Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
PROCESS FOR THE MODIFICATION OF A GLYCOPROTEIN USING A ΒETA-(1,4)-N-ACETYLGALACTOSAMINYLTRANSFERASE OR A MUTANT THEREOF
Document Type and Number:
WIPO Patent Application WO/2016/022027
Kind Code:
A1
Abstract:
The present invention relates to a process for the modification of a glycoprotein, using a β-(1,4)-N-acetylgalactosaminyltransferase or a mutant thereof.The process comprisesthe step of contacting a glycoprotein comprising a glycan comprising a terminal GlcNAc-moiety, in the presence of a β-(1,4)-N-acetylgalactosaminyl- transferase or a mutant thereof, with anon-natural sugar-derivative nucleotide.The non-natural sugar-derivative nucleotideis according to formula (3), wherein A is selected from the group consisting of -N3; -C(0)R3; -C=C-R4; -SH; -SC(0)R8; -SC(V)OR8, wherein V is O or S; -X wherein X is selected from the group consisting of F, CI, Br and I; -OS(0)2R5; an optionally substituted C2 - C24 alkyl group; an optionally substituted terminal C2 - C24 alkenyl group; and an optionally substituted terminal C3 - C24 allenyl group.

Inventors:
WASIEL ANNA AGNIESKA (NL)
VAN DELFT FLORIS LOUIS (NL)
VAN BERKEL SANDER SEBASTIAAN (NL)
Application Number:
PCT/NL2015/050567
Publication Date:
February 11, 2016
Filing Date:
August 04, 2015
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
SYNAFFIX BV (NL)
International Classes:
C07H19/10; C07K16/00; C12P21/00
Domestic Patent References:
WO2014065661A12014-05-01
WO2007095506A12007-08-23
WO2008029281A22008-03-13
WO2014065661A12014-05-01
WO2013037824A12013-03-21
WO2015057063A12015-04-23
WO2015112013A12015-07-30
Other References:
BRIAN M. ZEGLIS ET AL: "Enzyme-Mediated Methodology for the Site-Specific Radiolabeling of Antibodies Based on Catalyst-Free Click Chemistry", BIOCONJUGATE CHEMISTRY, vol. 24, no. 6, 19 June 2013 (2013-06-19), pages 1057 - 1067, XP055101895, ISSN: 1043-1802, DOI: 10.1021/bc400122c
CLARK P M ET AL: "Direct in-gel fluorescence detection and cellular imaging of O-GlcNAc-modified proteins", JOURNAL OF THE AMERICAN CHEMICAL SOCIETY, AMERICAN CHEMICAL SOCIETY, US, vol. 130, no. 35, 3 September 2008 (2008-09-03), pages 11576 - 11577, XP002723745, ISSN: 0002-7863, [retrieved on 20080807], DOI: 10.1021/JA8030467
MILLER ET AL., J. BIOL. CHERN., vol. 283, 2008, pages 1985
KAWAR ET AL., J. BIOL. CHERN., vol. 277, 2002, pages 34924
HOSKINS ET AL., SCIENCE, vol. 316, 2007, pages 1625
VADAIE ET AL., J. BIOL. CHEM., vol. 279, 2004, pages 33501
RAMAKRISHNAN ET AL., J. BIOL. CHEM., vol. 23, 2002, pages 20833
KHIDEKEL ET AL., J. AM. CHEM. SOC., vol. 125, 2003, pages 16162
CLARK ET AL., J. AM. CHEM. SOC., vol. 130, 2008, pages 11576
RAMAKRISHNAN ET AL., BIOCHEMISTRY, vol. 43, 2004, pages 12513
MERCER ET AL., BIOCONJ. CHEM., vol. 24, 2013, pages 144
BERTOZZI ET AL., ACS CHEM. BIOL., vol. 4, 2009, pages 1068
BURNHAM-MARUSICH ET AL., PLOS ONE, vol. 7, 2012, pages E49020
KAWAR ET AL., J. BIOL. CHEM., vol. 277, 2002, pages 34924
TARON ET AL., CARBOHYDR. RES., vol. 362, 2012, pages 62
"Computational Molecular Biology", 1988, OXFORD UNIVERSITY PRESS
"Biocomputing: Informatics and Genome Projects", 1993, ACADEMIC PRESS
"Computer Analysis of Sequence Data", 1994, HUMANA PRESS
HEINE, G.: "Sequence Analysis in Molecular Biology", 1987, ACADEMIC PRESS
"Sequence Analysis Primer, Gribskov", 1991, M STOCKTON PRESS
CARILLO, H.; LIPMAN, D., SIAM J. APPLIED MATH., vol. 48, 1988, pages 1073
DEVEREUX, J. ET AL., NUCLEIC ACIDS RESEARCH, vol. 12, no. 1, 1984, pages 387
ALTSCHUL, S. F. ET AL., J. MOL. BIOL., vol. 215, 1990, pages 403 - 410
ALTSCHUL, S. ET AL.: "BLAST Manual", NCBI NLM NIH BETHESDA
ALTSCHUL, S. ET AL., J. MOL. BIOL., vol. 215, 1990, pages 403 - 410
NEEDLEMAN; WUNSCH, J. MOL. BIOL., vol. 48, 1970, pages 443 - 453
HENTIKOFF; HENTIKOFF: "Comparison matrix: BLOSSUM62", PROC. NATL. ACAD. SCI. USA., vol. 89, 1992, pages 10915 - 10919
WONG ET AL., CHEM. REV., vol. 111, 2011, pages 4259
TARENTINO ET AL., GLYCOBIOLOGY, vol. 5, 1995, pages 599
COLLIN ET AL., EMBO J., vol. 20, 2001, pages 3046
KANDA ET AL., GLYCOBIOLOGY, vol. 17, 2006, pages 104 - 118
LINHARDT ET AL., J. ORG. CHEM., vol. 77, 2012, pages 1449 - 1456
BAISCH ET AL., BIOORG. MED. CHEM., vol. 5, 1997, pages 383 - 391
KISO ET AL., GLYCOCONJ. J., vol. 23, 2006, pages 565
RADEMANN ET AL., ANGEW. CHEM. INT. ED., vol. 51, 2012, pages 9441 - 9447
WU ET AL., GLYCOBIOLOGY, vol. 21, 2010, pages 723 - 733
R&D SYSTEMS, 31 July 2014 (2014-07-31), Retrieved from the Internet
POUILLY ET AL., ACS CHEM. BIOL., vol. 7, 2012, pages 753
GUAN ET AL., CHEM. EUR. J., vol. 16, 2010, pages 13343
HOSZTAFI ET AL., HELV. CHIM. ACTA,, vol. 79, 1996, pages 133 - 136
QASBA ET AL., J. BIOL. CHEM., vol. 277, 2002, pages 20833 - 20839
QASBA ET AL., PROT. EXPR. PUR., vol. 30, 2003, pages 219 - 76229
Attorney, Agent or Firm:
NEDERLANDSCH OCTROOIBUREAU (2502 LS The Hague, NL)
Download PDF:
Claims:
Claims

1. Process for the modification of a glycoprotein, the process comprising the step of contacting a glycoprotein comprising a glycan comprising a terminal GlcNAc moiety, with a sugar-derivative nucleotide Su(A)-Nuc, in the presence of a β-

(l,4)-N-acetylgalactosaminyltransf erase or a mutant thereof, wherein:

(i) the glycan comprising a terminal GlcNAc-moiety is according to formula (l) or (2):

1 2 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; and

(ϋ) the sugar-derivative nucleotide Su(A)-Nuc is according to formula (3):

wherein: a is 0 or 1 ;

Nuc is a nucleotide;

U is [C(R1)2]„ or [C(R1)2]p-0-[C(R1)2C(R1)20]o-[C(R1)2]q, wherein n is an integer in the range of 0 to 24; o is an integer in the range of 0 to 12; q and p are independently 0, 1 or 2; and R1 is independently selected from the group consisting of H, F, CI, Br, I and an optionally substituted Ci - C24 alkyl group;

T is a C3 - Ci2 (hetero)arylene group, wherein the (hetero)arylene group is optionally substituted; and

A is selected from the group consisting of:

(a) -N3

(b) -C(0)R3

wherein R3 is an optionally substituted Ci - C24 alkyl group;

(c) -C≡C-R4

wherein R4 is hydrogen or an optionally substituted Ci - C24 alkyl group;

(d) -SH

(e) -SC(0)R8

wherein R8 is an optionally substituted Ci - C24 alkyl group;

(f) -SC(V)OR8

wherein V is O or S, and R8 is an optionally substituted Ci - C24 alkyl group;

(g) -x

wherein X is selected from the group consisting of F, CI, Br and I;

(h) -OS(0)2R5

wherein R5 is selected from the group consisting of Ci - C24 alkyl groups, C6 - C24 aryl groups, C7 - C24 alkylaryl groups and C7 - C24 arylalkyl groups, the alkyl groups, aryl groups, alkylaryl groups and arylalkyl groups being optionally substituted;

(i) R11

wherein R11 is an optionally substituted C2 - C24 alkyl group; wherein R is an optionally substituted terminal C2 - C24 alkenyl group; and

(k) R13

wherein R13 is an optionally substituted terminal C3 - C24 allenyl group.

Process according to claim 1, wherein when U is [C(R1)2]n, n is an integer in the range of 1 to 24, and when U is [C(R1)2]p-0-[C(R1)2C(R1)20]0-[C(R1)2]q, o is an integer in the range of 1 to 12 and/or p is 1 or 2 and/or q is 1 or 2.

Process according to claim 1, wherein U is absent.

Process according to any one of the preceding claims, wherein the derivative nucleotide Su(A)-Nuc is according to formula (9) or (10):

10 wherein Nuc, A, U and T are as defined in claim 1. Process according to any one of the preceding claims, wherein T, if present, is selected from the group consisting of phenylene groups, pyridinylene groups, pyridiniumylene groups, pyrimidinylene groups, pyrimidiniumylene groups, pyrazinylene groups, pyradizinylene groups, pyrrolylene groups, pyrroliumylene groups, furanylene groups, thiophenylene groups, diazolylene groups, quinolinylene groups, imidazolylene groups, oxazolylene groups and oxazoliumylene groups, wherein said (hetero)arylene groups are optionally substituted.

Process according to any one of the preceding claims, wherein the sugar- derivative nucleotide is according to formula (11), (12), (13), (14), (15), (16), (34) or (35):

14 15 16

34 35

wherein:

A is as defined in claim 1; and

m is an integer in the range of 0 to 4;

R2 is independently selected from the group consisting of -F, -CI, -Br, -CN, -N02,

-C(0)R9, -C(0)OR9, -C(O)N(R10)2, Ci - C4 alkyl groups and Ci - C4 alkoxy groups, wherein R9 is a Ci - C12 alkyl group, and wherein R10 is independently selected from hydrogen and a Ci - C12 alkyl group; and

R6 is selected from the group consisting of H and optionally substituted Ci - C24 alkyl groups.

Process according to any one of the preceding claims, wherein the nucleotide UDP.

20 21 22 wherein:

X is F, CI, Br or I;

R8 is an optionally substituted Ci - C24 alkyl

V is O or S;

r is 0 or 1 ;

s is an integer in the range of 1 to 10;

t is an integer in the range of 1 to 10; and

u is an integer in the range of 0 to 10.

Process according to any one of the preceding claims, wherein the derivative nucleotide is according to formula (23), (24) or (25):

23 wherein X is F, CI, Br or I.

Process according to claim 8 or claim 9, wherein the sugar-derivative nucleotide is according to formula (17), (18), (19), (23) or (24), wherein (17), (18) and (19) are as defined in claim 8, wherein (23) and (24) are as defined in claim 9, and wherein X is CI or F.

11. Process according to any one of the preceding claims, wherein the glycan comprising a terminal GlcNAc moiety is according to formula (1), (26) or (27):

(Fuc)t

Man— GlcNAc

a— GlcNAc GlcNAc— Man /

Man— GlcNAc

27

b is as defined in claim 1

Process according to any one of the preceding claims, wherein the glycoprotein comprising a glycan comprising a terminal GlcNAc moiety is according to formula (7) or (8):

Pr-F-

7 wherein: b, d, e and G are as defined in claim 1;

y is an integer in the range of 1 to 24; and

Pr is a protein.

13. Process according to any one of the preceding claims, wherein the glycoprotein comprising a glycan comprising a terminal GlcNAc moiety is an antibody.

14. Process according to any one of the preceding claims, wherein the β-(1,4)-Ν- acetylgalactosaminyltransf erase is or is derived from an invertebrate β-(1,4)- GalNAcT enzyme.

15. Process according to any one of the preceding claims, wherein the β-(1,4)-Ν- acetylgalactosaminyltransferase is or is derived from a sequence selected from the group consisting of SEQ ID NO: 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22 and 23, or from the group consisting of SEQ ID NO: 25,

26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 71, 72 and 73.

16. Process according to any one of the preceding claims, wherein the β-(1,4)-Ν- acetylgalactosaminyltransferase has at least 50% identity to a sequence selected from the group consisting of SEQ ID NO: 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22 and 23, or from the group consisting of SEQ ID NO: 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 71, 72 and 73.

Description:
Process for the modification of a glycoprotein using a β-(1,4)-Ν- acetylgalactosaminyltransferase or a mutant thereof

Technical field of the invention

The present invention relates to a process for the enzymatic modification of a glycoprotein. More in particular, the invention relates to a process for the modification of a glycoprotein with a sugar-derivative nucleotide, using a β-(1,4)-Ν- acetylgalactosaminyltransferase, or a mutant thereof, and to the β-(1,4)-Ν- acetylgalactosaminyltransferase mutants.

Background of the invention

Glycosyltransferases constitute a superfamily of enzymes that are involved in the synthesis of complex carbohydrates present on glycoproteins and glycolipids. The fundamental role of a glycosyltransferase is to transfer the glycosyl moiety of a nucleotide derivative to a specific sugar acceptor. -l,4-Galactosyltransf erases ( 4Gal- Ts) (EC 2.4.1.38) constitute one of the subfamilies of glycosyltransferase superfamily - comprising at least seven members Gal-Tl to Gal-T7- which catalyze the transfer of galactose (Gal) from UDP-Gal to different sugar acceptors. A common motif resulting from a galactose transferase onto a terminal GlcNAc residue is the lactosamine sequence Gai 4GlcNAc-R (LacNAc or LN), which is subsequently modified in a variety of ways by the additions of other sugars and sulfate groups. The most common and important sugar structure of membrane glycoconjugates is poly-N- acetyllactosamine (poly-LN), which linked to proteins (or lipids), plays an important role in cellular communication, adhesion, and signalling and are key molecules in regulation of immune responses.

Another common terminal motif found in vertebrate and invertebrate glycoconjugates is the GalNAc 4GlcNAc-R (LacdiNAc or LDN) sequence. The LDN motif occurs in mammalian pituitary glycoprotein hormones, where the terminal GalNAc residues are 4-O-sulfated and function as recognition markers for clearance by the endothelial cell Man/S4GGnM receptor. However, non-pituitary mammalian glycoproteins also contain LDN determinants. In addition, LDN and modifications of LDN sequences are common antigenic determinants in many parasitic nematodes and trematodes. The biosynthesis of LDN involves the transfer of GalNAc to a terminal GlcNAc, a process executed by highly specific GalNAc-transf erases. For example it was reported by Miller et al. in J. Biol. Chem. 2008, 283, p. 1985, incorporated by reference, that two closely related i,4-N-acetylgalactosaminyltransf erases, 4GalNAc-T3 and 4GalNAc-T4, are thought to account for the protein-specific addition of i,4-linked GalNAc to Asn-linked oligosaccharides on a number of glycoproteins including the glycoprotein luteinizing hormone (LH) and carbonic anhydrase-6 (CA6).

β-(1, 4)- Acetylgalactosaminyltransf erases ( -(l,4)-GalNAcTs) have been identified in a range of organisms, including humans, Caenorhabditis elegans (Kawar et al, J. Biol. Chem. 2002, 277, 34924, incorporated by reference), Drosophila melanogaster (Hoskins et al. Science 2007, 316, 1625, incorporated by reference) and Trichoplusia ni (Vadaie et al, J. Biol. Chem. 2004, 279, 33501, incorporated by reference).

Finally, besides GalTs and GalNAcTs involved in N-glycoprotein modification, a non-related class of enzymes called UDP-N-acetylgalactosamine:polypeptide N- acetylgalactosaminyltransferases (also referred to as ppGalNAcTs) is responsible for the biosynthesis of mucin-type linkages (GalNAc-a-l-O-Ser/Thr). These enzymes transfer GalNAc from the sugar donor UDP-GalNAc to serine and threonine residues, forming an alpha anomeric linkage typical in O-glycoproteins. Despite the seeming simplicity of ppGalNAcTs catalytic function, it is estimated on the basis of in silico analysis that there are 24 unique ppGalNAcTs human genes alone. Because O-linked glycosylation proceeds step-wise, addition of GalNAc to serine or threonine represents the first committed step in mucin biosynthesis. Despite this seeming simplicity, multiple ppGalNAcTs family members appear to be necessary to fully glycosylate their protein substrates.

It has been shown that P-l,4-galactosyltransf erase 1 (P4Gal-Tl) is able to transfer, besides its natural substrate UDP-Gal, a range of unnatural galactose derivatives to an acceptor GlcNAc substrate. In particular the mutation of the Tyr289 residue to Leu289 in bovine P4Gal-Tl, as reported by Ramakrishnan et al. J. Biol. Chem. 2002, 23, 20833, incorporated by reference, creates a cavity in the catalytic pocket of the enzyme that can accommodate a UDP-Gal molecule carrying a chemical handle at C2, such as 2-keto-Gal. By a two-step procedure involving first transfer of the unnatural galactose moiety followed by oxime ligation onto the C-2 handle, this mutant enzyme, P4GalT(Y289L), has been used for in vitro detection of O-GlcNAc residues on proteins or the presence of a terminal GlcNAc moiety on the cell surface glycans of normal and malignant tumor tissues.

For example Khidekel et al, J. Am. Chem. Soc. 2003, 125, 16162, incorporated by reference, discloses chemoselective installation of an unnatural ketone functionality to O-GlcNAc modified proteins with p4GalT(Y289L). The ketone moiety serves as a unique marker to "tag" O-GlcNAc glycosylated proteins with biotin using oxime ligation. Once biotinylated, the glycoconjugates can be readily detected by chemiluminescence using streptavidin conjugated to horseradish peroxidase (HRP).

For example WO 2007/095506, WO 2008/029281 (both Invitrogen Corporation),

WO 2014/065661 (SynAffix B.V.) and Clark et al. J. Am. Chem. Soc. 2008, 130, 11576, all incorporated by reference, report a similar approach, using p4GalT(Y289L) and azidoacetyl variants of galactosamine, with similar success.

Mutant p4GalT(Y289L) has also been applied most recently in a preparative fashion for the site-selective radiolabeling of antibodies on the heavy chain glycans, as reported by Zeglis et al. in Bioconj. Chem. 2013, 24, 1057, incorporated by reference. In particular, the incorporation of azide-modified N-acetylgalactosamine monosaccharides (GalNAz) into the glycans of the antibody allowed the controlled labeling with 89 Zr upon after click chemistry introduction of the appropriate chelator.

Ramakrishnan et al. in Biochemistry 2004, 43, 12513, incorporated by reference, describe that the double mutant p4GalT(Y289L,M344H) loses 98% of its Mn 2+ - dependent activity, but nevertheless shows 25-30% activity in the presence of Mg 2+ , including a capability to transfer C-2 modified galactose substrates. The double mutant P4GalT(Y289L,M344H) was found useful for in vitro galactosylation assays, because the typical requirement of 5-10 mM Mn 2+ is known to have potential cytotoxic effects for the cells.

Mercer et al, Bioconj. Chem. 2013, 24, 144, incorporated by reference, describe that a double mutant Y289L-M344H- 4Gal-Tl enzyme transfers GalNAc and analogue sugars to the acceptor GlcNAc in the presence of Mg 2+ .

Attempts to employ a wild-type -(l,4)-N-acetylgalactosaminyltransferase, herein also referred to as -(l,4)-GalNAcT, for the transfer of C-2 modified GalNAc have met little success to date. Bertozzi et al. in ACS Chem. Biol. 2009, 4, 1068, incorporated by reference herein, applied the bioorthogonal chemical reporter technique for the molecular imaging of mucin-type O-glycans in live C. elegans. Worms were treated with the azido-sugar variant of N-acetylgalactosamine (GalNAz) allowing the in vivo incorporation of this unnatural sugar. Although metabolic incorporation of GalNAz into glycoproteins was observed, both chondroitinase ABC and peptide N-glycosidase F (PNGase F) digestion of C. elegans lysate, followed by the Staudinger ligation using a phosphine-Flag tag and subsequent probing of the glycoproteins by Western blotting utilizing an a-Flag antibody, indicated that the majority of GalNAz residues on glycoproteins were situated in other types of glycans than N-glycans. In addition, no detectable binding of azide-labeled glycoproteins to the N-glycan specific lectin concanavalin A (ConA) was observed, consistent with the hypothesis that a majority of labelled glycans are O-linked and not N-linked. Based on these observations, one may conclude that GalNAz does not metabolically incorporate onto N-GlcNAcylated proteins in this organism.

A similar conclusion was drawn most recently by Burnham-Marusich et al. in Plos One 2012, 7, e49020, incorporated by reference herein, where lack of signal reduction upon PNGAse treatment -indicating no apparent incorporation of GalNAz in N-glycoproteins- was also observed. Burnham-Marusich et al. describe a study using the Cu(I)-catalyzed azide-alkyne cycloaddition reaction of a terminal alkyne-probe with an azido-labeled glycoprotein to detect metabolically labelled glycoproteins. Results indicated that the majority of the GalNAz label is incorporated into glycan classes that are insensitive to pNGase F, hence are not N-glycoproteins.

High substrate specificity of a -(l,4)-GalNAcT for UDP-GalNAc becomes apparent from the poor recognition of UDP-GlcNAc, UDP-Glc and UDP-Gal, for which only 0.7%, 0.2% and 1% transferase activity remains, respectively, as was reported by Kawar et al, J. Biol. Chem. 2002, 277, 34924, incorporated by reference.

Based on the above, it is not surprising that no in vitro method for modification of glycoproteins has been reported by means of GalNAc-transferase of an unnatural GalNAc derivative such as a 2-keto or 2-azidoacetyl derivative.

Taron et al., Carbohydr. Res. 2012, 362, 62, incorporated by reference, describe the in vivo metabolic incorporation of GalNAz in GPI-anchors. Summary of the invention

The invention relates to a process for the modification of a glycoprotein, the process comprising the step of contacting a glycoprotein comprising a glycan comprising a terminal GlcNAc moiety, with a sugar-derivative nucleotide Su(A)-Nuc, in the presence of a -(l,4)-N-acetylgalactosaminyltransferase or a mutant thereof, wherein:

(i) the glycan comprising a terminal GlcNAc-moiety is according to formula (1) or (2):

;—

1 2 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; and

(ii) the sugar-derivative nucleotide Su(A)-Nuc is according to formula (3):

3 wherein:

a is 0 or 1 ; Nuc is a nucleotide;

U is [C(R 1 ) 2 ]„ or [C(R 1 ) 2 ]p-0-[C(R 1 )2C(R 1 ) 2 0]o-[C(R 1 )2] q , wherein n is an integer in the range of 0 to 24; o is an integer in the range of 0 to 12; p and q are independently 0, 1 or 2; and R 1 is independently selected from the group consisting of H, F, CI, Br, I and an optionally substituted Ci - C 24 alkyl group;

T is a C 3 - Ci 2 (hetero)arylene group, wherein the (hetero)arylene group is optionally substituted; and

A is selected from the group consisting of:

(a) -N 3

(b) -C(0)R 3

wherein R 3 is an optionally substituted Ci - C 24 alkyl group; (c) -C≡C-R 4

wherein R 4 is hydrogen or an optionally substituted Ci - C 24 alkyl group;

(d) -SH

(e) -SC(0)R 8

wherein R 8 is an optionally substituted Ci - C 24 alkyl group;

(f) -SC(V)OR 8

wherein V is O or S, and R 8 is an optionally substituted Ci - C 24 alkyl group;

(g) -x

wherein X is selected from the group consisting of F, CI, Br and I;

(h) -OS(0) 2 R 5

wherein R 5 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, the alkyl groups, aryl groups, alkylaryl groups and arylalkyl groups being optionally substituted;

(i) R 11

wherein R 11 is an optionally substituted C 2 - C 24 alkyl group.

G) R 12

wherein R 12 is an optionally substituted terminal C 2 - C 24 alkenyl group; and

(k) R 13 wherein R is an optionally substituted terminal C 3 - C24 allenyl group.

In a further aspect, the invention relates to the β-(1,4)-Ν- acetylgalactosaminyltransferase mutants that are suitable to be used in the process according to the invention.

Description of the figures

In Figure 1 examples of a glycoprotein comprising a glycan comprising a terminal GlcNAc moiety, that may be modified by the process according to the invention, are shown.

In Figure 2, an embodiment of the process for the modification of a glycoprotein, wherein the glycoprotein is an antibody, is shown. In this embodiment a sugar- derivative Su(A)-Nuc is attached to the terminal GlcNAc-moiety of an antibody glycan under the action a -(l,4)-N-acetylgalactosaminyltransferase to form a modified antibody.

Figure 3 shows different glycoforms of antibody glycans GO, Gl, G2, GOF, GIF and G2F.

Figure 4 shows a process for providing a glycoprotein comprising a glycan according to formula (27) by treatment of a mixture of glycoforms GO, Gl, G2, GOF, GIF and G2F with sialidase and galactosidase, and a process for providing a glycoprotein comprising a glycan according to formula (1) by treatment of a mixture of glycoforms GO, Gl, G2, GOF, GIF and G2F with an endoglycosidase. Incubation of the glycoproteins comprising a glycan according to formula (27) or (1) with an azido- modified UDP-GalNAc derivative, UDP-GalNAz, leads to an azido-modified glycoprotein (33) or (32), respectively.

Figure 5 shows the SDS-PAGE of a range of -(l,4)-GalNAc-Ts, crude after transient expression in CHO.

Figure 6 shows the non-reducing SDS-PAGE of a range of -(l,4)-CeGalNAc-T mutants.

