was washed with saturated aqueous NaHC03 (2 x 20 mL) and citric acid (1 x 10 mL). The organic layer was dried over Na2SC"4, filtrated and concentrated, to give the title compound as a crude product (630 mg, 2.4 mmol, 90%).
Example 46: Synthesis of 56
To a solution of galactosamine derivative 54 (167 mg, 0.43 mmol) in DCM/DMF (2 mL each) was added acetylmercapto-4-butanoic acid hydroxysuccinimide (200 mg, 0.77 mmol) and Et3N (88 μΕ, 0.63 mmol). After stirring overnight, the reaction mixture was concentrated under reduced pressure. Purification via flash column chromatography (EtOAc EtOAc:MeOH 30:70) yielded 56 (68 mg, 0.13 mmol, 30%). LRMS (EST) calcd for Ci8H27NOi3PS (M") 528.10, found 528.25
Example 47: Synthesis of 58
Next, galactosamine derivative 56 was coupled to UMP according to Baisch et al. Bioorg. Med. Chem., 1997, 5, 383-391).
Thus, a solution of D-uridine-5'-monophosphate disodium salt (1.49 g, 4.05 mmol) in H20 (15 mL) was treated with DOWEX 50Wx8 (H+ form) for 30 minutes and filtered. The filtrate was stirred vigorously at rt while tributylamine (0.966 mL, 4.05 mmol) was added dropwise. After 30 minutes of further stirring, the reaction mixture was lyophilized. The resulting tributylammonium uridine-5 '-monophosphate (78 mg, 0.15 mmol) was dissolved in dry DMF (2 mL) in an argon atmosphere. Carbonyldiimidazole (43 mg, 0.27 mmol) was added and the reaction mixture was stirred at r.t. for 30 min. Next, dry MeOH (6 μΐ.) was added and stirred for 15 min to remove the excess CDI. The leftover MeOH was removed under high vacuum for 15 min. Subsequently, compound 56 (68 mg, 0.13 mmol) and N-methylimidizole HC1 (118 mg, 0.72 mmol) were dissolved in dry DMF (2 mL) and added dropwise to the reaction mixture. The reaction was allowed to stir overnight before concentration in vacuo. Flash chromatography (9:2: 1-5:2: 1 EtOAc:MeOH:H20) afforded product 58 (53 mg, 0.06 mmol, 49%). LRMS (EST) calcd for C27H38N3O21P2S (M") 834.13, found 834.26.
Example 48: Synthesis of 60
Sugar 58 (53 mg, 0.06 mmol) was dissolved in a mixture of degassed water: MeOH :Et3N (3 :7:3, 3 mL) and stirred until full deprotection was achieved according to LCMS. The reaction was concentrated under reduced pressure and purified on flash chromatography (7:2: 1-3 :2: 1 EtOAc:MeOH:H20). One portion was stored, the other half was purified on anion exchange column (Q HITRAP, 1 x 5 mL column). First binding on the column was achieved via loading with buffer A (10 mM NaHC03) and the column was rinsed with 50 mL buffer A. Next, a gradient to 25% B (250 mM NaHC03) was performed to elute the product 60 (1 mg, 0.002 mmol, 5%). LRMS (ESI-) calcd for Ci9H3iN30i7P2S (M") 666.08, found 666.0.
1H-NMR (400 MHz, D20): δ 7.86 (d, J = 8.0 Hz, 1H), 5.88-5.86 (m, 2H), 5.48-5.45 (m, 1H), 4.27-4.10 (m, 7H), 3.94-3.86(m, 2H), 3.68-3.61 (m, 3H), 3.12-3.08 (m, 2H), 2.46- 2.35 (m, 2H), 1.20-1.17 (m, 2H).
Example 49. Synthesis of 64
Acetylmercaptoacetic acid pentafluorophenol ester (100 mg, 0.33 mmol) was dissolved in CH2C12 (3 mL) and benzylamine (69 μΕ, 0.5 mmol) and Et3N (44 μΕ, 0.40 mmol) were added. The mixture was stirred overnight and quenched by the addition of water (3 mL), followed by washing with water (3 x 3 mL). The organic layer was dried over Na2S04, filtrated and concentrated in vacuo. Flash chromatography (EtOAc: heptane 1 :4 1 : 1) afforded 61, which was dissolved in degassed methanol (7 mL). Subsequent degassed aqueous NaOH (1M, 3 mL) was added and the reaction was stirred under nitrogen atmosphere. After 2 h the reaction was quenched with 1 M HC1 (10 mL) and CH2CI2 (20 mL). The waterlayer was extracted with CH2CI2 (2 x 30 mL), dried over Na2SC"4, filtrated and concentrated in vacuo. The product 64 was used crude for the next reaction. 1H- MR (400 MHz, CDC13): δ 7.28-7.70 (m, 5H), 6.94 (bs, 1H), 4.40 (d, J = 5.6 Hz, 2H), 3.21 (d, J = 9.2 Hz, 2H), 1.81 (t, J = 9.2 Hz, 1H). LRMS (ESI+) calcd for C9H12NOS ( +H+) 182.06, found 182.60.
Example 50. Synthesis of 65
Acetylmercapto-3 -propanoic acid hydroxysuccinimide ester (100 mg, 0.41 mmol) was dissolved in CH2C12 (3 mL) and benzylamine (69 μΐ,, 0.5 mmol) and Et3N (44 μΐ,, 0.40 mmol) were added. The mixture was stirred overnight and quenched by the addition of water (3 mL), followed by washing with water (3 x 3 mL). The organic layer was dried over Na2SC"4, filtrated and concentrated in vacuo. Flash chromatography (EtOAc: heptane 1 :4→ 1 : 1) afforded product 62, which was dissolved in degassed methanol (7 mL). Subsequent degassed aqueous NaOH (1 M, 3 mL) was added and the reaction was stirred under nitrogen atmosphere. After 2h the reaction was quenched with 1 M HC1 (10 mL) and CH2C12 (20 mL). The waterlayer was extracted with CH2C12 (2 x 30 mL), dried over Na2S04, filtrated and concentrated in vacuo. The product 65 was used crude for the next reaction. LRMS (ESI+) calcd for C10H14NOS ( +H+) 196.07, found 196.60.
