Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
CARBOHYDRATE LIGANDS THAT BIND TO ANTIBODIES AGAINST GLYCOEPITOPES OF GLYCOSPHINGOLIPIDS
Document Type and Number:
WIPO Patent Application WO/2017/046172
Kind Code:
A1
Abstract:
The invention relates to carbohydrate ligands and moieties, respectively, mimicking glycoepitopes comprised by glycosphingolipids of the nervous system, particularly glycoepitopes comprised by glycosphingolipids of the cerebroside, the globoside-, the ganglioside- and the sulfoglucuronyl paragloboside type, which are bound by anti-glycan antibodies associated with neurological diseases. The invention further relates to the use of these carbohydrate ligands/moieties, in diagnosis as well as for the treatment of neurological diseases associated with anti-glycan antibodies. In particular, the invention relates to compounds of formula (I) and (II) and to therapeutically acceptable polymers comprising a multitude of these compounds, including polymers with loading of one compound of formula (I) or (II) or combinations of several compounds of formula (I), and/or (II). The compounds of formula (I) are defined as: formula (I), wherein RI1 is Z or (AA) or (BB) or (CC) or (DD); wherein RI2 is H, SO3H, or (EE) or (FF) or (GG) or (HH) or (JJ) or (KK) or (LL), wherein RI3 is H or (MM); wherein RI4 is H or (NN) or (OO), wherein RI5 and RI6 are independently H or (EE); wherein RI7 is H or (EE) or (FF) and compounds of formula (II) are defined as: formula (II), wherein RII1 is Z or (PP), wherein RII2 is Z or (QQ) or (RR) or (SS) or (TT), wherein Z is -N(Ra)-A-B-CH2-(CH2)q-SH, wherein Ra is H, C1-C4-alkyl, C1-C4-alkoxy, CH2C6H5, CH2CH2C6H5, OCH2C6H5, or OCH2CH2C6H5; A is C1-C7-alkylene, C1-C7-alkoxy, C1-C4-alkyl–(OCH2CH2)pO–C1-C4-alkyl, or C1-C7-alkoxy-Rb, wherein Rb is an optionally substituted aryl or an optionally substituted heteroaryl, and wherein p is 0 to 6, preferably p is 1, 2 or 3, and further preferably p is 1; B is NHC(O), S or CH2; q is 0 to 6, preferably q is 1, 2, 3 or 4, and further preferably q is 1 or 2.

Inventors:
HERRENDORFF RUBEN (CH)
ERNST BEAT (CH)
STECK ANDREAS (CH)
PFISTER HÉLÈNE (CH)
NAVARRA GIULIO (CH)
Application Number:
PCT/EP2016/071711
Publication Date:
March 23, 2017
Filing Date:
September 14, 2016
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
UNIV BASEL (CH)
International Classes:
C07H15/12; A61K31/7036; A61K31/715; A61P25/00; C07K2/00
Domestic Patent References:
WO2005054264A22005-06-16
Foreign References:
EP2727597A12014-05-07
US5155004A1992-10-13
Other References:
WILLISON HUGH J ET AL: "Peripheral neuropathies and anti-glycolipid antibodies", BRAIN, OXFORD UNIVERSITY PRESS, OXFORD, GB, vol. 125, no. 12, 1 December 2002 (2002-12-01), pages 2591 - 2625, XP009162997, ISSN: 0006-8950
H. J. WILLISON; N. YUKI, BRAIN, vol. 125, 2002, pages 2591 - 2625
K. A. SHEIKH; G. ZHANG, F1000 BIOLOGY REPORTS, vol. 2, 2010, pages 21
H. J. WILLISON; N. YUKI, BRAIN, 2002
A UEDA ET AL., MOL CELL NEUROSCI, vol. 45, no. 4, 2010, pages 355 - 62
T. ARIGA, J NEUROSCI RES, vol. 92, 2014, pages 1227 - 1242
H. J. WILLISON; C. S. GOODYEAR, CELL, vol. 34, 2013, pages 453 - 459
E. DELMONT; H. J. WILLISON, J NEUROM DIS., vol. 2, 2015, pages 107 - 112
E. NOBILE-ORAZIO, CLINICAL LYMPHOMA & MYELOMA, vol. 9, 2009, pages 107 - 109
Z. KADLECOVA ET AL., BIOMACROMOLECULES, vol. 13, 2012, pages 3127 - 3137
M.C. DALAKAS, CURR TREAT OPINIONS NEUROL, vol. 12, 2010, pages 71 - 83
O. BOHOROV ET AL., GLYCOBIOLOGY, vol. 16, 2006, pages 21 C - 27C
G. THOMA ET AL., J AM CHEM SOC, vol. 121, 1999, pages 5919 - 5929
I. UEDA ET AL., CHEM PHARM BULL (TOKYO), vol. 38, 1990, pages 3035 - 3041
M. NUMATA ET AL., CARBOHYDR RES, vol. 163, 1987, pages 209 - 225
T. FURUKAWA, TETRAHEDRON LETT, vol. 52, 2011, pages 5567 - 5570
K. T. AL-JAMAL ET AL., J DRUG TARGET, vol. 14, 2006, pages 405 - 412
BURGER ET AL., JOURNAL OF IMMUNOLOGICAL METHODS, vol. 140, 1991, pages 31 - 36
Attorney, Agent or Firm:
SPERRLE, Martin (CH)
Download PDF:
Claims:
Claims

1 . A compound comprising a carbohydrate moiety and a linker Z, wherein said carbohydrate moiety mimics a glycoepitope comprised by a glycosphingolipid of the nervous system, wherein

said linker Z is -N(Ra)-A-B-CH2-(CH2)q-SH, wherein

Ra is H, CrC4-alkyl, Ci-C4-alkoxy, CH2C6H5, CH2CH2C6H5, OCH2C6H5, or

A is Ci-C7-alkylene, Ci-C7-alkoxy, Ci-C4-alkyl-(OCH2CH2)pO-Ci-C4-alkyl, or

CrC7-alkoxy-Rb, wherein Rb is an optionally substituted aryl or an optionally substituted heteroaryl, and wherein p is 0 to 6, preferably p is 1 , 2 or 3, and further preferably p is 1 ;

B is NHC(O), S or CH2;

q is 0 to 6, preferably q is 1 , 2, 3 or 4, and further preferably q is 1 or 2; and wherein said linker Z is covalently bound via its -N(Ra)-group to the reducing end of said carbohydrate moiety.

2. The compound of claim 1 , wherein said compound is a compound of formula (I) or formula (II), wherein formula (I) is

wherein R11 is Z or

wherein R12 is H, S03H, or 108

wherein R15 and R16 are independently H or

wherein R17 is H or

and wherein formula (II)

wherein R is Z or

wherein R112 is Z or

wherein Z is -N(Ra)-A-B-CH2-(CH2)q-SH, wherein

Ra is H, CrC4-alkyl, Ci-C4-alkoxy, CH2C6H5, CH2CH2C6H5, OCH2C6H5, or

A is Ci-C7-alkylene, Ci-C7-alkoxy, Ci-C4-alkyl-(OCH2CH2)pO-Ci-C4-alkyl, or

CrC7-alkoxy-Rb, wherein Rb is an optionally substituted aryl or an optionally substituted heteroaryl, and wherein p is 0 to 6, preferably p is 1 , 2 or 3, and further preferably p is 1 ;

B is NHC(O), S or CH2;

q is 0 to 6, preferably q is 1 , 2, 3 or 4, and further preferably q is 1 or 2.

3. The compound of claim 1 or claim 2, wherein said compound is a compound of formula (I).

4. The compound of claim 1 or claim 2, wherein said compound is a compound of formula (II).

5. The compound of any one of claims 1 to 4, wherein said compound is a compound of formula 4*, 9*, 13*, 17*, 21 *, 25*, 29*, 33*, or any one 46*-60*.

13*

46* 112

55*

HO OH

ΗΟ -«-,^ --Ο

58*

wherein Z is -N(Ra)-A-B-CH2-(CH2)q-SH, wherein

Ra is H, CrC4-alkyl, Ci-C4-alkoxy, CH2C6H5! CH2CH2C6H5! OCH2C6H5! or

A is Ci-C7-alkylene, Ci-C7-alkoxy, Ci-C4-alkyl-(OCH2CH2)pO-Ci-C4-alkyl, or

CrC7-alkoxy-Rb, wherein Rb is an optionally substituted aryl or an optionally substituted heteroaryl, and wherein p is 0 to 6, preferably p is 1 , 2 or 3, and further preferably p is 1 ;

B is NHC(O), S or CH2;

q is 0 to 6, preferably q is 1 , 2, 3 or 4, and further preferably q is 1 or 2.

6. The compound of any one of claims 1 to 5, wherein

Ra is H, CH3, CH2CH3, CH2CH2CH3! CH(CH3)2, OCH3, OCH2CH3, OCH2CH2CH3, CH2C6H5, OCH2C6H5;

A is 0(CH2)pCH2, (CH2)PCH2, CH2(OCH2CH2)pOCH2! (OCH2CH2)pOCH2CH2 or

0(CH2)pC6H5;

B is NHC(O), S or CH2. 7. The compound of any one of claims 1 to 6, wherein said linker Z is of a formula selected from any one of the formula (a) to (g):

. SH

p " q

0 (a)

.N. .SH

P q (e)

wherein p is between 0 and 6, preferably 1 to 3, in particular 1, and q is between 0 and 6, preferably between 1, 2 and 4, in particular 1 or 2.

8. The compound of any one of claims 1 to 7, wherein said compound is a compound of formula 4, 9, 13, 17, 21, 25, 29, 33, 37, 41, 44, 56, 58 or 77.

37

77

9. A polymer comprising a multitude of compounds of any one of the claims 1 to 8 wherein said compounds are connected to the polymer backbone by way of said linker Z, and wherein said connection is effected via the SH-group of said linker Z, and wherein preferably said multitude of compounds is (i) a multitude of compounds of formula (I), (ii) a multitude of compounds of formula (II) or (iii) a multitude of compounds of formula (I) and of formula (II), wherein said compounds (I) and (II) are defined as in any one of the claims 2 to 8.

10. The polymer according to claim 9, wherein the polymer backbone is an a-amino acid polymer, an acrylic acid or methacrylic acid polymer or copolymer, a N-vinyl-2- pyrrolidone-vinyl alcohol copolymer, a chitosan polymer, or a polyphosphazene polymer, and wherein preferably the polymer backbone is an a-amino acid polymer, wherein further preferably the polymer backbone is an a-amino acid polymer and said a-amino acid of said a-amino acid polymer is lysine, ornithine, glutamic acid or aspartic acid.

1 1 . The polymer according to claim 9 or 10, wherein the polymer backbone is poly- lysine, and wherein preferably the molecular weight of the polymer backbone is 1 Ό00 Da to 300Ό00 Da.

12. The polymer according to any one of claims 9 to 1 1 , wherein the percentage of loading of the carbohydrate moiety of said compound onto the polymer backbone is between 10 and 90%, preferably between 20 and 70%, and in particular between 30 and 60%.

13. A pharmaceutical composition comprising a compound according to any one of claims 1 to 8, preferably a compound of formula (I) or formula (II) according to any one of claims 2 to 8, or a polymer according to any one of claims 9 to 12

14. A compound according to any one of claims 1 to 8, preferably a compound of formula (I) or formula (II) according to any one of claims 2 to 8, or a polymer according to any one of claims 9 to 12, or a pharmaceutical composition of claim 13 for use in a method of treating a neurological disease, wherein preferably said neurological disease is selected from multiple sclerosis, Parkinson's disease, Alzheimer's disease, dementia, and an immune-mediated neuropathy, wherein preferably said immune-mediated neuropathy is selected from Guillain-Barre syndrome (GBS), chronic inflammatory demyelinating polyneuropathy (CI DP), Miller-Fischer syndrome, Bickerstaff brainstem encephalitis, multifocal motor neuropathy or anti-MAG neuropathy. 15. A compound according to any one of claims 1 to 8, preferably a compound of formula (I) or formula (II) according to any one of claims 2 to 8, or a polymer according to any one of claims 9 to 12, or a pharmaceutical composition of claim 13 for use in a method of diagnosis of a neurological disease, wherein preferably said neurological disease is an immune-mediated neuropathy.

Description:
Carbohydrate ligands that bind to antibodies against glycoepitopes of glycosphingolipids

Field of the invention

The invention relates to carbohydrate ligands and moieties, respectively, that bind to antibodies against glycoepitopes of glycosphingolipids of the nervous system, polymers comprising these carbohydrate ligands, and to their use in diagnosis and therapy of neurological diseases.

Background of the invention

Various neurological diseases are associated with the presence or increased levels of anti-glycan antibodies. Anti-glycolipid antibodies, particularly anti-ganglioside antibodies have been detected in a variety of neuropathological conditions, e.g. in multiple sclerosis, Parkinson's disease, Alzheimer's disease, dementia, Amyotrophic Lateral Sclerosis (ALS) autoimmune-mediated neuropathies including chronic inflammatory demyelinating polyneuropathy (CI DP), Guillain-Barre-syndrome (GBS) (with subtypes acute motor axonal neuropathy (AMAN), acute motor and sensory axonal neuropathy (AMSAN) and acute inflammatory demyelinating polyneuropathy (AIDP)), Miller Fisher syndrome (MFS) and multifocal motor neuropathy (MMN) (K. Kollewe et al., Plos One 2015, 10).

There is evidence from cell culture, tissue culture and animal models that anti-glycan antibodies are involved in immune-mediated attack towards the nervous system. The anti- glycan antibodies target relevant antigens on neuronal or myelin cells and can lead to disruption of nerve fiber function, conduction failure, axonal degeneration and demyelination (H. J. Willison and N. Yuki, Brain, 2002, 125, 2591 -2625; K. A. Sheikh and G. Zhang, F1000 Biology Reports, 2010, 2, 21 ).

There are several mechanism that can explain the pathogenicity of the anti-glycan antibodies, including complement fixation and formation of membrane attack complex, disruption of signaling e.g. through sodium channel blockage (H. J. Willison and N. Yuki, Brain, 2002, loc. cit) or disruption of lipid rafts and interference with signaling pathways therein (A Ueda et al., Mol Cell Neurosci, 2010, 45(4), 355-62). Anti-ganglioside antibodies are also involved in dysfunction of the blood-brain barrier and thus contribute to progression of neurodegenerative diseases (T. Ariga, J Neurosci Res, 2014, 92, 1227- 1242). Interestingly, some anti-glycan antibodies involved in immune-mediated neuropathy do not recognize single glycans but glycan clusters, particularly glycolipid complexes (pattern-recognition antibodies). Thus anti-glycolipid antibodies with pattern recognition characteristics have been described recently in immune-mediated neuropathy where previously no antibodies could be identified. Such antibodies have been identified in GBS, e.g. in the GBS subtype AIDP (H. J. Willison and C. S. Goodyear, Cell, 2013, 34, 453-459).

A pathogenic role for the anti-glycan antibodies is not always clear, even if it is established in immune-mediated neuropathies of acute and chronic types. In this group of diseases specific anti-glycolipid antibodies and specific clinical serological patterns are associated with particular clinical phenotypes (H. J. Willison and N. Yuki, Brain, 2002, 125, 2591 - 2625). The anti-glycan antibodies are usually of the IgM, IgG or IgA type.

The carbohydrate epitopes relevant to immune-mediated neuropathies are predominantly glycolipids, mostly of the ganglioside type involving GM1 (GM1 a), GM1 b, GalNAc-GM1 b, Fucosyl-GM1 , GM2, GM3, GD2, GD3, GD1 a, GalNAc-GD1 a, GD1 b, GT1 a, GT1 b, GT1 aa, GQ1 b, GQ1 ba, LM1 , Hex-LM1 , furthermore carbohydrate antigens of the group of non- sialylated glycolipids such as sulfatide or asialo-GM1 / asialo-GM2, galactocerebroside, SGPG and SGLPG (HNK-1 epitope) (H. J. Willison and N. Yuki, Brain, 2002, 125, 2591 - 2625).

In the group of acute immune-mediated neuropathies, GBS encompasses several disease conditions that often involve autoantibodies against nerve glycoepitopes. The major subgroups among GBS are AMAN, AMSAN and AIDP, with AMAN predominantly affecting motor nerves compared to the other subtypes. GBS is associated with autoantibodies against gangliosides such as GM1 , GD1 a and structurally similar GM1 b and GalNAc-GD1 a, but also against ganglioside complexes, e.g. GM1 and GD1 a. The pharyngeal-cervical-brachial (PCB) variant of GBS correlates with autoantibodies against GT1 a alone or additionally GQ1 b. Another clinically distinct subgroup of GBS is the Miller Fisher syndrome, which is mainly associated with antibodies against the GQ1 b and the GT1 a epitope. The pathogenic autoantibodies in the group of acute neuropathies are mostly of the IgG isotype (E. Delmont, H. J. Willison, J Neurom Dis., 2015, 2, 107-1 12).