Figure 7 shows the activity plot of range of different -(l,4)-GalNAcTs in comparison with -(l,4)-GalT(Y289L) mutant for transfer of UDP-F 2 -GalNAz to GlcNAc, as determined by R&D systems glycosyltransferase activity kit. Figure 8 shows the activity plot of range of different -(l,4)-CeGalNAcT mutants Y257L, Y257M and Y257A for transfer of UDP-F 2 -GalNAz to GlcNAc, as determined by R&D systems glycosyltransferase activity kit. 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 element 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".

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. 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 C 2 ) 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 C 3 ) and may include monocyclic and bicyclic structures. Optionally, the heteroaiyl 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 heteroaiyl 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 C 2 - C 2 4 (hetero)aryl group is thus to be interpreted as including a C 2 - C 24 heteroaiyl group and a C 6 - C 24 aryl group. Similarly, 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, and 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.

Unless stated otherwise, alkyl groups, alkenyl groups, alkenes, alkynes, (hetero)aryl groups, (hetero)arylalkyl groups, alkyl(hetero)aryl groups, alkylene groups, alkenylene groups, cycloalkylene groups, (hetero)arylene groups, alkyl(hetero)arylene groups, (hetero)arylalkylene groups, alkenyl groups, alkynyl groups, cycloalkyl groups, alkoxy groups, alkenyloxy groups, (hetero)aryloxy groups, alkynyloxy groups and cycloalkyloxy groups may be 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, 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 2 ) 3 Si-, wherein R 2 is 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 and C 3 - C 12 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 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. 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 C n H 2n-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 alkylheteroaryl 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. annulated, 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 annulated 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. 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 (GlcNH 2 ), galactosamine (GalNH 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). 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-l 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 exist. Naturally occurring O- linked glycans typically feature a serine or threonine-linked α-0-GalNAc moiety, further substituted with another GalNAc, galactose, GlcNAc, sialic acid and/or fucose, preferably with galactose, GlcNAc, 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 exist. 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 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.

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, F(ab' )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. Identity/ similarity

In the context of the invention, a protein or a protein fragment is represented by an amino acid sequence.

It is to be understood that each protein or protein fragment or peptide or derived peptide or polypeptide as identified herein by a given Sequence Identity Number (SEQ ID NO) is not limited to this specific sequence as disclosed. "Sequence identity" is herein defined as a relationship between two or more amino acid (polypeptide or protein) sequences, as determined by comparing the sequences. In the art, "identity" also means the degree of sequence similarity between amino acid sequences, as the case may be, as determined by the match between strings of such sequences. Unless otherwise indicated herein, identity or similarity with a given SEQ ID NO means identity or similarity based on the full length of said sequence (i.e. over its whole length or as a whole).

"Similarity" between two amino acid sequences is determined by comparing the amino acid sequence and its conserved amino acid substitutes of one polypeptide to the sequence of a second polypeptide. "Identity" and "similarity" can be readily calculated by known methods, including but not limited to those described in (Computational Molecular Biology, Lesk, A. M., ed., Oxford University Press, New York, 1988; Biocomputing: Informatics and Genome Projects, Smith, D. W., ed., Academic Press, New York, 1993; Computer Analysis of Sequence Data, Part I, Griffin, A. M., and Griffin, H. G., eds., Humana Press, New Jersey, 1994; Sequence Analysis in Molecular Biology, von Heine, G., Academic Press, 1987; and Sequence Analysis Primer, Gribskov, M. and Devereux, J., eds., M Stockton Press, New York, 1991; and Carillo, H., and Lipman, D., SIAM J. Applied Math., 48: 1073 (1988).

Preferred methods to determine identity are designed to give the largest match between two or more sequences tested. Methods to determine identity and similarity are codified in publicly available computer programs. Preferred computer program methods to determine identity and similarity between two sequences include e.g. the GCG program package (Devereux, J., et al., Nucleic Acids Research 12 (1): 387 (1984)), BestFit, BLASTP, BLASTN, and FASTA (Altschul, S. F. et al, J. Mol. Biol. 215:403-410 (1990). The BLAST X program is publicly available from NCBI and other sources (BLAST Manual, Altschul, S., et al, NCBI NLM NIH Bethesda, MD 20894; Altschul, S., et al, J. Mol. Biol. 215:403-410 (1990). The well-known Smith Waterman algorithm may also be used to determine identity.

Preferred parameters for polypeptide sequence comparison include the following: Algorithm: Needleman and Wunsch, J. Mol. Biol. 48:443-453 (1970); Comparison matrix: BLOSSUM62 from Hentikoff and Hentikoff, Proc. Natl. Acad. Sci. USA. 89: 10915-10919 (1992); Gap Penalty: 12; and Gap Length Penalty: 4. A program useful with these parameters is publicly available as the "Ogap" program from Genetics Computer Group, located in Madison, WI. The aforementioned parameters are the default parameters for amino acid comparisons (along with no penalty for end gaps). Optionally, in determining the degree of amino acid similarity, the skilled person may also take into account so-called "conservative" amino acid substitutions, as will be clear to the skilled person. Conservative amino acid substitutions refer to the interchangeability of residues having similar side chains. For example, a group of amino acids having aliphatic side chains is glycine, alanine, valine, leucine, and isoleucine; a group of amino acids having aliphatic-hydroxyl side chains is serine and threonine; a group of amino acids having amide-containing side chains is asparagine and glutamine; a group of amino acids having aromatic side chains is phenylalanine, tyrosine, and tryptophan; a group of amino acids having basic side chains is lysine, arginine, and histidine; and a group of amino acids having sulphur-containing side chains is cysteine and methionine. Preferred conservative amino acids substitution groups are: valine-leucine-isoleucine, phenylalanine-tyrosine, lysine-arginine, alanine- valine, and asparagine-glutamine. Substitutional variants of the amino acid sequence disclosed herein are those in which at least one residue in the disclosed sequences has been removed and a different residue inserted in its place. Preferably, the amino acid change is conservative. Preferred conservative substitutions for each of the naturally occurring amino acids are as follows: Ala to Ser; Arg to Lys; Asn to Gin or His; Asp to Glu; Cys to Ser or Ala; Gin to Asn; Glu to Asp; Gly to Pro; His to Asn or Gin; He to Leu or Val; Leu to He or Val; Lys to Arg; Gin to Glu; Met to Leu or He; Phe to Met, Leu or Tyr; Ser to Thr; Thr to Ser; Trp to Tyr or His; Tyr to Trp or Phe; and, Val to He or Leu.

Process for the modification of a glycoprotein

The present invention relates to an in vitro process for the modification of a glycoprotein, in order to obtain a modified glycoprotein, said proces using a β-(1,4)-Ν- acetylgalactosaminyltransferase. Preferably, the process is an in vitro process. In particular, the invention relates to a process for the modification of a glycoprotein, the process comprising the step of contacting a glycoprotein comprising a glycan comprising a terminal GlcNAc moiety, with a sugar-derivative nucleotide Su(A)-Nuc, in the presence of, more particularly under the action of, a β-(1,4)-Ν- acetylgalactosaminyltransf erase or a mutant thereof, wherein:

(i) the glycan comprising a terminal GlcNAc-moiety is according to formula (1) or (2):

.—

1 2 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 compri to 20 sugar moieties; and

the sugar-derivative nucleotide Su(A)-Nuc is according to formula (3):

wherein:

a is 0 or 1 ;

Nuc is a nucleotide;

U is [C(R 1 ) 2 ]„ or [C(R 1 ) 2 ]p-0-[C(R 1 )2C(R 1 ) 2 0]o-[C(R 1 )2] q , wherein n is an integer in the range of 0 to 24; o is an integer in the range of 0 to 12; p and q are independently 0, 1 or 2; and R 1 is independently selected from the group consisting of H, F, CI, Br, I and an optionally substituted Ci - C 24 alkyl group; T is a C 3 - Ci 2 (hetero)arylene group, wherein the (hetero)arylene group is optionally substituted; and

A is selected from the group consisting of:

(a) -N 3

(b) -C(0)R 3

wherein R 3 is an optionally substituted Ci - C 24 alkyl group;

(c) -C≡C-R 4

wherein R 4 is hydrogen or an optionally substituted Ci - C 24 alkyl group;

(d) -SH

(e) -SC(0)R 8

wherein R 8 is an optionally substituted Ci - C 24 alkyl group;

(f) -SC(V)OR 8 wherein V is O or S and R is an optionally substituted Ci - C24 alkyl group;

(g) -x

wherein X is selected from the group consisting of F, CI, Br and I; (h) -OS(0) 2 R 5

wherein R 5 is selected from the group consisting of Ci - C24 alkyl groups, C 6 - C24 aryl groups, C 7 - C24 alkylaryl groups and C 7 - C24 arylalkyl groups, the alkyl groups, aryl groups, alkylaryl groups and arylalkyl groups being optionally substituted;

(i) R 11

wherein R 11 is wherein R 11 is an optionally substituted C2 - C24 alkyl group;

12

0) R

wherein R is an optionally substituted terminal C2 - C24 alkenyl group; and

(k) R 13 wherein R 13 is an optionally substituted terminal C3 - C24 allenyl group. As described above, the process according to the invention for the modification of a glycoprotein provides a modified glycoprotein. A modified glycoprotein is herein defined as a glycoprotein comprising a glycan according to formula (4) or (5):

-—

4 5

wherein:

b, d, e and G are as defined above; and

Su(A) is a sugar-derivative according to formula (6): wherein:

a, U, A and T are as defined above. In the modified glycoprotein glycan according to formula (4) and (5), CI of sugar-derivative Su(A) is attached to C4 of the GlcNAc moiety via a β- 1,4-0- glycosidic bond.

The process for the modification of a glycoprotein may further comprise the step of providing a glycoprotein comprising a glycan comprising a terminal GlcNAc- moiety. The invention therefore also relates to a process for the modification of a glycoprotein comprising the steps of:

(1) providing a glycoprotein comprising a glycan comprising a terminal GlcNAc moiety, wherein the glycan comprising a terminal GlcNAc-moiety is according to formula (1) or (2) as defined above; and

(2) contacting said glycoprotein with a sugar-derivative nucleotide Su(A)-Nuc, in the presence of a -(l,4)-N-acetylgalactosaminyltransf erase or a mutant thereof, wherein Su(A)-Nuc is according to formula (3) as defined above.

The -(l,4)-N-acetylgalactosaminyltransferase, the glycoprotein comprising a glycan comprising a terminal GlcNAc moiety, the sugar-derivative nucleotide Su(A)- Nuc and the modified glycoprotein, and preferred embodiments thereof, are described in more detail below. Enzyme

The process according to the invention comprises the step of contacting a glycoprotein comprising a glycan comprising a terminal GlcNAc moiety with a sugar- derivative nucleotide Su(A)-Nuc in the presence of, more particularly under the action of, a -(l,4)-N-acetylgalactosaminyltransferase, or a mutant thereof. In a second aspect, the invention concerns mutants of -(l,4)-N-acetylgalactosaminyltransferase as described herein, which are specifically designed for performing the process according to the invention. Mutants of -(l,4)-N-acetylgalactosaminyltransf erase are derived from naturally occurring β-(1,4)- N-acetylgalactosaminyltransf erase. A β-(1,4)-Ν- acetylgalactosaminyltransferase is herein also referred to as a -(l,4)-GalNAcT enzyme, or -(l,4)-GalNAcT, or GalNAcT. The term "a -(l,4)-N-acetyl- galactosaminyltransf erase, or a mutant thereof refers to a glycosyltransferase that is, or is derived from, a -(l,4)-N-acetylgalactosaminyltransf erase.

-(l,4)-N-Acetylgalactosaminyltransferases ( -(l,4)-GalNAcTs) are known in the art. Typically, a -(l,4)-GalNAcT is an enzyme that catalyzes the transfer of N- acetylgalactosamine (GalNAc) from uridine diphosphate-GalNAc (UDP-GalNAc, also referred to as GalNAc-UDP) to a terminal GlcNAc moiety of a glycoprotein glycan, wherein CI of the GalNAc moiety is attached to C4 of the GlcNAc moiety via a β-1,4- O-glycosidic bond. As described in more detail below, the GlcNAc moiety in a glycan according to formula (1) wherein b is 1, i.e. the GlcNAc moiety in a glycan consisting of a fucosylated GlcNAc, is herein also considered a terminal GlcNAc moiety.

In the process according to the invention, the -(l,4)-GalNAcT enzyme, or mutant thereof, catalyzes the transfer of sugar-derivative Su(A) from a sugar-derivative nucleotide Su(A)-Nuc to a terminal GlcNAc moiety of a glycoprotein glycan, wherein Su(A) is according to formula (6), Su(A)-Nuc is according to formula (3) and the glycan comprising a terminal GlcNAc-moiety is according to formula (1) or (2), as described above. In this process, CI of the Su(A) moiety is attached to C4 of the GlcNAc moiety via a -l,4-0-glycosidic bond.

Preferably, the -(l,4)-GalNAcT enzyme used in the process of the invention is or is derived from an invertebrate -(l,4)-GalNAcT enzyme, i.e. is or is derived from a -(l,4)-GalNAcT that originates from invertebrate animal species. The β-(1,4)- GalNAcT enzyme can be or can be derived from any invertebrate -(l,4)-GalNAcT enzyme known by a person skilled in the art. Preferably, the -(l,4)-GalNAcT enzyme is or is derived from a -(l,4)-GalNAcT enzyme that originates from the phylum of Nematoda, preferably of the class of Chromadorea or Secernentea, or of the phylum of Arthropoda, preferably of the class of Insecta. Preferably, the -(l,4)-GalNAcT enzyme is or is derived from a -(l,4)-GalNAcT enzyme that originates from Caenorhabditis elegans, Caenorhabditis remanei, Caenorhabditis briggsae, Ascaris suum, Trichoplusia ni, Drosophila melanogaster, Wuchereria bancrofti, Loa loa, Cerapachys biroi, Zootermopsis nevadensis, Camponotus floridanus, Crassostrea gigas or Danaus plexippus, preferably from Caenorhabditis elegans, Ascaris suum, Trichoplusia ni or Drosophila melanogaster. More preferably, the -(l,4)-GalNAcT enzyme is, or is derived from, a -(l,4)-GalNAcT enzyme that originates from Caenorhabditis elegans, Ascaris suum or Trichoplusia ni. In a further preferred embodiment, the -(l,4)-GalNAcT enzyme is, or is derived from, a -(l,4)-GalNAcT enzyme that originates from Ascaris suum. In another further preferred embodiment, the -(l,4)-GalNAcT enzyme is, or is derived from, a -(l,4)-GalNAcT enzyme that originates from Trichoplusia ni. In another further preferred embodiment, the β-(1,4)- GalNAcT enzyme is, or is derived from, a -(l,4)-GalNAcT enzyme that originates from Caenorhabditis elegans.

Caenorhabditis elegans is herein also referred to as Ce, Ascaris suum as As, Trichoplusia ni as Tn and Drosophila melanogaster as Dm.

Preferably, the -(l,4)-GalNAcT enzyme used in the process of the invention has at least 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or preferably 100% sequence identity to a sequence selected from the group consisting of SEQ ID NO: 2-5 and 15-23, more preferably to a sequence selected from the group consisting of SEQ ID NO: 2-5, i.e. SEQ ID NO: 2, 3, 4 or 5.

Preferably, the -(l,4)-GalNAcT enzyme used in the process of the invention is or is derived from any of the naturally occurring or wild type -(l,4)-GalNAcT enzymes selected from the group consisting of Caenorhabditis elegans β-(1,4)- GalNAcT denominated herein as CeGalNAcT (SEQ ID NO: 2), Ascaris suum β-(1,4)- GalNAcT denominated herein as AsGalNAcT (SEQ ID NO: 3), Trichoplusia ni β- (l,4)-GalNAcT denominated herein as TnGalNAcT (SEQ ID NO: 4), Drosophila melanogaster -(l,4)-GalNAcT denominated herein as DmGalNAcT (SEQ ID NO: 5), Caenorhabditis remanei -(l,4)-GalNAcT (SEQ ID NO: 15), Caenorhabditis briggsae -(l,4)-GalNAcT (SEQ ID NO: 16), Wuchereria barter ofti -(l,4)-GalNAcT (SEQ ID NO: 17), Loa loa -(l,4)-GalNAcT (SEQ ID NO: 18), Cerapachys biroi β-(1,4)- GalNAcT (SEQ ID NO : 19), Zootermopsis nevadensis β-( 1 ,4)-GalNAcT (SEQ ID NO : 20), Camponotus floridanus -(l,4)-GalNAcT (SEQ ID NO: 21), Crassostrea gigas β- (l,4)-GalNAcT (SEQ ID NO: 22) and Danaus plexippus -(l,4)-GalNAcT (SEQ ID NO: 23).

Further preferred is a -(l,4)-GalNAcT enzyme that is or is derived from a β- (l,4)-GalNAcT enzyme that originates from an invertebrate species of the phylum Nematode, preferably of the class Chromadorea, preferably of the order Rhabditida, preferably of the family Rhabditidae, preferably of the genus Caenorhabditis. Preferably, the -(l,4)-GalNAcT enzyme used in the process of the invention has at least 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%. 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or preferably 100% sequence identity to a sequence of the group consisting of SEQ ID NO: 2, 15 and 16. Most preferably, said invertebrate species is of Caenorhabditis Elegans. Preferably, the -(l,4)-GalNAcT enzyme used in the process of the invention has at least 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or preferably 100% sequence identity to SEQ ID NO: 2.

In another preferred embodiment the -(l,4)-GalNAcT enzyme used in the process of the invention is a -(l,4)-GalNAcT enzyme that is or is derived from a β- (l,4)-GalNAcT enzyme that originates from an invertebrate species of the phylum Nematode, preferably of the class Secementea, preferably of the order Ascaridida, preferably of the family Ascarididae, preferably of the genus Ascaris. More preferably, said invertebrate species is of Ascaris Sum. Preferably, the -(l,4)-GalNAcT enzyme used in the process of the invention has at least 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%. 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or preferably 100% sequence identity to a sequence of the group consisting of SEQ ID NO: 3.

In another preferred embodiment the -(l,4)-GalNAcT enzyme used in the process of the invention is a -(l,4)-GalNAcT enzyme that is or is derived from a β- (l,4)-GalNAcT enzyme that originates from an invertebrate species of the phylum Anthropoda, preferably of the class Insecta, preferably of the order Lepidoptera, preferably of the family Noctuidae, preferably of the genus Trichoplusia. More preferably, said invertebrate species is of Trichoplusia Ni. Trichoplusia Ni may sometimes also be referred to as Phytometra brassicae, Plusia innata or cabbage looper. Preferably, the -(l,4)-GalNAcT enzyme used in the process of the invention has at least 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%. 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or preferably 100% sequence identity to a sequence of the group consisting of SEQ ID NO: 4.

In another preferred embodiment the -(l,4)-GalNAcT enzyme used in the process of the invention is a -(l,4)-GalNAcT enzyme that is or is derived from a β- (l,4)-GalNAcT enzyme that originates from an invertebrate species of the phylum Anthropoda, preferably of the class Insecta, preferably of the order Diptera, preferably of the family Drosophilidae, preferably of the genus Drosophila. More preferably, said invertebrate species is of Drosophila melanogaster. Preferably, the -(l,4)-GalNAcT enzyme used in the process of the invention has at least 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%. 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or preferably 100% sequence identity to a sequence of the group consisting of SEQ ID NO: 5.

"Derived from" is to be understood herein as having an amino acid sequence that is altered from a naturally occurring -(l,4)-GalNAcT enzyme by substituting, inserting, deleting, or adding one or more, preferably 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 14, 16, 18, 20 or more amino acids, respectively. A -(l,4)-GalNAcT enzyme that is derived from a -(l,4)-GalNAcT enzyme is herein also referred to as a derived β-(1,4)- GalNAcT enzyme or a modified -(l,4)-GalNAcT enzyme or a -(l,4)-GalNAcT mutant enzyme or a -(l,4)-GalNAcT mutant.

Preferably, said derived -(l,4)-GalNAcT enzyme is modified by adding additional N- or C- terminal amino acids or chemical moieties or by deleting N- or C- terminal amino acids to increase stability, solubility, activity and/or ease of purification.

Preferably the -(l,4)-GalNAcT enzyme is modified by deleting the N-terminal cytoplasmic domain and transmembrane domain, which is denominated herein as a truncated enzyme. For instance, CeGalNAcT (30-383) is to be understood herein as a truncated Caenorhabditis elegans -(l,4)-GalNAcT enzyme consisting of the amino acid sequence represented by the amino acids on position 30-383 of SEQ ID NO: 2. Deletion of these domains is known in the art to result in an enzyme that shows an increased solubility in aqueous solutions.

Similarly, AsGalNAcT(30-383) is to be understood herein as a truncated Ascaris Sum -(l,4)-GalNAcT enzyme consisting of the amino acid sequence represented by the amino acids on position 30-383 of SEQ ID NO: 3, TnGalNAcT(33-421) is to be understood herein as a truncated Trichoplusia Ni -(l,4)-GalNAcT enzyme consisting of the amino acid sequence represented by the amino acids on position 33-421 of SEQ ID NO: 4, and DmGalNAcT(47-403) is to be understood herein as a truncated Drosophila melanogaster -(l,4)-GalNAcT enzyme consisting of the amino acid sequence represented by the amino acids on position 47-403 of SEQ ID NO: 5. Preferably, the -(l,4)-GalNAcT enzyme used in the process of the invention has at least 40%, 45%, 50%, 55% 60%, 65%, 70%, 75% 80%, 85% 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or preferably at least 100% sequence identity to any of the sequences of SEQ ID NO 6 - 9. More preferably, the -(l,4)-GalNAcT enzyme used in the process of the invention has at least 40%, 45%, 50%, 55% 60%, 65%, 70%, 75% 80%, 85% 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or preferably at least 100% sequence identity to any of the sequences of SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8 or SEQ ID NO: 9, more preferably to any of the sequences of SEQ ID NO: 6, SEQ ID NO: 7 or SEQ ID NO: 8, even more preferably to any of the sequences of SEQ ID NO: 7 or SEQ ID NO: 8, and even more preferably to the sequence SEQ ID NO: 8.

A -(l,4)-GalNAcT enzyme wherein one or more amino acid has been substituted, added or deleted is herein also referred to as a mutant -(l,4)-GalNAcT enzyme or a derived -(l,4)-GalNAcT enzyme. Preferably, the -(l,4)-GalNAcT enzyme is modified by deleting the N-terminal cytoplasmic domain and transmembrane domain, and mutated by substituting one or more amino acids. A substitution of one or more amino acids is herein also referred to as a mutation. An enzyme comprising one or more substituted amino acids is also referred to as a mutant enzyme.

In the process according to the invention, when the glycosyltransferase is derived from Caenorhabditis elegans -(l,4)-GalNAcT enzyme or truncated -(l,4)-GalNAcT enzyme, it is preferred that the enzyme further comprises one or more mutations. Preferred mutations include substitution of the isoleucine (He, also referred to as I) at position 257 by leucine (Leu, also referred to as L), methionine (Met, also referred to as M) or alanine (Ala, also referred to as A). Preferred mutations also include substitution of the methionine (Met, also referred to as M) at position 312 by histidine (His, also referred to as H). Consequently, when the glycosyltransferase is derived from CeGalNAcT or CeGalNAcT(30-383) it is preferred that the enzyme comprises a I257L, I257M or a I257A mutation, and/or a M312H mutation. It should be noted that the numbering of amino acid position is herein based on the numbering of amino acid position in the wild-type -(l,4)-GalNAcT enzyme. When a -(l,4)-GalNAcT enzyme is e.g. a truncated enzyme, the number used herein to indicate e.g. the position of an amino acid substitution corresponds to the numbering of amino acid position in the corresponding wild-type -(l,4)-GalNAcT enzyme.

As an example, in wild-type CeGalNAcT (SEQ ID NO: 2) an isoleucine (He, I) is present on amino acid position 257. In CeGalNAcT(I257L) the isoleucine amino acid at position 257 is substituted by a leucine amino acid (Leu, L). As described above, CeGalNAcT(30-383) is herein to be understood as a truncated CeGalNAcT enzyme consisting of the amino acid sequence represented by the amino acids on position 30- 383 of SEQ ID NO: 2, whereas CeGalNAcT(30-383) itself is represented by SEQ ID NO: 6. In CeGalNAcT(30-383; I257L), the number "257" in I257L indicates that it is the I amino acid on position 257 in the corresponding wild-type CeGalNAcT (i.e. number 257 of SEQ ID NO:2 that is substituted with an L amino acid. The isoleucine amino acid on position 257 SEQ ID NO:2 is represented by the isoleucine amino acid on position 228 of SEQ ID NO:6.

Preferred truncated Caenorhabditis elegans -(l,4)-GalNAcT mutant enzymes include CeGalNAcT(30-383; I257L) (SEQ ID NO: 10), CeGalNAcT(30-383; I257M) (SEQ ID NO: 11), CeGalNAcT(30-383; I257A) (SEQ ID NO: 12) and CeGalNAcT(30-383; M312H) (SEQ ID NO: 13).

In the process according to the invention, when the glycosyltransferase is derived from Trichoplusia Ni -(l,4)-GalNAcT enzyme or truncated Trichoplusia Ni β-(1,4)- GalNAcT enzyme, it is preferred that the enzyme further comprises one or more mutations. Preferred mutations include substitution of the tryptophan (Trp, also referred to as W) on position 336 by phenylalanine (Phe, also referred to as F), histidine (His, also referred to as H) or valine (Val, also referred to as V). Consequently, when the glycosyltransf erase is derived from TnGalNAcT or TnGalNAcT(33-421), it is preferred that the enzyme comprises a W336F, W336H or W336V mutation. Preferred mutations of TnGalNAcT or TnGalNAcT(33-421) also include substitution of the glutamic acid (Glu, also referred to as E) on position 339 by alanine (Ala, also referred to as A), aspartic acid (Asp, also referred to as D) or serine (Ser, also referred to as S). Consequently, when the glycosyltransferase is derived from TnGalNAcT or TnGalNAcT(33-421), it is preferred that the enzyme comprises a E339A, E339D or E339S mutation. Another preferred mutations of TnGalNAcT or TnGalNAcT(33-421) include substitution of leucine (Leu, also referred to as L) on position 302 by alanine (Ala, also referred to as A) or glycine (Gly, also referred to as G). Other preferred mutations includes substitution of the isoleucine (He, also referred to as I) on position 299 by methionine (Met, also referred to as M), alanine (Ala, also referred to as A) or glycine (Gly, also referred to as G). Another preferred mutation includes substitution of the isoleucine (He, also referred to as I) on position 311 by methionine (Met, also referred to as M). The most preferred mutant of TnGalNAcT or TnGalNAcT(33-421) comprises an L302A mutation.