Example 51. Synthesis of 66
Acetylmercapto-4-butanoic acid hydroxysuccinimide ester (100 mg, 0.38 mmol) was dissolved in CH2C12 (3 mL) and benzylamine (69 μΕ, 0.5 mmol) and Et3N (44 μΕ, 0.40 mmol) were added. The mixture was stirred overnight and quenched by the addition of water (3 mL), followed by washing with water (3 x 3 mL). The organic layer was dried over Na2S04, filtrated and concentrated in vacuo. Flash chromatography (EtOAc: heptane 1 :4 - 1 : 1) afforded product 63, which was dissolved in degassed methanol (7 mL). Subsequent degassed aqueous NaOH (1M, 3 mL) was added and the reaction was stirred under nitrogen atmosphere. After 2 h, the reaction was quenched with 1 M HC1 (10 mL) and CH2C12 (20 mL). The water-layer was extracted with CH2C12 (2 x 30 mL), dried over Na2SC"4, filtrated and concentrated in vacuo. The product 66 was used crude for the next reaction. LRMS (ESI+) calcd for CnHieNOS ( +H+) 210.09, found 210.60.
Example 52. Synthesis of succinimide adduct 67
To a solution of pyrene maleimide derivative 44 (10 mg, 0.02 mmol) in CH2CI2 was added 64 (8 mg, 0.044 mmol) and Et3N (10 μί, 0.09 mmol). After stirring for 1 h, the reaction was concentrated and flash chromatography (DCM → DCM:MeOH 9: 1) afforded 67 (4 mg, 0.01 mmol, 28%). LRMS (ESI-) calcd for C4oH43N407S ( +H+)
723.28, found 723.31.
Example 53. Synthesis of succinimide adduct 68
To a solution of pyrene maleimide derivative 44 (10 mg, 0.02 mmol) in CH2CI2 was added 65 (8 mg, 0.041 mmol) and Et3N (10 μί, 0.09 mmol). After stirring for 1 h, the reaction was concentrated and flash chromatography (DCM → DCM:MeOH 9: 1) afforded 68 (8 mg, 0.01 mmol, 54%). LRMS (ESI+) calcd for C4iH45N407S ( +H+)
737.29, found 736.29.
Example 54. Synthesis of succinimide adduct 69
To a solution of pyrene maleimide derivative 44 (10 mg, 0.02 mmol) in CH2CI2 was added 66 (8 mg, 0.04 mmol) and Et3N (10 μί, 0.09 mmol). After stirring for 1 h, the reaction was concentrated and flash chromatography (DCM → DGVLMeOH 9: 1) afforded 69 (6 mg, 0.01 mmol, 40%). LRMS (ESI+) calcd for C42H47N407S ( +H+) 751.31, found 751.38. Example 55. Stability studies of succinimides 67-69
One of succinimide conjugates 67-69 (1 mg) was dissolved in DMF (400 μΕ) followed by the addition of buffer solution 1, 2 or 3 (800 μΕ). Buffer solution 1 is PBS, buffer solution 2 is PBS + 1 mM glutathione (GSH, reduced) and buffer solution 3 is PBS + 1 mM glutathione (oxidized). Final concentration of glutathione (through dilution with DMF) is 0.71 mM. The samples were incubated at 37 °C and measured in time. Ratio of products was determined by LC-MS. Table 2. Relative ratio of starting succinimide (67-69) versus hydrolyzed succinimide versus glutathione adduct formed by reverse Michael, then GSH addition. Structures of compounds 67-69 are shown in Figure 12.
Figure imgf000094_0001
Example 56: Glycosyltransfer of UDP-sugar 59 (GalNProSH) to deglycosylated trastuzumab.
Deglycosylated trastuzumab (10 mg/mL) was incubated with 59 (1.3 mM) and β(1,4)- Gal-T1(Y289L,C342T) (0.2 mg/mL) in 10 mM MnCl2 and 50 mM Tris-HCl pH 6.0 for 16 hours at 30 °C.
Next, the functionalized trastuzumab was incubated with protein A agarose (40 μΐ. per mg IgG) for 1 hours at rt. The protein A agarose was washed three times with TBS (pH 6.0) and the IgG was eluted with 100 mM glycine-HCl pH 2.5. The eluted IgG was neutralized with 1 M Tris-HCl pH 7.0 and concentrated and washed with 50 mM Tris- HC1 pH 6.0 using an Amicon Ultra-0.5, Ultracel-10 Membrane (Millipore) to a concentration of 15-20 mg/mL. Spectral analysis after digestion with Fabricator™ (50 U in 10 μυ PBS pH 6.6) and subsequent wash with MiliQ using an Amicon Ultra-0.5, Ultracel-10 Membrane (Millipore) showed the formation of two products, the major product (24387 Da, expected mass 24388) of deglycosylated trastuzumab + GalNProSH (trastuzumab-(GalNProSH)2) and the minor (25037 Da, expected mass 25038) of deglycosylated trastuzumab + GalNProS-UDPGalNProS disulfide. The ratio between the products is about 60:40.
Example 57: Glycosyltransfer of UDP -sugar 60 (GalNBuSH) to deglycosylated trastuzumab.
Deglycosylated trastuzumab (10 mg/mL) was incubated with 60 (1.3 mM) and β(1,4)- Gal-T1(Y289M) (2 mg/mL) in 10 mM MnCl2 and 50 mM Tris-HCl pH 6.0 for 16 hours at 30 °C.
Spectral analysis after digestion with Fabricator™ (50 U in 10 μΙ_, PBS pH 6.6) and subsequent wash with MiliQ using an Amicon Ultra-0.5, Ultracel-10 Membrane (Millipore) showed 60% conversion of the starting material into two products, the minor product (24401 Da, expected mass 24402) of deglycosylated trastuzumab + GalNBuSH (trastuzumab-(GalNBuSH)2) and the major (25066 Da, expected mass 25068) of deglycosylated trastuzumab + GalNBuS-UDPGalNBuS disulfide. The ratio between the products is about 20:80.