In contrast to acute neuropathies the chronic immune-mediated neuropathies are mostly associated with IgM autoantibodies. Chronic inflammatory demyelinating polyneuropathy (CI DP) is the most common form of chronic demyelinating polyneuropathy. Subtypes of CIDP involve pathogenic anti-glycan antibodies (E. Delmont, H. J. Willison, J Neurom Dis., 2015, 2, 107-1 12). The two other major disease groups among the chronic inflammatory neuropathies are the anti-MAG neuropathy and multifocal motor neuropathy (MMN). The anti-MAG neuropathy mainly involves autoantibodies against the HNK-1 epitope, present on multiple myelin antigens such as MAG, SGPG, SGLPG, P0 and PMP22. MMN patients often show autoantibodies against the ganglioside GM1 (or the complex GM1 :GalC). Other, less frequent, chronic neuropathies encompass the chronic sensory axonal neuropathy with anti-sulfatide antibodies, the chronic motor neuropathy with GD1 a or GD1 b antibodies, and the CANOMAD (chronic ataxic neuropathy, opthalmoplegia, M-protein, Agglutination, Disialosyl antibodies) with antibodies against disialosyl gangliosides, such as GQ1 b and GD1 b (E. Nobile-Orazio, Clinical Lymphoma & Myeloma, 2009, 9, 107-109).

Summary of the invention

The invention relates to carbohydrate ligands and moieties, respectively, that bind to antibodies against glycoepitopes of glycosphingolipids of the nervous system, polymers comprising these carbohydrate ligands, and to their use in diagnosis and therapy of neurological diseases. In particular, the invention relates to carbohydrate ligands and moieties, respectively, mimicking glycoepitopes comprised by glycosphingolipids of the nervous system, particularly glycoepitopes comprised by glycosphingolipids of the cerebroside, the (neo)lacto-, the ganglio- and the sulfoglucuronyl paragloboside-type, which are bound by anti-glycan antibodies associated with neurological diseases. The invention relates to the use of these carbohydrate ligands and moieties respectively, in diagnosis as well as for the treatment of neurological diseases associated with anti-glycan antibodies.

In a first aspect, the present invention provides for a compound comprising a carbohydrate moiety and a linker Z, wherein said carbohydrate moiety mimics, or alternatively and preferably is, a glycoepitope comprised by a glycosphingolipid of the nervous system, wherein said linker Z is -N(R a )-A-B-CH2-(CH 2 ) q -SH, wherein R a is H, C C 4 -alkyl, C C 4 -alkoxy, CH 2 C 6 H 5 , CH 2 CH 2 C 6 H 5 , OCH 2 C 6 H 5 , or OCH 2 CH 2 C 6 H 5 ; A is d- C 7 -alkylene, C C 7 -alkoxy, Ci-C 4 -alkyl-(OCH 2 CH 2 )pO-Ci-C 4 -alkyl, or C C 7 -alkoxy-R b , wherein R b is an optionally substituted aryl or an optionally substituted heteroaryl, and wherein p is 0 to 6, preferably p is 1 , 2 or 3, and further preferably p is 1 ; B is NHC(O), S or CH 2 ; q is 0 to 6, preferably q is 1 , 2, 3 or 4, and further preferably q is 1 or 2; and wherein said linker Z is covalently bound via its -N(R a )-group to the reducing end of said carbohydrate moiety.

In a second aspect, the present invention provides for a compound of formula (I) or of formula (II), wherein formula (I) is

wherein R 11 is Z or

wherein R 12 is H, S0 3 H, or

wherein R is H or

wherein R 14 is H or

wherein R and R are independently H or

wherein R 17 is H or and wherein formula (II) is M

wherein R is Z or I2

wherein R is Z or

wherein said linker Z is -N(R a )-A-B-CH 2 -(CH 2 ) q -SH, wherein R a is H, C C 4 -alkyl, C C 4 - alkoxy, CH 2 C 6 H 5 , CH 2 CH 2 C 6 H 5! OCH 2 C 6 H 5 , or OCH 2 CH 2 C 6 H 5 ; A is CrC 7 -alkylene, C C 7 - alkoxy, Ci-C 4 -alkyl-(OCH 2 CH 2 ) p O-C 1 -C 4 -alkyl ! or CrC 7 -alkoxy-R b , wherein R b is an optionally substituted aryl or an optionally substituted heteroaryl, and wherein p is 0 to 6, preferably p is 1 , 2 or 3, and further preferably p is 1 ; B is NHC(O), S or CH 2 ; q is 0 to 6, preferably q is 1 , 2, 3 or 4, and further preferably q is 1 or 2; and wherein said linker Z is covalently bound via its -N(R a )-group to the reducing end of said carbohydrate moiety. Furthermore, the invention relates to therapeutically acceptable polymers comprising a multitude of substituents derived from the inventive compounds, wherein said compounds are connected to the polymer backbone by way of the linker Z, and wherein the connection is effected via the SH-moiety of linker Z.

Thus, in another aspect, the present invention provides for a polymer comprising a multitude of the inventive compounds, wherein said compounds are connected to the polymer backbone by way of said linker Z, and wherein said connection is effected via the SH-group of said linker Z.

In a further aspect, the present invention provides for a polymer comprising (i) a multitude of compounds of formula (I), (ii) a multitude of compounds of formula (II) or (iii) a multitude of compounds of formula (I) and of formula (II), wherein said compounds are connected to the polymer backbone by way of said linker Z, and wherein said connection is effected via the SH-group of said linker Z. Preferably said multitude of compounds of formula (I) and/or of formula (II) are either identical compounds of formula (I) and/or of formula (II) or different compounds selected from of formula (I) and/or of formula (II). The invention relates also to pharmaceutical compositions comprising these compounds, diagnostic kits containing these, and to the use of these compounds for the diagnosis and therapy of neurological diseases associated with anti-glycan antibodies.

Thus, in another aspect, the present invention provides for a pharmaceutical composition comprising said inventive compound, preferably said inventive compound of formula (I) or of formula (II), or comprising said inventive.

In another aspect, the present invention provides for said inventive compound, preferably said inventive compound of formula (I) or formula (II), or said inventive polymer, or said inventive pharmaceutical composition for use in a method of treating a neurological disease, wherein preferably said neurological disease is selected from multiple sclerosis, Parkinson's disease, Alzheimer's disease, dementia, and an immune-mediated neuropathy, wherein preferably said immune-mediated neuropathy is selected from Guillain-Barre syndrome (GBS), chronic inflammatory demyelinating polyneuropathy (CI DP), Miller-Fischer syndrome, Bickerstaff brainstem encephalitis, multifocal motor neuropathy or anti-MAG neuropathy, wherein further preferably said immune-mediated neuropathy is selected from Guillain-Barre syndrome (GBS), chronic inflammatory demyelinating polyneuropathy (CI DP), Miller-Fischer syndrome, Bickerstaff brainstem encephalitis or multifocal motor neuropathy. In another aspect, the present invention provides for said inventive compound, preferably said inventive compound of formula (I) or formula (II), or said inventive polymer, or said inventive pharmaceutical composition for use in a method of diagnosis of a neurological disease, wherein preferably said neurological disease is an immune-mediated neuropathy. In another aspect, the present invention provides for a diagnostic kit comprising said inventive compound, preferably said inventive compound of formula (I) or formula (II), or said inventive polymer.

In another aspect, the present invention provides for an use of said inventive compound, preferably said inventive compound of formula (I) or formula (II), or said inventive polymer for the diagnosis of a neurological disease, wherein preferably said neurological disease is an immune-mediated neuropathy.

In another aspect, the present invention provides for an use of said inventive compound, preferably said inventive compound of formula (I) or formula (II), or said inventive polymer, for the manufacture of a medicament for the treatment of a neurological disease, wherein preferably said neurological disease is selected from multiple sclerosis, Parkinson's disease, Alzheimer's disease, dementia, and an immune-mediated neuropathy, wherein preferably said immune-mediated neuropathy is selected from Guillain-Barre syndrome (GBS), chronic inflammatory demyelinating polyneuropathy (CIDP), Miller-Fischer syndrome, Bickerstaff brainstem encephalitis, multifocal motor neuropathy or anti-MAG neuropathy, wherein further preferably said immune-mediated neuropathy is selected from Guillain-Barre syndrome (GBS), chronic inflammatory demyelinating polyneuropathy (CIDP), Miller-Fischer syndrome, Bickerstaff brainstem encephalitis or multifocal motor neuropathy.

In another aspect, the present invention provides for a method of treatment of a neurological disease, wherein preferably said neurological disease is an immune- mediated neuropathy, wherein said method comprises administering said inventive compound, preferably said inventive compound of formula (I) or formula (II), or said inventive polymer in a quantity effective against said disease, to a warm-blooded animal, preferably to a human, requiring such treatment.

Brief Description of the Figures

FIG. 1 : Schematic representation of a competitive binding assay (a) Co-incubation of glycolipid-coated plates with neuropathy patient sera, containing anti- glycolipid antibodies of the IgG (and/or IgM) isotype, and glycopolymers. In this particular representative example GM1 a ganglioside-coated plates are co-incubated with anti-GMa IgG-containing serum and glycopolymer 6. (b) Wash step, (c) Incubation with anti-human IgG (or IgM) antibody coupled to horseradish peroxidase, (d) Wash step, (e) Addition of tetramethylbenzidin (TMB) substrate, (f) Addition of acidic stop solution and measurement of the optical density.

FIG. 2: Binding curves for compounds 6, 26, 34 and 86

FIG. 2A: The GM1 a-ganglioside-coated wells were co-incubated with compound 6 (1 mM highest concentration) and the two patient sera PP IgG Pos. (IgG), P21 (IgG). Compound 6 is a polylysine polymer (average of 250 repeating lysine units) with a defined percentage of lysine residues coupled to the GM1 a glycoepitope (4). The general abbreviation used is as follows: PL(glycoepitope) x with x defining the percentage of glycoepitope loading in %. In this case the polymer is PL(GM1 a) 2 8- Results are indicated as mean ± SD.

FIG. 2B: Co-incubation of GM1 a-coated wells with PL(GM1 a) 2 s polymer 6 (3 mM highest concentration) together with patient sera P3 (IgM) and P4 (IgM). Results are indicated as mean ± SD.

FIG. 2C: Co-incubation of GD1 b-coated wells with the PL(GD1 b) 20 polymer 26 (3 mM highest concentration) together with patient sera P22 (IgG). Results are indicated as mean ± SD. FIG. 2D: Co-incubation of GQ1 b-coated wells with the PL(GT1 a) 58 polymer 34 (3 mM highest concentration) together with patient sera EK-GCO 1803 (IgG), P23 (IgG). Results are indicated as mean ± SD.

FIG. 2E: Co-incubation of MAG-coated wells (MAG contains up to eight HNK-1 glycoepitopes) and the PL(HNK-1 mimetic(58)) 4 o polymer 86 (100 μΜ highest concentration) together with a mouse monoclonal anti-HNK-1 IgM antibody. Results are indicated as mean ± SD. FIG. 3: BALB/c wild type mice were immunized against the two glycosphingolipids SGPG and SGLPG, of which both bear the HNK-1 glycoepitope. Immunized mice showed high levels of anti-HNK-1 (anti-MAG) IgM antibodies at day 154 after immunization (0 h, pre- treatment). These induced mouse antibodies are a model for human anti-MAG IgM of anti- MAG neuropathy patients. An intravenous administration of the PL(HNK-1 mimetic(58)) 4 o polymer 86 (10 mg/kg) to immunized BALB/c mice (n = 6) led to a significant reduction of anti-HNK-1 (anti-MAG) IgM antibodies for up to a week (168h) after administration. Results are indicated as mean ± 95% CI (above) and mean ± SD (below). Results were analyzed by one-way ANOVA with Dunnett's multiple comparison posttest with a 0.05 confidence level accepted for statistical significance ( * p<0.05, ** p<0.01 , *** p<0.001 ).

Detailed description of the invention

The compounds of the present invention, and in particular the compounds of the present invention of formula (I) or (II), recognize anti-glycan antibodies against glycosphingolipid glycoepitopes of the nervous system, in particular glycoepitopes comprised by glycosphingolipids such as the cerebroside-, (neo)lacto-, and the ganglio-types. The carbohydrate ligands contain linkers that allow coupling to a polymer backbone for multivalent presentation. The glycopolymers resulting from the coupling are superior in the sequestration of anti-carbohydrate antibodies compared to the respective glycan- monomers. The glycopolymers are suitable diagnostic or therapeutic agents to detect and to bind anti-glycan antibodies in particular associated with neurological diseases.

The present invention provides for a compound comprising a carbohydrate moiety and a linker Z, wherein said carbohydrate moiety mimics, or alternatively and preferably is, a glycoepitope comprised by a glycosphingolipid of the nervous system, wherein said linker Z is -N(R a )-A-B-CH 2 -(CH 2 ) q -SH, wherein R a is H, C C 4 -alkyl, C C 4 -alkoxy, CH 2 C 6 H 5 , CH2CH2C 6 H 5 , OCH 2 C 6 H5, or OCH2CH2C 6 H 5 ; A is C C 7 -alkylene, C C 7 -alkoxy, C C 4 - alkyl-(OCH 2 CH 2 ) p O-Ci-C 4 -alkyl, or C C 7 -alkoxy-R b , wherein R b is an optionally substituted aryl or an optionally substituted heteroaryl, and wherein p is 0 to 6, preferably p is 1 , 2 or 3, and further preferably p is 1 ; B is NHC(O), S or CH 2 ; q is 0 to 6, preferably q is 1 , 2, 3 or 4, and further preferably q is 1 or 2; and wherein said linker Z is covalently bound via its -N(R a )-group to the reducing end of said carbohydrate moiety.

In a preferred embodiment, said glycosphingolipid of the nervous system is selected from the cerebroside-, (neo)lacto-, ganglio-, or sulfoglucuronyl paragloboside-type. In a further preferred embodiment, said glycosphingolipid of the nervous system is a ganglioside, wherein preferably said ganglioside is selected from GM1 (GM1a), GM1b, GalNAc-GM1b, Fucosyl-GM1, GM2, GM3, GD2, GD3, GD1a, GalNAc-GD1a, GD1b, GT1a, GT1b, GT1aa, GQ1 b, GQ1 ba, LM1 or Hex-LM1.

In particular, the present invention provides for a compound of formula (I) or of formula (II), wherein formula (I) is

wherein R' 2 is H, S0 3 H, or

wherein R is H or

wherein R is H or

wherein R 15 and R 16 are independently H or

wherein R 17 is H or

wherein R is Z or

wherein said linker Z is -N(R a )-A-B-CH2-(CH2) q -SH, wherein R a is H, CrC 4 -alkyl, C C 4 - alkoxy, CH 2 C 6 H5, CH 2 CH 2 C6H 5! OCH 2 C 6 H 5 , or OCH 2 CH 2 C 6 H 5 ; A is C C 7 -alkylene, C C 7 - alkoxy, Ci-C 4 -alkyl-(OCH 2 CH2) P 0-Ci-C4-alkyl, or C C 7 -alkoxy-R , wherein R is an optionally substituted aryl or an optionally substituted heteroaryl, and wherein p is 0 to 6, preferably p is 1 , 2 or 3, and further preferably p is 1 ; B is NHC(O), S or CH 2 ; q is 0 to 6, preferably q is 1 , 2, 3 or 4, and further preferably q is 1 or 2; and wherein said linker Z is covalently bound via its -N(R a )-group to the reducing end of said carbohydrate moiety.

In a further preferred embodiment, said compound is a compound of formula (I). The scope of the present invention comprises carbohydrate moieties mimicking glycoepitopes comprised by glycosphingolipid of the nervous system. Preferred compounds mimicking glycoepitopes comprised by glycosphingolipid of the nervous system in accordance with the present invention are compounds of the formula (I) as defined herein, wherein at least one of sialic acid moiety is replaced by a replacement moiety as shown and defined in formula (la) or formula (lb)

(la) (lb) wherein for said replacement moiety of formula (lb), R is H, CrC 8 -alkyl, Ci-C 8 -alkyl- cycloalkyl, CrC 8 -alkenyl, CrC 8 -alkynyl, aryl, substituted aryl, wherein preferably said substitution of said aryl is by halogen, Ci-C 8- alkoxy, Ci-C 8 -alkyl; heteroaryl, substituted heteroaryl, wherein preferably said substitution of said hetereoaryl is by halogen, Ci-C 8- alkoxy, CrC 8 -alkyl; arylalkyi, substituted arylalkyi, wherein preferably said substitution of said arylalkyi is by halogen, Ci-C 8- alkoxy, Ci-C 8 -alkyl; heteroarylalkyl, substituted heteroarylalkyl, wherein preferably said substitution of said heteroarylalkyl is by halogen, Ci-C 8- alkoxy, Ci-C 8 -alkyl; cycloalkyl, cycloalkyl-Ci-C 8 -alkyl, i-butyl, adamantyl, triazolyl all of which independently substituted with Ci-C 8 alkyl, aryl, heteroaryl, halogen.

In another preferred embodiment, said compound is a compound of formula (II).

In another preferred embodiment, said compound is a compound of formula 4 * , 9 * , 13 * , 17 * , 21 * , 25 * , 29 * , 33 * , or 46 * -60 * as depicted below.

4*

wherein said linker Z is -N(R a )-A-B-CH2-(CH2) q -SH, wherein R a is H , Ci-C 4 -alkyl, C C 4 - alkoxy, CH 2 C 6 H 5 , CH2CH2C6H5, OCH 2 C6H 5 , or OCH2CH2C6H5; A is Ci-C 7 -alkylene, C C 7 - alkoxy, Ci-C 4 -alkyl-(OCH 2 CH2)pO-Ci-C 4 -alkyl, or Ci-C 7 -alkoxy-R b , wherein R b is an optionally substituted aryl or an optionally substituted heteroaryl, and wherein p is 0 to 6, preferably p is 1 , 2 or 3, and further preferably p is 1 ; B is NHC(O), S or CH 2 ; q is 0 to 6, preferably q is 1 , 2, 3 or 4, and further preferably q is 1 or 2; and wherein said linker Z is covalently bound via its -N(R a )-group to the reducing end of said carbohydrate moiety.