The glycosyltransferase derived from TnGalNAcT or TnGalNAcT(33-421) may contain more than one mutation, such as a mutation at the 336 and at the 339 position, both as described above. In one embodiment, the glycosyltransferase derived from TnGalNAcT or TnGalNAcT(33-421) comprises a W336F, W336H or W336V mutation and a E339A, E339G, E339D or E339S mutation.

In a preferred embodiment of the process according to the invention, the glycosyltransferase that is, or is derived from, a -(l,4)-GalNAcT enzyme is a Trichoplusia Ni -(l,4)-GalNAcT enzyme selected from the group consisting of TnGalNAcT(33-421; W336F) (SEQ ID NO: 25), TnGalNAcT(33-421; W336H) (SEQ ID NO: 26), TnGalNAcT(33-421; W336V) (SEQ ID NO: 27), TnGalNAcT(33-421; E339A) (SEQ ID NO: 28), TnGalNAcT(33-421; E339D) (SEQ ID NO: 30); TnGalNAcT(33-421; E339S) (SEQ ID NO: 31); TnGalNAcT(33-421; L302A) (SEQ ID NO: 29); TnGalNAcT(33-421; L302G) (SEQ ID NO: 35); TnGalNAcT(33-421; I299M) (SEQ ID NO: 36); TnGalNAcT(33-421; I299A) (SEQ ID NO: 37); TnGalNAcT(33-421; I299G) (SEQ ID NO: 38); and TnGalNAcT(33-421; I311M) (SEQ ID NO: 39);

The glycosyltransferase that is, or is derived from, a -(l,4)-GalNAcT enzyme that is used in the process according to the invention may also contain more than one mutation o the Trichoplusia Ni -(l,4)-GalNAcT enzyme, such as TnGalNAcT(33-421; W336H, E339A) (SEQ ID NO: 32), TnGalNAcT(33-421; W336H, E339D) (SEQ ID NO: 33) and TnGalNAcT(33-421; W336H, E339S) (SEQ ID NO: 34).

In the process according to the invention, when the glycosyltransferase is derived from Ascaris Sum -(l,4)-GalNAcT enzyme or truncated Ascaris Sum β-(1,4)- GalNAcT enzyme, it is preferred that the enzyme further comprises one or more mutations. Preferred mutations include substitution of tryptophan (Trp, also referred to as W) on position 282 by histidine (His, also referred to as H), and/or substitution of glutamic acid (Glu, also referred to as E) on position 285 by aspartic acid (Asp, also referred to as D), and/or substitution of phenylalanine (Phe, also referred to as F) on position 248 by alanine (Ala, also referred to as A), and/or substitution of phenylalanine (Phe, also referred to as F) on position 248 by glycine (Gly, also referred to as G), and/or substitution of valine (Val, also referred to as V) on position 245 by methionine (Met, also referred to as M). Consequently, when the glycosyltransferase is derived from AsGalNAcT or AsGalNAcT(30-383) it is preferred that the enzyme comprises a W282H mutation, and/or a E285D mutation.

In another preferred embodiment of the process according to the invention, the glycosyltransferase that is or is derived from a -(l,4)-GalNAcT enzyme is & Ascaris Sum -(l,4)-GalNAcT selected from the group consisting of AsGalNAcT(30-383; F248A) (SEQ ID NO: 40), AsGalNAcT(30-383; F248G) (SEQ ID NO: 41) and AsGalNAcT(30-383; V245M) (SEQ ID NO: 42).

In a preferred embodiment of the process according to the invention, the glycosyltransferase that is or is derived from a -(l,4)-GalNAcT enzyme is & Ascaris Sum -(l,4)-GalNAcT selected from the group consisting of AsGalNAcT(30-383; W282H) (SEQ ID NO: 46) and AsGalNAcT(30-383; E285D) (SEQ ID NO: 47).

In a preferred embodiment, the -(l,4)-GalNAcT enzyme as defined herein comprises a sequence encoding a tag for ease of purification. Preferably, said tag is selected from, but is not limited to, the group consisting of a FLAG-tag, poly(His)-tag, HA-tag, Myc-tag, SUMO-tag, GST-tag, MBP-tag or CBP-tag, more preferably said tag is a 6xHis tag. Preferably, said tag is covalently linked to the -(l,4)-GalNAcT enzyme at the C-terminus of the enzyme. In another further preferred embodiment, said tag is covalently linked to the -(l,4)-GalNAcT enzyme at the N-terminus of the enzyme.

When the -(l,4)-GalNAcT enzyme is derived from C.Elegans β-(1,4)- GalNAcT, the His-tagged -(l,4)-GalNAcT enzyme is preferably linked to the β-(1,4)- GalNAcT enzyme at the C-terminus of the enzyme, denoted as CeGalNAcT(30-383)- His 6 (SEQ ID NO: 14).

In a preferred embodiment of the process according to the invention, when the β-

(l,4)-GalNAcT enzyme is, or is derived from, Trichoplusia Ni -(l,4)-GalNAcT, the His-tagged -(l,4)-GalNAcT enzyme is, or is derived from, His6-TnGalNAcT(33-421) (SEQ ID NO: 49).

In another preferred embodiment of the process according to the invention, when the -(l,4)-GalNAcT enzyme is, or is derived from, Trichoplusia Ni -(l,4)-GalNAcT, the His-tagged -(l,4)-GalNAcT enzyme is, or is derived from, His6-TnGalNAcT(33- 421; W336F) (SEQ ID NO: 50), His 6 -TnGalNAcT(33-421; W336H) (SEQ ID NO: 51, His 6 -TnGalNAcT(33-421; W336V) (SEQ ID NO: 52), His 6 -TnGalNAcT(33-421; 339A) (SEQ ID NO: 53), His 6 -TnGalNAcT(33-421; E339D) (SEQ ID NO: 55), His 6 - TnGalNAcT(33-421; E339S) (SEQ ID NO: 56), His 6 -TnGalNAcT(33-421; L302A) (SEQ ID NO: 43), His 6 -TnGalNAcT(33-421; L302G) (SEQ ID NO: 44), His 6 - TnGalNAcT(33-421; I299M) (SEQ ID NO: 45), His 6 -TnGalNAcT(33-421; 1299 A) (SEQ ID NO: 48), His 6 -TnGalNAcT(33-421; I299G) (SEQ ID NO: 54), His 6 - TnGalNAcT(33-421; I311M) (SEQ ID NO: 60).

In another preferred embodiment of the process according to the invention, when the -(l,4)-GalNAcT enzyme is, or is derived from, Ascaris Sum -(l,4)-GalNAcT, the His-tagged -(l,4)-GalNAcT enzyme is, or is derived from, His6-AsGalNAcT(30-383) (SEQ ID NO: 71).

In another preferred embodiment of the process according to the invention, when the -(l,4)-GalNAcT enzyme is, or is derived from, Ascaris Sum -(l,4)-GalNAcT, the His-tagged -(l,4)-GalNAcT enzyme is, or is derived from, His6-AsGalNAcT(30-383; W282H) (SEQ ID NO: 72), His 6 -AsGalNAcT(30-383; E285D) (SEQ ID NO: 73). In a preferred embodiment, the -(l,4)-N-acetylgalactosaminyltransf erase used in the process according to the invention is, or is derived from, a sequence selected from the group consisting of SEQ ID NO:2-23.

As described above, the term "derived from" comprises e.g. truncated enzymes, mutant enzymes and enzymes comprising a tag for ease of purification, and these modifications are described in more detail above. The term "derived from" also comprises enzymes comprising a combination of the modfications described in more detail above.

In another preferred embodiment, the -(l,4)-N-acetylgalactosaminyltransferase used in the process according to the invention has at least 50% identity to a sequence selected from the group consisting of SEQ ID NO: 2-23. More preferably the β-(1,4)- N-acetylgalactosaminyltransferase used in the process according to the invention has at least 50% sequence identity, preferably at least 55% 60%, 65%, 70%, 75% 80%, 85% 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or preferably 100% sequence identity, to a sequence selected from the group consisting of SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22 and SEQ ID NO: 23.

In another preferred embodiment of the process the β-(1,4)-Ν- acetylgalactosaminyltransf erase is or is derived from a wild-type -(l,4)-GalNAcT, preferably an invertebrate -(l,4)-GalNAcT. In another preferred embodiment of the process, the glycosyltransferase is or is derived from an invertebrate -(l,4)-GalNAcT. In a further preferred embodiment, the glycosyltransferase is or is derived from Caenorhabditis elegans -(l,4)-GalNAcT (CeGalNAcT), Ascaris Sum β-(1,4)- GalNAcT (AsGalNAcT) or Trichoplusia Ni -(l,4)-GalNAcT (TnGalNAcT). β-(1,4)- GalNAcTs that are or are derived from (CeGalNAcT), (AsGalNAcT) or (TnGalNAcT) are described in more detail above. In this embodiment it is particularly preferred that the -(l,4)-N-acetylgalactosaminyltransf erase used in the process is, or is derived from, a sequence selected from the group consisting of SEQ ID NO: 2-9, i.e. from the group consisting of SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8 and SEQ ID NO: 9. More preferably, the β- (l,4)-N-acetylgalactosaminyltransf erase is, or is derived from, a sequence selected from the group consisting of SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 7 and SEQ ID NO: 8, even more preferably from the group consisting of SEQ ID NO: 6, SEQ ID NO: 7 and SEQ ID NO: 8, yet even more preferably from the group consisting of SEQ ID NO: 7 and SEQ ID NO: 8. Most preferably the -(l,4)-N-acetylgalactosaminyltransf erase used in the process is, or is derived from SEQ ID NO: 8.

In another particularly preferred embodiment the β-(1,4)-Ν- acetylgalactosaminyltransferase used in the process has at least 50% sequence identity, preferably at least 55% 60%, 65%, 70%, 75% 80%, 85% 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or preferably 100% sequence identity, to a sequence selected from the group consisting of SEQ ID NO: 2-9, i.e. from the group consisting of SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8 and SEQ ID NO: 9. More preferably, the -(l,4)-N-acetyl- galactosaminyltransferase used in the process has at least 50% sequence identity, preferably at least 55% 60%, 65%, 70%, 75% 80%, 85% 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or preferably 100% sequence identity, to a sequence selected from the group consisting of SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 7 and SEQ ID NO: 8, more preferably from the group consisting of SEQ ID NO: 6, SEQ ID NO: 7 and SEQ ID NO: 8, even more preferably from the group consisting of SEQ ID NO: 7 and SEQ ID NO: 8. Most preferably the β- (l,4)-N-acetylgalactosaminyltransf erase used in the process has at least 50% sequence identity, preferably at least 55% 60%, 65%, 70%, 75% 80%, 85% 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or preferably 100% sequence identity, to SEQ ID NO: 8.

In another particularly preferred embodiment of the process according to the invention the glycosyltransferase is, or is derived from, Caenorhabditis elegans β- (l,4)-GalNAcT (CeGalNAcT). In another particularly preferred embodiment the CeGalNAcT is, or is derived from, SEQ ID NO: 2 or SEQ ID NO: 6. In another particularly preferred embodiment the CeGalNAcT used in the process is, or is derived from, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13 or SEQ ID NO: 14.

In another particularly preferred embodiment the CeGalNAcT used in the process has at least 50% sequence identity, preferably at least 55% 60%, 65%, 70%, 75% 80%, 85% 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or preferably 100% sequence identity, to SEQ ID NO: 2 or SEQ ID NO: 6. In another particularly preferred embodiment the CeGalNAcT used in the process has at least 50% sequence identity, preferably at least 55% 60%, 65%, 70%, 75% 80%, 85% 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or preferably 100% sequence identity, to a sequence SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13 or SEQ ID NO: 14.

In another particularly preferred embodiment of the process according to the invention the glycosyltransferase is, or is derived from, Trichoplusia Ni β-(1,4)- GalNAcT (TnGalNAcT). In a further preferred embodiment of the process the TnGalNAcT is or is derived from SEQ ID NO: 4 or SEQ ID NO: 8. In another further preferred embodiment the TnGalNAcT used in the process has at least 50% sequence identity, preferably at least 55% 60%, 65%, 70%, 75% 80%, 85% 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or preferably 100% sequence identity, to SEQ ID NO: 4 or SEQ ID NO: 8. In another preferred embodiment the TnGalNAcT used in the process is, or is derived from, a sequence selected from the group consisting of SEQ ID NO: 25, SEQ ID NO: 26, SEQ ID NO: 27, SEQ ID NO: 28, SEQ ID NO: 29, SEQ ID NO: 30, SEQ ID NO: 31, SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 34, SEQ ID NO: 35, SEQ ID NO: 36, SEQ ID NO: 37, SEQ ID NO: 38, SEQ ID NO: 39, SEQ ID NO: 43, SEQ ID NO: 44, SEQ ID NO: 45, SEQ ID NO: 48, SEQ ID NO: 49, SEQ ID NO: 50, SEQ ID NO: 51, SEQ ID NO: 52, SEQ ID NO: 53, SEQ ID NO: 54, SEQ ID NO: 55, SEQ ID NO: 56, SEQ ID NO: 57, SEQ ID NO: 58, SEQ ID NO: 59 and SEQ ID NO: 60. In another preferred embodiment, the TnGalNAcT used in the process has at least 50% sequence identity, preferably at least 55% 60%, 65%, 70%, 75% 80%, 85% 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or preferably 100% sequence identity, to a sequence selected from the group consisting of SEQ ID NO: 25, SEQ ID NO: 26, SEQ ID NO: 27, SEQ ID NO: 28, SEQ ID NO: 29, SEQ ID NO: 30, SEQ ID NO: 31, SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 34, SEQ ID NO: 35, SEQ ID NO: 36, SEQ ID NO: 37, SEQ ID NO: 38, SEQ ID NO: 39, SEQ ID NO: 43, SEQ ID NO: 44, SEQ ID NO: 45, SEQ ID NO: 48, SEQ ID NO: 49, SEQ ID NO: 50, SEQ ID NO: 51, SEQ ID NO: 52, SEQ ID NO: 53, SEQ ID NO: 54, SEQ ID NO: 55, SEQ ID NO: 56, SEQ ID NO: 57, SEQ ID NO: 58, SEQ ID NO: 59 and SEQ ID NO: 60.

In another particularly preferred embodiment of the process according to the invention the glycosyltransferase is, or is derived from, Ascaris Sum -(l,4)-GalNAcT (AsGalNAcT). In this embodiment it is further preferred that the AsGalNAcT is or is derived from SEQ ID NO: 3 or SEQ ID NO: 7. In another further preferred embodiment the AsGalNAcT used in the process has at least 50% sequence identity, preferably at least 55% 60%, 65%, 70%, 75% 80%, 85% 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or preferably 100% sequence identity, to SEQ ID NO: 3 or SEQ ID NO: 7. In another further preferred embodiment the AsGalNAcT used in the process is, or is derived from, a sequence selected from the group consisting of SEQ ID NO: 40, SEQ ID NO: 41, SEQ ID NO: 42, SEQ ID NO: 46, SEQ ID NO: 47, SEQ ID NO: 71, SEQ ID NO: 72 and SEQ ID NO: 73. In another further preferred embodiment of the process the AsGalNAcT used in the process has at least 50% sequence identity, preferably at least 55% 60%, 65%, 70%, 75% 80%, 85% 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or preferably 100% sequence identity, to a sequence selected from the group consisting of SEQ ID NO: 40, SEQ ID NO: 41, SEQ ID NO: 42, SEQ ID NO: 46, SEQ ID NO: 47, SEQ ID NO: 71, SEQ ID NO: 72 and SEQ ID NO: 73.

Preferably, the derived or wild type -(l,4)-GalNAcT enzyme used in the process of the invention has UDP-F 2 -GalNAz transfer activity. UDP-F 2 -GalNAz is a sugar- derivative nucleotide according to formula (18), and is described in more detail below. UDP-F 2 -GalNAz transfer activity is preferably assessed by a method as exemplified herein, i.e. by the method of the R&D Systems Glycosyltransferase Activity Kit (http://www.rndsystems.com/product_detail_objectname_glycotr ansferase_assayj3rinc iple.aspx, product number EA001).

In brief, the glycosyltransferase kit determines the activity of a particular glycosyltransferase by means of a coupled assay that detects the release of liberated UDP upon transfer of the donor sugar (from the sugar-UDP nucleotide) to an acceptor sugar. In more detail, the UDP that is liberated upon transfer of sugar is hydrolyzed by a particular enzyme (CD39L3/rectonucleoside triphosphate diphosphohydrolase-3, also known as NTPDase-3) thereby generating UMP and one equivalent of phosphate (Pi). The latter phosphate in turn is detected by malachite green, which is also added to the mixture. A green color develops in proportion to the amount of inorganic phosphate released and the absorbance of the color at 620 nm is measured as a direct measure of the activity of the gly cosy ltransf erase. In the present case, transfer of F 2 -GalNAz from the UDP-substrate to GlcNAc on the protein is accompanied by release of UDP, which is hydrolyzed by CD39L3 and the generated phosphate. Preferably, the derived or wild type -(l,4)-GalNAcT enzyme used in the process according to the invention has at least 30%, 33%, 50%, 75%, 100%, 150%, 200%, or more preferably at least 300% of the UDP-F 2 -GalNAz transfer activity as compared to the UDP-F 2 -GalNAz transfer activity of the -(l,4)-galactosyltransf erase mutant enzyme Bos taurus GalT-Y289L (SEQ ID NO: 1), wherein the transfer activity is assessed using the R&D Systems Glycosyltransferase Activity Kit and applying the conditions as indicated in detail in Example 18 and using equal amounts of the enzyme to be tested and β-(1,4)- galactosyltransferase mutant enzyme Bos taurus GalT-Y289L (SEQ ID NO: 1).

The mutants of -(l,4)-N-acetylgalactosaminyltransf erase according to the second aspect of the invention preferably have at least 50% sequence identity, more preferably at least 55% 60%, 65%, 70%, 75% 80%, 85% 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% and most preferably 100% sequence identity, to one of the sequences selected from the group consisting of SEQ ID NO: 1, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 25, SEQ ID NO: 26, SEQ ID NO: 27, SEQ ID NO: 28, SEQ ID NO: 29, SEQ ID NO: 30, SEQ ID NO: 31, SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 34, SEQ ID NO: 35, SEQ ID NO: 36, SEQ ID NO: 37, SEQ ID NO: 38, SEQ ID NO: 39, SEQ ID NO: 40, SEQ ID NO: 41, SEQ ID NO: 42, SEQ ID NO: 43, SEQ ID NO: 44, SEQ ID NO: 45, SEQ ID NO: 46, SEQ ID NO: 47, SEQ ID NO: 48, SEQ ID NO: 50, SEQ ID NO: 51, SEQ ID NO: 52, SEQ ID NO: 53, SEQ ID NO: 54, SEQ ID NO: 55, SEQ ID NO: 56, SEQ ID NO: 57, SEQ ID NO: 58, SEQ ID NO: 59, SEQ ID NO: 60, SEQ ID NO: 72 and SEQ ID NO: 73. Glycoprotein and modified glycoprotein

The glycoprotein 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 glycan comprising a terminal GlcNAc-moiety is according to formula (1) or (2):

1 2 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.

The glycoprotein to be modified may comprise more than one glycan comprising a terminal GlcNAc-moiety. The glycoprotein may also comprise additional glycans that do not comprise a terminal GlcNAc-moiety.

The core-GlcNAc-moiety, i.e. the GlcNAc-moiety that is attached to the protein, 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 said GlcNAc-moiety.

It should be noted that the GlcNAc-moiety of a glycan according to formula (1) wherein b is 1, i.e. the GlcNAc-moiety in a glycan consisting of a fucosylated GlcNAc, is herein also considered a terminal GlcNAc-moiety.

In one embodiment, the glycan comprising a terminal GlcNAc-moiety consists of one GlcNAc-moiety, and the glycan is a glycan according to formula (1) wherein b is 0.

In another embodiment, said glycan consists of a fucosylated GlcNAc-moiety, and the glycan is a glycan according to formula (1) wherein b is 1.

In another embodiment, said glycan is a glycan according to formula (2), wherein the core-GlcNAc, if present, is optionally fucosylated (b is 0 or 1). In a glycan according to formula (2), 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, 3, 4, 5, 6, 7 or 8, and most preferably 2, 3, 4, 5 or 6 sugar moieties. When G is a branched oligosaccharide, G may comprise one or more terminal GlcNAc- moieties. A glycan according to formula (2) may thus comprise more than one terminal GlcNAc-moiety. In glycan (2) it is preferred that when d is 0 then e is 1, and when e is 0 then d is 1. More preferably, in glycan (2) d is 1, and even more preferably d is 1 and e is 1.

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 and xylose (Xyl).

In a preferred embodiment of the process according to the invention, the glycan comprising a terminal GlcNAc-moiety is according to formula (1), as defined above. In another preferred embodiment, the glycan comprising a terminal GlcNAc-moiety is according to formula (2). When the glycan comprising a terminal GlcNAc-moiety is according to formula (2), it is further preferred that the glycan according to formula (2) is a glycan according to formula (26), (27), (28), (29) or (30):

27

28

wherein b is 0 or 1.

In a preferred embodiment of the process according to the invention, the glycan comprising a terminal GlcNAc-moiety is a glycan according to formula (1), (26), (27), (28), (29) or (30), more preferably an N-linked glycan according to formula (1), (26), (27), (28), (29) or (30). In a further preferred embodiment, the glycan comprising a terminal GlcNAc-moiety is a glycan according to formula (1), (26) or (27), more preferably an N-linked glycan according to formula (1), (26) or (27). Most preferably the glycan comprising a terminal GlcNAc-moiety is a glycan according to formula (1) or (27), more preferably an N-linked glycan according to formula (1) or (27).

The glycoprotein comprising a glycan comprising a terminal GlcNAc moiety is preferably according to formula (7) or (8):

Pr-|-

7 wherein:

b, d, e and G, and preferred embodiments thereof, are as defined above; y is an integer in the range of 1 to 24; and

Pr is a protein.

The glycoprotein to be modified in the process according to the invention comprises one or more glycans comprising a terminal GlcNAc-moiety (y is 1 to 24). Preferably y is an integer in the range of 1 to 12, more preferably an integer in the range of 1 to 10. More preferably, y is 1, 2, 3, 4, 5, 6, 7 or 8, and yet more preferably y is 1, 2, 3, 4, 5 or 6. Even more preferably, y is 1, 2, 3 or 4. As was describd above, the glycoprotein may further comprise one or more glycans not having a terminal GlcNAc- moiety.

When the glycoprotein to be modified in the process according to the invention is according to formula (7) or (8), it is also preferred that the glycan comprising a terminal GlcNAc-moiety is a glycan, preferably an N-linked glycan, according to formula (1), (26), (27), (28), (29) or (30) as described above, more preferably according to formula (1), (26) or (27) and most preferably according to formula (1) or (27). Most preferably the glycan comprising a terminal GlcNAc-moiety is an N-linked glycan according to formula (1) or (27).

In a preferred embodiment of the process according to the invention, the glycoprotein comprising a glycan comprising a terminal GlcNAc moiety is an antibody, more preferably an antibody according to formula (7) or (8), wherein the protein (Pr) is an antibody (Ab). Also when the glycoprotein to be modified is an antibody, it is preferred that the glycan comprising a terminal GlcNAc-moiety is a glycan according to formula (1), (26), (27), (28), (29) or (30) as defined above, more preferably according to formula (1), (26) or (27), even more preferably according to formula (1) or (27). In this embodiment it is further preferred that the glycan comprising a terminal GlcNAc-moiety is an N-linked glycan according to formula (1), (26), (27), (28), (29) or (30), more preferably an N-linked glycan according to formula (1), (26) or (27), and most preferably an N-linked glycan according to formula (1) or (27).

When the glycoprotein to be modified is an antibody, it is preferred that y is 1, 2, 3, 4, 5, 6, 7 or 8, more preferably y is 1, 2, 4, 6 or 8, even more preferably y is 1, 2 or 4, and most preferably y is 1 or 2.

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 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 IgGl, IgG2, IgG3 or IgG4 antibody, and most preferably said antibody is an IgGl antibody.

In the process according to the invention, a glycoprotein mixture comprising fucosylated as well as non-fucosylated glycans may be used as the starting glycoprotein. Said mixture may e.g. comprise glycoproteins comprising one or more fucosylated (b is 1) glycans (1) and/or (2) and/or one or more non-fucosylated (b is 0) glycans (1) and/or (2). Removal of fucose from a fucosylated glycan prior to the process according to the invention is therefore not necessary, but optional. 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 (7) wherein b is 1, and that the term "terminal non-reducing GlcNAc-glycan" includes a glycan of formula (1) wherein b is 1.

The terminal non-reducing GlcNAc-protein may comprise one or more linear and/or one or more branched terminal non-reducing GlcNAc-glycans. A glycan is bonded to the protein via CI of the glycan core-sugar-moiety, and said core-sugar- moiety preferably is a core-GlcN Ac-moiety. Consequently, when the terminal non- reducing GlcNAc-glycan bonded to the protein is a glycan according to formula (2), it is preferred that d is 1. More preferably, when the glycan is according to formula (2), d is 1 and e 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/or 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 of a protein.

When the glycoprotein is an antibody, it is preferred that the glycan comprising a terminal GlcNAc-moiety is attached to the conserved N-glycosylation site in the Fc- fragment at asparagine in the region 290-305, typically at N297.

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. The glycoprotein in Figure 1(A) may for example be obtained by regular expression followed by trimming with an endoglycosidase or a combination of endoglycosidases. Figure 1(B) shows a glycoprotein comprising a branched oligosaccharide glycan wherein one of the branches comprises a terminal GlcNAc- moiety (this glycan is also referred to as GnM 5 ). The core-GlcNAc moiety may optionally be fucosylated. The glycoprotein in Figure 1(B) may for example be obtained by expression of a glycoprotein in a mammalian system in the presence of swainsonine or by expression in an engineered host organism, e.g. Lecl CHO or Pichia. Figure 1(C) shows an antibody comprising a branched oligosaccharide glycan, wherein the core-GlcNAc moiety is optionally fucosylated and wherein all branches comprise a terminal GlcNAc-moiety. The glycoprotein in Figure 1(C) may for example be obtained by trimming of the regular mixture of antibody glycoforms (GO, Gl, G2, GOF, GIF and G2F) upon combined action of sialidase and galactosidase.