Claims

Claims
Process for the preparation of a modified glycoprotein, the process comprising contacting a glycoprotein comprising a glycan comprising a terminal GlcNAc- moiety with Su(A)x-P in the presence of a catalyst selected from the group consisting of /3(l,4)-galactosyltransferases, /3(l,3)-N-galactosyltransferases, /3(l,4)-galactosyltransf erases comprising a mutant catalytic domain and (1,3)-Ν- galactosyltransferases comprising a mutant catalytic domain; wherein Su(A)x is a sugar derivative Su comprising x functional groups A wherein x is 1, 2, 3 or 4 and wherein A is independently selected from the group consisting of a thiol group or a precursor thereof, a halogen, a sulfonyloxy group, a halogenated acetamido group, a mercaptoacetamido group and a sulfonated hydroxy acetamido group; wherein P is a nucleotide; and wherein a glycan comprising a terminal GlcNAc-moiety is a glycan according to formula (101) or (102):
Figure imgf000096_0001
wherein:
b is 0 or 1;
d is 0 or 1;
e is 0 or 1 ; and
G is a monosaccharide, or a linear or branched oligosaccharide comprising 2 to 20 sugar moieties.
Process according to claim 1, wherein the catalyst is selected from the group consisting of /3(l,4)-galactosyltransf erases comprising a mutant catalytic domain and /3(l,3)-N-galactosyltransferases comprising a mutant catalytic domain.
Process according to claim 1 or claim 2, wherein the catalyst is a catalyst comprising a mutant catalytic domain from a /3(l,4)-galactosyltransferase, selected from the group consisting of bovine ?(1,4)-Gal-Tl GalT Y289L, GalT Y289N, GalT Y289I, GalT Y289F, GalT Y289M, GalT Y289V, GalT Y289G and GalT Y289A.
Process according to any one of the previous claims, wherein the modified glycoprotein is a modified antibody.
Process according to any one of the previous claims, wherein the process further comprises a step of providing a glycoprotein comprising a glycan comprising a terminal GlcNAc-moiety at the non-reducing end.
Glycoprotein obtainable by the process according to any one of claims 1 - 5.
Glycoprotein according to claim 6, wherein the glycoprotein comprises a glycan according to formula (105) or (106):
Figure imgf000097_0001
wherein:
b is 0 or 1;
d is 0 or 1;
e is 0 or 1 ;
G is a monosaccharide, or a linear or branched oligosaccharide comprising 2 - 20 saccharide moieties; and
Su(A)x is a sugar derivative Su comprising x functional groups A wherein x is 1, 2, 3 or 4 and wherein A is independently selected from the group consisting a thiol group, a halogen, a sulfonyloxy group, a halogenated acetamido group, a mercaptoacetamido group and a sulfonated hydroxyacetamido group.
8. Glycoprotein according to claim 6 or claim 7, wherein the glycoprotein is an antibody. Process for the preparation of a glycoprotein-conjugate, comprising reacting a modified glycoprotein according to any one of claims 6 - 8 with a linker- conjugate, wherein said linker-conjugate comprises a functional group B and one or more molecules of interest, wherein said functional group B is a functional group that is capable of reacting with a functional group A on a glycan of the modified glycoprotein, and wherein functional group A is independently selected from the group consisting of a thiol group, a halogen, a sulfonyloxy group, a halogenated acetamido group, a mercaptoacetamido group and a sulfonated hydroxyacetamido group.
Process according to claim 9, wherein:
(a) when said modified glycoprotein is a halogen-modified glycoprotein or a halogenated acetamido-modified glycoprotein, functional group B comprises a thiol group, an alcohol group or an amine group; or
(b) when said modified glycoprotein is a thiol-modified glycoprotein or a mercaptoacetamido-modified glycoprotein, functional group B comprises an N-maleimide group or a halogenated acetamido group or an alkene group; or