In a further very preferred embodiment, said compound is a compound of formula 4*, 9*, 13*, 17*, 21 *, 25*, 29*, 33*, or 46*-60*, wherein at least one of sialic acid moiety is replaced by a replacement moiety as shown and defined in formula (la) or formula (lb)

(la) (lb) wherein for said replacement moiety of formula (lb), R 18 is H, CrC 8 -alkyl, Ci-C 8 -alkyl- cycloalkyl, CrC 8 -alkenyl, CrC 8 -alkynyl, aryl, substituted aryl, wherein preferably said substitution of said aryl is by halogen, Ci-C 8- alkoxy, Ci-C 8 -alkyl; heteroaryl, substituted heteroaryl, wherein preferably said substitution of said hetereoaryl is by halogen, Ci-C 8- alkoxy, CrC 8 -alkyl; arylalkyi, substituted arylalkyi, wherein preferably said substitution of said arylalkyi is by halogen, Ci-C 8- alkoxy, CrC 8 -alkyl; heteroarylalkyl, substituted heteroarylalkyl, wherein preferably said substitution of said heteroarylalkyl is by halogen, Ci-C 8- alkoxy, Ci-C 8 -alkyl; cycloalkyi, cycloalkyl-Ci-C 8 -alkyl, i-butyl, adamantyl, triazolyl all of which independently substituted with Ci-C 8 alkyl, aryl, heteroaryl, halogen.

Preferred embodiments of said linker Z are as follows. Thus, in one embodiment, R a is H, CH 3 , CH2CH 3 , CH2CH2CH 3 , CH(CH 3 )2, OCH 3 , OCH2CH 3 , OCH2CH2CI l3 i CH2C 6 H 5 , OCH 2 C 6 H 5 ; A is 0(CH 2 ) P CH 2 , (CH 2 ) P CH 2 , CH 2 (OCH 2 CH2)pOCH2, (OCH 2 CH2)pOCH 2 CH2 or 0(CH2)pC 6 H 5 ; and B is NHC(O), S or CH 2 . In a preferred embodiment, R a is CH 3 or OCH 3 ; A is 0(CH 2 ) P CH 2 , (CH 2 ) P CH 2 , CH2(OCH 2 CH 2 )pOCH2, (OCH 2 CH2)pOCH 2 CH2 or 0(CH 2 )pC 6 H 5 ; and B is NHC(O) or S. Preferably, when B is S, and A is (CH 2 )pCH 2 , then q is 1 to 5, preferably 1 , 2 or 3.

In a further preferred embodiment, R a is CH 3 or OCH 3 ; A is 0(CH 2 )pCH 2 , (CH 2 )pCH 2 , CH2(OCH 2 CH 2 )pOCH2, (OCH 2 CH2)pOCH 2 CH2 or 0(CH 2 )pC 6 H 5 ; and B is NHC(O). In a further preferred embodiment, R a is CH 3 ; A is 0(CH 2 )pCH 2 , (CH 2 )pCH 2 , CH2(OCH 2 CH 2 )pOCH2, (OCH 2 CH2)pOCH 2 CH2 or 0(CH 2 )pC 6 H 5 ; and B is NHC(O) or S. Preferably, when B is S and A is (CH 2 )pCH2, then q is 1 to 5, preferably 1 , 2 or 3.

In a further preferred embodiment, R a is CH 3 or OCH 3 ; A is 0(CH 2 )pCH 2 , (CH 2 )pCH 2 , CH2(OCH 2 CH 2 )pOCH2, (OCH 2 CH2)pOCH 2 CH2 or 0(CH 2 )pC 6 H 5 ; B is NHC(O) or S; and q is 1 to 5, preferably 1 , 2 or 3, preferably 2.

In a further preferred embodiment, said linker Z is of a formula selected from any one of the formula (a) to (g):

(d)

wherein p is between 0 and 6, preferably 1 to 3, in particular 1 , and q is between 0 and 6, preferably between 1 and 4, in particular 1 or 2. In one embodiment, when said linker Z is of formula (e), then p and q are independently 1 to 6, preferably 1 , 2 or 3; wherein, when p is 2, then q is 1 to 6, preferably 1 or 3 to 6, and when q is 2, then p is 3 to 6. In another embodiment, when said linker Z is of formula (e), then p and q are not both 2.

In said further preferred embodiment, and in light of the general formula of the present invention said linker Z is of a formula selected from any one of the formula (a) to

(9):

q (f) wherein p is between 0 and 6, preferably 1 to 3, in particular 1 , and q is between 0 and 6, preferably between 1 and 4, in particular 2 In one embodiment, when said linker Z is of formula (e), then p and q are independently 1 to 6, preferably 1 , 2 or 3; wherein, when p is 2, then q is 1 to 6, preferably 1 or 3 to 6, and when q is 2, then p is 3 to 6. In another embodiment, when said linker Z is of formula (e), then p and q are not both 2.

In a very preferred embodiment, said linker Z is -N(CH 3 )-0(CH2)2-NHC(0)-(CH 2 )3-SH.

In a further preferred embodiment, said carbohydrate moiety mimicking, or alternatively and preferably being, a glycoepitope comprised by a glycosphingolipid of the nervous system is a carbohydrate moiety comprised by a compound of formula (I), and said glycoepitope is a glycoepitope of the cerebroside-, (neo)lacto-, or ganglio-type, further preferably of a ganglioside.

In a further preferred embodiment, said carbohydrate moiety mimicking, or alternatively and preferably being, a glycoepitope comprised by a glycosphingolipid of the nervous system is a carbohydrate moiety comprised by a compound of formula (II), and said glycoepitope is a glycoepitope of a sulfoglucuronyl paragloboside and hereby in particular a glycoepitope such as the antigenic HNK-1 carbohydrate epitope.

In a further very preferred embodiment, said compound is a compound of formula 4, 9, 13, 17, 21 , 25, 29, 33, 37, 41 , 44, 56, 58 or 77. The formula are shown in the examples.

In a further very preferred embodiment, said compound is a compound of formula 4, 21 , 25, 33, 37, 41 or 44. The formula are shown in the examples.

In a further very preferred embodiment, said compound is a compound of formula 4, 9, 13, 17, 21 , 25, 29, 33, 37, 41 , 44, 56, 58 or 77, wherein at least one of sialic acid moiety is replaced by a replacement moiety as shown and defined in formula (la) or formula (lb)

(la) (lb) wherein for said replacement moiety of formula (lb), R is H, CrC 8 -alkyl, Ci-C 8 -alkyl- cycloalkyl, CrC 8 -alkenyl, CrC 8 -alkynyl, aryl, substituted aryl, wherein preferably said substitution of said aryl is by halogen, Ci-C 8- alkoxy, Ci-C 8 -alkyl; heteroaryl, substituted heteroaryl, wherein preferably said substitution of said hetereoaryl is by halogen, Ci-C 8- alkoxy, CrC 8 -alkyl; arylalkyi, substituted arylalkyi, wherein preferably said substitution of said arylalkyi is by halogen, Ci-C 8- alkoxy, Ci-C 8 -alkyl; heteroarylalkyl, substituted heteroarylalkyl, wherein preferably said substitution of said heteroarylalkyl is by halogen, Ci-C 8- alkoxy, Ci-C 8 -alkyl; cycloalkyl, cycloalkyl-Ci-C 8 -alkyl, i-butyl, adamantyl, triazolyl all of which independently substituted with Ci-C 8 alkyl, aryl, heteroaryl, halogen. In a further very preferred embodiment, said compound is a compound of formula 4, 21 , 25, 33, 37, 41 or 44, wherein at least one of sialic acid moiety is replaced by a replacement moiety as shown and defined in formula (la) or formula (lb)

(la) (lb) wherein for said replacement moiety of formula (lb), R is H, CrC 8 -alkyl, Ci-C 8 -alkyl- cycloalkyl, Ci-C 8 -alkenyl, Ci-C 8 -alkynyl, aryl, substituted aryl, wherein preferably said substitution of said aryl is by halogen, Ci-C 8- alkoxy, Ci-C 8 -alkyl; heteroaryl, substituted heteroaryl, wherein preferably said substitution of said hetereoaryl is by halogen, Ci-C 8- alkoxy, CrC 8 -alkyl; arylalkyi, substituted arylalkyi, wherein preferably said substitution of said arylalkyi is by halogen, Ci-C 8- alkoxy, Ci-C 8 -alkyl; heteroarylalkyl, substituted heteroarylalkyl, wherein preferably said substitution of said heteroarylalkyl is by halogen, Ci-C 8- alkoxy, Ci-C 8 -alkyl; cycloalkyl, cycloalkyl-Ci-C 8 -alkyl, i-butyl, adamantyl, triazolyl all of which independently substituted with Ci-C 8 alkyl, aryl, heteroaryl, halogen.

Furthermore the invention relates to therapeutically acceptable polymers comprising a multitude of substituents derived from the inventive compounds, wherein said compounds are connected to the polymer backbone by way of the linker Z, and wherein the connection is effected via the SH-moiety of linker Z. Typically, said inventive polymer further comprises spacer moieties for coupling of said SH-moieties of the linker Z to reactive moieties on the polymer backbone. Such spacer moieties are known to the skilled person in the art and preferred exmples are described herein. Thus, in another aspect, the present invention provides for a polymer comprising a multitude of the inventive compounds, wherein said compounds are connected to the polymer backbone by way of said linker Z, and wherein said connection is effected via the SH-group of said linker Z. Typically, said inventive polymer further comprises spacer moieties for coupling of said SH-moieties of the linker Z to reactive moieties on the polymer backbone. Preferred examples are described herein.

In a further aspect, the present invention provides for a polymer comprising (i) a multitude of compounds of formula (I), (ii) a multitude of compounds of formula (II) or (iii) a multitude of compounds of formula (I) and of formula (II), wherein said compounds are connected to the polymer backbone by way of said linker Z, and wherein said connection is effected via the SH-group of said linker Z. Preferably said multitude of compounds of formula (I) and/or of formula (II) are either identical compounds of formula (I) and/or of formula (II) or different compounds selected from of formula (I) and/or of formula (II). Typically, said inventive polymer further comprises spacer moieties for coupling of said SH-moieties of the linker Z to reactive moieties on the polymer backbone. Preferred examples are described herein.

In a further preferred embodiment, said polymer comprises (i) a multitude of compounds of formula (I), (ii) a multitude of compounds of formula (II) or (iii) a multitude of compounds of formula (I) and of formula (II), wherein said compounds are connected to the polymer backbone by way of said linker Z, and wherein said connection is effected via the SH- group of said linker Z, and wherein said linker Z is -N(R a )-A-B-CH 2 -(CH 2 ) q -SH, wherein R a is H, CrC 4 -alkyl, C C 4 -alkoxy, CH 2 C 6 H 5 , CH 2 CH 2 C 6 H 5 , OCH 2 C 6 H 5 , or OCH 2 CH 2 C 6 H 5 ; A is C C 7 -alkylene, C C 7 -alkoxy, CrC 4 -alkyl-(OCH 2 CH 2 ) p O-Ci-C 4 -alkyl, or C C 7 -alkoxy- R b , wherein R b is an optionally substituted aryl or an optionally substituted heteroaryl, and wherein p is 0 to 6, preferably p is 1 , 2 or 3, and further preferably p is 1 ; B is N HC(O), S or CH 2 ; q is 0 to 6, preferably q is 1 , 2, 3 or 4, and further preferably q is 1 or 2; and wherein said linker Z is covalently bound via its -N(R a )-group to the reducing end of said carbohydrate moiety. Preferably said multitude of compounds of formula (I) and/or of formula (II) are either identical compounds of formula (I) and/or of formula (II) or different compounds selected from of formula (I) and/or of formula (II). Typically, said inventive polymer further comprises spacer moieties for coupling of said SH-moieties of the linker Z to reactive moieties on the polymer backbone. Preferred examples are described herein.

Preferred embodiments of said linker Z are as follows. Thus, in one embodiment, R a is H , CH 3 , CH 2 CH 3 , CH 2 CH 2 CH 3 , CH(CH 3 ) 2 , OCH 3 , OCH 2 CH 3 , ΟΟΗ 2 ΟΗ 2 ΟΗ 3: CH 2 C 6 Hs, OCH 2 C 6 H 5 ; A is 0(CH 2 ) p CH 2! (CH 2 ) P CH 2! CH 2 (OCH 2 CH 2 ) p OCH 2! (OCH 2 CH 2 ) p OCH 2 CH 2 or 0(CH 2 ) p C 6 H 5 ; and B is NHC(O), S or CH 2 . In a preferred embodiment, R a is CH 3 or OCH 3 ; A is 0(CH 2 ) p CH 2 , (CH 2 ) P CH 2 , CH 2 (OCH 2 CH 2 ) p OCH 2 , (OCH 2 CH 2 ) p OCH 2 CH 2 or 0(CH 2 ) p C 6 H 5 ; and B is NHC(O) or S. Preferably, when B is S, and A is (CH 2 ) P CH 2 , then q is 1 to 6, preferably 1 , 2 or 3.

In a further preferred embodiment of said linker comprised by said inventive polymer, R a is CH 3 or OCH 3 ; A is 0(CH 2 ) p CH 2 , (CH 2 ) P CH 2 , CH 2 (OCH 2 CH 2 ) p OCH 2 , (OCH 2 CH 2 ) p OCH 2 CH 2 or 0(CH 2 ) p C 6 H 5 ; and B is NHC(O). In another preferred embodiment of said linker comprised by said inventive polymer, R a is CH 3 ; A is 0(CH 2 ) p CH 2 , (CH 2 ) P CH 2 , CH 2 (OCH 2 CH 2 ) p OCH 2 , (OCH 2 CH 2 ) p OCH 2 CH 2 or 0(CH 2 ) p C 6 H 5 ; and B is NHC(O) or S. Preferably, when B is S and A is (CH 2 ) P CH 2 , then q is 1 to 6, preferably 1 , 2 or 3. In a further preferred embodiment of said linker comprised by said inventive polymer, R a is CH 3 or OCH 3 ; A is 0(CH 2 ) p CH 2 , (CH 2 ) P CH 2 , CH 2 (OCH 2 CH 2 ) p OCH 2 , (OCH 2 CH 2 ) p OCH 2 CH 2 or 0(CH 2 ) p C 6 H 5 ; B is NHC(O) or S; and q is 1 to 6, preferably 1 , 2, 3, 4 or 5, preferably 2 or 4, further preferably 2.

Preferably, said linker Z is of a formula selected from any one of the formula (a) to (g):

(g) wherein p is between 0 and 6, preferably 1 to 3, in particular 1 , and q is between 0 and 6, preferably between 2 and 4, in particular 2 In one embodiment, when said linker Z is of formula (e), then p and q are independently 1 to 6, preferably 1 , 2 or 3; wherein, when p is 2, then q is 1 to 6, preferably 1 or 3 to 6, and when q is 2, then p is 3 to 6. In another embodiment, when said linker Z is of formula (e), then p and q are not both 2.

Preferably, and in light of the general formula of the present invention, said linker Z is of a formula selected from any one of the formula (a) to (g):

OMe

I

. N . . SH

P q (e)

wherein p is between 0 and 6, preferably 1 to 3, in particular 1 , and q is between 0 and 6, preferably between 2 and 4, in particular 2 In one embodiment, when said linker Z is of formula (e), then p and q are independently 2 to 6, preferably 2, 3 or 4; wherein, when p is 2, then q is 1 to 6, preferably 1 or 3 to 6, and when q is 2, then p is 3 to 6. In another embodiment, when said linker Z is of formula (e), then p and q are not both 2. In a very preferred embodiment, said linker Z is -N(CH 3 )-0(CH2)2-NHC(0)-(CH 2 )3-SH. The invention further particularly relates to compounds of formula (I) and (II) and to therapeutically acceptable polymers comprising a multitude of these compounds, including polymers with loading of a multitude of one identical compound of formula (I) or (II) or a multitude being a combination of several different compounds of formula (I) or (II). Preferred polymers in said context are polymers with loading of one or several of compounds of formula (I) or (II), wherein said compounds of formula (I) or (II) are preferably selected from 4 * , 9 * , 13 * , 17 * , 21 * , 25 * , 29 * or 33 * , and 46 * -60 * .

The inventive polymer comprising the multitude of identical or different compounds of formula (I) and/or (II) wherein the SH-group of said linker Z connects said compounds to the polymer backbone, is preferably an oarmino acid polymer, and hereby typically and preferably a homomeric or heteromeric oarmino acid polymer, an acrylic acid or methacrylic acid polymer or copolymer, or a N-vinyl-2-pyrrolidone-vinylalcohol copolymer, a chitosan polymer, or a polyphosphazene polymer.

In a preferred embodiment, the polymer backbone is an oarmino acid polymer, an acrylic acid or methacrylic acid polymer or copolymer, a N-vinyl-2-pyrrolidone-vinyl alcohol copolymer, a chitosan polymer, or a polyphosphazene polymer.

In another preferred embodiment, the polymer backbone is an oamino acid polymer.

In a further preferred embodiment, the polymer backbone is an oamino acid polymer and said oamino acid of said oamino acid polymer is lysine, ornithine, glutamic acid, aspartic acid or serine.

In a very preferred embodiment, the polymer backbone is poly-lysine, and wherein preferably the molecular weight of said poly-lysine is 1 Ό00 Da to 300Ό00 Da.