In Figure 2 an embodiment of the process for the modification of a glycoprotein, wherein the glycoprotein is an antibody, is shown. In this embodiment a sugar- derivative Su(A) is transferred from Su(A)-Nuc to a terminal GlcNAc-moiety of an antibody glycan, using a -(l,4)-N-acetylgalactosaminyltransferase, to form a modified antibody.

As was described above, the process according to the invention for the modification of a glycoprotein may further comprise the step of providing a glycoprotein comprising a glycan comprising a terminal GlcNAc-moiety, and the invention therefore also relates to a process for the modification of a glycoprotein comprising the steps of:

(1) providing a glycoprotein comprising a glycan comprising a terminal GlcNAc moiety, wherein the glycan comprising a terminal GlcNAc-moiety is according to formula (1) or (2) as defined above; and

(2) contacting said glycoprotein with a sugar-derivative nucleotide Su(A)-Nuc, in the presence of a -(l,4)-N-acetylgalactosaminyltransf erase or a mutant thereof, wherein Su(A)-Nuc is according to formula (3) as defined above.

When for example the glycoprotein to be modified in the process according to the invention comprises a glycan according to formula (1), in step (1) of the process the glycoprotein to be modified may be provided by a process comprising the step of trimming a glycoprotein comprising an oligosaccharide glycan by the action of a suitable enzyme, preferably an endo-glycosidase.

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 can be trimmed in order to obtain a glycoprotein comprising a glycan according to formula (1). Trimming occurs in between said two GlcN Ac-residue s .

A "suitable enzyme" is defined as an enzyme wherefore the glycan that is to be trimmed is a substrate. The preferred type of enzyme that is to be used in step (1) 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 (1) of this particular embodiment of the process is selected from the group of endo- glycosidases.

Endoglycosidases are capable of cleaving internal glycosidic linkages in glycan structures, which provides a benefit to remodeling and synthetic endeavors. For example, endoglycosidases 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/complex-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, 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, also known as Endo S2, 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 (1) 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 S49, Endo Fl, Endo F2, Endo F3, Endo H, Endo M and Endo A, or a 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 and Endo F3, or a combination thereof.

When the glycoprotein is an antibody and 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-β-Ν- acetylglucosaminidase is preferably selected from the group consisting of Endo S, Endo S49, Endo Fl, Endo F2 and Endo F3, or a combination thereof, more preferably from the group consisting of Endo S and Endo S49, or a combination thereof.

When the glycoprotein is an antibody and 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-P-N-acetylglucosaminidase is preferably selected from the group consisting of Endo Fl, Endo F2 and Endo F3, or 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.

Therefore, when the glycoprotein to be modified in the process according to the invention comprises a glycan according to formula (1), in step (1) of the process the glycoprotein to be modified is preferably provided by a process comprising the step of 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 according to formula (1).

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, EndoH, Endo Fl, Endo F2, Endo F3 and any combination thereof. Most preferably, the endo- -N-acetylglucosaminidase is Endo S or Endo S49.

The process for providing a glycoprotein comprising a glycan according to formula (1) by treatment of a mixture of glycoforms GO, Gl, G2, G0F, GIF and G2F with an endoglycosidase is shown in Figure 4. Figure 4 shows that treatment of a glycoprotein, in this case an antibody, comprising a mixture of glycoforms GO, Gl, G2, G0F, GIF and G2F (said glycoforms are shown shown in Figure 2) with an endoglycosidase, followed by transfer of for example N-azidoacetylgalactosamine (GalNAz) from UDP-GalNAz using a -(l,4)-GalNAcT enzyme, results in a modified antibody according to formula (32).

When for example the glycoprotein to be modified in the process according to the invention comprises a glycan according to formula (26), the glycoprotein comprising an optionally fucosylated glycan of formula (26), also referred to as "GnM5", may be provided in various ways. In this embodiment, it is preferred that the glycoprotein is provided by a expression of hybrid N-glycoprotein in the presence of swainsonine, as for example described in Kanda et al, Glycobiology 2006, 17, 104 - 118, incorporated by reference, and if necessary followed by sialidase/galactosidase treatment). An alternative approach includes the genetic engineering of a host organism. For example, Lecl CHO is a knock-out CHO cel-line lacking the gene for expression of Mns-II. As a consequence, biosynthesis of the N-glycan inevitable stops at the GnM 5 -stage of the glycan, which can be isolated pure from the supernatant. A more extensive approach entails the engineering of host organisms not normally programmed to produce hybrid or complex N-glycans, such as yeast or insect cells. However, it has been amply demonstrated that these non-mammalian host cells (e.g. Glycoswitch™) can also be employed for the selective expression of a single glycoform of a particular N- glycoprotein, including glycans of the GnM 5 -type and of the M 5 -type.

Therefore, when the glycoprotein to be modified in the process according to the invention comprises a glycan according to formula (26), in step (1) of the process the glycoprotein comprising an optionally fucosylated glycan of formula (26) is preferably provided by a process comprising expression of the glycoprotein in a host organism, in the presence of swainsonine. Preferably, said host organism is a mammalian cell line, e.g. HEK293 or NS0 or a CHO-cell line. The resulting glycoproteins may be obtained as a mixture of proteins comprising a glycan of the formula (26) (also referred to as GnM 5 ), a glycan referred to as GalGnM5, a sialylated glycan referred to as SiaGalGnM 5 and/or a mixture thereof. The non-reducing sialic acid and/or galactose moiety, if present, may be removed by processing of the glycoprotein with sialidase (removal of the sialic acid moiety) and/or β-galactosidase (removal of galactose moiety), whereby a glycoprotein comprising a glycan of formula (26) is obtained. Preferably, treatment with sialidase and β-galactosidase occurs in a single step in (lb). In this embodiment it is further preferred that in step (1) of the process the glycoprotein to be modified isprovided by a process comprising the steps of:

(la) expression of a glycoprotein in a host organism in the presence of swainsonine; and

(lb) treatment of the obtained glycoprotein with sialidase and/or β-galactosidase in order to obtain a glycoprotein comprising a glycan of formula (26). When the glycoprotein to be modified in the process according to the invention comprises a glycan according to formula (27), in step (1) of the process the glycoprotein to be modified may for example be provided by a process comprising a treatment of a mixture of glycoforms GO, Gl, G2, GOF, GIF and G2F of the glycoprotein with sialidase and galactosidase. In Figure 3 the glycoforms GO, Gl, G2, GOF, GIF and G2F of an antibody comprising a biantennary glycan are shown.

Figure 4 shows a process for providing a glycoprotein, in this case an antibody, comprising a glycan according to formula (27) by treatment of a mixture of glycoforms GO, Gl, G2, GOF, GIF and G2F with sialidase and galactosidase, followed by transfer of for example N-azidoacetylgalactosamine (GalNAz) from UDP-GalNAz, using a β- (l,4)-GalNAcT, providing a modified antibody according to formula (33).

Sugar-derivative nucleotide Su(A)-Nuc

In the process for the modification of a glycoprotein according to the invention, a glycoprotein comprising a glycan according to formula (1) or (2) is contacted, under the action of a (mutant) -(l,4)-acetylgalactosaminyltransferase, with a sugar- derivative nucleotide Su(A)-Nuc. The sugar-derivative nucleotide Su(A)-Nuc is according to formula (3):

wherein:

a is O or l; Nuc is a nucleotide;

U is [C(R 1 ) 2 ]„ or [C(R 1 ) 2 ]p-0-[C(R 1 )2C(R 1 ) 2 0]o-[C(R 1 )2] q , wherein n is an integer in the range of 0 to 24; o is an integer in the range of 0 to 12; p and q are independently 0, 1 or 2; and R 1 is independently selected from the group consisting of H, F, CI, Br, I and an optionally substituted Ci - C 24 alkyl group;

T is a C 3 - Ci 2 (hetero)arylene group, wherein the (hetero)arylene group is optionally substituted; and

A is selected from the group consisting of:

(a) -N 3

(b) -C(0)R 3

wherein R 3 is an optionally substituted Ci - C 24 alkyl group;

(c) -C≡C-R 4

wherein R 4 is hydrogen or an optionally substituted Ci - C 24 alkyl group;

(d) -SH

(e) -SC(0)R 8

wherein R 8 is an optionally substituted Ci - C 24 alkyl group;

(f) -SC(V)OR 8

wherein V is O or S, and R 8 is an optionally substituted Ci - C 24 alkyl group;

(g) -x

wherein X is selected from the group consisting of F, CI, Br and I;

(h) -OS(0) 2 R 5

wherein R 5 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, the alkyl groups, aryl groups, alkylaryl groups and arylalkyl groups being optionally substituted;

(i) R 11

wherein R 11 is an optionally substituted C 2 - C 24 alkyl group;

G) R 12

wherein R 12 is an optionally substituted terminal C 2 - C 24 alkenyl group; and (k) R 13

wherein R 13 is an optionally substituted terminal C 3 - C 24 allenyl group. Nuc is herein defined as a nucleotide. Nuc is preferably selected from the group consisting of a nucleoside monophosphate and a nucleoside diphosphate, more preferably from the group consisting of uridine diphosphate (HDP), 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) and cytidine diphosphate (CDP). Most preferably, Nuc is uridine diphosphate (UDP). Therefore, in a preferred embodiment, Su(A)-Nuc (3) is Su(A)-UDP (31):

wherein U, a, T and A are as defined above.

In one embodiment, A is an azido group -N 3 .

In another embodiment, A is a keto group -C(0)R 3 , wherein R 3 is an optionally substituted Ci - C 24 alkyl group, preferably an optionally substituted Ci - C 12 alkyl group, and more preferably an optionally substituted Ci - C 6 alkyl group. Even more preferably, R 3 is methyl, ethyl, n-propyl, i-propyl, n-butyl, s-butyl or t-butyl, and most preferably, R 3 is methyl.

In another embodiment, A is an alkynyl group. In a preferred embodiment, the alkynyl group is a (hetero)cycloalkynyl group, preferably a (hetero)cyclooctynyl group. In another preferred embodiment, the alkynyl group is -C≡C-R 4 , wherein R 4 is hydrogen or an optionally substituted Ci - C 2 4 alkyl group, preferably hydrogen or an optionally substituted Ci - Ci 2 alkyl group, and more preferably hydrogen or an optionally substituted Ci - C 6 alkyl group. Most preferably, R 4 is hydrogen, methyl, ethyl, n-propyl, i-propyl, n-butyl, s-butyl or t-butyl. In this embodiment, it is further preferred that the alkynyl group is a terminal alkynyl group, i.e. R 4 is preferably hydrogen.

In another embodiment, A is a thiol group -SH.

In another embodiment, A is a precursor of a thiol group -SC(0)R 8 , wherein R 8 is an optionally substituted Ci - C 24 alkyl group. Preferably, R 8 is an optionally substituted Ci - Ci 2 alkyl group, more preferably R 8 is an optionally substituted Ci - C 6 alkyl group, and even more preferably R 8 is methyl, ethyl, n-propyl, i-propyl, n-butyl, s-butyl or t-butyl. Most preferably, R 8 is methyl. In the process according to the invention for the modification of a glycoprotein, a sugar-derivative nucleotide wherein A is a precursor of a thiol group may be used. During the process, the thiol-precursor is converted to a thiol group.

In another embodiment, A is -SC(V)OR 8 , wherein V is O or S, and R 8 is an optionally substituted Ci - C 24 alkyl group. In a preferred embodiment, A is -SC(0)OR 8 . In another preferred embodiment, A is -SC(S)OR 8 . Both when A is -SC(0)OR 8 and when A is -SC(S)OR 8 , R 8 is preferably an optionally substituted Ci - Ci 2 alkyl group, more preferably R 8 is an optionally substituted Ci - C 6 alkyl group, and even more preferably R 8 is methyl, ethyl, n-propyl, i-propyl, n-butyl, s-butyl or t- butyl. Most preferably, R 8 is methyl.

In another embodiment, A is a halogen X. X is selected from the group consisting of F, CI, Br and I, preferably from the group consisting of CI, Br and I, more preferably from the group consisting of CI and Br. Most preferably, X is CI.

In another embodiment, A is a sulfonyloxy group -OS(0) 2 R 5 , wherein R 5 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, the alkyl groups, aryl groups, alkylaryl groups and arylalkyl groups being optionally substituted. Preferably, R 5 is a Ci - Ci 2 alkyl group, C 6 - Ci 2 aryl group, C 7 - Ci 2 alkylaryl group or a C 7 - Ci 2 arylalkyl group. More preferably R 5 is selected from the group consisting of -CH 3 , - C 2 H 5 , a C 3 linear or branched alkyl group, a C 4 linear or branched alkyl group, a C 6 - Cio aryl group and a C 7 alkylaryl group. Even more preferably, R 5 is a methyl group, an ethyl group, a phenyl group or a p-tolyl group. Most preferably 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 )).

In another embodiment, A is R 11 , wherein R 11 is an optionally substituted C 2 - C 2 4 alkyl group, preferably an optionally substituted C 2 - C 12 alkyl group, and more preferably an optionally substituted C 2 - C 6 alkyl group. Even more preferably, R 11 is ethyl, n-propyl, i-propyl, n-butyl, s-butyl or t-butyl, and most preferably, R 11 is ethyl.

In another embodiment, A is R 12 , wherein R 12 is an optionally substituted terminal C 2 - C 24 alkenyl group. The term "terminal alkenyl group" herein refers to an alkenyl group wherein the carbon-carbon double bond is situated at a terminus of the alkenyl group. The terminal C 2 - C 24 alkenyl group thus ends with a C=CH 2 moiety. Preferably R 12 is an optionally substituted terminal C 2 - C 12 alkenyl group, and more preferably an optionally substituted terminal C 2 - C 6 alkenyl group. More preferably, the terminal alkenyl group is a linear alkenyl group, preferably an unsubstituted linear alkenyl group. Even more preferably R 12 is selected from the group consisting of -C(H)=CH 2 , -CH 2 -C(H)=CH 2 , -CH 2 -CH 2 -C(H)=CH 2 , -CH 2 -CH 2 -CH 2 -C(H)=CH 2 and -CH 2 -CH 2 -CH 2 -CH 2 -C(H)=CH 2 . Yet even more preferably R 12 is selected from the group consisting of -C(H)=CH 2 , -CH 2 -C(H)=CH 2 and -CH 2 -CH 2 -C(H)=CH 2 . Most preferably, R 12 is -C(H)=CH 2 .

In another embodiment, A is R 13 , wherein R 13 is an optionally substituted terminal C 3 - C 24 allenyl group. The term "terminal allenyl group" herein refers to an allenyl group wherein the C=C=C moiety is situated at a terminus of the allenyl group. The terminal C 3 - C 24 alkenyl group thus ends with a -C=C=CH 2 moiety. Preferably R 13 is an optionally substituted terminal C 3 - C 12 alkenyl group, and more preferably an optionally substituted terminal C 3 - C 6 alkenyl group. More preferably, the terminal allenyl group is a linear allenyl group, preferably an unsubstituted linear allenyl group. Even more preferably R 13 is selected from the group consisting of -C(H)=C=CH 2 , -CH 2 -C(H)=C=CH 2 , -CH 2 -CH 2 -C(H)=C=CH 2 and -CH 2 -CH 2 -CH 2 -C(H)=C=CH 2 . Yet even more preferably R 13 is selected from the group consisting of -C(H)=C=CH 2 and -CH 2 -C(H)=C=CH 2 . Most preferably, R 13 is -C(H)=C=CH 2 . When A is R 13 , it is particularly preferred that in Su(A)-Nuc (3), both U and T are absent, i.e. it is particularly preferred that a is 0, and when U is[C(R 1 ) 2 ] n then n is 0, and when U is [C(R 1 ) 2 ] p -0-[C(R 1 ) 2 C(R 1 ) 2 0] 0 -[C(R 1 ) 2 ] q then o, p and q are all 0. In Su(A)-Nuc (3), T is a C 3 - C 12 (hetero)arylene group, wherein the (hetero)arylene group is optionally substituted. In a preferred embodiment, T is absent (a is 0). In another preferred embodiment, T is present (a is 1). When a is 1, (hetero)arylene group T in (3) is substituted with A, wherein A is as defined above.

(Hetero)arylene group T is optionally further substituted with one or more substituents R 2 , wherein R 2 is independently selected from the group consisting of halogen (-F, -CI, -Br, -I, preferably -F, -CI, -Br), -CN, -N0 2 , -C(0)R 9 , -C(0)OR 9 , -C(O)N(R 10 ) 2 , Ci - Ci 2 alkyl groups, C 2 - Ci 2 alkenyl groups, C 2 - Ci 2 alkynyl groups, C 3 - Ci 2 cycloalkyl groups, C 5 - Ci 2 cycloalkenyl groups, C 8 - Ci 2 cycloalkynyl groups, Ci - Ci 2 alkoxy groups, C 2 - Ci 2 alkenyloxy groups, C 2 - Ci 2 alkynyloxy groups, C 3 - Ci 2 cycloalkyloxy groups, amino groups (preferably -N(R 10 ) 2 ), oxo groups and -Si(R 7 ) 3 groups, wherein the alkyl groups, alkenyl groups, alkynyl groups, cycloalkyl groups, cycloalkenyl groups, cycloalkynyl groups, alkoxy groups, alkenyloxy groups, alkynyloxy groups and cycloalkyloxy groups are optionally interrupted by one of more hetero-atoms selected from the group consisting of O, N and S, and wherein R 7 is independently selected from the group consisting of Ci - Ci 2 alkyl groups, C 2 - Ci 2 alkenyl groups, C 2 - Ci 2 alkynyl groups, C 3 - Ci 2 cycloalkyl groups, Ci - Ci 2 alkoxy groups, C 2 - Ci 2 alkenyloxy groups, C 2 - Ci 2 alkynyloxy groups and C 3 - Ci 2 cycloalkyloxy groups wherein the alkyl groups, alkenyl groups, alkynyl groups, cycloalkyl groups, alkoxy groups, alkenyloxy groups, alkynyloxy groups and cycloalkyloxy groups are optionally substituted, wherein R 9 is a Ci - Ci 2 alkyl group, and wherein R 10 is independently selected from hydrogen and a Ci - Ci 2 alkyl group. Preferably, R 9 is a Ci - C 6 alkyl group, even more preferably a Ci - C 4 alkyl group, and most preferably a methyl, ethyl, n-propyl, i-propyl, n-butyl, s-butyl or a t-butyl group. Preferably, R 10 is a hydrogen or a Ci - C 6 alkyl group, more preferably hydrogen or a Ci - C 4 alkyl group, and most preferably R 10 is hydrogen, a methyl, ethyl, n-propyl, i- propyl, n-butyl, s-butyl or a t-butyl group.

When R 2 is a -Si(R 7 ) 3 group, preferably R 7 is, independently, a Ci - Ci 2 alkyl group, more preferably independently a Ci - C 6 alkyl group, even more preferably independently a Ci - C 4 alkyl group, and most preferably R 7 is, independently, a methyl, ethyl, n-propyl, i-propyl, n-butyl, s-butyl or a t-butyl group.

Preferably, R 2 , when present, is independently selected from the group consisting of -F, -CI, -Br, -I, -CN, -N0 2 , -C(0)R 9 , -C(0)OR 9 , -C(O)N(R 10 ) 2 , Ci - C u alkyl groups, Ci - Ci 2 alkoxy groups, amino groups (-N(R ) 2 ), oxo groups and -Si(R ) 3 groups, wherein R 7 , R 9 , R 10 and preferred embodiments of R 7 , R 9 , R 10 are as defined above.

More preferably, R 2 , when present, is independently selected from the group consisting of -F, -CI, -Br, -CN, -N0 2 , -C(0)R 9 , -C(0)OR 9 , -C(O)N(R 10 ) 2 , Ci - C 6 alkyl groups, Ci - C 6 alkoxy groups, amino groups, oxo groups and -Si(R 7 ) 3 groups, wherein R 7 , R 9 , R 10 and preferred embodiments of R 7 , R 9 , R 10 are as defined above.

Even more preferably, R 2 , when present, is independently selected from the group consisting of -F, -CI, -Br, -CN, -N0 2 , -C(0)R 9 , -C(0)OR 9 , -C(O)N(R 10 ) 2 , Ci - C 4 alkyl groups and Ci - C 4 alkoxy groups, wherein R 9 and R 10 , and preferred embodiments of R 9 and R 10 , are as defined above.

Yet even more preferably, R 2 , when present, is independently selected from the group consisting of -F, -CI, -Br, -CN, -N0 2 , methyl, methoxy, ethyl, ethoxy, n-propyl, n-propoxy, i-propyl, i-propoxy, n-butyl, n-butoxy, s-butyl, s-butoxy, t-butyl and t- butoxy. Most preferably, R 2 , when present, is independently selected from the group consisting of -F, -CI, -Br, -CN, -N0 2 , methyl and methoxy.

In a preferred embodiment, the (hetero)arylene group in (3) is unsubstituted. In another preferred embodiment, the (hetero)arylene group in (3) comprises one or more substituents R 2 , wherein R 2 and preferred embodiments of R 2 are defined above.

The term "(hetero)arylene group" herein refers to arylene groups as well as to heteroarylene groups. The term "(hetero)arylene group" herein refers to monocyclic (hetero)arylene groups as well as to bicyclic (hetero)arylene groups. The (hetero)arylene group in Su(A)-Nuc (3) may be any arylene group or any heteroarylene group.

In a preferred embodiment of the process according to the invention,

(hetero)arylene group T in (3) is selected from the group consisting of phenylene groups, naphthylene groups, anthracylene groups, pyrrolylene groups, pyrroliumylene groups, furanylene groups, thiophenylene groups (i.e. thiofuranylene groups), pyrazolylene groups, imidazolylene groups, pyrimidiniumylene groups, imidazoliumylene groups, isoxazolylene groups, oxazolylene groups, oxazoliumylene groups, isothiazolylene groups, thiazolylene groups, 1,2,3-triazolylene groups, 1,3,4- triazolylene groups, diazolylene groups, l-oxa-2,3-diazolylene groups, l-oxa-2,4- diazolylene groups, l-oxa-2,5-diazolylene groups, l-oxa-3,4-diazolylene groups, 1- thia-2,3-diazolylene groups, l-thia-2,4-diazolylene groups, l-thia-2,5-diazolylene groups, l-thia-3,4-diazolylene groups, tetrazolylene groups, pyridinylene groups, pyridazinylene groups, pyrimidinylene groups, pyrazinylene groups, pyradizinylene groups, pyridiniumylene groups, pyrimidiniumylene groups, benzofuranylene groups, benzothiophenylene groups, benzimidazolylene groups, indazolylene groups, benzotriazolylene groups, pyrrolo[2,3-b]pyridinylene groups, pyrrolo[2,3- c] pyridinylene groups, pyrrolo[3,2-c]pyridinylene groups, pyrrolo[3,2-b]pyridinylene groups, imidazo[4,5-b]pyridinylene groups, imidazo[4,5-c]pyridinylene groups, pyrazolo[4,3-d]pyridinylene groups, pyrazolo[4,3-c]pyridinylene groups, pyrazolo[3,4- c]pyridinylene groups, pyrazolo[3,4-b]pyridinylene groups, isoindolylene groups, indazolylene groups, purinylene groups, indolininylene groups, imidazo[l,2- a]pyridinylene groups, imidazo[l,5-a]pyridinylene groups, pyrazolo[l,5-a]pyridinylene groups, pyrrolo[l,2-b]pyridazinylene groups, imidazo[l,2-c]pyrimidinylene groups, quinolinylene groups, isoquinolinylene groups, cinnolinylene groups, quinazolinylene groups, quinoxalinylene groups, phthalazinylene groups, 1,6-naphthyridinylene groups, 1,7-naphthyridinylene groups, 1,8-naphthyridinylene groups, 1,5-naphthyridinylene groups, 2,6-naphthyridinylene groups, 2,7-naphthyridinylene groups, pyrido[3,2- d] pyrimidinylene groups, pyrido[4,3-d]pyrimidinylene groups, pyrido[3,4- d]pyrimidinylene groups, pyrido[2,3-d]pyrimidinylene groups, pyrido[2,3- b]pyrazinylene groups, pyrido[3,4-b]pyrazinylene groups, pyrimido[5,4- d]pyrimidinylene groups, pyrazino[2,3-b]pyrazinylene groups and pyrimido[4,5- d]pyrimidinylene groups, all groups optionally substituted with one or more substituents R 2 , wherein R 2 and preferred embodiments of R 2 are as defined above.

In a further preferred embodiment, (hetero)arylene group T is selected from the group consisting of phenylene groups, pyridinylene groups, pyridiniumylene groups, pyrimidinylene groups, pyrimidiniumylene groups, pyrazinylene groups, pyradizinylene groups, pyrrolylene groups, pyrroliumylene groups, furanylene groups, thiophenylene groups (i.e. thiofuranylene groups), diazolylene groups, quinolinylene groups, imidazolylene groups, pyrimidiniumylene groups, imidazoliumylene groups, oxazolylene groups and oxazoliumylene groups, all groups optionally substituted with one or more substituents R 2 , wherein R 2 and preferred embodiments of R 2 are as defined above. Even more preferably, (hetero)arylene group T is selected from the group consisting of phenylene groups, pyridinylene groups, pyridiniumylene groups, pyrimidinylene groups, pyrimidiniumylene groups, imidazolylene groups, pyrimidiniumylene groups, imidazoliumylene groups, pyrrolylene groups, furanylene groups and thiophenylene groups, all groups optionally substituted with one or more substituents R 2 , wherein R 2 and preferred embodiments of R 2 are as defined above.

Most preferably, (hetero)aryl group T is selected from the group consisting of phenylene groups, imidazolylene groups, imidazoliumylene groups, pyrimidiniumylene groups, pyridinylene groups and pyridiniumylene groups, all groups optionally substituted with one or more substituents R 2 , wherein R 2 and preferred embodiments of R 2 are as defined above.