(c) when said modified glycoprotein is a sulfonyloxy-modified glycoprotein or a sulfonated hydroxyacetamido-modified glycoprotein, functional group B comprises thiol group, an alcohol group or an amine group.
Process according to claim 9, wherein said modified glycoprotein is a thiol- modified glycoprotein or a mercaptoacetamido-modified glycoprotein, and functional group B comprises an alleneamide group.
Glycoprotein-conjugate obtainable by the process according to claim 9 or claim 10.
Glycoprotein-conjugate obtainable by the process according to claim 11 Glycoprotein-conjugate according to claim 12, wherein the glycoprot according to formula (121) or (122):
Figure imgf000099_0001
121
Figure imgf000099_0002
122
wherein:
Pr represents a protein;
L is a Linker;
D is a molecule of interest;
r is 1 to 20;
x is 1, 2, 3 or 4;
y is 1 to 20;
b is 0 or 1;
d is 0 or 1;
e is 0 or 1 ;
p is 0 or 1;
Q is -N(H)C(0)CH2- or CH2;
Su is a sugar or sugar derivative; and
G is a monosaccharide, or a linear or branched oligosaccharide comprising 2 20 sugar moieties. Glycoprotein-conjugate according to claim 12, wherein the glycoprot according to formula (123) or (124):
Figure imgf000100_0001
123
Figure imgf000100_0002
124 wherein Pr, L, D, r, x, y, b, d, e, p, Q, Su and G are as defined in claim 14 and R9 is selected from the group consisting of L(D)r, hydrogen, Ci - C24 alkyl groups, C6 - C24 aryl groups C7 - C24 alkylaryl groups and C7 - C24 arylalkyl groups, the Ci - C24 alkyl groups, C6 - C24 aryl groups C7 - C24 alkylaryl groups and C7 - C24 arylalkyl groups optionally being substituted.
16. Glycoprotein-conjugate according to claim 13, wherein the glycoprotein is according to formula (125) or (126): ( iuc )t
-GlcNAc Su-
Figure imgf000101_0001
125
Figure imgf000101_0002
126 wherein Pr, L, D, r, x, y, b, d, e, p, Q, Su and G are as defined in claim 14 and R is as defined in claim 15.
17. Glycoprotein-conjugate according to any one of claims 12, 14 or 15, wherein the glycoprotein-conjugate is an antibody-conjugate.
18. Glycoprotein-conjugate according to claim 13 or claim 16, wherein the glycoprotein-conjugate is an antibody-conjugate.
Antibody-conjugate according to claim 17, wherein an antibody is conjugated to a molecule of interest D via a linker L, wherein D is selected from the group consisting of pharmaceutically active substances.
Antibody-conjugate according to claim 18, wherein an antibody is conjugated to a molecule of interest D via a linker L, wherein D is selected from the group consisting of pharmaceutically active substances.
21. Antibody-conjugate according to claim 19, for use as a medicament. Antibody-conjugate according to claim 20, for use as a medicament.
PCT/NL2014/050714 2013-10-14 2014-10-14 Modified glycoprotein, protein-conjugate and process for the preparation thereof WO2015057063A1 (en)

Priority Applications (5)

Application Number Priority Date Filing Date Title
CN201480068041.2A CN105814213B (en) 2013-10-14 2014-10-14 Modified glycoproteins, protein-conjugates and methods of making the same
EP14790774.5A EP2935608A1 (en) 2013-10-14 2014-10-14 Modified glycoprotein, protein-conjugate and process for the preparation thereof
JP2016547822A JP2016538877A (en) 2013-10-14 2014-10-14 Modified glycoproteins, protein conjugates and methods for their preparation
US15/029,123 US10072096B2 (en) 2013-10-14 2014-10-14 Modified glycoprotein, protein-conjugate and process for the preparation thereof
US16/115,261 US20190225706A1 (en) 2013-10-14 2018-08-28 Modified glycoprotein, protein-conjugate and process for the preparation thereof

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
EP13188514.7 2013-10-14
EP13188585.7 2013-10-14
EP13188514 2013-10-14
EP13188585 2013-10-14
EP14165581.1 2014-04-23
EP14165563.9 2014-04-23
EP14165581 2014-04-23
EP14165563 2014-04-23