In a further preferred embodiment, the percentage of loading of the carbohydrate moiety of said compound onto the polymer backbone is between 10 and 90%, preferably between 20 and 70%, and in particular between 30 and 60%. The latter means that 30 to 60% of the reactive polymer side chains and, if applicable the spacer moiety, are reacted with the -SH group of said linker Z. The percentage of loading of the carbohydrate moiety of said compound onto the polymer backbone is typically and preferably determined by NMR spectroscopy and refers to % mole/mole. Further particular examples of polymers of the invention are

(A) a poly-oamino acid, wherein the amino acid carries a side chain aminoalkyl function, such as in poly-lysine, in particular poly-L-lysine or poly-D-lysine, and the amino group is connected via a spacer moiety to the SH-group of said linker Z. A typical and preferred spacer moiety comprises a terminal CH 2 -group, wherein said terminal CH 2 -group of said spacer moiety is connected to the S-of said linker Z. A preferred spacer moiety is an acetyl group. (B) a poly-oamino acid (D- and L- form), wherein the amino acid carries a side chain carbonylalkyl function, such as in poly-aspartic acid, poly-glutamic acid, poly-asparagine or poly-glutamine, and the carbonyl group (which corresponds to the original carboxy group in aspartic acid and glutamic acid, respectively) is connected via a spacer moiety to the SH-group of said linker Z. A typical and preferred spacer moiety comprises a terminal CH 2 -group, wherein said terminal CH 2 -group of said spacer moiety is connected to the S- of said linker Z.

(C) a poly-oamino acid (D- and L- form), wherein the amino acid carries a side chain hydroxyalkyl or hydroxyaryl function, such as in poly-serine, poly-threonine, poly-tyrosine, or poly-hydroxyproline, and the hydroxy group is connected via a spacer moiety to the SH-group of said linker Z. A typical and preferred spacer moiety comprises a terminal CH 2 -group, wherein said terminal CH 2 -group of said spacer moiety is connected to the S- of said linker Z.

(D) a poly-oamino acid, wherein the amino acid carries a side chain thiolalkyl function, such as in poly-cysteine, wherein the terminal CH 2 group of the amino acid side-chain (next to the thiol) is connected to the terminal SH group of linker Z, typically and preferably as a thioether;

(E) Co-polymers of two or more different oamino acids connected via a spacer moiety to the SH-group of said linker Z, as described in (A)-(D);

(F) poly-acrylic acid, poly-methacrylic acid or a copolymer of acrylic and methacrylic acid, wherein the carboxy group is connected via a spacer moiety to the SH-group of said linker Z. A typical and preferred spacer moiety comprises a terminal CH 2 -group, wherein said terminal CH 2 -group of said spacer moiety is connected to the S-of said linker Z.

(G) a copolymer of N-vinyl-2-pyrrolidone and vinyl alcohol, wherein the hydroxy group of the vinyl alcohol part of the copolymer is connected via a spacer moiety to the SH-group of said linker Z. A typical and preferred spacer moiety comprises a terminal CH 2 -group, wherein said terminal CH 2 -group of said spacer moiety is connected to the S-of said linker Z. (H) chitosan, wherein the amino group is connected via a spacer moiety to the SH-group of said linker Z. A typical and preferred spacer moiety comprises a terminal CH 2 -group, wherein said terminal CH 2 -group of said spacer moiety is connected to the S-of said linker Z; and

(I) a polyphosphazene polymer, wherein the terminal ester group is connected via a spacer moiety to the SH-group of said linker Z. A typical and preferred spacer moiety comprises a terminal CH 2 -group, wherein said terminal CH 2 -group of said spacer moiety is connected to the S-of said linker Z. A preferred spacer moiety is an acetyl group.

In a particular embodiment, a polymer (A) comprises the partial formula (III)

wherein

R 1 is an aminoalkyi substituent connected to said linker Z, wherein the SH-group of of said linker Z is connected to the terminal amino group of R 1 via a spacer moiety, wherein typically and preferably said spacer moiety is an acetyl group.,

R 2 is 2,3-dihydroxypropylthioacetyl-aminoalkyl, which is a capped amino function having a solubilizing substituent, and the relation between the two bracketed entities with R 1 and R 2 , respectively, in the polymer indicates the relation of carbohydrate loading to capped amino function.

For example, R 1 is of formula (Ilia)

and R 2 is of formula (lllb) wherein o is between 1 and 6, preferably 3 or 4 and m is between 1 and 6, preferably between 1 and 2, in particular 1.

When o is 3, substituent R 1 represents a side chain of poly-ornithine, and when o is 4, substituent R 1 represents a side chain of poly-lysine, connected to said SH-group of said linker Z which linker Z is comprised by the inventive compounds, and preferably by the inventive compounds of formula (I) or (II),

The poly-amino acid can be linear, hyperbranched or dendritic, as described by Z. Kadlecova et al., Biomacromolecules 2012, 13:3127-3137, for poly-lysine as follows:

The poly-lysine used to prepare polymer (A) of formula (III) has preferably a molecular weight between 1 Ό00 and 300Ό00 Da, in particular 30Ό00 to 70Ό00 Da, and such polymers further connected via the SH-group of the linker Z to compounds of formula (I) and/or (II) and with a capping 2,3-dihydroxypropylthio-acetylylaminoalkyl residue are preferred. For example, the polylysine polymer is first functionalized by chloroacetylation. Reaction of the chloroacetylated polymer with said linker Z comprising the terminal thiol functionality by nucleophilic substitution gives access to the desired polymers.

In a particular embodiment, a polymer (B) comprises the partial formula (III)

wherein

R 1 is a carbonylalkyl substituent connected to said linker Z, wherein the SH-group of said linker Z is connected to the -CH 2 -group of R 1 ,

R 2 is 2,3-dihydroxypropylthio-carbonylalkyl, and the relation between the two bracketed entities with R 1 and R 2 , respectively, in the polymer indicates the relation of carbohydrate loading to capped carbonyl or carboxy function.

For example, R 1 is of formula (lllc)

and R 2 is of formula (Hid)

wherein X is either oxygen or nitrogen, o is between 1 and 6, preferably 1 or 2, m is between 1 and 6, preferably between 1 and 2, in particular 1 . When o is 1 and X is O, substituent R 1 represents a side chain of poly-aspartic acid, and when o is 2 and X is O, substituent R 1 represents a side chain of poly-glutamic acid, when o is 1 and X is N, substituent R 1 represents a side chain of poly-asparagine, and when o is 2 and X is N, substituent R 1 represents a side chain of poly-glutamine, connected to said SH-group of said linker Z which linker Z is comprised by the inventive compounds, and preferably by the inventive compounds of formula (I) or (II), and R 2 is 2,3-dihydroxypropylthio-carbonylalkyl, i.e. a capped carboxy or amide function having a solubilizing substituent.

The poly-aspartic acid used to prepare polymer (B) of formula (IV) has preferably a molecular weight between 1 Ό00 and 300Ό00 Da, in particular 30Ό00 to 70Ό00 Da, and such polymers further connected via the SH-group of said linker Z to compounds of formula (I) and/or (II) and with a capping 2,3-dihydroxypropylthio-carbonylalkyl residue are preferred. For example, polyaspartic acid is first functionalized by esterification. Reaction of the chloroacetylated polymer with said linker Z comprising the terminal thiol functionality by nucleophilic substitution gives access to the desired polymers.

In case of poly-aspartic acid or poly-glutamic acid the polymer can be linear, hyperbranched or dendritic.

In a particular embodiment, a polymer (C) comprises the partial formula (III)

(Hi), wherein

R 1 is a hydroxyalkyl or hydroxyaryl substituent connected to said linker Z, wherein the SH-group of said linker Z is connected to the -CH 2 -group of R 1 ,

R 2 is 2,3-dihydroxypropylthioacetyl-hydroxyalkyl (or -hydroxyaryl), and the relation between the two bracketed entities with R 1 and R 2 , respectively, in the polymer indicates the relation of carbohydrate loading to capped hydroxy function.

For example, in the case of poly-serine and analogs, R 1 is of formula (Hie) and R 2 is of formula (lllf)

(lllf) wherein o is between 1 and 6, preferably 1 or 2, in partcular 1 , m is between 1 and 6, preferably between 1 and 2, in particular 1 .

When o is 1 , substituent R 1 represents a side chain of poly-serine, connected to said SH- group of said linker Z, which linker Z is comprised by the inventive compounds, and preferably by the inventive compounds of formula (I) or (II), and R 2 is 2,3-dihydroxy- propylthio-hydroxyalkyl, i.e. a capped hydroxy function having a solubilizing substituent. The poly-serine (and other hydroxy-functionalized a-amino acid side-chains) used to prepare polymer (C) of formula (III) has preferably a molecular weight between 1 Ό00 and 300Ό00 Da, in particular 30Ό00 to 70Ό00 Da, and such polymers further connected via the SH-group of said linker Z to compounds of formula (I) and/or (II) and with a capping 2,3-dihydroxypropylthio-hydroxyalkyl residue are preferred. For example, polyserine is first functionalized by esterification. Reaction of the chloroacetylated polymer with said linker Z comprising the terminal thiol functionality by nucleophilic substitution gives access to the desired polymers.

In a particular embodiment, a polymer (D) comprises the partial formula (IV)

wherein

R 1 is a thioalkyl substituent connected to said linker Z, wherein the SH-group of said linker Z is connected to the -CH 2 -group of R 1 ,

R 2 is 2,3-dihydroxypropylthioalkyl, and the relation between the two bracketed entities with R 1 and R 2 , respectively, in the polymer indicates the relation of carbohydrate loading to capped thiol function.

For example, R 1 is of formula (lllg) and R 2 is of formula (lllh)

wherein o is between 1 and 6, preferably 1 or 2, in particular 1 .

When o is 1 , substituent R 1 represents a side chain of poly-cysteine, connected to said SH-group of said linker Z, which linker Z is comprised by the inventive compounds, and preferably by the inventive compounds of formula (I) or (II), and hereby connected to the - CH 2 -group of R 1 , and R 2 is 2,3-dihydroxypropylthio-alkyl, i.e. a capped thiol function having a solubilizing substituent.

The poly-cysteine used to prepare polymer (D) of formula (III) has preferably a molecular weight between 1 Ό00 and 300Ό00 Da, in particular 30Ό00 to 70Ό00 Da, and such polymers further connected via the SH-group of said linker Z to compounds of formula (I) and/or (II) with a capping 2,3-dihydroxypropylthio-thioalkyl residue are preferred. For example, the polycysteine polymer is reacted with a compound containing a terminal alkene group via a radical reaction.

In a particular embodiment, a polymer (F) comprises the partial formula (IV)

wherein

R 1 is an aminoalkyl substituent connected to said linker Z, wherein the SH-group of said linker Z is connected to the -CH 2 -group of R 1 (IVa).

R 2 is 2,3-dihydroxypropylthio-acetylaminoalkylamino or a related amino substituent, and R is hydrogen or methyl; and the relation between the two bracketed entities with R 1 and R 2 , respectively, in the polymer indicates the relation of carbohydrate loading to capped amide function.

For example, R 1 is of formula (IVa)

and R 2 is of formula (IVb), R 3 is of formula (IVc)

In another embodiment R 1 is of formula (IVd)

and R 2 is of formula (IVe)

wherein m is between 1 and 10, preferably between 1 and 4. In another embodiment R 1 is of formula (IVf)

. (CH 2 ) r — Z

' N '

H (IVf) wherein r is between 1 and 6, preferably between 1 and 4, in particular 2, and R 2 is of formula (IVc) (above).

The poly-acrylic acid used to prepare polymer (F) of formula (IV) has preferably a molecular weight between 1 Ό00 and 400Ό00 Da, in particular 30Ό00 to 160Ό00 Da, and such polymers further connected via the SH-group of said linker Z to compounds of formula (I) and/or (II) and with a capping 2,3-dihydroxypropylthio-acetylaminoalkylamino residue are preferred.

In a particular embodiment, a polymer (G) comprises the partial formula (V)

wherein

R 1 is an aminoalkyi substituent connected to said linker Z, wherein the SH-group of said linker Z is connected to the -CH 2 -group of R 1 (Va).

R 2 is 2,3-dihydroxypropylthio-acetylaminoalkylaminocarbonyl or a related aminocarbonyl substituent, and the relation between the two bracketed entities with R 1 and R 2 , respectively, in the polymer indicates the relation of carbohydrate loading to capped hydroxy function.

For example, R 1 is of formula (Va)

and R 2 is of formula (Vb)

In another embodiment R 1 is of formula (Vc) and R 2 is of formula (Vd)

wherein m is between 1 and 10, preferably between 1 and 4.

In another embodiment R 1 is of formula (Ve) and R 2 is of formula (Vf)

wherein r is between 1 and 6, preferably between 1 and 4, in particular 2.

The copolymer used to prepare polymer (G) of formula (VI) has preferably a molecular weight between 1 Ό00 and 400Ό00 Da, in particular 30Ό00 to 160Ό00 Da, and such polymers further connected via the SH-group of said linker Z to compounds of formula (I) and/or (II) and with a capping 2,3-dihydroxypropylthio-carbonylaminoalkylaminocarbonyl residue are preferred.

In a particular embodiment, a polymer (H) comprises the partial formula (VI)

wherein

R 1 is an aminoalkyl substituent connected to said linker Z, wherein the SH-group of said linker Z is connected to the -CH 2 -group of R 1 .

R 2 is 2,3-dihydroxypropylthio-acetylamine, and the relation between the two bracketed entities with R 1 and R 2 , respectively, in the polymer indicates the relation of carbohydrate loading to capped amino function. For example, R 1 is of formula (Via) and R 2 is of formula (Vlb) wherein o is between 1 and 6, preferably 3 or 4 and m is between 1 and 6, preferably between 1 and 2, in particular 1. The chitosan used to prepare polymer (H) of formula (VI) has preferably a molecular weight between 1 Ό00 and 300Ό00 Da, in particular 30Ό00 to 70Ό00 Da, and such polymers connected via the SH-group of said linker Z to compounds of formula (I) and/or (II) and connected to a capping 2,3-dihydroxypropylthio-acetylamine residue are preferred. For example, the chitosan polymer is first functionalized by chloroacetylation of the amino groups. Reaction of the chloroacetylated polymer with said linker Z comprising the terminal thiol functionality by nucleophilic substitution gives access to the desired polymers.

In a particular embodiment, a polymer (I) comprises the partial formula (VII)

(VII), wherein

R 1 is a carbonylalkyl or carbonylaryl substituent connected to said linker Z, wherein the SH-group of said linker Z is connected to the -CH 2 -group of R 1 ,

R 2 is 2,3-dihydroxypropylthio-carbonylalkyl or carbonylaryl, and the relation between the two bracketed entities with R 1 and R 2 , respectively, in the polymer indicates the relation of carbohydrate loading to capped carboxy function.

For example, R 1 is of formula (Vila)

and R 2 is of formula (Vllb)

wherein m is between 1 and 6, preferably between 1 and 2, in particular 1.

The polyphosphazen used to prepare polymer (I) of formula (VII) has preferably a molecular weight between 1 Ό00 and 300Ό00 Da, in particular 30Ό00 to 70Ό00 Da, and such polymers further connected via the SH-group of said linker Z to compounds of formula (I) and/or (II) and with a capping 2,3-dihydroxypropylthio-carbonylalkyl or carbonylaryl residue are preferred. For example, the polyphosphazene is first functionalized by esterification. Reaction of the chloroacetylated polymer with said linker Z comprising the terminal thiol functionality by nucleophilic substitution gives access to the desired polymers.

From the group of polymers (A) - (I), preferred polymers are a-amino acid polymers (D- and L-form) or combinations (co-polymers) of different a-amino acids (A) - (D). More preferred are a-amino acid polymers consisting of poly-lysine, poly-ornithine, poly-aspartic acid, poly-glutamic acid. Particularly preferred among these a-amino acid polymers is poly-L-lysine.

In a further very preferred embodiment, said polymer is a polymer of formula 6, 10, 14, 18, 22, 26, 30, 34, 38, 42, 45, 78, 86, 89, 93, 100 or 102, wherein said formulas are shown in the experimental section, and wherein for each of said polymer n is independently 20- 1200, preferably 100-1 100, further preferably 200-500, and wherein for each of said polymer x is independently 10-90, preferably 30-60, and further preferably 40-50.

In a further very preferred embodiment, said polymer is a polymer of formula 6, 10, 14, 18, 22, 26, 30, 34, 38, 42, 45, 78, 86, 89, 93, 100 or 102, wherein said formulas are shown in the experimental section, and wherein for each of said polymer n is independently 100- 1 100, preferably 200-500, and wherein for each of said polymer x is independently 30-60, and further preferably 40-50.

In a further very preferred embodiment, said polymer is a polymer of formula 6, 22, 26, 34, 38, 42, 45, wherein said formulas are shown in the experimental section, and wherein for each of said polymer n is independently 20-1200, preferably 100-1 100, further preferably 200-500, and wherein for each of said polymer x is independently 10-90, preferably 30-60, and further preferably 40-50. In a further very preferred embodiment, said polymer is a polymer of formula 6, 22, 26, 34, 38, 42, 45, wherein said formulas are shown in the experimental section, and wherein for each of said polymer n is independently 100-1 100, preferably 200-500, and wherein for each of said polymer x is independently 30-60, and further preferably 40-50. The general terms used hereinbefore and hereinafter preferably have within the context of this disclosure the following meanings, unless otherwise indicated:

Where the plural form is used for compounds and the like, this is taken to mean also a single compound, or the like.