In Su(A)-Nuc (3), U is [C(R 1 ) 2 ]„ or [C(R 1 ) 2 ] p -0-[C(R 1 ) 2 C(R 1 ) 2 0] 0 -[C(R 1 ) 2 ] q , wherein n is an integer in the range of 0 to 24; o is an integer in the range of 0 to 12; p and q are independently 0, 1 or 2; and R 1 is independently selected from the group consisting of H, F, CI, Br, I and an optionally substituted Ci - C 24 alkyl group. Preferably, U is [C(R 1 ) 2 ]„.

In a preferred embodiment of the process according to the invention, U is absent, i.e. n, p, o and q are all 0.

In another preferred embodiment of the process according to the invention, U is present, i.e. n, p, o and q are not all 0. Consequently, in this embodiment, when U is [C(R 1 ) 2 ] n , n is an integer in the range of 1 to 24, and when U is [C(R 1 ) 2 ] p -0- [C(R 1 ) 2 C(R 1 ) 2 0]o-[C(R 1 ) 2 ] q , o is an integer in the range of 1 to 12 and/or p is 1 or 2 and/or q is 1 or 2. In other words, when U is [C(R 1 ) 2 ] p -0-[C(R 1 ) 2 C(R 1 ) 2 0] 0 -[C(R 1 ) 2 ] q , at least one of o, p and q is not 0.

When U is [C(R 1 ) 2 ] n , n is an integer in the range of 0 to 24. In a preferred embodiment, n is an integer in the range of 1 to 24, preferably in the range of 1 to 12. In this embodiment, more preferably n is 1, 2, 3, 4, 5, 6, 7 or 8, even more preferably n is 1 , 2, 3, 4, 5 or 6, yet even more preferably n is 1 , 2, 3 or 4, yet even more preferably n is 1, 2 or 3, yet even more preferably, n is 1 or 2 and most preferably n is 1. In another preferred embodiment, n is 0. It is particularly preferred that n is 0 or 1.

When U is [C(R 1 ) 2 ]„ and n is 1 or more, R 1 is independently selected from the group consisting of H, F, CI, Br, I and an optionally substituted Ci - C 24 alkyl group, preferably from the group consisting of H, F, CI, Br, I and an optionally substituted Ci - Ci2 alkyl group, and more preferably from the group consisting of H, F, CI, Br, I and an optionally substituted Ci - C 6 alkyl group. Even more preferably, R 1 is independently selected from the group consisting of H, F, CI, Br, I, a methyl group, an ethyl group, an n-propyl group, an i-propyl group, an n-butyl group, an s-butyl group or a t-butyl group. Even more preferably, R 1 is independently selected from the group consisting of H, F, CI and methyl, and most preferably, R 1 is independently selected from the group consisting of H and F. When U is [QR 1 )^ and n is 1 or 2, preferred examples of the -[C(R 1 )2] n - moiety in Su(A)-Nuc include -(CH 2 )-, -(CF 2 )-, -(CC1 2 )-, -(CBr 2 )-, -(CMe 2 )-, -(CH 2 CH 2 )-, -(CH 2 CF 2 )-, -(CH 2 CC1 2 )-, -(CH 2 CBr 2 )-, -(CH 2 CI 2 )-, -(CH 2 CMe 2 )-, -(CF 2 CF 2 )-, -(CC1 2 CC1 2 )-, -(CBr 2 CBr 2 )- and -(CMe 2 CMe 2 )-, more preferably -(CH 2 )-, -(CF 2 )-, -(CH 2 CH 2 )-, -(CH 2 CF 2 )- and -(CF 2 CF 2 )-.

When U is [C(R 1 ) 2 ]„ and n is 3 or more, preferred examples of the -[C(R 1 ) 2 ] n - moiety in Su(A)-Nuc include -(C„H 2n )-, -(C„F 2n )-, -(C„C1 2 „)-, -(C„Br 2n )-, -(C (n -i ) H 2(n- i ) CF 2 )-, -(C (n -i ) H 2(n- i ) CCl 2 )-, -(C (n -i ) H 2(n- i ) CBr 2 )- and -(C (n -i ) H 2(n- i ) CMe 2 )-, for example -(C 3 H 6 )-, -(C 3 F 6 )-, -(C 3 C1 6 )-, -(C 3 Br 6 )-, -(CH 2 CH 2 CF 2 )-, -(CH 2 CH 2 CC1 2 )-, -(CH 2 CH 2 CBr 2 )- and -(C 4 H 8 )-. More preferred examples include -(C n H 2n )-, -(C n F 2n )-, e.g. -(C 3 H 6 )-, -(C 4 H 8 )-, -(C 3 F 6 )- and -(C 4 F 8 )-.

When U is [C(R 1 ) 2 ] p -0-[C(R 1 ) 2 C(R 1 ) 2 0] 0 -[C(R 1 ) 2 ] q , o is an integer in the range of 0 to 12 and p and q are independently 0, 1 or 2. Preferably, o is an integer in the range of 1 to 10, more preferably o is 1, 2, 3, 4, 5, 6, 7 or 8, even more preferably o is 1, 2, 3, 4, 5 or 6, yet even more preferably o is 1 , 2, 3 or 4, yet even more preferably o is 1, 2 or 3, yet even more preferably, o is 1 or 2 and most preferably o is 1. In another preferred embodiment, o is 0. It is particularly preferred that o is 0, 1 or 2. When o is 0, it is further preferred that when p is 0, q is 1 or 2, and that when q is 0, p is 1 or 2.

When U is [C(R 1 ) 2 ] p -0-[C(R 1 ) 2 C(R 1 ) 2 0] 0 -[C(R 1 ) 2 ] q and o and/or p and/or q are 1 or more, R 1 is independently selected from the group consisting of H, F, CI, Br, I and an optionally substituted Ci - C 24 alkyl group, preferably from the group consisting of H, F, CI, Br, I and an optionally substituted Ci - Ci 2 alkyl group, and more preferably from the group consisting of H, F, CI, Br, I and an optionally substituted Ci - C 6 alkyl group. Even more preferably, R 1 is independently selected from the group consisting of H, F, CI, Br, I, a methyl group, an ethyl group, an n-propyl group, an i-propyl group, an n-butyl group, an s-butyl group or a t-butyl group. Even more preferably, R 1 is independently selected from the group consisting of H, F, CI and methyl. Most preferably, R 1 is H.

When U is [C(R 1 ) 2 ]p-0-[C(R 1 )2C(R 1 ) 2 0]o-[C(R 1 )2] q , preferred examples of the -[C(R 1 ) 2 ] p -0-[C(R 1 ) 2 C(R 1 ) 2 0] 0 -[C(R 1 ) 2 ] q - moiety in Su(A)-Nuc include -CH 2 -0-, -(CH 2 ) 2 -0-, -0-CH 2 -, -0-(CH 2 ) 2 -, -CH 2 -0-(CH 2 CH 2 0)o-, -(CH 2 ) 2 -0-(CH 2 CH 2 0) 0 -, -0-(CH 2 CH 2 0)o-, -0-(CH 2 CH 2 0)o-CH 2 -, -0-(CH 2 CH 2 0) 0 -(CH 2 ) 2 -, -CH 2 - 0-(CH 2 CH 2 0)o-CH 2 -, -CH 2 -0-(CH 2 CH 2 0)o-(CH 2 ) 2 -, -(CH 2 ) 2 -0-(CH 2 CH 2 0) 0 -CH 2 - and -(CH 2 ) 2 -0-(CH 2 CH 2 0) 0 -(CH 2 ) 2 -, wherein o is 1 , 2, 3, 4, 5 or 6, preferably o is 1 , 2, 3 or 4, more preferably o is 1 or 2 and most preferably o is 1.

In the process according to the invention, it is preferred that a, n, o, p and q are not all 0. Thus, when U is [C(R 1 ) 2 ] p -0-[C(R 1 ) 2 C(R 1 ) 2 0] 0 -[C(R 1 ) 2 ] q and o, p and q are 0, a is preferably not 0, and/or when U is [C(R 1 ) 2 ] n and n is 0, a is preferably not 0. In other words, in the absence of U (i.e. when o, p and q are 0 and n is 0), it is preferred that a is not 0.

In a preferred embodiment of the process according to the invention, a is 0, U is [C(R 1 ) 2 ] n and n is an integer in the range of 1 to 24. In this embodiment, the sugar- derivative nucleotide Su(A)-Nuc is according to formula (9) as defined below, wherein U is [C(R 1 ) 2 ] n . In this embodiment it is further preferred that a is 0 and n is in the range of 1 to 12, more preferably a is 0 and n is 1 , 2, 3, 4, 5, 6, 7 or 8, even more preferably a is 0 and n is 1, 2, 3, 4, 5 or 6, yet even more preferably a is 0 and n is 1, 2, 3 or 4, yet even more preferably a is 0 and n is 1 or 2, and most preferably a is 0 and n is 1.

Preferred examples of [C(R 1 ) 2 ] n are as described in more detail above.

In another preferred embodiment of the process according to the invention, a is 0,

U is [C(R 1 ) 2 ] p -0-[C(R 1 ) 2 C(R 1 ) 2 0]o-[C(R 1 ) 2 ] q and p, o and q are not all 0, i.e. o is an integer in the range of 1 to 12 and/or p is 1 or 2 and/or q is 1 or 2. In this embodiment, the sugar-derivative nucleotide Su(A)-Nuc is according to formula (9) as defined below, wherein U is [C(R 1 ) 2 ] p -0-[C(R 1 ) 2 C(R 1 ) 2 0] 0 -[C(R 1 ) 2 ] q . In this embodiment it is further preferred that a is 0 and o is in the range of 1 to 12, more preferably a is 0 and o is in the range of 1 to 10, even more preferably a is 0 and o is 1 , 2, 3, 4, 5, 6, 7 or 8, yet even more preferably a is 0 and o is 1, 2, 3, 4, 5 or 6, yet even more preferably a is 0 and o is 1 , 2, 3 or 4, yet even more preferably a is 0 and o is 1 or 2, and most preferably a is 0 and o is 1. Also in this embodiment, p and q are independently 0, 1 or 2. Preferred examples of [C(R 1 ) 2 ]p-0-[C(R 1 )2C(R 1 )20]o-[C(R 1 ) 2 ] q are as described in more detail above.

In yet another preferred embodiment, a is 1, U is [C(R 1 ) 2 ] n and n is an integer in the range of 1 to 24. In this embodiment it is further preferred that a is 1 and n is in the range of 1 to 12, more preferably a is 1 and n is 1, 2, 3, 4, 5, 6, 7 or 8, even more preferably a is 1 and n is 1, 2, 3, 4, 5 or 6, yet even more preferably a is 1 and n is 1, 2, 3 or 4, yet even more preferably a is 1 and n is 1 or 2, and most preferably a is 1 and n is 1. Preferred examples of [C(R 1 )2] n are as described in more detail above.

In yet another preferred embodiment, a is 1, U is [C(R 1 ) 2 ]p-0-[C(R 1 )2C(R 1 ) 2 0]o-

[C(R 1 ) 2 ] q , o is an integer in the range of 1 to 12 and p and q are independently 0, 1 or 2. In this embodiment it is further preferred that a is 1, U is [C(R 1 ) 2 ] p -0- [C(R 1 )2C(R 1 )20] 0 -[C(R 1 )2]q and o is in the range of 1 to 10, more preferably a is 1, U is [C(R 1 ) 2 ]p-0-[C(R 1 )2C(R 1 ) 2 0]o-[C(R 1 )2] q and o is 1, 2, 3, 4, 5, 6, 7 or 8, even more preferably a is 1, U is [C(R 1 ) 2 ]p-0-[C(R 1 )2C(R 1 ) 2 0]o-[C(R 1 )2] q and o is 1, 2, 3, 4, 5 or 6, yet even more preferably a is 1, U is [C(R 1 ) 2 ]p-0-[C(R 1 )2C(R 1 ) 2 0]o-[C(R 1 )2] q and o is 1, 2, 3 or 4, yet even more preferably a is 1, U is [C(R 1 ) 2 ]p-0-[C(R 1 )2C(R 1 ) 2 0]o- [C(R 1 ) 2 ] q and o is 1 or 2, and most preferably a is 1, U is [C(R 1 ) 2 ] p -0- [C(R 1 )2C(R 1 )20] 0 -[C(R 1 )2] q and o is 1. Also in this embodiment, p and q are independently 0, 1 or 2. Preferred examples of [C(R 1 ) 2 ]p-0-[C(R 1 )2C(R 1 ) 2 0]o-[C(R 1 )2] q are as described in more detail above.

In a preferred embodiment of the process according to the invention, a is 0 and U is present, i.e. a is 0 and, when U is [C(R 1 ) 2 ] n , n is an integer in the range of 1 to 24, or a is 0 and, when U is [C(R 1 ) 2 ]p-0-[C(R 1 )2C(R 1 ) 2 0]o-[C(R 1 )2] q , o is an integer in the range of 1 to 12 and/or p is 1 or 2 and/or q is 1 or 2. In other words, in this embodiment when U is [C(R 1 ) 2 ]p-0-[C(R 1 )2C(R 1 ) 2 0]o-[C(R 1 )2] q , at least one of o, p and q is not 0, and a is 0.

In yet another preferred embodiment of the process according to the invention, a is 1 and U is absent. U is absent when U is [C(R 1 ) 2 ]p-0-[C(R 1 )2C(R 1 ) 2 0]o-[C(R 1 )2] q and p, o and q are all 0, or when U is [C(R 1 ) 2 ] n and n is 0. In this embodiment, the sugar- derivative nucleotide Su(A)-Nuc is according to formula (10) as defined below.

In a preferred embodiment of the process according to the invention, the sugar- derivative nucleotide Su(A)-Nuc is according to formula (9) or (10):

10 wherein Nuc, A, T and U, and preferred embodiments thereof, are as defined above.

When Su(A)-Nuc is according to formula (9), U may be present or absent. As was described above, U is [C(R 1 ) 2 ]„ or [C(R 1 ) 2 ]p-0-[C(R 1 )2C(R 1 ) 2 0]o-[C(R 1 )2] q , and U is absent when n, p, o and q are all 0.

When Su(A)-Nuc is according to formula (9) and U is absent, it is preferred that A is selected from the group consisting of R 12 and R 13 . Preferred embodiments of R 12 and R 13 are described in more detail above. When U is absent in (9), it is particularly preferred that R 12 is -C(H)=CH 2 and that R 13 is -C(H)=C=CH 2 . Nuc is preferably UDP. In a particularly preferred embodiment, Su(A)-Nuc is according to formula (9), U is absent and A is R 13 , wherein R 13 is -C(H)=C=CH 2 . In a particularly preferred embodiment, Su(A)-Nuc is thus according to formula (44):

44 When Su(A)-Nuc is according to formula (9) and U is present, it is preferred that U is [C(R 1 )2] n - In this embodiment it is preferred that n is an integer in the range of 1 to 24, and, as described above, it is further preferred that n is in the range of 1 to 12, more preferably n is 1, 2, 3, 4, 5, 6, 7 or 8, even more preferably n is 1, 2, 3, 4, 5 or 6, yet even more preferably n is 1, 2, 3 or 4, yet even more preferably n is 1 or 2, and most preferably n is 1.

Also when Su(A)-Nuc is according to formula (9) and when U is [C(R 1 )2] n , preferred examples of the -[C(R 1 )2] n - moiety in (9) include, when n is 1 or 2, -(CH 2 )-, -(CF 2 )-, -(CC1 2 )-, -(CBr 2 )-, -(CMe 2 )-, -(CH 2 CH 2 )-, -(CH 2 CF 2 )-, -(CH 2 CC1 2 )-, -(CH 2 CBr 2 )-, -(CH 2 CI 2 )-, -(CH 2 CMe 2 )-, (CF 2 CF 2 )-, -(CC1 2 CC1 2 )-, -(CBr 2 CBr 2 )- and -(CMe 2 CMe 2 )-, and when n is 3 or more, -(C„H 2 „)-, -(C„F 2 „)-, -(C„C1 2 „)-, -(C„Br 2 „)-, -(C(n-i)H2(n-i)CF2)-, -(C( n -i)H2( n -i)CCl2)-, -(C( n -i)H2( n -i)CBr2)- and -(C( n -i)H2( n -i)CMe2)-, e.g. -(C 3 ¾)-, -(C 3 F 6 )-, -(C 3 C1 6 )-, -(C 3 Br 6 )-, -(CH 2 CH 2 CF 2 )-, -(CH 2 CH 2 CC1 2 )-, -(CH 2 CH 2 CBr 2 )-, -(C 4 H 8 )-, -(C 4 F 8 )-, -(C 4 C1 8 )- and -(C 4 Br 8 )-.

In a particularly preferred embodiment, when U is [C(R 1 ) 2 ] n , n is 1 or 2 and R 1 is H or F. Consequently, in a particularly preferred embodiment, U is [C(R 1 ) 2 ] n and the -[C(R 1 ) 2 ]„- moiety in (9) is -(CH 2 )-, -(CF 2 )-, -(CH 2 CH 2 )-, -(CF 2 CF 2 )- or -(CH 2 CF 2 )-.

When Su(A)-Nuc is according to formula (9), both when U is [C(R 1 ) 2 ] n and when U is [C(R 1 ) 2 ]p-0-[C(R 1 )2C(R 1 ) 2 0]o-[C(R 1 )2] q , it is preferred that A is selected from the group consisting of -N 3 , -C≡C-R 4 , -SH, -SC(0)R 8 , -SC(V)OR 8 , -X and -OS(0) 2 R 5 , wherein V, R 4 , R 5 , R 8 , and preferred embodiments thereof, are as defined above. X is F, CI, Br or I, and when A is X, it is preferred that X is CI or Br, and most preferably X is CI. More preferably, A is selected from the group consisting of -N 3 , -SH, -SC(0)CH 3 and -X, wherein X is preferably CI or Br, more preferably CI. It is further preferred that U is [C(R 1 ) 2 ]„.

Also when Su(A)-Nuc is according to formula (9), both when U is [C(R 1 ) 2 ] n and when U is [C(R 1 ) 2 ]p-0-[C(R 1 )2C(R 1 ) 2 0]o-[C(R 1 )2] q , it is preferred that Nuc is UDP. It is further preferred that U is [C(R 1 ) 2 ] n .

Several particularly preferred sugar-derivative nucleotides according to formula

(9) are shown below. In a preferred embodiment of the process according to the invention, Su(A)-Nuc is therefore according to formula (17), (18), (19), (20), (21) or (22):

20 21 22 wherein:

X is F, CI, Br or I;

V is O or S;

R 8 is an optionally substituted Ci - C24 alkyl group;

r is 0 or 1 ;

s is an integer in the range of 1 to 10;

t is an integer in the range of 1 to 10; and

u is an integer in the range of 0 to 10.

In (19), X is selected from the group consisting of F, CI, Br and I, preferably X is CI or Br, and more preferably X is CI.

In (20), both when r is 0 and when r is 1, preferably s is 1, 2, 3, 4, 5 or 6. More preferably, both when r is 0 and when r is 1, s is 1, 2, 3 or 4, even more preferably, both when r is 0 and when r is 1, s is 1, 2 or 3, and more preferably, both when r is 0 and when r is 1, s is 2 or 3. In particular when s is 2 or 3, subsequent conjugation of the modified glycoprotein obtained by this embodiment of the process according to the invention via a maleimide results in a particularly stable maleimide conjugate. Both when r is 0 and when r is 1, preferably, R 8 is an optionally substituted Ci - C 12 alkyl group, more preferably, both when r is 0 and when r is 1, R 8 is an optionally substituted Ci - C 6 alkyl group, and even more preferably, both when r is 0 and when r is 1, R 8 is methyl, ethyl, n-propyl, i-propyl, n-butyl, s-butyl or t-butyl. Most preferably, both when r is 0 and when r is 1, R 8 is methyl. In (20), it is particularly preferred, both when r is 0 and when r is 1, that R 8 is methyl and s is 1, 2, 3 or 4, more particularly preferred, both when r is 0 and when r is 1, that R 8 is methyl and s is 1, 2 or 3, and most preferred, both when r is 0 and when r is 1, that R 8 is methyl and s is 1. It is further preferred that r is 0.

In (21), preferably t is 1, 2, 3, 4, 5 or 6. More preferably, t is 1, 2, 3 or 4, even more preferably t is 1, 2 or 3 and most preferably t is 2 or 3. In particular when t is 2 or 3, subsequent conjugation of the modified glycoprotein obtained by this embodiment of the process according to the invention via a maleimide results in a particularly stable maleimide conjugate.

In (22), preferably u is 1, 2, 3, 4, 5 or 6. More preferably, u is 1, 2, 3 or 4, and most preferably u is 1 or 2.

When Su(A)-Nuc is according to formula (9) and U is [C(R 1 ) 2 ] p -0- [C(R 1 )2C(R 1 ) 2 0]o-[C(R 1 )2]q, several preferred sugar-derivative nucleotides according to formula (9) are shown below. In a preferred embodiment of the process according to the invention, Su(A)-Nuc is therefore according to formula (36), (37), (38), (39), (40), (41), (42) or (43): 61

When Su(A)-Nuc is according to formula (36), (37), (38), (39), (40), (41), (42) or (43), more preferably p and q are 1 or 2.

In another embodiment of the process according the invention, the sugar- derivative nucleotide Su(A)-Nuc (3) is according to formula (45), (46) or (47):

45 46 47 In this embodiment, A comprises an optionally substituted cyclopropenyl group or an optionally substituted cyclopropenylene group. More preferably, A comprises an optionally substituted C 3 - C 12 cyclopropenyl group or an optionally substituted C 3 - Ci2 cyclopropenylene group, and even more preferably an optionally substituted C 3 - C 8 cyclopropenyl group or an optionally substituted C 3 - C 8 cyclopropenylene group. Even more preferably, A comprises an optionally substituted C 4 - C 12 cyclopropenyl group or an optionally substituted C 4 - C 12 cyclopropenylene group, and yet even more preferably an optionally substituted C 4 - C 8 cyclopropenyl group or an optionally substituted C 4 - C 8 cyclopropenylene group.

When Su(A)-Nuc is according to formula (10) it is also preferred that Nuc is UDP. (Hetero)arylene group T and preferred embodiments of T in (10) are as described in more detail above for (3). Also in (10), T is optionally substituted with one or more, independently selected substituents R 2 . R 2 and preferred embodiments thereof in (10) are as decribed in more detail above for (3).

When Su(A)-Nuc is according to formula (10) it is preferred that A is selected from the group consisting of -N 3 , -C≡C-R 4 , -SH, -SC(0)R 8 , -SC(V)OR 8 and -OS(0) 2 R 5 , wherein V, R 4 , R 5 , R 8 , and preferred embodiments thereof, are as defined above. More preferably, A is selected from the group consisting of -N 3 , -SH and -SC(0)R 8 .

Several particularly preferred sugar-derivative nucleotides according to formula (10) are shown below. In a preferred embodiment of the process according to the invention, Su(A)-Nuc is therefore according to formula (11), (12), (13), (14), (15), (16), (34) or (35):

wherein:

Nuc, A and R 2 , and preferred embodiments thereof, are as defined above for (3); m is 0, 1, 2, 3 or 4; and

R 6 is selected from the group consisting of H and optionally substituted Ci - C 24 alkyl groups. In (11), m is 0, 1, 2, 3 or 4; in (12), m is 0, 1, 2 or 3; in (13), m is 0, 1, 2 or 3; in

(14), m is 0, 1 or 2; in (15), m is 0, 1 or 2; in(16), m is 0 or 1.

In (11), (12), (13), (14), (15) or (16) it is preferred that R 2 , when present, is independently selected from the group consisting of -F, -CI, -Br, -CN, -N0 2 , -C(0)R 9 , -C(0)OR 9 , -C(O)N(R 10 ) 2 , Ci - C 4 alkyl groups and Ci - C 4 alkoxy groups, wherein R 9 is a Ci - Ci 2 alkyl group, and wherein R 10 is independently selected from hydrogen and a Ci - Ci 2 alkyl group. Preferably, R 9 is a Ci - C 6 alkyl group, even more preferably a Ci - C 4 alkyl group, and most preferably a methyl, ethyl, n-propyl, i-propyl, n-butyl, s- butyl or a t-butyl group. Preferably, R 10 is a hydrogen or a Ci - C 6 alkyl group, more preferably hydrogen or a Ci - C 4 alkyl group, and most preferably R 10 is hydrogen, a methyl, ethyl, n-propyl, i-propyl, n-butyl, s-butyl or a t-butyl group. More preferably, R 2 , when present, is independently selected from the group consisting of -F, -CI, -Br, -CN, -N0 2 , methyl, methoxy, ethyl, ethoxy, n-propyl, n-propoxy, i-propyl, i-propoxy, n-butyl, n-butoxy, s-butyl, s-butoxy, t-butyl and t-butoxy. Even more preferably, R 2 , when present, is independently selected from the group consisting of -F, -CI, -Br, -CN, -N0 2 , methyl and methoxy. Most preferably, R 2 , when present is selected from the group consisting of F and CI. In a further preferred embodiment, m is 1 or 2. In another preferred embodiment, m is 0.

In (11), (12), (13), (14), (15) or (16) it is also preferred that A is selected from the group consisting of -N 3 , -C≡C-R 4 , -SH, -SC(0)R 8 , -SC(V)OR 8 and -OS(0) 2 R 5 , wherein V, R 4 , R 5 , R 8 , and preferred embodiments thereof, are as defined above. More preferably, A is selected from the group consisting of -N 3 , -SH and -SC(0)R 8 . It is furthermore preferred that Nuc is UDP.

In (13), it is preferred that R 6 is H or an optionally substituted Ci - Ci 2 alkyl group, more preferably R 6 is H or an optionally substituted Ci - C 6 alkyl group, even more preferably R 6 is H, a methyl group, an ethyl group, an n-propyl group, an i-propyl group, an n-butyl group, an s-butyl group or a t-butyl group, and most preferably R 6 is H or a methyl group. In a particularly preferred embodiment of Su(A)-Nuc (10), A is -N 3 , Nuc is UDP, m is 0, 1, 2, 3 or 4 and R 2 (when m is 1, 2, 3 or 4) is X, wherein X is F, CI, Br or I. In this embodiment it is further preferred that X is F or CI.

In a particularly preferred embodiment of the process according to the invention, Su(A)-Nuc is according to formula (23), (24) or (25):

23 24 25

X = F, CI, Br or I X = F, CI, Br or I

In (24) and (25), X may be the same or different. Preferably, X is the same. It is further preferred that X is F, CI or Br, more preferably F or CI. In a particularly preferred embodiment, in (24) and (25) X is F. In another particularly preferred embodiment, in (24) and (25) X is CI.