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US15/029,123 A-371-Of-International US10072096B2 (en) 2013-10-14 2014-10-14 Modified glycoprotein, protein-conjugate and process for the preparation thereof
US16/115,261 Continuation US20190225706A1 (en) 2013-10-14 2018-08-28 Modified glycoprotein, protein-conjugate and process for the preparation thereof

Publications (1)

Publication Number Publication Date
WO2015057063A1 true WO2015057063A1 (en) 2015-04-23

Family

ID=51842729

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/NL2014/050714 WO2015057063A1 (en) 2013-10-14 2014-10-14 Modified glycoprotein, protein-conjugate and process for the preparation thereof

Country Status (5)

Country Link
US (2) US10072096B2 (en)
EP (1) EP2935608A1 (en)
JP (1) JP2016538877A (en)
CN (1) CN105814213B (en)
WO (1) WO2015057063A1 (en)

Cited By (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016022027A1 (en) 2014-08-04 2016-02-11 Synaffix B.V. Process for the modification of a glycoprotein using a βeta-(1,4)-n-acetylgalactosaminyltransferase or a mutant thereof
WO2017137457A1 (en) * 2016-02-08 2017-08-17 Synaffix B.V. Antibody-conjugates with improved therapeutic index for targeting cd30 tumours and method for improving therapeutic index of antibody-conjugates
WO2017137458A1 (en) * 2016-02-08 2017-08-17 Synaffix B.V. Antibody-conjugates with improved therapeutic index for targeting cd30 tumours and method for improving therapeutic index of antibody-conjugates
JP2017197512A (en) * 2016-02-08 2017-11-02 シンアフィックス ビー.ブイ. Novel antibody-conjugates with improved therapeutic index for targeting cd30 tumors and methods for improving therapeutic index of antibody-conjugates
JP2017200902A (en) * 2016-02-08 2017-11-09 シンアフィックス ビー.ブイ. Novel antibody-conjugate with improved therapeutic index for targeting HER2 tumor and method for improving therapeutic index of antibody-conjugate
GB201901608D0 (en) 2019-02-06 2019-03-27 Vib Vzw Vaccine adjuvant conjugates
CN112135837A (en) * 2018-03-14 2020-12-25 勇·巴 Pegylated antifreeze proteins and methods of making and using same
US10874746B2 (en) 2016-02-08 2020-12-29 Synaffix B.V. Sulfamide linkers for use in bioconjugates
US10905678B2 (en) 2014-04-08 2021-02-02 University Of Georgia Research Foundation, Inc. Site-specific antibody-drug glycoconjugates and methods
US10973922B2 (en) 2013-05-02 2021-04-13 Glykos Finland Oy Glycoprotein-toxic payload conjugates
US10973920B2 (en) 2014-06-30 2021-04-13 Glykos Finland Oy Saccharide derivative of a toxic payload and antibody conjugates thereof
WO2021144315A1 (en) 2020-01-13 2021-07-22 Synaffix B.V. Conjugates of antibodies an immune cell engagers
WO2021144314A1 (en) 2020-01-13 2021-07-22 Synaffix B.V. Via cycloaddition bilaterally functionalized antibodies
US11085062B2 (en) 2016-12-29 2021-08-10 Development Center For Biotechnology Processes for preparing glycoprotein-drug conjugates
US11338043B2 (en) 2016-02-08 2022-05-24 Synaffix B.V. Antibody-conjugates with improved therapeutic index for targeting HER2 tumours and method for improving therapeutic index of antibody-conjugates
WO2022167689A1 (en) 2021-02-08 2022-08-11 Synaffix B.V. Multifunctional antibodies
WO2022187591A1 (en) 2021-03-05 2022-09-09 Go Therapeutics, Inc. Anti-glyco-cd44 antibodies and their uses
WO2023014863A1 (en) 2021-08-05 2023-02-09 Go Therapeutics, Inc. Anti-glyco-muc4 antibodies and their uses
US11583590B2 (en) 2017-09-29 2023-02-21 Daiichi Sankyo Company, Limited Antibody-pyrrolobenzodiazepine derivative conjugate and method of use thereof for treating a tumor
WO2023086028A3 (en) * 2021-11-10 2023-08-10 Nanyang Technological University Allenamide linkers for antibody-drug conjugate
WO2023161296A1 (en) 2022-02-22 2023-08-31 Adc Therapeutics Sa Conjugation method involving a transglutaminase at the fc region comprising a trimmed n-glycan
US11746124B2 (en) 2016-09-01 2023-09-05 Forschungsverbund Berlin E.V. Chemoselective thiol-conjugation with alkene or alkyne-phosphonamidates
US11951175B2 (en) 2016-02-08 2024-04-09 Synaffix B.V. Bioconjugates containing sulfamide linkers for use in treatment

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10523731B2 (en) 2014-10-20 2019-12-31 Lg Electronics Inc. Apparatus for transmitting broadcast signal, apparatus for receiving broadcast signal, method for transmitting broadcast signal and method for receiving broadcast signal
MX2023007126A (en) 2020-12-23 2023-09-04 Univ Muenchen Ludwig Maximilians Improved cd30 targeting antibody drug conjugates and uses thereof.
CN115247160B (en) * 2022-07-28 2024-03-15 格进(杭州)生物技术有限责任公司 Detection method for protein core fucosylation modification