The term "glycoepitope", as used herein, refers to the carbohydrate moiety that is recognized by an antibody or by a lectin-like glycan-binding protein. Preferably, the term "glycoepitope", as used herein, refers to a carbohydrate moiety comprised by a glycosphingolipid expressed in the nervous system. Glycosphingolipids are known to the skilled person in the art and are a subset of glycolipids defined by their content of sphingosine and are particularly relevant to the nervous system. Subtypes of glycosphingolipids are cerebrosides (single carbohydrate attached to the lipid part), (neo)lacto-, ganglio-, or sulfoglucuronyl paragloboside-type (sialylated or non-sialylated oligosaccharide attached to the lipid part). Preferably, the term "glycoepitope", as used herein, refers to the carbohydrate moiety that is recognized by an antibody or by a lectin- like glycan-binding protein, wherein said glycoepitope is comprised by a glycosphingolipid that is expressed in the nervous system and wherein said a glycosphingolipid is selected from cerebrosides, (neo)lactosides, gangliosides, sulfoglucuronyl paraglobosides or carbohydrate moieties comprised by compounds of formula I or formula II.

Thus, in a preferred embodiment, said glycoepitope comprised by said glycosphingolipid of the nervous system is selected from the cerebroside-, (neo)lacto-, ganglio-, or sulfoglucuronyl paragloboside-type or a carbohydrate moiety comprised by a compound of formula (I) or formula (II).

In a further preferred embodiment, said glycoepitope comprised by said glycosphingolipid of the nervous system is selected from the cerebroside-, (neo)lacto-, or ganglio-type. In another preferred embodiment, said glycoepitope comprised by said glycosphingolipid of the nervous system is selected from a carbohydrate moiety comprised by a compound of formula (I). In another preferred embodiment, said glycoepitope comprised by said glycosphingolipid of the nervous system is selected from a carbohydrate moiety comprised by a compound of formula (II). The term "reducing end", as used herein in the context of the glycoepitope of the present invention and of the specific inventive compounds, refers to the terminal monosaccharide of the glycoepitope with a free anomeric carbon that is not involved in a glycosidic bond, wherein said free anomeric carbon bears a hemiacetal group. The term "Ci-C 4 -alkyl", as used herein refers to straight or branched chain of 1 to 4 carbon atoms and includes butyl, such as n-butyl, sec-butyl, / ' so-butyl, ie f-butyl, propyl, such as n-propyl or / ' so-propyl, ethyl or methyl. Preferably the term "CrC 4 -alkyl", refers to methyl or ethyl, n-propyl or / ' so-propyl. Further preferably, the term "CrC 4 -alkyl", refers to methyl. Correspondingly, the term "Ci-C 8 -alkyl", as used herein refers to straight or branched chain of 1 to 8 carbon atoms. The term "Ci-C 4 -alkyl-(OCH 2 CH2)pO-Ci-C 4 -alkyr', as used herein, and when referring to the linker Z defined as -N(R a )-A-B-CH2-(CH 2 )q-SH, and when referring to A within said linker Z, should refer, as evident from the description and examples herein, to a bivalent "Ci-C 4 -alkyl-(OCH 2 CH2) p O-Ci-C 4 -alkyr' group including groups such as -(CH2)n-(OCH2CH2)pO-(CH 2 )n- with n requal 1 to 4. The term "CrC 7 -alkylene", as used herein, refers to a straight or branched bivalent alkyl chain, preferably to a straight or branched bivalent alkyl chain of 1 to 7 carbon atoms, and includes, for example, -CH 2 -, -CH 2 -CH 2 -, -CH(CH 3 )-, -CH 2 -CH 2 -CH 2 -, -CH(CH 3 )-CH 2 -, or - CH(CH 2 CH 3 )-.

The term "CrC 7 -alkoxy", as used herein, refers to an alkoxy with a straight or branched chain of 1 to 7 carbon atoms. The term "CrC 4 -alkoxy", as used herein, refers to an alkoxy with a straight or branched chain of 1 to 4 carbon atoms and includes methoxy, ethoxy, propoxy, / ' so-propoxy, n-butoxy, sec-butoxy and ie f-butoxy. Preferably, the term "Ci-C 4 - alkoxy", as used herein, refers to methoxy, ethoxy, propoxy. Further preferably, the term "CrC 4 -alkoxy", as used herein, refers to methoxy. The term "CrC 7 -alkoxy", as used herein, and when referring to the linker Z defined as -N(R a )-A-B-CH2-(CH 2 )q-SH, and when referring to A within said linker Z, should refer, as evident from the description and examples herein, to a bivalent d-C 7 -alkoxy group including groups such as -(CH 2 ) n O- or - 0(CH 2 ) n - with n requal 1 to 7, typically and very preferably to groups such as -0(CH 2 ) n - forming with the N(R a ) of the linker Z a preferred bonding N(R a )-0(CH 2 ) n -. The term "Ci-C 8 -alkenyl", as used herein, refers to is a straight or branched chain containing one or more, e.g. two or three, double bonds, and is preferably CrC 4 -alkenyl, such as 1 - or 2-butenyl, 1 -propenyl, allyl or vinyl. Double bonds in principle can have E- or Z-configuration. The compounds of this invention may therefore exist as isomeric mixtures or single isomers. If not specified both isomeric forms are intended.

The term "C-i-Cs-alkynyl", as used herein, refers to is a straight or branched chain comprising one or more, preferably one triple bond. Preferred are CrC 4 - alkynyl, such as propargyl or acetylenyl.

Any asymmetric carbon atoms may be present in the (R)-, (S)- or (R,S)-configuration, preferably in the (R)- or (S ^configuration. The compounds may thus be present as mixtures of isomers or as pure isomers, preferably as enantiomer-pure diastereomers. The term "aryl", as used herein, refers to a mono- or bicyclic fused ring aromatic group with 5 to 10 carbon atoms optionally carrying substituents, such as phenyl, 1 -naphthyl or 2-naphthyl, or also a partially saturated bicyclic fused ring comprising a phenyl group, such as indanyl, indolinyl, dihydro- or tetrahydronaphthyl, all optionally substituted. Preferably, aryl is phenyl, indanyl, indolinyl or tetrahydronaphthyl, in particular phenyl. The term "heteroaryl", as used herein, refers to an aromatic mono- or bicyclic ring system containing at least one heteroatom, and preferably up to three heteroatoms selected from nitrogen, oxygen and sulfur as ring members. Heteroaryl rings do not contain adjacent oxygen atoms, adjacent sulfur atoms, or adjacent oxygen and sulfur atoms within the ring. Monocyclic heteroaryl preferably refers to 5 or 6 membered heteroaryl groups and bicyclic heteroaryl preferably refers to 9 or 10 membered fused-ring heteroaryl groups. Examples of heteroaryl include pyrrolyl, thienyl, furyl, pyrazolyl, imidazolyl, triazolyl, tetrazolyl, oxazolyl, isoxazolyl, oxadiazolyl, thiazolyl, isothiazolyl, thiadiazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, and benzo or pyridazo fused derivatives of such monocyclic heteroaryl groups, such as indolyl, benzimidazolyl, benzofuryl, quinolinyl, isoquinolinyl, quinazolinyl, pyrrolopyridine, imidazopyridine, or purinyl, all optionally substituted.

Preferably, the term "heteroaryl" refers to a 5- or 6-membered aromatic monocyclic ring system containing at least one heteroatom, and preferably up to three heteroatoms selected from nitrogen, oxygen and sulfur as ring members. Preferably, heteroaryl is pyridyl, pyrimdinyl, pyrazinyl, pyridazinyl, thienyl, pyrazolyl, imidazolyl, thiazolyl, oxadiazolyl, triazolyl, oxazolyl, isoxazolyl, isothiazolyl, pyrrolyl, indolyl, pyrrolopyridine or imidazopyridine; in particular pyridyl, pyrimidinyl, pyrazinyl, pyridazinyl, pyrazolyl, imidazolyl, thiazolyl, oxadiazolyl, triazolyl, indolyl, pyrrolopyridine or imidazopyridine The term ..optionally substituted aryl", as used herein, refers to aryl substituted by up to four substituents, preferably up to two substituents. In optionally substituted aryl, preferably in optionally substituted phenyl, substituents are preferably and independently selected from CrC 4 -alkyl, Ci-C 4 -alkoxy, amino-Ci-C 4 -alkyl, acylamino-Ci-C 4 -alkyl, aryl-Cr C 4 -alkyl hydroxy, carboxy, CrC 4 -alkoxycarbonyl, aminocarbonyl, hydroxylaminocarbonyl, tetrazolyl, hydroxysulfonyl, aminosulfonyl, halo, or nitro, in particular Ci-C 4 -alkyl, Ci-C 4 - alkoxy, amino-Ci-C 4 -alkyl, acylamino-Ci-C 4 -alkyl, carboxy, CrC 4 -alkoxycarbonyl, aminocarbonyl, hydroxylaminocarbonyl, tetrazolyl, or aminosulfonyl.

The term ..optionally substituted heteroaryl", as used herein, refers to heteroaryl substituted by up to three substituents, preferably up to two substituents. In optionally substituted heteroaryl, substituents are preferably and independently selected from Ci-C 4 - alkyl, Ci-C 4 -alkoxy, halo- CrC 4 -alkyl, hydroxy, CrC 4 -alkoxycarbonyl, aminocarbonyl, hydroxylaminocarbonyl, tetrazolyl, aminosulfonyl, halo, aryl-Ci-C 4 -alkyl, or nitro.

Cycloalkyl has preferably 3 to 7 ring carbon atoms, and may be unsubstituted or substituted, e.g. by Ci-C 4 -alkyl or Ci-C 4 -alkoxy. Cycloalkyl is, for example and preferably, cyclohexyl, cyclopentyl, methylcyclopentyl, or cyclopropyl, in particular cyclopropyl.

Acyl designates, for example, alkylcarbonyl, cycloalkylcarbonyl, arylcarbonyl, aryl- Ci-C 4 - alkylcarbonyl, or heteroarylcarbonyl. Ci-C 4 -acyl is preferably lower alkylcarbonyl, in particular propionyl or acetyl. Ac stands for acetyl. Hydroxyalkyl is especially hydroxy- CrC 4 -alkyl, preferably hydroxy methyl, 2-hydroxyethyl or 2-hydroxy-2-propyl.

Haloalkyl is preferably fluoroalkyl, especially trifluoromethyl, 3,3,3-trifluoroethyl or pentafluoroethyl.

Halogen is fluorine, chlorine, bromine, or iodine. ArylalkyI includes aryl and alkyl as defined hereinbefore, and is e.g. benzyl, 1 -phenethyl or 2-phenethyl.

Heteroarylalkyl includes heteroaryl and alkyl as defined hereinbefore, and is e.g. 2-, 3- or 4-pyridylmethyl, 1 - or 2-pyrrolylmethyl, 1 -pyrazolylmethyl, 1 -imidazolylmethyl, 2-(1 - imidazolyl)ethyl or 3-(1 -imidazolyl)propyl. In substituted amino, the substituents are preferably those mentioned as substituents hereinbefore. In particular, substituted amino is alkylamino, dialkylamino, optionally substituted arylamino, optionally substituted arylalkylamino, lower alkylcarbonylamino, benzoylamino, pyridylcarbonylamino, lower alkoxycarbonylamino or optionally substituted aminocarbonylamino.

Particular salts considered are those replacing the hydrogen atoms of the sulfate group and the carboxylic acid function. Suitable cations are, e.g., sodium, potassium, calcium, magnesium or ammonium cations, or also cations derived by protonation from primary, secondary or tertiary amines containing, for example, CrC 4 -alkyl, hydroxy- CrC 4 -alkyl or hydroxy- CrC 4 -alkoxy- CrC 4 -alkyl groups, e.g., 2-hydroxyethylammonium, 2-(2-hydroxy- ethoxy)ethyldimethylammonium, diethylammonium, di(2-hydroxyethyl)ammonium, trimethylammonium, triethylammonium, 2-hydroxyethyldimethylammonium, or di(2- hydroxyethyl)methylammonium, also from correspondingly substituted cyclic secondary and tertiary amines, e.g., N-methylpyrrolidinium, N-methylpiperidinium, N-methyl- morpholinium, N-2-hydroxyethylpyrrolidinium, N-2-hydroxyethylpiperidinium, or N-2- hydroxyethylmorpholinium, and the like.

In view of the close relationship between the novel compounds in free form and those in the form of their salts, including those salts that can be used as intermediates, for example in the purification or identification of the novel compounds, any reference to the free compounds hereinbefore and hereinafter is to be understood as referring also to the corresponding salts, and vice versa, as appropriate and expedient

A preferred polymer backbone in the inventive polymers comprising a multitude of compounds of formula (I) or formula (II) is polylysine, in particular poly-L-lysine.

Preferably the molecular weight of the polylysine is 1 Ό00 to 300Ό00 kD, preferably 10Ό00 to 200Ό00 kD. Particularly preferred is a molecular weight of approximately 50Ό00 kD, 85Ό00 kD, 125Ό00 kD or 200Ό00 kD. Most preferred is a molecular weight of approximately 50Ό00 kD. In particular the invention relates to such polymers wherein the relative loading of polymer backbone with the carbohydrate moiety of said compound of formula (I) and/or (II) is 10 - 90 %, meaning that 10 - 90 % of all lysine side chains in the polymer are connected to said SH-group of said linker Z, which linker Z is comprised by the inventive compounds, and preferably by the inventive compounds of formula (I) or (II), the remaining amino functions being capped. Preferably the loading of the polymer is 20 - 70 %, more preferably 30 - 60 %. Further preferred polymers in said context are polymers with loading of one or several of compounds of formula (I) or (II), wherein said compounds of formula (I) or (II) are selected from 4 * , 9 * , 13 * , 17 * , 21 * , 25 * , 29 * or 33 * , and 46 * -60 * . The polymers of the present invention which comprises the inventive compounds comprising a carbohydrate moieties and linkers Z, wherein said carbohydrate moieties mimic glycoepitopes comprised by glycosphingolipids of the nervous system allow straightforward coupling of said carbohydrate moieties such as ganglioside glycoepitopes to biodegradable poly-L-lysine and other functionalized biodegradable polymers without loosing the integrity of the carbohydrate moieties at their reducing end. This is in particular important since the monosaccharide with the reducing end comprised the carbohydrate moieties can also contribute to binding affinity to antibodies or other targets, and thus chemical linkage methods that leave this carbohydrate ring intact are preferable. Thus, the resulting inventive chemically defined glycoconjugates/glycopolymers based on biodegradable polymer backbones can be used in a clinical context, either therapeutic and diagnostic, to detect or neutralize pathogenic anti-glycan antibodies. Moreover, the multivalent presentation of the carbohydrate moieties mimicking glycoepitopes comprised by glycosphingolipids of the nervous system, on, preferably, poly-L-lysine, can substantially increase their binding affinity towards binding partners.

In a particularly preferred embodiment, the invention relates to polymers comprising a multitude of compounds of formula (I), and/or (II) wherein the polymer is poly-L-lysine and wherein said polymer further comprises said linker Z connecting said compounds to the polymer backbone. Poly-L-lysine is biodegradable and therefore in particular suitable for therapeutical application.

The compounds of the invention have valuable pharmacological properties. The invention also relates to compounds as defined hereinbefore for use as medicaments. A compound according to the invention shows prophylactic and therapeutic efficacy especially against neurological diseases associated with anti-glycan antibodies, particularly immune- mediated neuropathies.

One or multiple compounds of formula (I), and/or (II) or polymers comprising these, can be administered alone or in combination with one or more other therapeutic agents, possible combination therapy taking the form of fixed combinations, or the administration of a compound of the invention and one or more other therapeutic agents being staggered or given independently of one another, or the combined administration of fixed combinations and one or more other therapeutic agents.

Therapeutic agents for possible combination are especially immunosuppressive agents/ therapies. Examples are purine analogues such as fludarabine and/or cladribine, plasmapheresis, intravenous immunoglobulins, furthermore the chimeric monoclonal antibody rituximab (M.C. Dalakas, Curr Treat Opinions Neurol, 2010, 12, 71 -83).

In another particular embodiment, the invention relates to the use of the compounds of the invention in a diagnostic assay for neurological diseases, particularly immune-mediated neuropathies. In particular, the invention relates to kits comprising the compounds of formula (I), and/or (II) as defined above, and also polymers of the invention comprising such compounds as substituents.

The present invention relates to a method of diagnosis of neurological diseases, particularly immune-mediated neuropathies, wherein the level of antibodies (e.g. IgM/lgG) against glycans of the nervous system, particularly glycolipids, is determined in a body fluid sample, e.g. serum, and a high level is indicative of the development and the severity of a particular neurological condition.

Other body fluids than serum are useful for determination of antibodies against glycosphingolipid glycoepitopes and are, e.g., whole blood, cerebrospinal fluid or extracts from solid tissue.

Any known method may be used for the determination of the level of antibodies against glycosphingolipid glycoepitopes in body fluids. Methods considered are, e.g., ELISA, RIA, EIA, or microarray analysis.