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, containing a suitable concentration of Mn 2+ or Mg 2+ ions. 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 15 to about 40 °C and most preferable in the range of about 20 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.

In a particular embodiment, when the process according to the invention is performed using a particular mutant -(l,4)-GalNAcT, e.g. CeGalNAcT(M312H), the process may also be performed in a suitable buffer solution containing Mg 2+ ions instead of Mn 2+ ions. The process according to the present invention has several advantages. First of all, the process is performed using a -(l,4)-GalNAcT enzyme. The enzyme may be a wild-type -(l,4)-GalNAcT, or a mutant thereof. A wide range of a -(l,4)-GalNAcTs, from different organisms, is available in nature. In addition, a wide range of β-(1,4)- GalNAcTs may be readily obtained from CHO by transient expression followed by a straightforward cation exchange column purification. In this way, enzymes are typically obtained with a purity of at least 75%.

In addition, the transferase activity of a -(l,4)-GalNAcT for the transfer of an unnatural UDP-GalNAc derivative, such as the sugar-derivatives Su(A) as described in more detail above, may be higher, or even significantly higher, than the transferase activity of the -(l,4)-galactosyltransf erase GalT(Y289L) mutant known from prior art.

For example, it was determined that in the process according to the invention, the transferase activity of several -(l,4)-GalNAcTs and mutants thereof is (significantly) higher for several sugar-derivatives Su(A). In particular CeGalNAcT was found to display a 10 times higher activity for the transfer of an azido-GalNAc derivative to a terminal GlcNAc-containing glycoprotein than GalT(Y289L), as becomes clear from Figure 7.

Figure 7 shows the activity plot of a range of different -(l,4)-GalNAcTs in comparison with the GalT(Y289L) mutant for the transfer of F 2 -GalNAz, from the sugar-derivative Su(A) according to formula (18), to GlcNAc, as determined by R&D systems glycosyltransferase activity kit as described in more detail above. From Figure

7 it can be clearly seen that CeGalNAcT, CeGalNAcT-His and TnGalNAcT have a transferase activity that is significantly higher than the transferase activity of GalT(Y289L) for the same process. Similarly, Figure 8 shows the activity plot of a range of different CeGalNAcT mutants, i.e. CeGalNAcT(Y257L), CeGalNAcT (Y257M) and CeGalNAcT(Y257A) for the transfer of F 2 -GalNAz from UDP-F 2 -GalNAz (18) to GlcNAc, as determined by R&D systems glycosyltransferase activity kit. Figure 8 clearly shows that also mutant -(l,4)-GalNAcTs CeGalNAcT(Y257L), CeGalNAcT (Y257M) and CeGalNAcT(Y257A) have a transferase activity that is significantly higher than the transferase activity of GalT(Y289L) for the same process.

Examples

Example 1. Selection and design of GalNAc-transf erases

Five specific sequences were selected for initial evaluation, in particular Uniprot accession numer: Q9GUM2 (C. elegans; identified herein as SEQ ID NO: 2), U1MEV9 (A. suum; identified herein as SEQ ID NO: 3), Q6J4T9 (T. ni; identified herein as SEQ ID NO: 4), Q7KN92 (D. melanogaster; identified herein as SEQ ID NO: 5) and Q6L9W6 (H. sapiens).

The following polypeptides were constructed based on deletion of the predicted cytoplasmatic domain and transmembrane domain. These polypeptides comprise the predicted :

C. elegans (CeGalNAcT [30-383] identified by SEQ ID NO: 6)

KIPSLYENLTIGSSTLIADVDAMEAVLGNTASTSDDLLDTWNSTFSPISEVNQTS FMEDIRPILFPDNQTLQFCNQTPPHLVGPIRVFLDEPDFKTLEKIYPDTHAGGHG MPKDCVARHRVAIIVPYRDREAHLRF LHNLHSLLAKQQLDYAIFIVEQVANQ TFNRGKLMNVGYDVASRLYPWQCFIFHDVDLLPEDDRNLYTCPIQPRHMSVAI DKFNYKLPYSAIFGGISALTKDHLKKINGFSNDFWGWGGEDDDLATRTSMAGL KVSRYPTQIARYKMIKHSTEATNPVNKCRYKFMGQTKRRWTRDGLSNLKYKL VNLELKPLYTRAVVDLLEKDCRRELRRDFPTCF A. suum (AsGalNAcT [30-383] identified by SEQ ID NO: 7)

DYSFWSPAFIISAPKTLTTLQPFSQSTSTNDLAVSALESVEFSMLDNSSILHASDN WTNDELVMRAQNENLQLCPMTPPALVGPIKVWMDAPSFAELERLYPFLEPGG HGMPTACRARHRVAIVVPYRDRESHLRTFLHNLHSLLTKQQLDYAIFVVEQTA NETFNRAKLMNVGYAEAIRLYDWRCFIFHDVDLLPEDDRNLYSCPDEPRHMS VAVDKFNYKLPYGSIFGGISALTREQFEGINGFS DYWGWGGEDDDLSTRVTL AGYKISRYPAEIARYKMIKHNSEKK PVNRCRYKLMSATKSRWR DGLSSLSY DLISLGRLPL YTHIK VDLLEKQ SRRYLRTHGFPTC T. ni (TnGalNAcT [33-421] identified by SEQ ID NO: 8)

SPLRT YL YTPL YNATQPTLRNVERL AANWPKKIP SNYIED SEEYSIKNISL SNHT TRASVVHPPSSITETASKLDKNMTIQDGAFAMISPTPLLITKLMDSIKSYVTTED GVKKAEAVVTLPLCDSMPPDLGPITLNKTELELEWVEKKFPEVEWGGRYSPPN CTARHRVAIIVPYRDRQQHLAIFLNHMHPFLMKQQIEYGIFIVEQEGNKDFNRA KLMNVGF VESQKL VAEGWQCF VFHDIDLLPLDTRNL YSCPRQPRHMS ASIDKL HFKLPYEDIFGGVSAMTLEQFTRVNGFSNKYWGWGGEDDDMSYRLKKINYHI ARYKMSI AR Y AMLDHKK S TPNPKRYQLL SQ T SKTF QKDGL S TLE YEL VQ V VQ YHLYTHILVNIDERS D. melanogaster (DmGalNAcT [47-403] identified by SEQ ID NO: 9)

HKYAHIYGNASSDGAGGSEASRLPASPLALSKDRERDQELNGGPNSTIRTVIAT ANF T SIPQDLTRFLLGTKKFLPPRQK S T S ALL ANC TDPDPRDGGPITPNTTLE SL DVIEAELGPLLRPGGAFEPENCNAQHHVAIVVPFRDRYAHLLLFLRNIHPFLMK QRIAYRIFIVEQTNGKPFNRAAMMNIGYLEALKLYQWDCFIFHDVDLLPLDDR NLYNCPRQPRHMSVAIDTLNFRLPYRSIFGGVSAMTREHFQAVNGFSNSFFGW GGEDDDMSNRLKHANLFISRYPVNIARYKMLKHQKEKANPKRYENLQNGMS KIEQDGINSIKYSIYSIKQFPTFTWYLAELKNSERKS

H. sapiens (HuGalNAcT [57-998] identified by SEQ ID NO: 24)

RYGSWRELAKALASRNIPAVDPHLQFYHPQRLSLEDHDIDQGVSSNSSYLKWN KPVPWLSEFRGRANLHVFEDWCGSSIQQLRRNLHFPLYPHIRTTLRKLAVSPK WTNYGLRIFGYLHPFTDGKIQFAIAADDNAEFWLSLDDQVSGLQLLASVGKTG KEWTAPGEFGKFRSQISKPVSLSASHRYYFEVLHKQNEEGTDHVEVAWRRNDP GAKFTIIDSLSLSLFTNETFLQMDEVGHIPQTAASHVDSSNALPRDEQPPADMLR PDPRDTLYRVPLIPKSHLRHVLPDCPYKPSYLVDGLPLQRYQGLRFVHLSFVYP ND YTRL SHMETHNKCF YQENAYYQDRF SFQEYIKIDQPEKQGLEQPGFEENLL EESQYGEVAEETPASNNQNARMLEGRQTPASTLEQDATDYRLRSLRKLLAQPR EGLLAPFSKRNSTASFPGRTSHIPVQQPEKRKQKPSPEPSQDSPHSDKWPPGHPV KNLPQMRGPRPRPAGDSPRKTQWLNQVESYIAEQRRGDRMRPQAPGRGWHG EEEVVAAAGQEGQVEGEEEGEEEEEEEDMSEVFEYVPVFDPVVNWDQTFSAR NLDFQ ALRTD WIDL S CNT S GNLLLPEQE ALE VTRVFLKKLNQRSRGR YQLQRI VNVEKRQDQLRGGRYLLELELLEQGQRVVRLSEYVSARGWQGIDPAGGEEVE ARNLQGLWDPHNRRRQVLNTRAQEPKLCWPQGFSWSHRAVVHFVVPVKNQ ARWVQQFIKDMENLFQVTGDPHFNIVITDYSSEDMDVEMALKRSKLRSYQYV KLSGNFERSAGLQAGIDLVKDPHSIIFLCDLHIHFPAGVIDAIRKHCVEGKMAFA PMVMRLHCGATPQWPEGYWEVNGFGLLGIYKSDLDRIGGMNTKEFRDRWGG EDWELLDRILQGLDVERLSLRNFFHHFHSKRGMWSRRQMKTL

In addition, polypeptide variants containing an N-terminal His-tag were constructed for AsGalNAcT(30-383): (His 6 -AsGalNAcT(30-383) identified by SEQ ID NO: 71) and for TnGalNAcT(33-421) (His 6 -TnGalNAcT(33-421) identified by SEQ ID NO: 49). Example 2. Design of C. elegans GalNAcT mutants of 1257

Based on a sequence alignment of CeGalNAcT with GalT, three active site mutants were designed by mutation of isoleucine257 to leucine, methionine or alanine (underlined). CeGalNacT(30-3 3; I257L) identified by SEQ ID NO: 10

KIPSLYENLTIGSSTLIADVDAMEAVLGNTASTSDDLLDTWNSTFSPISEVNQTS FMEDIRPILFPDNQTLQFCNQTPPHLVGPIRVFLDEPDFKTLEKIYPDTHAGGHG MPKDCVARimVAIIWYRDREAHLRIMLHNLHSLLAKQQLDYAIFIVEQVANQ TFNRGKLMNVGYDVASRLYPWQCFIFHDVDLLPEDDRNLYTCPIQPRHMSVAI DKFNYKLPYSALFGGISALTKDHLKKINGFSNDFWGWGGEDDDLATRTSMAG LKVSRYPTQIARYKMIKHSTEATNPVNKCRYKIMGQTKRRWTRDGLSNLKYK LVNLELKPLYTRAVVDLLEKDCRRELRRDFPTCF

CeGalNAcT(30-3S3; I257M) identified by SEQ ID NO: 11

KIPSLYENLTIGSSTLIADVDAMEAVLGNTASTSDDLLDTWNSTFSPISEVNQTS FMEDIRPILFPDNQTLQFCNQTPPHLVGPIRVFLDEPDFKTLEKIYPDTHAGGHG MPKDCVARHRVAIIVPYRDREAHLRIMLHNLHSLLAKQQLDYAIFIVEQVANQ TFNRGKLMNVGYDVASRLYPWQCFIFHDVDLLPEDDRNLYTCPIQPRHMSVAI DKFNYKLPYSAMFGGISALTKDHLKKINGFS DFWGWGGEDDDLATRTSMAG

LKVSRYPTQIARYKMIKHSTEAT PV KCRYKF GQTKRRWTRDGLS LKYK

LVNLELKPLYTRAVVDLLEKDCRRELRRDFPTCF CeGalNacTft 0-383; 1257 A ) identified by SEQ ID NO: 12

KIPSLYE LTIGSSTLIADVDAMEAVLGNTASTSDDLLDTWNSTFSPISEVNQTS FMEDIRPILFPDNQTLQFCNQTPPHLVGPIRVFLDEPDFKTLEKIYPDTHAGGHG MPKDCVARHRVAIIVPYRDREAHLRIMLHNLHSLLAKQQLDYAIFIVEQVANQ TF RGKLMNVGYDVASRLYPWQCFIFHDVDLLPEDDR LYTCPIQPRHMSVAI DKFNYKLPYSAAFGGISALTKDHLKKINGFS DFWGWGGEDDDLATRTSMAG LKVSRYPTQIARYKMIKHSTEAT PV KCRYKF GQTKRRWTRDGLS LKYK LVNLELKPLYTRAVVDLLEKDCRRELRRDFPTCF

Example 3. Design of C. elegans GalNAcT mutant ofM312

A CeGalNAcT mutant was designed by mutation of methionines 12 to histidine.

CeGalNacT(30-383 M312H) identified by SEQ ID NO: 13

KIPSLYE LTIGSSTLIADVDAMEAVLGNTASTSDDLLDTWNSTFSPISEVNQTS FMEDIRPILFPDNQTLQFCNQTPPHLVGPIRVFLDEPDFKTLEKIYPDTHAGGHG MPKDCVARHRVAIIVPYRDREAHLRIMLHNLHSLLAKQQLDYAIFIVEQVANQ TF RGKLMNVGYDVASRLYPWQCFIFHDVDLLPEDDR LYTCPIQPRHMSVAI DKFNYKLPYSAIFGGISALTKDHLKKINGFS DFWGWGGEDDDLATRTSMAGL KVSRYPTQIARYKHIKHSTEAT PVNKCRYKIMGQTKRRWTRDGLS LKYKL VNLELKPLYTRAVVDLLEKDCRRELRRDFPTCF

Example 4. Design CeGalNAcT with C-terminal Hisg-tag

A CeGalNAcT with a C-terminal His 6 -tag was designed (CeGalNAcT-Hise).

CeGalNAcT(30-383)-His 6 ) identified by SEQ ID NO: 14

KIPSLYENLTIGSSTLIADVDAMEAVLGNTASTSDDLLDTWNSTFSPISEVNQTS FMEDIRPILFPDNQTLQFCNQTPPHLVGPIRVFLDEPDFKTLEKIYPDTHAGGHG MPKDCVARHRVAIIVPYRDREAHLRIMLHNLHSLLAKQQLDYAIFIVEQVANQ TFNRGKLMNVGYDVASRLYPWQCFIFHDVDLLPEDDRNLYTCPIQPRHMSVAI DKFNYKLPYSAIFGGISALTKDHLKKINGFS DFWGWGGEDDDLATRTSMAGL

KVSRYPTQIARYKMIKHSTEAT PVNKCRYKF GQTKRRWTRDGLS LKYKL

VNLELKPLYTRAVVDLLEKDCRRELRRDFPTCFHHHHHH Example 5. Transient expression of enzymes in CHO

Proteins were transiently expressed in CHO Kl cells by Evitria (Zurich, Switzerland) at 20 mL scale. All GalNAcTs except HuGalNAcTs were successfully expressed, as visualized in Figure 5 and Figure 6.

Figure 5 shows the SDS-PAGE of a range of -(l,4)-GalNAcTs, crude after transient expression in CHO. Lanes 2-6 reduced enzymes, lanes 7-11, same enzymes non- reduced. Lane 1 : Marker, Biorad precision plus protein standard; MW from top to bottom: 250kDa, 150kDa, lOOkDa, 75kDa, 50kDa, 37kDa, 25kDa, 20kDa. Lane 2+7: TnGalNAc. Lane 3+8: DmGalNAcT. Lane 4+9: AsGalNAcT. Lane 5+10: CeGalNAcT with C-terminal His 6 -tag. Lane 6+11 : huGalNAcT. Protein bands visible between 50 and 55 kDa are the desired GalNAcTs, all variants expression successful except huGalNAcT.

Figure 6 shows the non-reducing SDS-PAGE of a range of -(l,4)-CeGalNAcT mutants. Lane 1 : Marker: GE rainbow molecular weight marker; MW from top to bottom: 225kDa, 150kDa, 102kDa, 76kDa, 52kDa, 38kDa, 33 kDa, 24kDa, 17kDa. Lane 2: CeGalNAcT(M312H). Lane 3 : CeGalNAcT(M312H). Lane 4: CeGalNAcT(I257A). Lane 5: CeGalNAcT(I257A). Lane 6: CeGalNAcT(I257L). Lane 7: CeGalNAcT(I257L). Lane 8: CeGalNAcT(I257M). Lane 9: CeGalNAcT(I257L). Lane 10: CeGalNAcT with His 6 -tag. Protein bands visible at ~52kDa are the monomeric CeGalNAcT species and the bands visible at ~102kDa are the dimeric CeGalNAcT species. Protein bands at higher MW (>225kDa) were not characterized.

Example 6. Purification protocol for the non-His tagged GalNAcT proteins

Purification protocol was based on cation exchange on a SP column (GE Healthcare) followed by size exclusion.

In a typical purification experiment, CHO-produced supernatant containing the expressed GalNAcT was dialyzed against 20 mM Tris buffer, pH 7.5. The supernatant (typically 25 mL) was filtered through a 0.45 μΜ-pore diameter filter and subsequently purified over a cation exchange column (SP column, 5 mL, GE Healthcare), which was equilibrated with 20 mM Tris buffer, pH 7.5 prior to use. Purification was performed on an AKTA Prime chromatography system equipped with an external fraction collector. Samples were loaded from system pump A. The non-bound proteins were eluted from the column by washing the column with 10 column volumes (CV) of 20 mM Tris buffer, pH 7.5. Retained protein was eluted with elution buffer (20 mM Tris, 1 NaCl, pH 7.5; 10 mL). Collected fractions were analyzed by SDS-PAGE on polyacrylamide gels (12%), and fractions containing the target protein were combined and concentrated using spin filtration to a volume of 0.5 mL. Next the protein was purified on a preparative Superdex size exclusion chromatography column, on an AKTA purifier system (UNICORN v6.3). This purification step led to the identification and separation of a dimer, and a monomer fraction of target protein. Both fractions were analyzed by SDS-PAGE and stored at -80 °C prior to further use

Example 7. Purification of CeGalNAcT-Hise

In a typical purification experiment, CHO supernatant was filtered through a 0.45 μΜ- pore diameter filter and applied to a Ni-NTA column (GE Healthcare, 5 mL), which was equilibrated with buffer A (20 mM Tris buffer, 20 mM imidazole, 500 mM NaCl, pH 7.5) prior to use. Before filtration, imidazole was added to the CHO supernatant to a final concentration of 20 mM in order to minimize unspecific binding to the column. The column was first washed with buffer A (50 mL). Retained protein was eluted with buffer B (20 mM Tris, 500 mM NaCl, 250 mM imidazole, pH 7.5, 10 mL). Fractions were analyzed by SDS-PAGE on polyacrylamide gels (12%), and the fractions that contained purified target protein were combined and the buffer was exchanged against 20 mM Tris (pH 7.5) by dialysis performed overnight at 4 °C. The purified protein was stored at -80 °C prior to further use. Note: for the identification of the monomelic and dimeric CeGalNAcT-His 6 species an additional SEC purification was performed (as described above).

Example 8. Synthesis of ethyl 2-azido-2,2-difluoroacetate

To a solution of ethyl 2-bromo-2,2-difluoroacetate (950 mg, 4.68 mmol) in dry DMSO (5 mL) was added sodium azide (365 mg, 5.62 mmol). After stirring overnight at room temperature, the reaction mixture was poured out into water (150 mL). The layers were separated, dichloromethane was added to the organic layer and the layer was dried over sodium sulfate. After filtration, the solvent was removed under reduced pressure (300 mbar) at 35 °C affording the crude ethyl 2-azido-2,2-difluoroacetate (250 mg, 1.51 mmol, 32%).

1H- MR (300 MHz, CDC1 3 ): δ 4.41 (q, J= 7.2 hz, 2H), 1.38 (t, J= 6.9 Hz, 3H).

Example 9. Synthesis of a-2-azido-2-deoxy-3, 4, 6-tri-O-acetyl-D-galactose 1- phosphate

2-Azido-2-deoxy-3,4,6-tri-O-acetyl-D-galactose 1-phosphate was prepared from D- galactosamine according to procedures described for D-glucosamine in Linhardt et al, J. Org. Chem. 2012, 77, 1449-1456, incorporated by reference.

1H- MR (300 MHz, CD 3 OD): δ 5.69 (dd, J= 7.2, 3.3 Hz, 1H), 5.43-5.42 (m, 1H), 5.35 (dd, J = 11.1, 3.3 Hz, 1H), 4.53 (t, J = 7.2 Hz, 1H), 4.21-4.13 (m, 1H), 4.07-4.00 (m, 1H), 3.82 (dt, J= 10.8, 2.7 Hz, 1H), 2.12 (s, 3H), 2.00 (s, 3H), 1.99 (s, 3H).

LRMS (ESI-) calcd for Ci 2 Hi 7 N 3 0nP ( -H + ) 410.06, found 410.00.

Example 10. Synthesis of a-2-amino-3, 4, 6-tri-O-acetyl-D-galactose 1-phosphate To a solution of a-2-azido-2-deoxy-3,4,6-tri-0-acetyl-D-galactose (105 mg, 0.255 mmol) in MeOH (3 mL) was added Pd/C (20 mg). The reaction was stirred under a hydrogen 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 the free amine (94 mg, 0.244 mmol, 96%).

1H- MR (300 MHz, D 2 0): 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 (ESI-) calcd for Ci 2 Hi 9 NOnP ( -H + ) 384.07, found 384.10.

Example 11. Synthesis of -(2'-azido-2',2'-difluoroacetamido)-3, 4, 6-tri-O-acetyl-D- galactose 1-phosphate

To a solution of a-2-amino-3,4,6-tri-0-acetyl-D-galactose 1-phosphate (94 mg, 0.244 mmol) in dry DMF (3 mL), were added ethyl difluoroazidoacetate (48 mg, 0.293 mmol) and Et 3 N (68 μΐ., 0.488 mmol). The reaction was stirred for 6 h, followed by concentration in vacuo to afford the crude product. Flash chromatography (100:0 - 50:50 EtOAc:MeOH) afforded a-(2'-azido-2',2'-difluoroacetamido)-3,4,6-tri-0-acetyl- D-galactose 1-phosphate (63 mg, 0.125 mmol, 51%). Example 12. Synthesis of UDP-a-(2'-azido-2',2'-difluoroacetamido)-3,4,6-tri-0-acetyl- D-galactose

a-(2'-Azido-2',2'-difluoroacetamido)-3,4,6-tri-0-acetyl-D -galactose 1 -phosphate was coupled to UMP according to Baisch et al. Bioorg. Med. Chem., 1997, 5, 383-391, incorporated by reference).

Thus, a solution of D-uridine-5 '-monophosphate disodium salt (98 mg, 0.266 mmol) in H 2 0 (1 mL) was treated with DOWEX 50Wx8 (H + form) for 40 minutes and filtered. The filtrate was stirred vigorously at r.t. while tributylamine (63 μΐ., 0.266 mmol) was added dropwise. After 30 minutes of further stirring, the reaction mixture was lyophilized and further dried over P 2 0 5 under vacuum for 5 h.

The resulting tributylammonium uridine-5 '-monophosphate was dissolved in dry DMF (15 mL) under an argon atmosphere. Carbonyl diimidazole (35 mg, 0.219 mmol) was added and the reaction mixture was stirred at r.t. for 30 min. Next, dry MeOH (4.63 was added and stirred for 15 min to remove the excess carbonyl diimidazole. The leftover MeOH was removed under high vacuum for 15 min. Subsequently, N- methylimidazole, HC1 salt (61 mg, 0.52 mmol) was added to the reaction mixture and the resulting compound (63 mg, 0.125 mmol) was dissolved in dry DMF (15 mL) and added dropwise to the reaction mixture. The reaction was allowed to stir at r.t. for o.n. 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:H 2 0) afforded UDP-a-(2'-azido-2',2'-difluoroacetamido)-3,4,6-tri-0-acetyl- D-galactose. 1H-NMR (300 MHz, D 2 0): δ 7.87 (d, J= 8.1 Hz, 1H), 5.913-5.85 (m, 2H), 5.67 (dd, J = 6.6, 2.7 Hz, 1H), 5.56-5.50 (m, 1H), 5.47-5.43 (m, 1H), 5.31-5.25 (m, 2H), 4.61-4.43 (m, 2H), 4.31-4.05 (m, 5H), 2.16 (s, 3H), 2.02 (s, 3H), 1.94 (s, 3H).

LRMS (ESI-) calcd for C 2 3H 29 F 2 N 6 0 2 oP 2 ( -H + ) 809.09, found 809.1.

1H-NMR (300 MHz, CD 3 OD): δ 5.64 (m, 1H), 5.47 (d, J = 2.4 Hz, 1H), 5.35 (dd, J = 11.4, 3.0 Hz, 1H), 4.58-4.48 (m, 2H), 4.25-4.15 (m, 1H), 4.09-4.00 (m, 1H), 2.14 (s, 3H), 2.00 (s, 3H), 1.93 (s, 3H).

LRMS (ESI-) calcd for Ci 4 Hi 8 F 2 N 4 0i 2 P ( -H + ) 503.06, found 503.0. Example 13. Synthesis of a-UDP-2-(2'-azido-2',2'-difluoroacetamido)-2-deoxy-D- galactose (UDP-F 2 -GalNAz, 18)

Deacetylation of UDP-a-(2'-azido-2',2'-difluoroacetamido)-3,4,6-tri-0-acetyl- D- galactose was performed according to Kiso et al, Glycoconj. J, 2006, 23, 565, incorporated by reference.

Thus, UDP-a-(2'-azido-2',2'-difluoroacetamido)-3,4,6-tri-0-acetyl- D-galactose was dissolved in H 2 0 (1 mL) and triethylamine (1 mL) and MeOH (2.4 mL) were added. The reaction mixture was stirred for 2 h and then concentrated in vacuo. Flash chromatography (7:2: 1-5:2: 1 EtOAc:MeOH:H 2 0) afforded a-UDP-2-(2'-azido-2',2'- difluoroacetamido)-2-deoxy-D-galactose (18).

1H- MR (300 MHz, D 2 0): δ 7.86 (d, J= 8.1 Hz, 1H), 5.91-5.85 (m, 2H), 5.54 (dd, J =

6.6, 3.6 Hz, 1H), 4.31-3.95 (m, 9H), 3.74-3.62 (m, 2H).