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004063344A2 (en) 2003-01-10 2004-07-29 Government Of The United States Of America As Represented By The Sercretary Of The Department Of Health And Human Services National Institutes Of Health CATALYTIC DOMAINS OF β/1,4)-GALACTOSYLTRANSFERASE I HAVING ALTERED DONOR AND ACCEPTOR SPECIFICITIES, DOMAINS THAT PROMOTE IN VITRO PROTEIN FOLDING, AND METHODS FOR THEIR USE
WO2007095506A1 (en) 2006-02-10 2007-08-23 Invitrogen Corporation Oligosaccharide modification and labeling of proteins
WO2009025646A1 (en) 2007-08-22 2009-02-26 Government Of The U.S.A, As Represented By The Secretary, Department Of Health & Human Services Alpha 1-3 n-galactosyltransferase with altered donor specificities
WO2009102820A2 (en) 2008-02-11 2009-08-20 Government Of The U.S A., As Represented By The Secretary, Department Of Health And Human Services Modified sugar substrates and methods of use
WO2013037824A1 (en) 2011-09-13 2013-03-21 Genovis Ab Endoglycosidase from streptococcus pyogenes and methods using it
WO2014065661A1 (en) * 2012-10-23 2014-05-01 Synaffix B.V. Modified antibody, antibody-conjugate and process for the preparation thereof

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2009531289A (en) * 2006-02-10 2009-09-03 ライフ テクノロジーズ コーポレーション Method for modifying and labeling proteins with oligosaccharides
JP2009544327A (en) * 2006-07-21 2009-12-17 ノヴォ ノルディスク アー/エス Glycosylation of peptides with O-linked glycosylation sequences
PE20130643A1 (en) * 2010-07-12 2013-06-07 Covx Technologies Ireland Ltd CONJUGATES OF MULTIFUNCTIONAL ANTIBODIES
CN103608038A (en) * 2011-03-31 2014-02-26 弗·哈夫曼-拉罗切有限公司 Methods of administering beta7 integrin antagonists

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004063344A2 (en) 2003-01-10 2004-07-29 Government Of The United States Of America As Represented By The Sercretary Of The Department Of Health And Human Services National Institutes Of Health CATALYTIC DOMAINS OF β/1,4)-GALACTOSYLTRANSFERASE I HAVING ALTERED DONOR AND ACCEPTOR SPECIFICITIES, DOMAINS THAT PROMOTE IN VITRO PROTEIN FOLDING, AND METHODS FOR THEIR USE
WO2007095506A1 (en) 2006-02-10 2007-08-23 Invitrogen Corporation Oligosaccharide modification and labeling of proteins
WO2008029281A2 (en) 2006-02-10 2008-03-13 Invitrogen Corporation Labeling and detection of post translationally modified proteins
WO2009025646A1 (en) 2007-08-22 2009-02-26 Government Of The U.S.A, As Represented By The Secretary, Department Of Health & Human Services Alpha 1-3 n-galactosyltransferase with altered donor specificities
WO2009102820A2 (en) 2008-02-11 2009-08-20 Government Of The U.S A., As Represented By The Secretary, Department Of Health And Human Services Modified sugar substrates and methods of use
WO2013037824A1 (en) 2011-09-13 2013-03-21 Genovis Ab Endoglycosidase from streptococcus pyogenes and methods using it
WO2014065661A1 (en) * 2012-10-23 2014-05-01 Synaffix B.V. Modified antibody, antibody-conjugate and process for the preparation thereof

Non-Patent Citations (36)