A preferred method for the determination of antibodies against glycosphingolipid glycoepitopes in human body fluids, e.g. in serum, is an ELISA. In such an embodiment, microtiter plates are coated with compounds of formula (I), and/or (II) or preferably polymers of the invention comprising such compounds as substituents. The plates are then blocked and the sample or a standard solution is loaded. After incubation, an anti- IgM/lgG antibody is applied, e.g. an anti-lgM or anti-lgG antibody directly conjugated with a suitable label, e.g. with an enzyme for chromogenic detection. Alternatively, a polyclonal rabbit (or mouse) anti-lgM / anti-lgG antibody is added. A second antibody detecting the particular type of the anti-lgM / anti-lgG antibody, e.g. an anti-rabbit (or anti-mouse) antibody, conjugated with a suitable label, e.g. the enzyme for chromogenic detection as above, is then added. Finally the plate is developed with a substrate for the label in order to detect and quantify the label, being a measure for the presence and amount of antibodies against glycosphingolipid glycoepitopes of the nervous system. If the label is an enzyme for chromogenic detection, the substrate is a colour-generating substrate of the conjugated enzyme. The colour reaction is then detected in a microplate reader and compared to standards.

It is also possible to use antibody fragments. Suitable labels are chromogenic labels, i.e. enzymes which can be used to convert a substrate to a detectable colored or fluorescent compound, spectroscopic labels, e.g. fluorescent labels or labels presenting a visible color, affinity labels which may be developed by a further compound specific for the label and allowing easy detection and quantification, or any other label used in standard ELISA.

Other preferred methods of detection of antibodies against glycosphingolipid glycoepitopes are radioimmunoassay or competitive immunoassay and chemiluminescence detection on automated commercial analytical robots. Microparticle enhanced fluorescence, fluorescence polarized methodologies, or mass spectrometry may also be used. Detection devices, e.g. microarrays, are useful components as readout systems for antibodies against glycosphingolipid glycoepitopes.

In a further embodiment the invention relates to a kit suitable for an assay as described above, in particular an ELISA, comprising compounds of formula (I), and/or (II) or polymers comprising such compounds as substituents. The kits further contain anti-lgM / anti-lgG antibodies (or anti-lgM/lgG antibody fragments) carrying a suitable label, or anti- lgM / anti-lgG antibodies and second antibodies carrying such a suitable label, and reagents or equipment to detect the label, e.g. reagents reacting with enzymes used as labels and indicating the presence of such a label by a colour formation or fluorescence, standard equipment, such as microtiter plates, pipettes and the like, standard solutions and wash solutions.

The ELISA can be also designed in a way that patient blood or serum samples are used for the coating of microtiter plates with the subsequent detection of anti-glycan antibodies with labelled compounds of formula (I), and/or (II) or labelled polymers comprising such compounds as substituents. The label is either directly detectable or indirectly detectable via an antibody.

The polymer carrying compounds of formula (I), and/or (II) of the invention binds to the pathogenic anti-glycan antibodies and potentially downregulates the anti-glycan IgM or IgG antibody production. It allows an antigen-specific treatment for neurological diseases involving anti-glycan antibodies against glycosphingolipid glycoepitopes. Furthermore the invention relates to a pharmaceutical composition comprising a compound of formula (I), and/or (II) or a polymer carrying compounds of formula (I), and/or (II) of the invention.

Pharmaceutical compositions for parenteral administration, such as subcutaneous, intravenous, intrahepatic or intramuscular administration, to warm-blooded animals, especially humans, are considered. The compositions comprise the active ingredient(s) alone or, preferably, together with a pharmaceutically acceptable carrier. The dosage of the active ingredient(s) depends upon the age, weight, and individual condition of the patient, the individual pharmacokinetic data, and the mode of administration. For parenteral administration preference is given to the use of suspensions or dispersions of the carbohydrate polymer of the invention, especially isotonic aqueous dispersions or suspensions which, for example, can be made up shortly before use. The pharmaceutical compositions may be sterilized and/or may comprise excipients, for example preservatives, stabilizers, wetting agents and/or emulsifiers, solubilizers, viscosity- increasing agents, salts for regulating osmotic pressure and/or buffers and are prepared in a manner known per se, for example by means of conventional dissolving and lyophilizing processes.

Suitable carriers for enteral administration, such as nasal, buccal, rectal or oral administration, are especially fillers, such as sugars, for example lactose, saccharose, mannitol or sorbitol, cellulose preparations, and/or calcium phosphates, for example tricalcium phosphate or calcium hydrogen phosphate, and also binders, such as starches, for example corn, wheat, rice or potato starch, methylcellulose, hydroxypropyl methylcellulose, sodium carboxymethylcellulose, and/or polyvinylpyrrolidone, and/or, if desired, disintegrators, such as the above-mentioned starches, also carboxymethyl starch, crosslinked polyvinylpyrrolidone, alginic acid or a salt thereof, such as sodium alginate. Additional excipients are especially flow conditioners and lubricants, for example silicic acid, talc, stearic acid or salts thereof, such as magnesium or calcium stearate, and/or polyethylene glycol, or derivatives thereof.

Tablet cores can be provided with suitable, optionally enteric, coatings through the use of, inter alia, concentrated sugar solutions which may comprise gum arabic, talc, polyvinylpyrrolidone, polyethylene glycol and/or titanium dioxide, or coating solutions in suitable organic solvents or solvent mixtures, or, for the preparation of enteric coatings, solutions of suitable cellulose preparations, such as acetylcellulose phthalate or hydroxypropyl- methylcellulose phthalate. Dyes or pigments may be added to the tablets or tablet coatings, for example for identification purposes or to indicate different doses of active ingredient(s).

Pharmaceutical compositions for oral administration also include hard capsules consisting of gelatin, and also soft, sealed capsules consisting of gelatin and a plasticizer, such as glycerol or sorbitol. The hard capsules may contain the active ingredient in the form of granules, for example in admixture with fillers, such as corn starch, binders, and/or glidants, such as talc or magnesium stearate, and optionally stabilizers. In soft capsules, the active ingredient is preferably dissolved or suspended in suitable liquid excipients, such as fatty oils, paraffin oil or liquid polyethylene glycols or fatty acid esters of ethylene or propylene glycol, to which stabilizers and detergents, for example of the polyoxy- ethylene sorbitan fatty acid ester type, may also be added.

Pharmaceutical compositions suitable for rectal administration are, for example, suppositories that consist of a combination of the active ingredient and a suppository base. Suitable suppository bases are, for example, natural or synthetic triglycerides, paraffin hydrocarbons, polyethylene glycols or higher alkanols.

The mentioned pharmaceutical compositions according to the invention may contain separate tablets, granules or other forms of orally acceptable formulation of the active ingredients, or may contain a mixture of active ingredients in one suitable pharmaceutical dosage form, as described above. In particular the separate orally acceptable formulations or the mixture in one suitable pharmaceutical dosage form may be slow release and controlled release pharmaceutical compositions.

The pharmaceutical compositions comprise from approximately 1 % to approximately 95% active ingredient or mixture of active ingredients, single-dose administration forms comprising in the preferred embodiment from approximately 20% to approximately 90% active ingredient(s) and forms that are not of single-dose type comprising in the preferred embodiment from approximately 5% to approximately 20% active ingredient(s).

The invention also relates to the mentioned pharmaceutical compositions as medicaments in the treatment of neurological diseases associated with anti-glycan antibodies, particularyl immune-mediated neuropathies. The present invention relates furthermore to a method of treatment of neurological diseases associated with anti-glycan antibodies, particularyl immune-mediated neuropathies, which comprises administering a composition according to the invention in a quantity effective against said disease, to a warm-blooded animal requiring such treatment. The pharmaceutical compositions can be administered prophylactically or therapeutically, preferably in an amount effective against the said diseases, to a warmblooded animal, for example a human, requiring such treatment. In the case of an individual having a bodyweight of about 70 kg the daily, weekly or monthly dose administered is from approximately 0.01 g to approximately 5 g, preferably from approximately 0.1 g to approximately 1 .5 g, of the active ingredients in a composition of the present invention.

The following Examples serve to illustrate the invention without limiting the invention in its scope. Examples

General Methods

NMR spectra were obtained on a Bruker Avance DMX-500 (500 MHz) spectrometer. Assignment of 1 H and 13 C NMR spectra was achieved using 2D methods (COSY, HSQC and HMBC). Chemical shifts are expressed in ppm using residual CHCI 3 , CHD 2 OD, DMSO-d 6 or HDO as references. IR spectra were recorded using a Perkin-Elmer Spectrum One FT-IR spectrometer. Electron spray ionization mass spectra (ESI-MS) were obtained on a Waters micromass ZQ. HRMS analysis was carried using an Agilent 1 100LC equipped with a photodiode array detector and a micromass QTOF I equipped with a 4 GHz digital-time converter. Reactions were monitored by ESI-MS and TLC using glass plates coated with silica gel 60 F 25 4 (Merck) and visualized by using UV light and/or by charring with mostain (a 0.02 M solution of ammonium cerium sulfate dihydrate and ammonium molybdate tetrahydrate in 10% aq H 2 S0 4 ). Column chromatography was performed on silica gel (Redisep normal phase silica gel column 35/70) or RP-18 (Merck LiChroprep® RP-18 40/63). Dichloromethane (DCM) and MeOH were dried by filtration over Al 2 0 3 (Fluka, type 5016A basic). Dimethylformamide (DMF) was purchased from Acros (99.8%, extra dry, over molecular sieves). Molecular sieves (MS, 4 A) were activated in vacuo at 400°C for 30 min immediately before use. Size-exclusion chromatography was performed on polyacrylamide gel (Biogel P-2 Fine). Dialysis was performed on a Biotech Cellulose Ester (CE) Membrane (SpectrumLabs, molecular weight cutoff: 100-500 Da). Centrifugations were carried out with an Eppendorf Centrifuge 5804 R. rt = room temperature.

Seventeen glycopolymers were synthesized (6, Scheme 1 ; 10, Scheme 2; 14, Scheme 3; 18, Scheme 4; 22, Scheme 5; 26, Scheme 6; 30, Scheme 7; 34, Scheme 8; 38, Scheme 9; 42, Scheme 10; 45, Scheme 1 1 ; 78, Scheme 16; 86, Scheme 18; 89, Scheme 19; 93, Scheme 20; 100, Scheme 22; 102, Scheme 23) for biological evaluation. Polylysine glycoconjugates 6, 10, 14, 18, 22, 26, 30, 34 all bear the same linker but differ by their carbohydrate moiety (respectively GM1 a, GM1 b, asialo GM1 , GM2, GD1 a, GD1 b, GD3 and GT1 a). Polylysine glycoconjugates 6, 38, 42 and 45 bear the same carbohydrate (GM1 a) but differ by their linker moiety. Polylysine glycoconjugate 78 bears a GM4 mimetic. Polylysine glycoconjugate 86 bears the H0 3 S-p-D-GlcpA-(1→3)-p-D-Galp (HNK- 1 ) disaccharide. The above-mentioned glycoconjugates (6, 10, 14, 18, 22, 26, 30, 34, 38, 45, 78, 86) are all poly-L-lysine conjugates. Conjugates 89 and 93 bear the same HNK-1 disaccharide but differ by their polymer backbones (poly-L-lysine dendrimer and poly-L- ornithine respectively). Conjugates 100 and 102 bear the same lactose disaccharide but differ by their polymer backbones (chitosan and poly-L-glutamic acid respectively). The synthesis of the HNK-1 disaccharide 58 functionalized by Iinker5 72 is described in Scheme 17. The synthesis of the lactose disaccharide 56 functionalized by Iinker5 72 is described in Scheme 121. The synthesis of linkers 35, 39, 43 and 72 is described in Scheme 12, 13, 14 and 15 respectively.

All reagents were bought from Sigma Aldrich, Acros, Alfa-Aesar, Elicityl or Alamanda Polymers. Linker 2 and compound 66 were synthesized according to a published procedure (O. Bohorov, et al. Glycobiology, 2006, 16, 21 C-27C). Chloroacetylated poly-L- lysine 5 (250 lysine repeating units) was synthesized from commercial poly-L-lysine polymer according to a published procedure (G. Thoma et al., J Am Chem Soc 1999, 121 , 5919-5929). Derivatives 68, 73, 74, 80, 87 and 98 were synthesized according to published procedures (respectively I. Ueda, et al. Chem Pharm Bull (Tokyo), 1990, 38, 3035-3041 ; M. Numata, et al. Carbohydr Res, 1987, 163, 209-225; J. L. Magnani, Preparation of oligosaccharide glycomimetic antagonists as E- and P-selectin modulators, WO 2005054264A2, June 16, 2005; T. Furukawa, Tetrahedron Lett, 2011 , 52, 5567-5570; K. T. Al-Jamal, et al. J Drug Target, 2006, 14, 405-412; T. Kojima, Chitosan or chitin derivative and method for processing silver halide photographic material by using the same, US 005155004A, Oct 13, 1992). Scheme 1 : Synthesis of the GM1 a polymer 6

HN„

b)

4 [GM1a-N(Me)0(CH 2 ) 2 NHC(0)(CH 2 ) 3 SH]

6 Reagents and conditions: a) 2, sodium acetate buffer, 91 %; b) DL-dithiothreitol, Y-thiobutyrolactone, Et 3 N, DMF, 54%; c) i. 5, DBU, DMF/H 2 0; ii. thioglycerol, Et 3 N, 84%

/V-(/V-Methyl-0-[2-aminoethyllhvdroxylamino)-p-D-galactop yranosyl-(1 ^3)-2-acetamido-p- D-galactopyranosyl-(1→4H5-acetyl-a-neuraminic acid-(2→3VI-p-D-galactopyranosyl- (1→4)-p-D-glucopyranoside (3):

To a solution of hemiacetal 1 (5.0 mg, 4.90 μηιοΙ) in NaOAc/AcOH buffer (0.1 M, pH 4.5, 50 μΙ_) was added oxyamine 2 (4.4 mg, 49 μηηοΙ, 10 equiv). The reaction mixture was stirred for 24-48 h at 25-40°C. Purification by dialysis gave compound 3 (4.97 mg, 4.55 μηηοΙ, 91 %) as a white fluffy solid. 1 H-NMR (500 MHz, D 2 0): δ 4.80 (d, 1 H), 4.57 (d, 1 H), 4.57 (d,1 H), 4.24 (d, 1 H), 4.22-4.1 1 (m, 2H), 4.06 (dd, 1 H), 4.04-3.96 (m, 3H), 3.94 (d, 1 H), 3.89 (dd, 1 H), 3.86-3.74 (m, 12H), 3.73-3.57 (m, 10H), 3.54 (dd, 1 H), 3.52 (dd, 1 H), 3.39 (dd, 1 H), 3.28-3.26 (m, 2H), 2.81 (s, 3H), 2.68 (dd, 1 H), 2.05, 2.03 (2s, 6H), 1.94 (t, 1 H).

HRMS (ESI + ): m/z 1071.4132 (calc for C 4 oH 71 N 4 029 + [M+H] + : m/z 1071.4198). /V-(/\/-lv1ethyl-0-[2-(2-mercaptobutanamido)ethyllhvdroxylam ino)-p-D-galactopyranosyl- (1→3)-2-acetamido-p-D-galactopyranosyl-(1→4)-[5-acetyl-a -neuraminic acid-(2→3VI-p-D- galacto-pyranosyl-d→4)-p-D-glucopyranoside (4):

To a suspension of amine 3 (4.97 mg, 4.55 μηηοΙ) in anhyd DMF (90 μΙ_) were successively added DL-dithiothreitol (1.2 mg, 8.2 μηηοΙ, 1 .8 equiv), γ-thiobutyrolactone (3.9 μΐ, 46 μηιοΙ, 10 equiv) and Et 3 N (6.3 μΐ, 46 μηιοΙ, 10 equiv). The reaction mixture was stirred for 12-24 h at 25-40°C. After that time, the reaction mixture was concentrated and the solvents co-evaporated with xylene. Purification by P2 size-exclusion chromatography gave compound 4 (2.8 mg, 2.33 μηηοΙ, 54%) as a white fluffy solid.

1 H-NMR (500 MHz, D 2 0): δ 4.79 (d, 1 H), 4.56 (m, 2H), 4.22-3.31 (m, 32H), 2.75 (s, 3H), 2.68 (m, 1 H), 2.57 (t, 2H), 2.41 (t, 2H), 2.05, 2.03 (2s, 6H), 1.91 (m, 3H).

MS (ESI-): m/z 1 171.59 (calc for C 44 H 75 N 4 0 3 oS " [M-Na] " : m/z 1 171.42). GM1 a Polymer (6):

To a solution of 5 (1 .2 mg, 5.83 μηιοΙ) in DMF (60 μΐ) were subsequently added compound 4 (2.8 mg, 2.33 μηιοΙ, 0.4 equiv), water (3 μΐ) and a solution of DBU (1 .3 μΐ, 8.74 μηιοΙ, 1 .5 equiv) in DMF (10 μΙ_). After stirring for 1 -24 h at rt, thioglycerol (1.5 μΙ_, 17.5 μηιοΙ, 3.0 equiv) and Et 3 N (2.4 μΙ_, 17.5 μηιοΙ, 3.0 equiv) were added. The reaction mixture was stirred at rt for another 12-24 h. The product was precipitated by slow addition to a stirring solution of EtOH/Et 2 0 (1 :1 , 1 ml_). The precipitate was filtered off, washed with EtOH and dried. Further purification was achieved by ultrafiltration (Sartorius Stedim Vivaspin tubes, 6 ml_, molecular weight cutoff 10 kDa, 5500 rpm). Freeze-drying gave GM1 a polymer 6 (2.88 mg, 84%) as a white solid. According to 1 H NMR, the product contained approximately 28% of the lysine side-chains substituted by the carbohydrate epitope 4. In this particular embodiment, the GM1 a epitope 4 carrying the linker Z with the terminal sulfhydryl function was synthesized and reacted in a substochiometric amount with the activated (chloroacetylated) lysine polymer 5. The carbohydrate loading (28%) of the obtained glycopolymer 6 was determined by 1 H NMR. The starting polylysine hydrobromide had an average molecular weight (MW) of 52 kDa (250 repeating lysine units), whereas the final polymer 6 with 28% GM1 a epitope loading had a calculated average MW of 145 kDa.