LRMS (ESI-) calcd for Ci7H 23 F 2 N 6 Oi7P 2 ( -H + ) 683.06, found 683.10. Example 14. Synthesis of a-UDP-2-azido-2-deoxy-3,4, 6-tri-0-acetyl-D-galactose a-2-Azido-2-deoxy-3,4,6-tri-0-acetyl-D-galactose 1-phosphate, as prepared in example 9, was attached 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 H 2 0 (15 mL) was treated with DOWEX 50Wx8 (H + form) for 30 minutes and filtered. The filtrate was stirred vigorously at room temperature 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 P 2 0 5 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 μΐ,) was added and stirred for 15 min to remove the excess carbonyldiimidazole. The leftover MeOH was removed under high vacuum for 15 min. The resulting compound (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:H 2 0) afforded a-UDP-2-azido-2-deoxy- 3,4,6-tri-O-acetyl-D-galactose (1.08 g, 1.51 mmol, 37%). 1H- MR (300 MHz, D 2 0): 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, 1 1.0, 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.

Example 15. Synthesis of a-UDP-2-azido-2-deoxy-D-galactose

Deacetylation of a-UDP-2-azido-2-deoxy-3,4,6-tri-O-acetyl-D-galactose, as prepared in example 14, was performed according to Kiso et al, Glycoconj. J, 2006, 23, 565. Thus, a-UDP-2-azido-2-deoxy-3,4,6-tri-0-acetyl-D-galactose (222 mg, 0.309 mmol) was dissolved in H 2 0 (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 a-UDP-2-azido-2-deoxy-D-galactose. 1 H- MR (300 MHz, D 2 0): 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 (EST) calcd for Ci5H 23 N 5 Oi 6 P 2 ( -H + ) 590.05, found 590.2.

Example 16. Synthesis of a-UDP-D-galactosamine (Gal-NH 2 )

To a solution of a-UDP-2-azido-2-deoxy-D-galactose, as prepared in example 15, in H 2 0:MeOH 1 : 1 (4 mL) was added Lindlar' s catalyst (50 mg). The reaction was stirred under a hydrogen atmosphere for 5 h and filtered over celite. The filter was rinsed with H 2 0 (10 ml) and the filtrate was concentrated in vacuo to afford the a-UDP-D- galactosamine (UDP-Gal H 2 ) (169 mg, 0.286 mmol, 92% yield over two steps). 1H- MR (300 MHz, D 2 0): 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 Ci5H 25 N 3 Oi 6 P 2 ( -H + ) 564.06, found 564.10.

Example 17. Synthesis of -UDP-N-(4'-azido-3',5'-difluorobenzoyl)-D-galactosamine (24 with X=F)

4-Azido-3,5-difluorobenzoic acid succinimidyl ester was prepared according to the procedure for pent-4-ynoic acid succinimidyl ester according to Rademann et al , Angew. Chem. Int. Ed , 2012, 51, 9441-9447, incorporated by reference. Thus, to a solution of 4-azido-3,5-difluorobenzoic acid was added dicyclohexylcarbodiimide (1.1 equiv) and N-hydroxysuccinimide (1.2 equiv) and the resulting suspension was stirred overnight followed by vacuum filtration. The filtrate was concentrated and dissolved in EtOAc followed by washing with saturated NaHC0 3 and brine. The organic layer was dried over Na 2 SC"4, filtrated and concentrated in vacuo to use crude in the next reaction.

1H- MR (300 MHz, CDC1 3 ): δ 7.74-7.66 (m, 2H), 2.91 (s, 4H).

Next, UDP-GalNH 2 as prepared in example 16 (30 mg, 0.0531 mmol) was dissolved in 0.1 M NaHC0 3 (0.2 M) and the N-hydroxysuccinimide ester of 4-azido-3,5- difluorobenzoic acid (31 mg, 0.106 mmol, 2 equiv.), dissolved in DMF (0.2 M), was added. The reaction was stirred overnight at r.t. and concentrated in vacuo. Flash chromatography (7:2: 1-5:2: 1 EtOAc:MeOH:H 2 0) afforded the product 24 (with X=F) (8 mg, 0.0107 mmol, 20 %).

1H-NMR (300 MHz, D 2 0): δ 7.73 (d, J= 8.4 Hz, 1H), 7.52-7.31 (m, 2H), 5.87-5.71 (m, 2H), 5.65-5.57 (m, 1H), 5.47-5.33 (m, 1H), 4.43-3.96 (m, 8H), 3.76-3.60 (m, 2H). LRMS (EST) calcd for C 22 H 25 F 2 N 6 Oi 7 P 2 ( -H + ) 745.07, found 744.9.

Example 18. Determination of specific activities of enzymes

Specific activity of enzymes was determined by a coupled glycosyltransferase procedure as described by Wu et al. in Glycobiology 2010, 27, 723-733, incorporated by reference, and commercially available as a kit from R&D systems: http://www.rndsystems.com/Products/EA001 (accessed July 31st, 2014).

In short, a glycosyltransferase reaction was carried out in 50 μΙ_, of reaction buffer (25 mM Tris, 150 mM NaCl, 5 mM MgCl 2 and 5 mM MnCl 2 , pH 7.5) in a 96-well plate at room temperature for 20 min. To determine the kinetic parameters of a glycosyltransferase, multiple reactions with varying amounts of the enzyme were carried out simultaneously (0.01, 0.02, 0.04, 0.06, 0.08, 0.10 and 0.12 μg of enzyme) in the presence of fixed amounts of all other components (GlcNAc: 20 mM, UDP-F 2 - GalNAz: 500 μΜ), including the coupling phosphatase 1 (ENTPD3/CD39L3) (2.5 μΐ, of 20 ng/μΕ solution). A well containing all components except for the enzyme served as a blank control. The reactions were initiated by adding the substrates and phosphatase to the enzyme and terminated by the addition of 30 μΙ_, of Malachite reagent A and 100 μΙ_, of water to each well. The color was developed by the addition of 30 μΙ_, of Malachite reagent B to each well followed by gentle mixing and incubation at room temperature for 20 min. Following color development, the plate was read at 620 nm with a multi-well plate reader. A phosphate standard curve was also performed to determine the conversion factor between the absorbance and the inorganic phosphate contents. Preparation of UDP-F 2 -GalNAz is described in Example 13.

The specific activity of the GalNAcTs described herein were compared to GalT(Y289L), an enzyme known to transfer UDP-GalNAz as previously disclosed in WO 2007/095506 and WO 2008/029281 (both Invitrogen Corporation).

The data collected from the determination of the specific activity of several enzymes is shown in Table 1 and Table 2.

Table 1: Data from the determination of the specific activity of CeGalNAcT, CeGalNAcT-His, TnGalNAcT and GalT(Y289L).

Table 2: Data from the determination of the specific activity of CeGalNAcT(I257L), CeGalNAcT(I257A) and CeGalNAcT (I257M).

enzyme ^g) CeGalNAcT CeGalNAcT CeGalNAcT

(I257L) (I257A) (I257M)

0.01 0.02 0.009 0.0130

0.02 0.038 0.032 0.0230

0.04 0.081 0.049 0.0470

0.06 0.136 0.078 0.0680 0.08 0.153 0.097 0.0880

0.1 0.205 0.119 0.1220

0.12 0.245 0.152

specific activity

381 240 146

(pmol/min^g)

A graph depicting the conversion (in pmol/min^g) plotted against the amount of enzyme is shown in Figure 7 (for Table 1) and Figure 8 (for Table 2). From these plots specific activities were calculated by linear regression.

Example 19. Activity determination of CeGalNAcT 4312H)

Activity of CeGalNAcT(M312H) was also measured with the same procedure as described in example 14, however in this cases Mg 2+ was used instead of Mn 2+ . In this case, a specific activity of 15 pmol/min^g was determined.

Example 20. Trimming of IgG glycans with Endo S (general protocol)

Trimming of IgG glycans was performed using Endo S from Streptococcus pyogenes (commercially available from Genovis, Lund, Sweden). The IgG (10 mg/mL) was incubated with Endo S (40 U/mL) in 25 mM Tris pH 8.0 for approximately 16 hours at 37 °C. The deglycosylated IgG was concentrated and washed with 10 mM MnCl 2 and 25 mM Tris-HCl pH 8.0 using an Amicon Ultra-0.5, Ultracel-10 Membrane (Millipore).

Example 21. Trimming of trastuzumab

Trastuzumab was subjected to the trimming protocol above. Analysis of the trimmed antibody was performed on a JEOL AccuToF equipped with an Agilent 1100 UPLC. Samples (2 μΐ.) were reduced with DTT (2 μΐ.) during 10 min and subsequently diluted with H 2 0 (40 μΐ.) prior to injection. 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 (49496 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. This is an example of a glycoprotein comprising a glycan according to formula (1). Example 22. Expression of trastuzumab in presence of swainsonine

Trastuzumab was transiently expressed in CHO Kl cells by Evitria (Zurich, Switzerland) in the presence of 10 or 25 μg/mL swainsonine (commercially available from Sigma-Aldrich), purified using protein A sepharose and analyzed by mass spectrometry. Both concentrations of swainsonine gave three major heavy chain products of trastzumab which correspond to the trastuzumab heavy chain substituted with GlcNAc-Man 5 -GlcNAc-GlcNAc(Fuc)- (c=d=0, 50712 Da, ±20% of total heavy chain product), Gal-GlcNAc-Man 5 -GlcNAc-GlcNAc(Fuc)- (c=l, d=0, 50874 Da, ±35% of total heavy chain product), and Sial-Gal-GlcNAc-Man 5 -GlcNAc-GlcNAc(Fuc)- (c=d=l, 51164 Da, ±35% of total heavy chain product).

Example 23. Trimming with sialidase/galactosidase to give trast-MansGlcNAc

Trastuzumab transiently expressed in the presence of swainsonine as described in Example 18 (10 mg/mL) was incubated with neuraminidase (0.5 mU/mg IgG) from Vibrio cholerae (commercially available from Sigma-aldrich) in 100 mM sodium acetate pH 6.0 and 2 mM CaCl 2 for 16 hrs, which led to complete removal of the sialic acid (two major heavy chain products of 50712 and 50874 Da which correspond to approximately 20 and 70% of the total heavy chain products). When the same reaction was performed in the presence of P(l,4)-galactosidase (3 mU/mg IgG) from Streptococcus pneumoniae (commercially available from Calbiochem), a single major heavy chain product was observed corresponding to trastuzumab with a GlcNAc-Man 5 - GlcNAc-GlcNAc(Fuc)-substituted heavy chain (c=d=0, 50712 Da, ±90% of total heavy chain product, and minor heavy chain products between 50700 and 50900 Da).

This is an example of a glycoprotein comprising a glycan according to formula (26).

Example 24. Trimming with galactosidase to give trast-Man3GlcNAc2

Trastuzumab (10 mg/mL) in 50 mM sodium phosphate pH 6.0 and P(l,4)-galactosidase (3 mU/mg IgG) from Streptococcus pneumoniae (commercially available from Calbiochem) was stirred for 16 hrs at 37 °C prior to analysis by MS. A single major heavy chain product was observed corresponding to trastuzumab with a GlcNAc 2 - Man 3 -GlcNAc-GlcNAc(Fuc)-substituted heavy chain (c=d=0, 50592 Da).

This is an example of a glycoprotein comprising a glycan according to formula (27). General protocol for mass spectral analysis of IgG

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 μΐ. 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.

Glycosyltransfer of galactose derivative (e.g. azidosugar) with GalNAcT (general protocol)

Enzymatic introduction of galactose derivative (e.g. azido-containing sugar) onto IgG was effected with a GalNAc-transferase or a mutant thereof. The deglycosylated IgG (prepared as described above, 10 mg/mL) was incubated with a modified UDP- galactose derivative (e.g. an azido-modified sugar-UDP derivative) (0.4 mM) and GalNAcT (1 mg/mL) in 10 mM MnCl 2 and 25 mM Tris-HCl pH 8.0 for 16 hours at 30 °C. The functionalized IgG (e.g. azido-functionalized 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 15-20 mg/mL.

Example 25. Trastuzumab(GalNAz) 2

Trimmed trastuzumab was subjected to the glycosyltransfer protocol with UDP-N- azidoacetylgalactosamine (UDP-GalNAz) and CeGalNAcT. After protein A affinity purification, a small sample was reduced with DTT and subsequently subjected to MS analysis indicating the formation of a one major product of (49713 Da, 90% of total heavy chain), resulting from GalNAz transfer to core GlcNAc(Fuc) substituted trastuzumab, and a minor product (49566 Da, ±10% of total heavy chain), resulting from GalNAz transfer to core GlcNAc substituted trastuzumab. Example 26. Trastuzumab(F 2-GalNAz) 2

Trimmed trastuzumab was subjected to the glycosyltransfer protocol with UDP-N- azidodifluoroacetylgalactosamine (UDP-F 2 -GalNAz) and a GalNAcT, with the GalNAcT selected from CeGalNAcT (or a mutant thereof as described above), AsGalNAcT, TnGalNAcT or DmGalNAcT. After protein A affinity purification a small sample was reduced with DTT and subsequently subjected to MS analysis indicating the formation of one major heavy chain product (49865 Da, approximately 90% of total heavy chain), resulting from F 2 -GalNAz transfer to core GlcNAc(Fuc)- substituted trastuzumab which has reacted with DTT during sample preparation.

Example 27. Trastuzumab-(F 2-GalNBAz) 2

Trimmed trastuzumab (10 mg/mL, 6.6 nmol), obtained by Endo S treatment of trastuzumab as in Formula 1, was incubated with UDP-F 2 Gal BAz (24 with X=F, 7 mM) and CeGalNAcT (2 mg/mL) in 10 mM MnCl 2 and 25 mM Tris-HCl pH 8.0 at 30 °C overnight. Mass spectral analysis of the reduced sample indicated the formation of a one major product (49815 Da, approximately 90% of total heavy chain), resulting from F 2 -GalNBAz transfer to core GlcNAc(Fuc) substituted trastuzumab heavy chain. Example 28. Trastuzumab-(MansGlcNAc-GalNAz)2

GlcNAc-Man 5 -GlcNAc-GlcNAc(Fuc)-substituted trastuzumab (as in Formula 26, obtained by transient expression of trastuzumab in the presence of swainsonine and trimmed with neuraminidase and galactosidase as described in Example 19) was subjected to the glycosyltransfer protocol with UDP-GalNAz and GalNAcT. The trimmed antibody was incubated with UDP-GalNAz (0.5 mM) (commercially available from Glycohub, Inc) and CeGalNAcT (0.1 mg/mL) in 10 mM MnCl 2 and 25 mM Tris- HCl pH 8.0 for 16 hours at 30 °C, which led to complete conversion into GalNAz- GlcNAc-Man 5 -GlcNAc-GlcNAc(Fuc)-substituted trastuzumab (major heavy chain product of 50929 Da, ±90% of the total heavy chain products).

Example 29. Trastuzumab-(Mani(GlcNAc-GalNAz) 2) 2

Trastuzumab obtained after trimming with galactosidase (as in Formula 27, preparation described in Example 24) was subjected to the glycosyltransfer protocol with UDP- GalNAz and GalNAcT. The trimmed antibody was incubated with UDP-GalNAz (0.5 mM) (commercially available from Glycohub, Inc) and CeGalNAcT (0.1 mg/mL) in 10 mM MnCl 2 and 25 mM Tris-HCl pH 8.0 for 16 hours at 22 °C, which led to complete conversion into (GalNAz-GlcNAc) 2 -Man 3 -GlcNAc-GlcNAc(Fuc)-substituted trastuzumab (major heavy chain product of 51027 Da, ±90% of the total heavy chain products).

Example 30. Trastuzumab (F ^-GalNAz) '2 using CeGalNAcT(M312H) andMg 2+

Trimmed trastuzumab was subjected to the glycosyltransfer protocol with UDP-N- azidodifluoroacetylgalactosamine (UDP-F 2 -GalNAz) and a CeGalNAcT(M312H), with the GalNAcT (1 mg/mL) in the presence of 10 mM MgCl 2 . After protein A affinity purification, a small sample was reduced with DTT and subsequently subjected to MS analysis indicating the formation of one major heavy chain product (49873 Da, approximately 25% of total heavy chain, rest is remaining trimmed antibody), resulting from F 2 -GalNAz transfer to core GlcNAc(Fuc)-substituted trastuzumab which has reacted with DTT during sample preparation.

Synthesis of additional sugar-derivative nucleotides Su(A)-Nuc (3)

Additional sugar-derivative nucleotides Su(A)-Nuc according to formula (3) were prepared a.o. according to procedures as disclosed in e.g. WO 2014/065661 (SynAffix B. V., Pouilly et al, ACS Chem. Biol. 2012, 7, 753 and Guan et al, Chem. Eur. J. 2010, 16, 13343, all incorporated by reference.

Example 31. Synthesis of UDP-GalNAcSAc ((20), wherein Vis O, r is 0 andR 8 is CH 3 ) UDP-D-galactosamine (Example 16) (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 (28 mg, 0.041 mmol, 52%).

1H- MR (300 MHz, D 2 0): 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 Ci 9 H 29 N 3 Oi 8 P 2 S ( -H + ) 680.06, found 680.1. Example 32. Synthesis of UDP-GalNAcCl ((19), wherein X is CI)

UDP-D-galactosamine (Example 16) (42 mg, 0.074 mmol) was dissolved in 0.1 M NaHC0 3 (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.t., 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:H 2 0) afforded UDP-GalNAcCl (25 mg, 0.039 mmol, 53%).

1H-NMR (300 MHz, D 2 0): 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 Ci 7 H 26 ClN 3 Oi 7 P 2 ( -H + ) 640.03 (100%), 642.03 (32%), found 640.1 (100%), 642.2 (35%). Example 33. Synthesis of UDP-GalNAcBr ((19), wherein Xis Br)

UDP-D-galactosamine (Example 16) (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 (28 mg, 0.048 mmol, 65%).

1H-NMR (300 MHz, D 2 0): 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 Ci 7 H 26 BrN 3 Oi 7 P 2 ( -H + ) 683.98 (100%), 685.98 (98%), found 687.1 (100%), 688.0 (92%), 686.0 (85%), 689.0 (72%).

Example 34. Design ofT. ni GalNAcT mutants and A. suum GalNAcT mutants

Mutants of TnGalNAcT and AsGalNAcT were designed based on the crystal structure for bovine P(l,4)-Gal-Tl in complex with UDP-N-acetyl-galactosamine (PDB entry 10QM) and the p(l,4)-Gal-Tl(Y289L) mutant reported by Qasba et al. (J. Biol. Chem. 2002, 277: 20833-20839, incorporated by reference). Mutants of TnGalNAcT and AsGalNAcT were designed based on a sequence alignment of TnGalNAcT and AsGalNAcT with bovine P(l,4)Gal-Tl . The corresponding amino acid residues between these proteins are shown in Table 3.

Table 3. Numbers of corresponding amino acids in different GalNAcT/GalT species

Example 35. Site directed mutagenesis of Hise-TnGalNAcT(33-421) mutants

A pET15b-vector containing the codon optimized sequence encoding residues 33-421 of TnGalNAcT (identified by SEQ ID NO: 8) between the Ndel-Bamffl sites was obtained from Genscript, resulting in His 6 -TnGalNAcT(33-421) (identified by SEQ ID NO: 49). The TnGalNacT mutant genes were amplified from the above described construct using a set of overlapping primers by a linear amplification PCR. The overlapping primer sets used for each mutant are shown in table 4. For the construction of His 6 -TnGalNAcT(33-421; W336F) (identified by SEQ ID NO: 50) the DNA fragment was amplified with a pair of primers defined herein as SEQ ID NO: 79 and SEQ ID NO: 80. For the construction of His 6 -TnGalNAcT(33-421 ; W336H) (identified by SEQ ID NO: 51) the DNA fragment was amplified with a pair of primers defined herein as SEQ ID NO: 81 and SEQ ID NO: 82. For the construction of His 6 - TnGalNAcT(33-421; W336V) (identified by SEQ ID NO: 52) the DNA fragment was amplified with a pair of primers defined herein as SEQ ID NO: 83 and SEQ ID NO: 84. For the construction of His 6 -TnGalNAcT(33-421; E339A) (identified by SEQ ID NO: 53) the DNA fragment was amplified with a pair of primers defined herein as SEQ ID NO: 85 and SEQ ID NO: 86. For the construction of His-TnGalNAcT(33-421; E339D) (identified by SEQ ID NO: 55) the DNA fragment was amplified with a pair of primers defined herein as SEQ ID NO: 88 and SEQ ID NO: 89. For the construction of His 6 - TnGalNAcT(33-421; I299M) (identified by SEQ ID NO: 45) the DNA fragment was amplified with a pair of primers defined herein as SEQ ID NO: 61 and SEQ ID NO: 62. For the construction of His 6 -TnGalNAcT(33-421; I299A) (identified by SEQ ID NO: 48) the DNA fragment was amplified with a pair of primers defined herein as SEQ ID NO: 63 and SEQ ID NO: 64. For the construction of His 6 -TnGalNAcT(33-421; I299G) (identified by SEQ ID NO: 54) the DNA fragment was amplified with a pair of primers defined herein as SEQ ID NO: 65 and SEQ ID NO: 66. For the construction of His 6 - TnGalNAcT(33-421; L302A) (identified by SEQ ID NO: 43) the DNA fragment was amplified with a pair of primers defined herein as SEQ ID NO: 67 and SEQ ID NO: 68. For the construction of His 6 -TnGalNAcT(33-421; L302G) (identified by SEQ ID NO: 44) the DNA fragment was amplified with a pair of primers defined herein as SEQ ID NO: 69 and SEQ ID NO: 70. For the construction of His 6 -TnGalNAcT(33-421; I311M) (identified by SEQ ID NO: 60) the DNA fragment was amplified with a pair of primers defined herein as SEQ ID NO: 74 and SEQ ID NO: 87.After the PCR amplification, the reaction mixture was treated with Dpnl to digest template DNA followed by transformation into NEB 10-beta competent cells (obtained from New England Biolabs). DNA was isolated and sequences were confirmed by sequence analysis for the mutants His 6 -TnGalNAcT(33-421; W336F) (identified by SEQ ID NO: 50), His 6 - TnGalNAcT(33-421; W336V) (identified by SEQ ID NO: 52), His 6 -TnGalNAcT(33- 421; E339A) (identified by SEQ ID NO: 53), His 6 -TnGalNAcT(33-421; I299M) (identified by SEQ ID NO: 45), His 6 -TnGalNAcT(33-421; I299A) (identified by SEQ ID NO: 48), His 6 -TnGalNAcT(33-421; I299G) (identified by SEQ ID NO: 54), His 6 - TnGalNAcT(33-421; L302A) (identified by SEQ ID NO: 43), His 6 -TnGalNAcT(33- 421; L302G) (identified by SEQ ID NO: 44) and His 6 -TnGalNAcT(33-421; I311M) (identified by SEQ ID NO: 60).

Table 4. Sequence identification of the primers used. Codons corresponding to the mutated amino acid are in bold.

SEQ ID Name Nucleotide sequence

NO

SEQ ID W336F, fwd C TCG AAT AAA TAT TGG GGT TTT GGC NO: 79 GGT GAA GAT GAC GAT ATG

SEQ ID W336F, rev CAT ATC GTC ATC TTC ACC GCC AAA ACC NO: 80 CCA ATA TTT ATT CGA G

SEQ ID W336H, fwd CG AAT AAA TAT TGG GGT CAC GGC GGT NO: 81 GAA GAT GAC G

SEQ ID W336H, rev C GTC ATC TTC ACC GCC GTG ACC CCA NO: 82 ATA TTT ATT CG

SEQ ID W336V, fwd CG AAT AAA TAT TGG GGT GTG GGC GGT NO: 83 GAA GAT GAC G

SEQ ID W336V, rev C GTC ATC TTC ACC GCC CAC ACC CCA NO: 84 ATA TTT ATT CG

SEQ ID E339A, fwd G GGT TGG GGC GGT GCG GAT GAC GAT NO: 85 ATG AGC

SEQ ID E339A, rev GCT CAT ATC GTC ATC CGC ACC GCC CCA NO: 86 ACC C

SEQ ID E339D, fwd G GGT TGG GGC GGT GAT GAT GAC GAT NO: 88 ATG AGC

SEQ ID E339D, rev GCT CAT ATC GTC ATC ATC ACC GCC CCA NO: 89 ACC C

SEQ ID I299M, fwd GT CAC ATG TCA GCC AGC ATG GAC NO: 61 AAA CTG CAC TTT AAA C

SEQ ID I299M, rev G TTT AAA GTG CAG TTT GTC CAT NO: 62 GCT GGC TGA CAT GTG AC

SEQ ID I299A, fwd G CGT CAC ATG TCA GCC AGC GCC NO: 63 GAC AAA CTG CAC TTT AAA C

SEQ ID I299A, rev G TTT AAA GTG CAG TTT GTC GGC NO: 64 GCT GGC TGA CAT GTG ACG C

SEQ ID I299G, fwd G CGT CAC ATG TCA GCC AGC GGC NO: 65 GAC AAA CTG CAC TTT AAA C

SEQ ID I299G, rev G TTT AAA GTG CAG TTT GTC GCC NO: 66 GCT GGC TGA CAT GTG ACG C

SEQ ID L302A, fwd C AGC CAG CAT CGAC AAA GCG CAC NO: 67 TTT AAA CTG CCG

SEQ ID L302A, rev CGG CAG TTT AAA GTG CGC TTT GTC GAT NO: 68 GCT GGC TG

SEQ ID L302G, fwd CA GCC AGC ATC GAC AAA GGG CAC NO: 69 TTT AAA CTG CCG

SEQ ID L302G, rev CGG CAG TTT AAA GTG CCC TTT GTC GAT NO: 70 GCT GGC TG SEQ ID 1311M, fwd C GAA GAT ATG TTC GGC GGT GTC NO: 74 TCA GCC ATG

SEQ ID I311M, rev CAT GGC TGA GAC ACC GCC GAA CAT ATC NO: 87 TTC G

Example 36. Expression and refolding of Hise-TnGalNAcT(33-421), Hise- TnGalNAcT(33-421; W336F), His 6 -TnGalNAcT(33-421; W336V), His 6 -

TnGalNAcT(33-421; I299M), His 6 -TnGalNAcT(33-421; I299A), His 6 -TnGalNAcT(33- 421; I299G), His 6 -TnGalNAcT(33-421; L302A), His 6 -TnGalNAcT(33-421; L302G), His 6 -TnGalNAcT(33-421; I311M), His 6 -TnGalNAcT(33-421; E339D) and His 6 - TnGalNAcT(33-421; E339A) in E. coli

His 6 -TnGalNAcT(33-421), His 6 -TnGalNAcT(33-421; W336F), His 6 -TnGalNAcT(33- 421; W336V), His 6 -TnGalNAcT(33-421; I299M), His 6 -TnGalNAcT(33-421; I299A), His 6 -TnGalNAcT(33-421; I299G), His 6 -TnGalNAcT(33-421; L302A), His 6 - TnGalNAcT(33-421; L302G), His 6 -TnGalNAcT(33-421; I311M), His 6 - TnGalNAcT(33-421; E339D) and His 6 -TnGalNAcT(33-421; E339A) were expressed from the corresponding pET15b-constructs which are obtained as described in Example 35. Expression, inclusion body isolation and refolding was performed according to the reported procedure by Qasba et al. (Prot. Expr. Pur. 2003, 30, 219-76229, incorporated by reference). After refolding, the insoluble protein was removed by centrifugation (10 minutes at 4 °C at 14.000 x g) followed by filtration through a 0.45 μΜ-pore diameter filter. The soluble protein was purified and concentrated using a HisTrap HP 5 mL column (GE Healthcare). The column was first washed with buffer A (5 mM Tris buffer, 20 mM imidazole, 500 mM NaCl, pH 7.5). Retained protein was eluted with buffer B (20 mM Tris, 500 mM NaCl, 500 mM imidazole, pH 7.5, 10 mL). Fractions were analyzed by SDS-PAGE on polyacrylamide gels (12%), and the fractions that contained purified target protein were combined and the buffer was exchanged against 20 mM Tris pH 7.5 and 500 mM NaCl by dialysis performed overnight at 4 °C. The purified protein was concentrated to at least 2 mg/mL using an Amicon Ultra-0.5, Ultracel-10 Membrane (Millipore) and stored at -80 °C prior to further use. Example 37. Design ofT. ni GalNAcT mutants of W336

Based on a sequence alignment of TnGalNAcT with GalT, three active site mutants were designed by mutation of tryptophanes 36 to phenylalanine, histidine or valine (underlined).