* Cited by examiner, † Cited by third party
Title
ABBAS ET AL., ANGEW. CHEM. INT. ED., vol. 53, 2014, pages 7491 - 7494
AGNEW ET AL: "Evaluating options for hard to label antibodies", LIFE TECHNOLOGIES / ABRF ARG USER'S STUDY: 2012-2013, 4 April 2013 (2013-04-04), pages 1 - 12, XP002723746, Retrieved from the Internet <URL:http://www.abrf.org/Other/ABRFMeetings/ABRF2013/RG%20presentations/RG3_ARG_Agnew.pdf> [retrieved on 20140429] *
BIOCONJUGATE CHEM, vol. 20, 2009, pages 1228
BRIK ET AL: "Sugar-assisted ligation for the synthesis of glycopeptides", CHEMISTRY - A EUROPEAN JOURNAL, vol. 13, 2007, pages 5670 - 5675, XP009136145 *
CLARK ET AL: "Direct in-gel fluorescence detection and cellular imaging of O-GlcNAc-modified proteins", JOURNAL OF THE AMERICAN CHEMICAL SOCIETY, vol. 130, 2008, pages 11576 - 11577, XP002723745 *
COLLIN ET AL., EMBO J., vol. 20, 2001, pages 3046
ELLING ET AL., CHEMBIOCHEM, vol. 2, 2001, pages 884
EMBO J., vol. 12, 2001, pages 3046
HONG ET AL., ANGEW. CHEM. INT. ED. ENGL., vol. 48, 2009, pages 9879
HOSZTAFI ET AL., HELV. CHIM. ACTA, vol. 79, 1996, pages 133 - 136
J. BIOL. CHEM., vol. 277, 2002, pages 20833
KIM ET AL: "Chemical arsenal for the study of O-GlcNAc", MOLECULES, vol. 16, 2011, pages 1987 - 2022, XP002720161 *
KISO ET AL., GLYCOCONJ. J., vol. 23, 2006, pages 565 - 573
KUNZ ET AL., ANGEW. CHEM. INT. ED., vol. 46, 2007, pages 5226 - 5230
LINHARDT ET AL., J. ORG. CHEM., vol. 77, 2012, pages 1449 - 1456
MERCER ET AL: "Use of novel mutant galactosyltransferase for the bioconjugation of terminal N-acetylglucosamine (GlcNAc) residues on live cell surface", BIOCONJUGATE CHEMISTRY, vol. 24, 22 December 2012 (2012-12-22), pages 144 - 152, XP055061203 *
N.M. OKELEY ET AL., BIOCONJ. CHEM., vol. 24, 2013, pages 1650
OKELEY ET AL: "Metabolic engineering of monoclonal antibody carbohydrates for antibody-drug conjugation", BIOCONJUGATE CHEMISTRY, vol. 24, 19 September 2013 (2013-09-19), pages 1650 - 1655, XP002734768 *
PANNECOUCKE ET AL., TETRAHEDRON LETT., vol. 49, 2008, pages 2294
PILLER ET AL., ACS CHEM. BIOL., vol. 7, 2012, pages 753
PILLER ET AL., BIOORG. MED. CHEM. LETT., vol. 15, 2005, pages 5459 - 5462
POUILLY ET AL: "Evaluation of analogues of GalNAc as substrates for enzymes of the mammalian GalNAc salvage pathway", ACS CHEMICAL BIOLOGY, vol. 7, 2012, pages 753 - 760, XP055101993 *
QASBA ET AL., BIOCONJUGATE CHEM., vol. 20, 2009, pages 1228
QASBA ET AL., GLYCOBIOLOGY, vol. 12, 2002, pages 691
QASBA ET AL., J. BIOL. CHEM., vol. 277, 2002, pages 20833
QASBA ET AL., PROT. EXPR. PUR., vol. 30, 2003, pages 219 - 229
QASBA ET AL., PROTO EXPR. PUR., vol. 30, 2003, pages 219
QASBA ET AL: "Site-specific linking of biomolecules via glycan residues using glycosyltransferases", BIOTECHNOLOGY PROGRESS, vol. 24, 2008, pages 520 - 526, XP009107098 *
QU ET AL., J. IMMUNOL. METHODS, vol. 213, 1998, pages 131
SATOH ET AL., GLYCOBIOLOGY, vol. 17, 2006, pages 104 - 118
SHABAT ET AL., SOFT MATTER, vol. 6, 2012, pages 1073
SHARMA ET AL: "Design and synthesis of LNA based mercaptoacetamido-linked nucleoside dimers", CARBOHYDRATE NEWS LETTER (INDIA), vol. 14, December 2013 (2013-12-01), pages 15, XP002734784 *
TARENTINO ET AL., GLYCOBIOLOGY, vol. 5, 1995, pages 599
WANG ET AL., CHEM. EUR. J., vol. 16, 2010, pages 13343 - 13345
WONG ET AL., CHEM. REV., vol. 111, 2011, pages 4259
ZEGLIS ET AL: "Enzyme-mediated methodology for the site-specific radiolabeling of antibodies based on catalyst-free click chemistry", BIOCONJUGATE CHEMISTRY, vol. 24, 17 May 2013 (2013-05-17), pages 1057 - 1067, XP055101895 *