Scheme 2: Synthesis of the GM1 b polymer 10

HN „ NH 2

b)

10 Reagents and conditions: a) 2, sodium acetate buffer, 71 %; b) DL-dithiothreitol, Y-thiobutyrolactone, Et 3 N, DMF, 65%; c) i. 5, DBU, DMF/H 2 0; ii. thioglycerol, Et 3 N, 61 %

(1→4)-p-D-glucopyranoside (8):

To a solution of hemiacetal 7 (10 mg, 9.80 μηιοΙ) in NaOAc/AcOH buffer (0.1 M, pH 4.5, 98 μΙ_) was added oxyamine 2 (8.8 mg, 98.0 μηηοΙ, 10 equiv). The reaction mixture was stirred for 24-48 h at 25-40°C. Purification by dialysis gave compound 8 (7.6 mg, 6.95 μηιοΙ, 71 %) as a white fluffy solid. 1 H-NMR (500 MHz, D 2 0): δ 4.72 (d, 1 H), 4.54 (d, 1 H), 4.46 (d, 1 H), 4.24 (d, 1 H), 4.17 (d, 1 H), 4.13 (d, 1 H), 4.09 (dd, 1 H), 4.06-3.97 (m, 4H), 3.95 (d, 1 H), 3.91 (dd, 1 H), 3.89-3.81 (m, 7H), 3.81 -3.60 (m, 12H), 3.78 (dd, 1 H), 3.62-3.57 (m, 1 H), 3.59 (dd, 1 H), 3.57 (dd, 1 H), 3.44 (dd, 1 H), 3.29-3.27 (m, 2H), 2.81 (s, 3H), 2.77 (dd, 1 H), 2.06, 2.05 (2s, 6H), 1.81 (dd, 1 H). MS (ESP): m/z 1069.62 (calc for C 4 oH 69 N 4 029 " [M-Na] " : m/z 1069.41 ).

/V-(/V-Methyl-0-[2-(2-mercaptobutanamido)ethyllhvdroxylam ino)-5-acetyl-a-neuraminic acid-(2→'3)-p-D-galactopyranosyl-(1→'3)-2-acetamido-p-D- galactopyranosyl-(1→'4)-p-D- galactopyranosyl-d→4)-p-D-glucopyranoside (9):

To a suspension of amine 8 (7.6 mg, 6.95 μηηοΙ) in anhyd DMF (140 μΙ_) were successively added DL-dithiothreitol (spatula tip), γ-thiobutyrolactone (6.0 μΙ_, 69.5 μηιοΙ, 10 equiv) and Et 3 N (9.7 μΙ_, 69.5 μηηοΙ, 10 equiv). The reaction mixture was stirred for 12-24 h at 25-40°C. After that time, the reaction mixture was concentrated and the solvents co-evaporated with xylene. Purification by P2 size-exclusion chromatography gave compound 9 (5.4 mg, 4.52 μηιοΙ, 65%) as a white fluffy solid. 1 H-NMR (500 MHz, D 2 0): δ 4.72 (d, 1 H), 4.54 (d, 1 H), 4.47 (d, 1 H), 4.19 (d, 1 H), 4.17 (d, 1 H), 4.13 (d, 1 H), 4.09 (dd, 1 H), 4.04 (dd, 1 H), 4.00 (dd, 1 H), 3.96 (d, 1 H), 3.93-3.80 (m, 10H), 3.80-3.58 (m, 13H), 3.58-3.52 (m, 1 H), 3.56 (dd, 1 H), 3.55 (dd, 1 H), 3.47-3.40 (m, 1 H), 3.44 (dd, 1 H), 2.81 -2.74 (m, 5H), 2.57 (t, 2H), 2.40 (t, 2H), 2.06, 2.05 (2s, 6H), 1.94- 1 .89 (m, 2H), 1 .81 (dd, 1 H). HRMS (ESI-): m/z 1 171.01 (calc for C 44 H 75 N 4 0 3 oS " [M-Na] " : m/z 1 171.42). GM1 b Polymer (10):

To a solution of 5 (1.86 mg, 9.04 μηιοΙ) in DMF (60 μΙ_) were subsequently added compound 9 (5.4 mg, 4.52 μηιοΙ, 0.5 equiv), water (40 μΙ_) and a solution of DBU (2.0 μΙ_, 13.6 μηιοΙ, 1 .5 equiv) in DMF (15 μΙ_). After stirring for 1 -24 h at rt, thioglycerol (2.3 μΙ_, 27.1 μηιοΙ, 3.0 equiv) and Et 3 N (3.8 μΙ_, 27.1 μηιοΙ, 3.0 equiv) were added. The reaction mixture was stirred at rt for another 12-24 h. The product was precipitated by slow addition to a stirring solution of EtOH/Et 2 0 (1 :1 , 1 ml_). The precipitate was filtered off, washed with EtOH and dried. Further purification was achieved by ultrafiltration (Sartorius Stedim Vivaspin tubes, 6 ml_, molecular weight cutoff 10 kDa, 5500 rpm). Freeze-drying gave GM1 b polymer 10 (4.2 mg, 61 %) as a white solid. According to 1 H NMR, the product contained approximately 45% of the lysine side-chains substituted by the carbohydrate epitope 9.

Scheme 3: Synthesis of the asialo GM1 polymer 14

HN „ , NH 2

O

12

b)

13 [aGM1 -N(Me)0(CH 2 )2NHC(0)(CH2) 3 SH]

14 Reagents and conditions: a) 2, sodium acetate buffer, quant; b) DL-dithiothreitol, Y-thiobutyrolactone, Et 3 N, DMF, 80%; c) i. 5, DBU, DMF/H 2 0; ii. thioglycerol, Et 3 N, 71 %

/V-(/V-Methyl-0-[2-aminoethyllhvdroxylamino)-p-D-galactop yranosyl-(1 ^3)-2-acetamido-p- p-galactopyranosyl-d→4)-p-D-galactopyranosyl-(1→4)-p-D-g lucopyranoside (12): To a solution of hemiacetal 11 (10.0 mg, 14.1 μηιοΙ) in NaOAc/AcOH buffer (0.1 M, pH 4.5, 141 μΙ_) was added oxyamine 2 (12.7 mg, 141 μηηοΙ, 10 equiv). The reaction mixture was stirred for 24-48 h at 25-40°C. Purification by P2 size-exclusion chromatography gave compound 12 (10.9 mg, 14.0 μηηοΙ, quant) as a white fluffy solid.

1 H-NMR (500 MHz, D 2 0): δ 4.71 (d, 1 H), 4.47 (d, 1 H), 4.46 (d, 1 H), 4.24 (d, 1 H), 4.18 (d, 1 H), 4.13 (d, 1 H), 4.06-4.00 (m, 3H), 4.04 (dd, 1 H), 3.93 (d, 1 H), 3.90 (dd, 1 H), 3.87-3.75 (m, 8H), 3.76-3.65 (m, 4H), 3.64 (dd, 1 H), 3.61 -3.58 (m, 2H), 3.60 (dd, 1 H), 3.55 (dd, 1 H), 3.43 (dd, 1 H), 3.28 (t, 2H), 2.81 (s, 3H), 2.06 (s, 3H).

MS (ESI + ): m/z 780.46 (calc for [M+H] + : m/z 780.32).

/V-(/\/-lv1ethyl-0-[2-(2-mercaptobutanamido)ethyllhvdroxy lamino)-p-D-galactopyranosyl- (1→3)-2-acetamido-p-D-galactopyranosyl-(1→4)-p-D-galacto pyranosyl-(1→4)-β-ρ- glucopyranoside (13):

To a suspension of amine 12 (1 1 mg, 14.1 μηηοΙ) in anhyd DMF (282 μΙ_) were successively added DL-dithiothreitol (tip of spatula), γ-thiobutyrolactone (12.2 μΙ_, 141 μηηοΙ, 10 equiv) and Et 3 N (19.7 μΙ_, 141 μηηοΙ, 10 equiv). The reaction mixture was stirred for 12-24 h at 25-40°C. After that time, the reaction mixture was concentrated and the solvents co-evaporated with xylene. Purification by P2 size-exclusion chromatography gave compound 13 (10.0 mg, 1 1.3 μηιοΙ, 80%) as a white fluffy solid.

1 H-NMR (500 MHz, D 2 0): δ 4.72 (d, 1 H), 4.47 (d, 2H), 4.19 (d, 1 H), 4.19-4.16 (m, 1 H), 4.14-4.1 1 (m, 1 H), 4.04 (dd, 1 H), 4.01 -3.97 (m, 1 H), 3.94-3.91 (m, 1 H), 3.92-3.85 (m, 3H), 3.85-3.71 (m, 8H), 3.76-3.63 (m, 4H), 3.64 (dd, 1 H), 3.60 (dd, 1 H), 3.57-3.52 (m, 3H), 3.46-3.38 (m, 3H), 2.78-2.75 (m, 2H), 2.76 (s, 3H), 2.40 (t, 2H), 2.06 (s, 3H), 2.03-2.00 (m, 2H).

MS (ESI + ): m/z 904.05 (calc for C 33 H 59 N 3 0 22 SNa + [M+Na] + : m/z 904.32). Asialo GM1 Polymer (14):

To a solution of 5 (1 .3 mg, 6.25 μηιοΙ) in DMF (60 μΙ_) were subsequently added compound 13 (3.7 mg, 4.19 μηιοΙ, 0.4 equiv), water (5 μΙ_) and a solution of DBU (2.3 μΙ_, 15.7 μηηοΙ, 1 .5 equiv) in DMF (105 μΙ_). After stirring for 1 -24 h at rt, thioglycerol (2.7 μΙ_, 31.4 μηιοΙ, 3.0 equiv) and Et 3 N (4.4 μΙ_, 31 .4 μηιοΙ, 3.0 equiv) were added. The reaction mixture was stirred at rt for another 12-24 h. The product was precipitated by slow addition to a stirring solution of EtOH/Et 2 0 (1 :1 , 1 ml_). The precipitate was filtered off, washed with EtOH and dried. Further purification was achieved by ultrafiltration (Sartorius Stedim Vivaspin tubes, 6 ml_, molecular weight cutoff 10 kDa, 5500 rpm). Freeze-drying gave the asialo GM1 polymer 14 (4.6 mg, 71 %) as a white solid. According to 1 H NMR, the product contained approximately 44% of the lysine side-chains substituted by the carbohydrate epitope 13.

Scheme 4: Synthesis of the GM2 polymer 18

HN . , NH,

b)

17 [GM2-N(Me)0( 2 )2NHC(0)(CH2) 3 SH]

18 Reagents and conditions: a) 2, sodium acetate buffer, 76%; b) DL-Dithiothreitol, Y-thiobutyrolactone, Et 3 N, DMF, 52%; c) i. 5, DBU, DMF/H 2 0; ii. thioglycerol, Et 3 N, 56%

/V-(/V-Methyl-0-[2-aminoethyllhvdroxylamino)-2-acetamido- p-D-galactopyranosyl-(1 ^4)- [5-acetyl-g-neuraminic acid-(2→3)l-p-D-galactopyranosyl-(1→4)-p-D-glucopyranosi de (16):

To a solution of hemiacetal 15 (12.0 mg, 14.0 μηιοΙ) in NaOAc/AcOH buffer (0.1 M, pH 4.5, 140 μΙ_) was added oxyamine 2 (12.6 mg, 1 14 μηηοΙ, 10 equiv). The reaction mixture was stirred for 24-48 h at 25-40°C. Purification by dialysis gave compound 16 (9.9 mg, 10.6 μηιοΙ, 76%) as a white fluffy solid. 1 H NMR (500 MHz, D 2 0) δ 4.79 (d, 1 H), 4.55 (d, 1 H), 4.24 (d, 1 H), 4.20-4.09 (m, 2H), 4.04-4.00 (dd 2H), 3.97-3.87 (m, 3H), 3.91 -3.68 (m, 14H), 3.66-3.56 (m, 5H), 3.50 (dd, 1 H), 3.41 -3.34 (t, 1 H), 3.31 -3.26 (t, 2H), 2.81 (s, 3H), 2.72-2.63 (m, 1 H), 2.05 (s, 3H), 2.04 (s, 3H), 2.00-1 .88 (m, 1 H).

MS (ESP): m/z 907.56 (calc for C 3 4H 59 N 4 024 " [M-Na] " : m/z 907.35). /V-(/\/-lv1ethyl-0-[2-(2-mercaptobutanamido)ethyllhvdroxylam ino)-2-acetamido-p-D-galacto- pyranosyl-d→4)-[5-acetyl-g-neuraminic acid-(2→3)l-p-D-galactopyranosyl-(1→4)-β-ρ- glucopyranoside (17):

To a suspension of amine 16 (9.9 mg, 10.6 μηηοΙ) in anhyd DMF (250 μΙ_) were successively added DL-dithiothreitol (tip of spatula), γ-thiobutyrolactone (9.2 μΙ_, 106 μηιοΙ, 10 equiv) and Et 3 N (14.8 μΙ_, 106 μηιοΙ, 10 equiv). The reaction mixture was stirred for 12-24 h at 25-40°C. After that time, the reaction mixture was concentrated and the solvents co-evaporated with xylene. Purification by P2 size-exclusion chromatography gave compound 17 (5.7 mg, 5.52 μηηοΙ, 52%) as a white fluffy solid.

1 H-NMR (500 MHz, D 2 0): δ 4.76 (d, 1 H), 4.56 (d, 1 H), 4.19 (d, 1 H), 4.17 (dd, 1 H), 4.15 (d, 1 H), 4.01 (dd, 1 H), 3.94 (d, 1 H), 3.94 (dd, 1 H), 3.90-3.75 (m, 1 1 H), 3.75-3.60 (m, 5H), 3.62 (dd, 1 H), 3.58-3.53 (m, 1 H), 3.56 (dd, 1 H), 3.50 (dd, 1 H), 3.45-3.40 (m, 2H), 3.38 (dd, 1 H, H-2 Ga i), 2.77 (s, 3H), 2.68 (dd, 1 H), 2.57 (t, 2H), 2.40 (t, 2H), 2.05, 2.04 (2s, 6H), 1 .97- 1 .89 (m, 3H).

MS (ESP): m/z 1009.54 (calc for C 38 H 65 N 4 0 25 S " [M-Na] " : m/z 1009.37). GM2 Polymer (18):

To a solution of 5 (2.27 mg, 1 1.04 μηιοΙ) in DMF (1 10 μΙ_) were subsequently added compound 17 (5.7 mg, 5.52 μηιοΙ, 0.5 equiv), water (25 μΙ_) and a solution of DBU (2.5 μΙ_, 16.55 μη-ιοΙ, 1 .5 equiv) in DMF (22 μΙ_). After stirring for 1 -24 h at rt, thioglycerol (2.9 μΙ_, 33.1 1 μηιοΙ, 3.0 equiv) and Et 3 N (4.6 μΙ_, 33.1 1 μηιοΙ, 3.0 equiv) were added. The reaction mixture was stirred at rt for another 12-24 h. The product was precipitated by slow addition to a stirring solution of EtOH/Et 2 0 (1 :1 , 1 ml_). The precipitate was filtered off, washed with EtOH and dried. Further purification was achieved by ultrafiltration (Sartorius Stedim Vivaspin tubes, 6 ml_, molecular weight cutoff 10 kDa, 5500 rpm). Freeze-drying gave GM2 polymer 18 (4.5 mg, 56%) as a white solid. According to 1 H NMR, the product contained approximately 49% of the lysine side-chains substituted by the carbohydrate epitope 17.

Scheme 5: Synthesis of the GD1 a polymer 22

HN

b)

21 [GD1a-N(Me)0(CH 2 )2NHC(0)(CH2) 3 SH]

22 Reagents and conditions: a) 2, sodium acetate buffer, 87%; b) DL-dithiothreitol, Y-thiobutyrolactone, Et 3 N, DMF, 78%; c) i. 5, DBU, DMF/H 2 0; ii. thioglycerol, Et 3 N, 59%

galactopyranosyl-d ^^-acetamido-p-D-galactopyranosyl-d ^VfS-acetyl-a-neuraminic acid-(2→3)l-p-D-galactopyranosyl-(1→4)-p-D-glucopyranosi de (20):

To a solution of hemiacetal 19 (5.0 mg, 3.75 μηιοΙ) in NaOAc/AcOH buffer (0.1 M, pH 4.5, 35 μΙ_) was added oxyamine 2 (3.4 mg, 38 μηηοΙ, 10 equiv). The reaction mixture was stirred for 24-48 h at 25-40°C. Purification by dialysis gave compound 20 (5.0 mg, admixed with 1 .2 equiv of oxyamine 2, 3.27 μηηοΙ, corrected yield 87%) as a white fluffy solid.