T. ni (Hise-TnGalNAcT [33-421; W336F] identified by SEQ ID NO: 50)

MGSSHHHHHHSSGLVPRGSHMSPLRTYLYTPLYNATQPTLRNVERLAANWPK KIPSNYIEDSEEYSIKNISLSNHTTRASVVHPPSSITETASKLDKNMTIQDGAFAM ISPTPLLITKLMDSn SYVTTEDGVKKAEAVVTLPLCDSMPPDLGPITLNKTELE LEWVEKKFPEVEWGGRYSPPNCT ARHRVAIIVP YRDRQQHL AIFLNHMHPFLM KQQIEYGIFIVEQEGNKDFNRAKLMNVGFVESQKLVAEGWQCFVFHDIDLLPL DTRNLYSCPRQPRHMSASIDKLHFKLPYEDIFGGVSAMTLEQFTRVNGFSNKY WGFGGEDDDMSYRLKKINYHIARYKMSIARYAMLDHKKSTPNPKRYQLLSQT SKTF QKD GL S TLE YEL VQ V VQ YHL YTHIL VNIDERS

T. ni (Hise-TnGalNAcT [33-421; W336H] identified by SEQ ID NO: 51)

MGSSHHHHHHSSGLVPRGSHMSPLRTYLYTPLYNATQPTLRNVERLAANWPK KIPSNYIEDSEEYSIKNISLSNHTTRASVVHPPSSITETASKLDKNMTIQDGAFAM ISPTPLLITKLMDSn SYVTTEDGVKKAEAVVTLPLCDSMPPDLGPITLNKTELE LEWVEKKFPEVEWGGRYSPPNCTARimVAIIWYRDRQQHLAIFLNHMHPFLM KQQIEYGIFIVEQEGNKDFNRAKLMNVGFVESQKLVAEGWQCFVFHDIDLLPL DTRNLYSCPRQPRHMSASIDKLHFKLPYEDIFGGVSAMTLEQFTRVNGFSNKY WGHGGEDDDMSYRLKKINYHIARYKMSIARYAMLDHKKSTPNPKRYQLLSQT SKTFQKDGLSTLEYELVQVVQYHL YTHIL VNIDERS

T. ni (Hise-TnGalNAcT [33-421; W336V] identified by SEQ ID NO: 52)

MGSSHHHHHHSSGLVPRGSHMSPLRTYLYTPLYNATQPTLRNVERLAANWPK KIPSNYIEDSEEYSIKNISLSNHTTRASVVHPPSSITETASKLDKNMTIQDGAFAM ISPTPLLITKLMDSn SYVTTEDGVKKAEAVVTLPLCDSMPPDLGPITLNKTELE LEWVEKKFPEVEWGGRYSPPNCT ARHRVAIIVP YRDRQQHL AIFLNHMHPFLM KQQIEYGIFIVEQEGNKDFNRAKLMNVGFVESQKLVAEGWQCFVFHDIDLLPL DTRNLYSCPRQPRHMSASIDKLHFKLPYEDIFGGVSAMTLEQFTRVNGFSNKY WGVGGEDDDMSYRLKKINYHIARYKMSIARYAMLDHKKSTPNPKRYQLLSQT SKTF QKD GL S TLE YEL VQ V VQ YHL YTHIL VNIDERS

Example 38. Design ofT. ni GalNAcT mutants ofL302

Based on a sequence alignment of TnGalNAcT with GalT, two active site mutants were designed by mutation of leucine30 2 to glycine or alanine (underlined).

T. ni (Hise-TnGalNAcT [33-421; L302G] identified by SEQ ID NO: 44)

MGSSHHHHHHSSGLVPRGSHMSPLRTYLYTPLYNATQPTLRNVERLAANWPK KIPSNYIEDSEEYSIKNISLSNHTTRASVVHPPSSITETASKLDKNMTIQDGAFAM ISPTPLLITKLMDSn SYVTTEDGVKKAEAVVTLPLCDSMPPDLGPITLNKTELE LEWVEKKFPEVEWGGRYSPPNCTARHRVAIIWYRDRQQHLAIFLNHMHPFLM KQQIEYGIFIVEQEGNKDFNRAKLMNVGFVESQKLVAEGWQCFVFHDIDLLPL DTRNLYSCPRQPRHMSASIDKGHFKLPYEDIFGGVSAMTLEQFTRVNGFSNKY WGWGGEDDDM S YRLKKIN YHI AR YKM S I AR Y AMLDHKK S TPNPKR YQLL S Q TSKTFQKDGLSTLEYELVQVVQYHL YTHIL VNIDERS

T. ni (Hise-TnGalNAcT [33-421; L302A] identified by SEQ ID NO: 43)

MGSSHHHHHHSSGLVPRGSHMSPLRTYLYTPLYNATQPTLRNVERLAANWPK KIPSNYIEDSEEYSIKNISLSNHTTRASVVHPPSSITETASKLDKNMTIQDGAFAM ISPTPLLITKLMDSn SYVTTEDGVKKAEAVVTLPLCDSMPPDLGPITLNKTELE LEWVEKKFPEVEWGGRYSPPNCTARimVAIIWYRDRQQHLAIFLNHMHPFLM KQQIEYGIFIVEQEGNKDFNRAKLMNVGFVESQKLVAEGWQCFVFHDIDLLPL DTRNLYSCPRQPRHMSASIDKAHFKLPYEDIFGGVSAMTLEQFTRVNGFSNKY WGWGGEDDDMSYRLKKINYHIARYKMSIARYAMLDHKKSTPNPKRYQLLSQ TSKTFQKDGLSTLEYELVQVVQYHL YTHIL VNIDERS

Example 39. Design of T. ni GalNAcT mutants o/E339

Based on a sequence alignment of TnGalNAcT with GalT, two active site mutants were designed by mutation of glutamic acid 3 39 to glycine, alanine or aspartic acid (underlined).

T. ni (Hise-TnGalNAcT [33-421; E339A] identified by SEQ ID NO: 53) MGSSHHHHHHSSGLVPRGSHMSPLRTYLYTPLYNATQPTLRNVERLAANWPK KIPSNYIEDSEEYSIKNISLS HTTRASVVHPPSSITETASKLDK MTIQDGAFAM ISPTPLLITKLMDSIKSYVTTEDGVKKAEAVVTLPLCDSMPPDLGPITLNKTELE LEWVEKKFPEVEWGGRYSPPNCTARHRVAIIVPYRDRQQHLAIFL HMHPFLM KQQIEYGIFIVEQEG KDF RAKLMNVGFVESQKLVAEGWQCFVFHDIDLLPL DTR LYSCPRQPRHMSASIDKLHFKLPYEDIFGGVSAMTLEQFTRVNGFS KY WGWGG ADDDM S YRLKKIN YHI AR YKM S I AR Y AMLDHKK S TP PKR YQLL S Q TSKTFQKDGLSTLEYELVQVVQYHLYTHILVNIDERS T. ni (Hise-TnGalNAcT [33-421; E339D] identified by SEQ ID NO: 55)

MGSSHHHHHHSSGLVPRGSHMSPLRTYLYTPLYNATQPTLRNVERLAANWPK KIPSNYIEDSEEYSIKNISLSNHTTRASVVHPPSSITETASKLDKNMTIQDGAFAM ISPTPLLITKLMDSn SYVTTEDGVKKAEAVVTLPLCDSMPPDLGPITLNKTELE LEWVEKKFPEVEWGGRYSPPNCTARHRVAIIVPYRDRQQHLAIFLNHMHPFLM KQQIEYGIFIVEQEGNKDFNRAKLMNVGFVESQKLVAEGWQCFVFHDIDLLPL DTRNLYSCPRQPRHMSASIDKLHFKLPYEDIFGGVSAMTLEQFTRVNGFSNKY WGWGGDDDDM S YRLKKIN YHI AR YKM S I ARY AMLDFDCK S TPNPKR YQLL S Q TSKTFQKDGLSTLEYELVQVVQYHLYTHILVNIDERS Example 40. Design o/T. ni GalNAcT mutant I299M

Based on a sequence alignment of TnGalNAcT with GalT, three active site mutants were designed by mutation of isoleucine299 to methionine, alanine or glycine (underlined). T. ni (Hise-TnGalNAcT [33-421; I299M] identified by SEQ ID NO: 45)

MGSSHHHHHHSSGLVPRGSHMSPLRTYLYTPLYNATQPTLRNVERLAANWPK KIPSNYIEDSEEYSIKNISLSNHTTRASVVHPPSSITETASKLDKNMTIQDGAFAM ISPTPLLITKLMDSn SYVTTEDGVKKAEAVVTLPLCDSMPPDLGPITLNKTELE LEWVEKKFPEVEWGGRYSPPNCTARHRVAIIVPYRDRQQHLAIFLNHMHPFLM KQQIEYGIFIVEQEGNKDFNRAKLMNVGF VESQKL VAEGWQCF VFHDIDLLPL DTRNLYSCPRQPRHMSASMPKLHFKLPYEDIFGGVSAMTLEQFTRVNGFSNKY WGWGGEDDDM S YRLKKIN YHI ARYKMSI ARY AMLDHKK S TPNPKR YQLL S Q TSKTFQKDGLSTLEYELVQVVQYHLYTHILVNIDERS T. ni (Hise-TnGalNAcT [33-421; 1299 A] identified by SEQ ID NO: 48)

MGSSHHHHHHSSGLVPRGSHMSPLRTYLYTPLYNATQPTLRNVERLAANWPK KIPSNYIEDSEEYSIKNISLSNHTTRASVVHPPSSITETASKLDKNMTIQDGAFAM ISPTPLLITKLMDSn SYVTTEDGVKKAEAVVTLPLCDSMPPDLGPITLNKTELE LEWVEKKFPEVEWGGRYSPPNCTARHRVAIIVPYRDRQQHLAIFLNHMHPFLM KQQIEYGIFIVEQEGNKDFNRAKLMNVGFVESQKLVAEGWQCFVFHDIDLLPL DTRNLYSCPRQPRHMSASADKLHFKLPYEDIFGGVSAMTLEQFTRVNGFSNKY WGWGGEDDDM SYRLKKINYHI ARYKMSI ARY AMLDtfKK S TPNPKR YQLL S Q TSKTFQKDGLSTLEYELVQVVQYHLYTHILVNIDERS

T. ni (Hise-TnGalNAcT [33-421; I299G] identified by SEQ ID NO: 54)

MGSSHHHHHHSSGLVPRGSHMSPLRTYLYTPLYNATQPTLRNVERLAANWPK

KIPSNYIEDSEEYSIKNISLSNHTTRASVVHPPSSITETASKLDKNMTIQDGAFAM ISPTPLLITKLMDSIKS YVTTEDGVKKAEAVVTLPLCDSMPPDLGPITLNKTELE LEWVEKKFPEVEWGGRYSPPNCTARHRVAIIVPYRDRQQHLAIFLNHMHPFLM KQQIEYGIFIVEQEGNKDFNRAKLMNVGFVESQKLVAEGWQCFVFHDIDLLPL DTRNLYSCPRQPRHMSASGDKLHFKLPYEDIFGGVSAMTLEQFTRVNGFSNKY WGWGGEDDDM SYRLKKINYHI ARYKMSI ARY AMLDHKK S TPNPKR YQLL S Q TSKTFQKDGLSTLEYELVQVVQYHLYTHILVNIDERS

Example 41. Design o/T. ni GalNAcT mutant 1311M

A TnGalNAcT mutant was designed by mutation of isoleucine3ii to methionine. T. ni (TnGalNAcT [33-421; 1311M] identified by SEQ ID NO: 60)

SPLRT YL YTPL YNATQPTLRNVERL AANWPKKIP SNYIED SEEYSIKNISL SNHT TRASVVHPPSSITETASKLDKNMTIQDGAFAMISPTPLLITKLMDSIKSYVTTED GVKKAEAVVTLPLCDSMPPDLGPITLNKTELELEWVEKKFPEVEWGGRYSPPN CTARHRVAIIVPYRDRQQHLAIFLNHMHPFLMKQQIEYGIFIVEQEGNKDFNRA KLMNVGFVESQKLVAEGWQCFVFHDIDLLPLDTRNLYSCPRQPRHMSASIDKL HFKLP YEDMF GGVS AMTLEQF TRVNGF SNK YWGWGGEDDDM S YRLKKIN YH I ARYKM SI ARY AMLDHKK S TPNPKR YQLL SQT SKTF QKDGL S TLE YEL VQ V VQ YHLYTHILVNIDERS Example 42. Synthesis of UDP-GalNPropN3 ((9), wherein U is [CH 2 J 2 and A = N3) UDP-Gal PropN 3 , a sugar-derivative nucleotide Su(A)-Nuc according to formula (3), was prepared a.o. according to procedures as disclosed in e.g. WO 2014/065661 (SynAffix B.V.), Pouilly et al, ACS Chem. Biol. 2012, 7, 753 and Guan et al, Chem. Eur. J. 2010, 16, 13343, all incorporated by reference.

Example 43. Synthesis of UDP-GalNButNe ((9), wherein U is [CH 2 ]3 and A = N3) UDP-Gal ButN 3 , a sugar-derivative nucleotide Su(A)-Nuc according to formula (3), was prepared a.o. according to procedures as disclosed in e.g. WO 2014/065661 (SynAffix B.V.), Pouilly et al, ACS Chem. Biol. 2012, 7, 753 and Guan et al, Chem. Eur. J. 2010, 16, 13343, all incorporated by reference.

Example 44. Synthesis of UDP-GalNProSH ((21), wherein t = 2)

UDP-GalNProSH, a sugar-derivative nucleotide Su(A)-Nuc according to formula (3), was prepared a.o. according to procedures as disclosed in e.g. WO 2015/057063 (SynAffix B.V.), Pouilly et al, ACS Chem. Biol. 2012, 7, 753 and Guan et al, Chem. Eur. J. 2010, 16, 13343, all incorporated by reference. Example 45. Synthesis of UDP-GalNBzN 3 ((23), wherein X is H)

UDP-Gal BzN 3 , a sugar-derivative nucleotide Su(A)-Nuc according to formula (3), was prepared a.o. according to procedures as disclosed in e.g. WO 2015/112013 (SynAffix B.V.), Pouilly et al, ACS Chem. Biol. 2012, 7, 753 and Guan et al, Chem. Eur. J. 2010, 16, 13343, all incorporated by reference.

Example 46. Synthesis of UDP-GalNPyrNs ((12), wherein A is N3 andR 2 is H)

UDP-GalNPyrN 3 , a sugar-derivative nucleotide Su(A)-Nuc according to formula (3), was prepared a.o. according to procedures as disclosed in e.g. WO 2015/112013 (SynAffix B.V.), Pouilly et al, ACS Chem. Biol. 2012, 7, 753 and Guan et al, Chem. Eur. J. 2010, 16, 13343, all incorporated by reference. Glycosyltransfer of galactose derivative (e.g. thiosugar) with GalNAcT (general protocol)

Enzymatic introduction of galactose derivative (e.g. thio-containing sugar 21) onto IgG was effected with a GalNAc-transferase or a mutant thereof. The deglycosylated IgG (prepared as described above, 10 mg/mL) was incubated with a modified UDP- galactose derivative (e.g. an thio-modified sugar-UDP derivative) (2 mM) and GalNAcT (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 IgG (e.g. thio-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- HC1 pH 2.5. The eluted IgG was neutralized with 1 M Tris-HCl pH 7.0 and concentrated and washed with 50 mM Tris-HCl pH 6.0 using an Amicon Ultra-0.5, Ultracel-10 Membrane (Millipore) to a concentration of 15-20 mg/mL. Example 47. Preparation of trastuzumab(GalNProSH) 2 via glycosyltransfer of UDP- GalNProSH ((21), wherein t = 2) to deglycosylated trastuzumab with CeGalNAcT Trimmed trastuzumab was subjected to the glycosyltransfer protocol with UDP- Gal ProSH (2 mM) and CeGalNAcT (1 mg/mL). After incubation overnight, a small sample was subjected to spectral analysis after digestion with Fabricator™ (50 U in 10 iL PBS pH 6.6) and subsequent wash with MiliQ using an Amicon Ultra-0.5, Ultracel- 10 Membrane (Millipore). Full conversion of the starting material into two products was observed. The major product (24387 Da, expected mass 24388) corresponded to deglycosylated trastuzumab + GalNProSH (trastuzumab-(GalNProSH) 2 ) whereas the minor (25037 Da, expected mass 25038) corresponded to deglycosylated trastuzumab + GalNProS-UDPGalNProS disulfide. The ratio between the two products is about 60:40.

Example 48. Preparation of trastuzumab(GalNProSH) 2 via glycosyltransfer of UDP- GalNProSH ((21), wherein t = 2) to deglycosylated trastuzumab with TnGalNAcT Trimmed trastuzumab was subjected to the glycosyltransfer protocol with UDP- GalNProSH (2 mM) and TnGalNAcT (0.2 mg/mL). After incubation overnight, a small sample was subjected to spectral analysis after digestion with Fabricator™ (50 U in 10 PBS pH 6.6) and subsequent wash with MiliQ using an Amicon Ultra-0.5, Ultracel- 10 Membrane (Millipore). Full conversion of the starting material into two products was observed. The major product (24387 Da, expected mass 24388) corresponded to deglycosylated trastuzumab + GalNProSH (trastuzumab-(GalNProSH) 2 ) whereas the minor (25037 Da, expected mass 25038) corresponded to deglycosylated trastuzumab + GalNProS-UDP-GalNProS disulfide. The ratio between the two products is about 60:40.

Example 49. Preparation of trastuzumab(GalNProSH) 2 via glycosyltransfer of UDP- GalNProSH ((21), wherein t = 2) to deglycosylated trastuzumab with AsGalNAcT Trimmed trastuzumab was subjected to the glycosyltransfer protocol with UDP- GalNProSH (2 mM) and AsGalNAcT (0.2 mg/mL). After incubation overnight,a small sample was reduced with DTT and subsequently subjected to MS analysis indicating the formation of a one major product of (49755 Da, 95% of total heavy chain) resulting from GalNProSH transfer to core GlcNAc(Fuc) substituted trastuzumab. Example 50. Preparation of trastuzumab (GalPyrNe) 2 via glycosyltransfer of UDP- GalNPyrNs ((12), wherein A is N3 and R 2 is H)) to deglycosylated trastuzumab with TnGalNAcT

According to the general protocol for glycosyltransfer, trimmed trastuzumab was treated with TnGalNAcT (0.5 mg/mL) in the presence of UDP-GalNPyrN 3 ((12), wherein A is N 3 and R 2 is H)) (4 mM). After incubation overnight, a small sample was subjected to spectral analysis after digestion with Fabricator™ (50 U in 10 iL PBS pH 6.6) and subsequent wash with MiliQ using an Amicon Ultra-0.5, Ultracel-10 Membrane (Millipore). About 50% conversion was observed. The formed product (24445 Da, expected mass 24445) results from GalNPyrN 3 transfer to core GlcNAc(Fuc) substituted trastuzumab.

Example 51. Preparation of trastuzumab (F2-GalNAz) 2 via glycosyltransfer of IIDP-F2- GalNAz (18) to deglycosylated trastuzumab with TnGalNAcT

According to the general protocol for glycosyltransfer, trimmed trastuzumab was treated with TnGalNAcT(wt), TnGalNAcT(W336V), TnGalNAcT(W336F), TnGalNAcT(E339 A) or TnGalNAcT(L302 A) at a concentration of 0.25 mg/mL in the presence of UDP-F 2 -GalNAz (18, 1 mM). After incubation overnight, a small sample was subjected to spectral analysis after digestion with Fabricator™ (50 U in 10 iL PBS pH 6.6) and wash with MiliQ using an Amicon Ultra-0.5, Ultracel-10 Membrane (Millipore). MS analysis indicated the formation of a major product (MW = 24420 Da) resulting from F 2 -GalNAz transfer to core GlcNAc(Fuc) substituted trastuzumab, and a minor product (MW = 24273 Da) resulting from F 2 -GalNAz transfer to core GlcNAc substituted trastuzumab. Observed cumulative conversions of the major and minor product for TnGalNAcT(wt), TnGalNAcT(W336V), TnGalNAcT(W336F), TnGalNAcT(E339 A) or TnGalNAcT(L302 A) were 95%, 27%, 24%, 5% and 95%, respectively. Example 52. Preparation of trastuzumab (GalBzNi) 2 via glycosyltransfer of UDP- GalNBzN 3 ((23), wherein Xis H) to deglycosylated trastuzumab with TnGalNAcT According to the general protocol for glycosyltransfer, trimmed trastuzumab was treated with TnGalNAcT (0.7 mg/mL) in the presence of UDP-GalNBzN 3 (4 mM) ((23), wherein X is H). After incubation overnight, a small sample was subjected to spectral analysis after digestion with Fabricator™ (50 U in 10 iL PBS pH 6.6) and subsequent wash with MiliQ using an Amicon Ultra-0.5, Ultracel-10 Membrane (Millipore). About 70% conversion was observed. The formed product (24444 Da, expected mass 24443) results from GalNBzN 3 transfer to core GlcNAc(Fuc) substituted trastuzumab.

Example 53. Preparation of trastuzumab(F 2 -GalNBzNs) 2 via glycosyltransfer of UDP- F 2 -GalNBzN 3 (24) to deglycosylated trastuzumab with TnGalNAcT

According to the general protocol for glycosyltransfer, trimmed trastuzumab was treated with TnGalNAcT(wt), TnGalNAcT(W336H), TnGalNAcT(I299M), TnGalNAcT(L302 A) or TnGalNAcT(L302G) at a concentration of 0.5 mg/mL in the presence of UDP-F 2 -GalNBzN 3 (24, 1 mM). After incubation overnight, a small sample was digested with Fabricator™ (50 U in 10 μΙ_, PBS pH 6.6) and subsequent washed with MiliQ using an Amicon Ultra-0.5, Ultracel-10 Membrane (Millipore). MS analysis indicating the formation of a major product (MW = 24479 Da), resulting from F 2 -GalNBzN 3 transfer to core GlcNAc(Fuc) substituted trastuzumab, and a minor product (MW = 24332 Da) resulting from F 2 -GalNBzN 3 transfer to core GlcNAc substituted trastuzumab. Observed cumulative conversions of the major and minor product for TnGalNAcT(wt), TnGalNAcT(W336H), TnGalNAcT(I299M), TnGalNAcT(L302 A) or TnGalNAcT(L302G) were 14%, 62%, 26%, 37% and 14%, respectively.

Example 54. Preparation of trastuzumab(GalNPropNs) 2 via glycosyltransfer of UDP- GalNPropNs ((31), wherein U is [CH 2 ] 2 and A = N3) to deglycosylated trastuzumab with AsGalNAcT

According to the general protocol for glycosyltransfer, trimmed trastuzumab was treated with AsGalNAcT at a concentration of 0.2 mg/mL in the presence of UDP- GalNPropN 3 ((31), wherein U is [CH 2 ] 2 and A = N 3 , 0.7 mM). After incubation overnight, a small sample was reduced with DTT and subsequently subjected to MS analysis indicating the formation of a one major product of (49759 Da, 70% of total heavy chain) resulting from GalNPropN 3 transfer to core GlcNAc(Fuc) substituted trastuzumab. Example 55. Preparation of trastuzumab(GalNButNs) 2 via glycosyltransfer of UDP- GalNButNs ((31) wherein U is [CH 2 ]3 and A = N3) to deglycosylated trastuzumab with AsGalNAcT

According to the general protocol for glycosyltransfer, trimmed trastuzumab was treated with AsGalNAcT(wt) at a concentration of 0.2 mg/mL in the presence of UDP- GalNButN 3 ((31), wherein U is [CH 2 ] 3 and A = N 3 , 0.7 mM). After incubation overnight, a small sample was reduced with DTT and subsequently subjected to MS analysis indicating the formation of a one major product of (49772 Da, 50% of total heavy chain) resulting from GalNButN 3 transfer to core GlcNAc(Fuc) substituted trastuzumab.