Cited By (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10973922B2 (en) 2013-05-02 2021-04-13 Glykos Finland Oy Glycoprotein-toxic payload conjugates
US10905678B2 (en) 2014-04-08 2021-02-02 University Of Georgia Research Foundation, Inc. Site-specific antibody-drug glycoconjugates and methods
US11872215B2 (en) 2014-04-08 2024-01-16 University Of Georgia Research Foundation, Inc. Site-specific antibody-drug glyconjugates and methods
US10973920B2 (en) 2014-06-30 2021-04-13 Glykos Finland Oy Saccharide derivative of a toxic payload and antibody conjugates thereof
EP4148138A1 (en) 2014-08-04 2023-03-15 SynAffix B.V. Process for the modification of a glycoprotein using a beta-(1,4)-n-acetylgalactosaminyltransferase or a mutant thereof
WO2016022027A1 (en) 2014-08-04 2016-02-11 Synaffix B.V. Process for the modification of a glycoprotein using a βeta-(1,4)-n-acetylgalactosaminyltransferase or a mutant thereof
JP2017200902A (en) * 2016-02-08 2017-11-09 シンアフィックス ビー.ブイ. Novel antibody-conjugate with improved therapeutic index for targeting HER2 tumor and method for improving therapeutic index of antibody-conjugate
US11951175B2 (en) 2016-02-08 2024-04-09 Synaffix B.V. Bioconjugates containing sulfamide linkers for use in treatment
US10874746B2 (en) 2016-02-08 2020-12-29 Synaffix B.V. Sulfamide linkers for use in bioconjugates
US11957763B2 (en) 2016-02-08 2024-04-16 Synaffix B.V. Sulfamide linkers for use in bioconjugates
CN108883191A (en) * 2016-02-08 2018-11-23 西纳福克斯股份有限公司 Antibody-conjugate with improved therapeutic index for targeting CD30 tumour and the method for improving antibody-conjugate therapeutic index
US11590239B2 (en) 2016-02-08 2023-02-28 Synaffix B.V. Antibody-conjugates with improved therapeutic index for targeting CD30 tumours and method for improving therapeutic index of antibody-conjugates
JP2017197512A (en) * 2016-02-08 2017-11-02 シンアフィックス ビー.ブイ. Novel antibody-conjugates with improved therapeutic index for targeting cd30 tumors and methods for improving therapeutic index of antibody-conjugates
WO2017137458A1 (en) * 2016-02-08 2017-08-17 Synaffix B.V. Antibody-conjugates with improved therapeutic index for targeting cd30 tumours and method for improving therapeutic index of antibody-conjugates
US11338043B2 (en) 2016-02-08 2022-05-24 Synaffix B.V. Antibody-conjugates with improved therapeutic index for targeting HER2 tumours and method for improving therapeutic index of antibody-conjugates
WO2017137457A1 (en) * 2016-02-08 2017-08-17 Synaffix B.V. Antibody-conjugates with improved therapeutic index for targeting cd30 tumours and method for improving therapeutic index of antibody-conjugates
US11746124B2 (en) 2016-09-01 2023-09-05 Forschungsverbund Berlin E.V. Chemoselective thiol-conjugation with alkene or alkyne-phosphonamidates
US11085062B2 (en) 2016-12-29 2021-08-10 Development Center For Biotechnology Processes for preparing glycoprotein-drug conjugates
US11583590B2 (en) 2017-09-29 2023-02-21 Daiichi Sankyo Company, Limited Antibody-pyrrolobenzodiazepine derivative conjugate and method of use thereof for treating a tumor
US11628223B2 (en) 2017-09-29 2023-04-18 Daiichi Sankyo Company, Limited Antibody-drug conjugates comprising substituted benzo[e]pyrrolo[1,2-α][1,4]diazepines
CN112135837A (en) * 2018-03-14 2020-12-25 勇·巴 Pegylated antifreeze proteins and methods of making and using same
GB201901608D0 (en) 2019-02-06 2019-03-27 Vib Vzw Vaccine adjuvant conjugates
WO2021144314A1 (en) 2020-01-13 2021-07-22 Synaffix B.V. Via cycloaddition bilaterally functionalized antibodies
WO2021144315A1 (en) 2020-01-13 2021-07-22 Synaffix B.V. Conjugates of antibodies an immune cell engagers
WO2022167689A1 (en) 2021-02-08 2022-08-11 Synaffix B.V. Multifunctional antibodies
WO2022187591A1 (en) 2021-03-05 2022-09-09 Go Therapeutics, Inc. Anti-glyco-cd44 antibodies and their uses
WO2023014863A1 (en) 2021-08-05 2023-02-09 Go Therapeutics, Inc. Anti-glyco-muc4 antibodies and their uses
WO2023086028A3 (en) * 2021-11-10 2023-08-10 Nanyang Technological University Allenamide linkers for antibody-drug conjugate
WO2023161296A1 (en) 2022-02-22 2023-08-31 Adc Therapeutics Sa Conjugation method involving a transglutaminase at the fc region comprising a trimmed n-glycan

Also Published As

Publication number Publication date
JP2016538877A (en) 2016-12-15
US20160280797A1 (en) 2016-09-29
US10072096B2 (en) 2018-09-11
EP2935608A1 (en) 2015-10-28
CN105814213A (en) 2016-07-27
US20190225706A1 (en) 2019-07-25
CN105814213B (en) 2021-05-04

Similar Documents

Publication Publication Date Title
US20190225706A1 (en) Modified glycoprotein, protein-conjugate and process for the preparation thereof
EP3057618B1 (en) Glycoengineered antibody, antibody-conjugate and methods for their preparation
EP2935611B1 (en) Glycoengineered antibody, antibody-conjugate and methods for their preparation
EP3058083B1 (en) Modified glycoprotein, protein-conjugate and process for the preparation thereof
JP7167071B2 (en) Modified Antibodies, Antibody Conjugates and Methods of Preparing Them
CN107810273B (en) Method for modifying glycoproteins with a glycosyltransferase that is or is derived from a beta- (1,4) -N-acetylgalactosamine transferase
WO2015112013A1 (en) Process for the attachment of a galnac moiety comprising a (hetero)aryl group to a glcnac moiety, and product obtained thereby
WO2022037665A1 (en) Site-specific antibody conjugates and the methods for preparation of the same

Legal Events

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

Ref document number: 14790774

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 2014790774

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2016547822

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 15029123

Country of ref document: US

NENP Non-entry into the national phase

Ref country code: DE