1 H-NMR (500 MHz, D 2 0): δ 4.79 (m, 1 H), 4.63 (d, 1 H), 4.55 (d, 1 H), 4.24 (d, 1 H), 4.18 (d, 1 H), 4.18-4.14 (m, 1 H), 4.15-4.10 (m, 1 H), 4.1 1 (dd, 1 H), 4.09-4.05 (m, 2H), 4.06-4.02 (m, 1 H), 4.04-3.96 (m, 1 H), 3.99-3.95 (m, 1 H), 3.93-3.87 (m, 1 H), 3.92-3.85 (m, 2H), 3.89- 3.80 (m, 2H), 3.87-3.80 (m, 1 H), 3.86-3.70 (m, 7H), 3.82-3.52 (m, 12H), 3.67-3.60 (m, 3H), 3.58 (dd, 1 H), 3.54 (dd, 1 H), 3.41 (dd, 1 H), 3.30-3.26 (m, 2H), 2.81 (s, 3H), 2.77 (dd, 1 H), 2.70 (dd, 1 H), 2.05 (s, 6H), 2.03 (s, 3H), 1 .93 (t, 1 H), 1.82 (t, 1 H).

MS (ESP): m/z 679.83 (calc for C 5 iH 85 N 5 0 3 7 2" [M-2Na] 2" : m/z 679.75).

/V-(/V-Methyl-0-[2-(2-mercaptobutanamido)ethyllhvdroxylam ino)-5-acetyl-a-neuraminic acid-(2→'3)-p-D-galactopyranosyl-(1→'3)-2-acetamido-p-D- galactopyranosyl-(1→'4)-[5- acetyl-g-neuraminic acid-(2→3)l-p-D-galactopyranosyl-(1→4)-p-D-glucopyranosi de (21 ):

To a suspension of amine 20 (5.0 mg, 3.27 μηηοΙ) in anhyd DMF (65 μΙ_) were successively added DL-dithiothreitol (tip of spatula), γ-thiobutyrolactone (2.8 μΙ_, 32.7 μηιοΙ, 10 equiv) and Et 3 N (4.6 μΙ_, 32.7 μηιοΙ, 10 equiv). The reaction mixture was stirred for 12-24 h at 25-40°C. After that time, the reaction mixture was concentrated and the solvents co-evaporated with xylene. Purification by P2 size-exclusion chromatography gave compound 21 (3.8 mg, 2.52 μηηοΙ, 78%) as a white fluffy solid.

1 H-NMR (500 MHz, D 2 0): δ 4.79 (m, 1 H), 4.63 (d, 1 H), 4.56 (d, 1 H), 4.19 (d, 1 H), 4.18- 4.13 (m, 3H), 4.1 1 (dd, 1 H), 4.06 (m, 1 H), 4.02-3.96 (m, 1 H), 3.97 (d, 1 H), 3.94-3.85 (m, 3H), 3.94-3.51 (m, 12H), 3.93-3.84 (m, 2H), 3.88-3.80 (m, 2H), 3.87-3.68 (m, 7H), 3.86- 3.81 (m, 1 H), 3.68-3.61 (m, 2H), 3.66-3.60 (m, 1 H), 3.58-3.54 (m, 1 H), 3.56-3.50 (m, 1 H), 3.45-3.38 (m, 3H), 2.80-2.75 (m, 1 H), 2.77 (s, 3H), 2.70 (dd, 1 H), 2.57 (t, 2H), 2.40 (t, 2H), 2.05 (s, 6H), 2.03 (s, 3H), 1 .96-1.89 (m, 3H), 1.82 (t, 1 H).

MS (ESI-): m/z 730.97 (calc for C 55 H 91 N 5 0 3 8S 2" [M-2Na] 2" : m/z 730.75). GD1 a Polymer (22): To a solution of 5 (1 .4 mg, 6.63 μιτιοΙ) in DMF (67 μΙ_) were subsequently added compound 21 (2.4 mg, 1 .59 μηιοΙ, 0.4 equiv), water (15 μΙ_) and a solution of DBU (1 .5 μΙ_, 9.9 μηηοΙ, 1 .5 equiv) in DMF (13 μΙ_). After stirring for 1 -24 h at rt, thioglycerol (1 .7 μΙ_, 19.9 μηιοΙ, 3.0 equiv) and Et 3 N (2.8 μΙ_, 19.9 μηιοΙ, 3.0 equiv) were added. The reaction mixture was stirred at rt for another 12-24 h. The product was precipitated by slow addition to a stirring solution of EtOH/Et 2 0 (1 :1 , 1 ml_). The precipitate was filtered off, washed with EtOH and dried. Further purification was achieved by ultrafiltration (Sartorius Stedim Vivaspin tubes, 6 ml_, molecular weight cutoff 10 kDa, 5500 rpm). Freeze-drying gave GD1 a polymer 22 (3.6 mg, 59%) as a white solid. According to 1 H NMR, the product contained approximately 46% of the lysine side-chains substituted by the carbohydrate epitope 21.

Scheme 6: Synthesis of the GD1 b polymer 26

26 Reagents and conditions: a) 2, sodium acetate buffer, 70%; b) DL-dithiothreitol, Y-thiobutyrolactone, Et 3 N, DMF, 77%; c) i. 5, DBU, DMF/H 2 0; ii. thioglycerol, Et 3 N, 40%

/V-(/V-Methyl-0-[2-aminoethyllhvdroxylamino)-p-D-galactop yranosyl-(1 ^3)-2-acetamido-p- D-galactopyranosyl-d→4)-[5-acetyl-g-neuraminic acid-(2→8)-5-acetyl-a-neuraminic acid- (2→3)l-p-D-galactopyranosyl-(1→4)-p-D-glucopyranoside (24):

To a solution of hemiacetal 23 (19.3 mg, 15.0 μηιοΙ) in NaOAc/AcOH buffer (0.1 M, pH 4.5, 150 μΙ_) was added oxyamine 2 (313.5 mg, 150 μηιοΙ, 10 equiv). The reaction mixture was stirred for 24-48 h at 25-40°C. Purification by dialysis gave compound 24 (14.2 mg, 10.1 μηιοΙ, 70%) as a white fluffy solid 1 H-NMR (500 MHz, D 2 0): δ 4.80 (d, 1 H), 4.55-4.53 (t, 2H), 4.23 (d, 1 H), 4.21 -3.40 (m, 38H), 3.29-3.27 (m, 2H), 2.81 (s, 3H), 2.78-2.69 (m, 2H), 2.09, 2.06, 2.05 (3s, 9H), 1 .82- 1 .73 (m, 2H).

MS (ESP): m/z 1382.67 (calc for C 5 iH 85 N 5 0 3 7 " [M-Na] " : m/z 1382.48).

/V-(/\/-Methyl-0-[2-(2-mercaptobutanamido)ethyllhvdroxyla mino)-p-D-galactopyranosyl- (1→3)-2-acetamido-p-D-galactopyranosyl-(1→4)-[5-acetyl-g -neuraminic acid-(2→8)-5- acetyl-g-neuraminic acid-(2→3)l-p-D-galactopyranosyl-(1→4)-p-D-glucopyranosi de (25):

To a suspension of amine 24 (4.9 mg, 3.51 μηηοΙ) in anhyd DMF (70 μΙ_) were successively added DL-dithiothreitol (1 .0 mg, 6.31 μηηοΙ, 1 .8 equiv), γ-thiobutyrolactone (3.0 μΙ_, 35.0 μηιοΙ, 10 equiv) and Et 3 N (4.9 μΙ_, 32.0 μηιοΙ, 10 equiv). The reaction mixture was stirred for 12-24 h at 25-40°C. After that time, the reaction mixture was concentrated and the solvents co-evaporated with xylene. Purification by P2 size-exclusion chromatography gave compound 25 (4.1 mg, 2.72 μηηοΙ, 77%) as a white fluffy solid.

1 H-NMR (500 MHz, D 2 0): δ 4.79 (d, 1 H), 4.55-4.53 (m, 2H), 4.21 -3.40 (m, 41 H), 2.77 (s, 3H), 2.80-2.67 (m, 4H), 2.41 (t, 2H), 2.09, 2.05 (2s, 1 1 H), 1.84-1 .73 (m, 2H). MS (ESP): m/z 1484.86 (calc for C 55 H 91 N 5 0 38 NaS " [M-Na] " : m/z 1484.50). GD1 b Polymer (26):

To a solution of 5 (0.59 mg, 2.89 μηιοΙ) in DMF (30 μΙ_) were subsequently added compound 25 (2.1 mg, 1 .45 μηιοΙ, 0.5 equiv), water (3 μΙ_) and a solution of DBU (0.6 μΙ_, 4.34 μηηοΙ, 1 .5 equiv) in DMF (6 μΙ_). After stirring for 1 -24 h at rt, thioglycerol (0.75 μΙ_, 8.7 μηιοΙ, 3.0 equiv) and Et 3 N (1 .21 μΙ_, 8.7 μηιοΙ, 3.0 equiv) were added. The reaction mixture was stirred at rt for another 12-24 h. The product was precipitated by slow addition to a stirring solution of EtOH/Et 2 0 (1 :1 , 1 ml_). The precipitate was filtered off, washed with EtOH and dried. Further purification was achieved by ultrafiltration (Sartorius Stedim Vivaspin tubes, 6 ml_, molecular weight cutoff 10 kDa, 5500 rpm). Freeze-drying gave GD1 b polymer 26 (0.68 mg, 40%) as a white solid. According to 1 H NMR, the product contained approximately 20% of the lysine side-chains substituted by the carbohydrate epitope 25.

Scheme 7: Synthesis of the GD3 polymer 30

Reagents and conditions: a) 2, sodium acetate buffer, 50%; b) DL-dithiothreitol, γ-thiobutyrolactone, Et 3 N, DMF, 66%; c) i. 5, DBU, DMF/H 2 0; ii. thioglycerol, Et 3 N, 18% /V-(/V-Methyl-0-[2-aminoethyllhvdroxylamino)-5-acetyl-a-neur aminic acid-(2→8)-5-acetyl- g-neuraminic acid-(2→3)-p-D-galactopyranosyl-(1→4)-p-D-glucopyranosid e (28): To a solution of hemiacetal 27 (10 mg, 10.3 μηιοΙ) in NaOAc/AcOH buffer (0.1 M, pH 4.5, 103 μΙ_) was added oxyamine 2 (9.3 mg, 103 μηηοΙ, 10 equiv). The reaction mixture was stirred for 24-48 h at 25-40°C. Purification by dialysis gave compound 28 (5.4 mg, 5.18 μηιοΙ, 50%) as a white fluffy solid. 1 H-NMR (500 MHz, D 2 0): δ 4.55 (d, 1 H), 4.24-3.57 (m, 28 H), 4.23 (d, 1 H), 3.27 (m, 2H), 2.84-2.77 (m, 1 H), 2.81 (s, 3H), 2.70 (dd, 1 H), 2.09, 2.05 (2s, 6H), 1 .76 (t, 2H).

MS (ESP): m/z 497.36 (calc for C 3 7H 62 N 4 027 2" [M-2Na] 2" : m/z 497.18).

/V-(/V-Methyl-0-[2-(2-mercaptobutanamido)ethyllhvdroxylam ino)-5-acetyl-a-neuraminic acid-(2→8)-5-acetyl-a-neuraminic acid-(2→3)-p-D-galactopyranosyl-(1→4)-p-D- glucopyranoside (29):

To a suspension of amine 28 (3.2 mg, 3.07 μηηοΙ) in anhyd DMF (61 μΙ_) were successively added DL-dithiothreitol (tip of spatula), γ-thiobutyrolactone (2.7 μΙ_, 30.7 μηιοΙ, 10 equiv) and Et 3 N (4.3 μΙ_, 30.7 μηιοΙ, 10 equiv). The reaction mixture was stirred for 12-24 h at 25-40°C. After that time, the reaction mixture was concentrated and the solvents co-evaporated with xylene. Purification by P2 size-exclusion chromatography gave compound 29 (2.3 mg, 2.01 μηιοΙ, 66%) as a white fluffy solid.

1 H-NMR (500 MHz, D 2 0): δ 4.55 (d, 1 H), 4.21 -3.55 (m, 28 H), 4.20 (d, 1 H), 3.43 (m, 2H), 2.80-2.75 (m, 1 H), 2.77 (s, 3H), 2.71 -2.68 (m, 1 H), 2.57 (t, 2H), 2.40 (t, 2H), 2.09, 2.05 (2s, 6H), 1 .92-1 .89 (m, 2H), 1 .76 (t, 2H). MS (ESP): m/z 548.26 (calc for C 4 iH 68 N 4 0 28 S 2" [M-2Na] 2" : m/z 548.19). GD3 Polymer (30):

To a solution of 5 (0.75 mg, 3.67 μηιοΙ) in DMF (37 μΙ_) were subsequently added compound 29 (2.1 mg, 1 .83 μηηοΙ, 0.5 equiv), water (10 μΙ_) and a solution of DBU (0.8 μΙ_, 5.5 μηιοΙ, 1 .5 equiv) in DMF (7 μΙ_). After stirring for 1 -24 h at rt, thioglycerol (1 .0 μΙ_, 1 1.0 μηιοΙ, 3.0 equiv) and Et 3 N (1 .5 μΙ_, 1 1 .0 μηιοΙ, 3.0 equiv) were added. The reaction mixture was stirred at rt for another 12-24 h. The product was precipitated by slow addition to a stirring solution of EtOH/Et 2 0 (1 :1 , 1 ml_). The precipitate was filtered off, washed with EtOH and dried. Further purification was achieved by ultrafiltration (Sartorius Stedim Vivaspin tubes, 6 ml_, molecular weight cutoff 10 kDa, 5500 rpm). Freeze-drying gave GD3 polymer 30 (0.3 mg, 18%) as a white solid. According to 1 H NMR, the product contained approximately 17% of the lysine side-chains substituted by the carbohydrate epitope 29.

Scheme 8: Synthesis of the GT1 a polymer 34

33 [GT1a-N(Me)0(CH 2 ) 2 NH

Reagents and conditions: a) 2, sodium acetate buffer, 84%; b) DL-dithiothreitol, γ-thiobutyrolactone, Et 3 N, DMF, 24%; c) i. 5, DBU, DMF/H 2 0; ii. thioglycerol, Et 3 N, 25% /V-(/V-Methyl-0-[2-aminoethyllhvdroxylamino)-5-acetyl-a-neur ami acid-(2→8)-5-acetyl- g-neuraminic acid-(2→'3)-p-D-galactopyranosyl-(1→'3)-2-acetamido-p-D- galactopyranosyl- (1→4)-[5-acetyl-g-neuraminic acid-(2→3)l-p-D-galactopyranosyl-(1→4)-β-ρ- glucopyranoside (32): To a solution of hemiacetal 31 (5.0 mg, 3.04 μηιοΙ) in NaOAc/AcOH buffer (0.1 M, pH 4.5, 30 μΙ_) was added oxyamine 2 (2.7 mg, 30 μηηοΙ, 10 equiv). The reaction mixture was stirred for 24-48 h at 25-40°C. Purification by dialysis gave compound 32 (4.38 mg, 2.55 μηηοΙ, 84%) as a white fluffy solid.

1 H-NMR (500 MHz, D 2 0): δ 4.80 (d, 1 H), 4.64, 4.55 (2d, 2H), 4.24 (d, 1 H), 4.20-3.40 (m, 45H), 3.29-3.27 (m, 2H), 2.81 (s, 3H), 2.78-2.69 (m, 3H), 2.08, 2.05 (2s, 12H), 1 .85-1.65 (m, 3H).

MS (ESP): m/z 836.33 (calc for C 6 2H 10 iN 6 O 45 Na 2" [M-2Na] 2" : m/z 836.29).

/V-(/V-Methyl-0-[2-(2-mercaptobutanamido)ethyllhvdroxylam ino)-5-acetyl-a-neuraminic acid-(2→8)-5-acetyl-a-neuraminic acid-(2→3)-p-D-galactopyranosyl-(1→3)-2-acetamido- p-D-galactopyranosyl-(1→4)-[5-acetyl-a-neuraminic acid-(2→3)l-p-D-galactopyranosyl- (1→4)-p-D-glucopyranoside (33):

To a suspension of amine 32 (3.2 mg, 1.86 μηηοΙ) in anhyd DMF (37 μΙ_) were successively added DL-dithiothreitol (0.5 mg, 3.35 μηιοΙ, 1 .8 equiv), γ-thiobutyrolactone (1 .6 μΙ_, 18.6 μηιοΙ, 10 equiv) and Et 3 N (2.6 μΙ_, 18.6 μηιοΙ, 10 equiv). The reaction mixture was stirred for 12-24 h at 25-40°C. After that time, the reaction mixture was concentrated and the solvents co-evaporated with xylene. Purification by P2 size-exclusion chromatography gave compound 33 (0.82 mg, 0.45 μηηοΙ, 24%) as a white fluffy solid.

1 H-NMR (500 MHz, D 2 0): δ 4.79 (d, 1 H), 4.64, 4.55 (2d, 2H), 4.20-3.40 (m, 39H), 2.77 (s, 3H), 2.79-2.66 (m, 3H), 2.57 (t, 2H), 2.40 (t, 2H), 2.08, 2.05 (2s, 14H), 1.94-1 .74 (m, 2H). MS (ESP): m/z 583.80 (calc for C 66 H 10 7N 6 O 46 SNa 3" [M-3Na] 3" : m/z 583.87). GT1 a Polymer (34):

To a solution of 5 (0.19 mg, 0.90 μηιοΙ) in DMF (9 μΙ_) were subsequently added compound 33 (0.82 mg, 0.45 μηιοΙ, 0.5 equiv), water (1 μΙ_) and a solution of DBU (0.2 μΙ_, 1 .36 μηιοΙ, 1 .5 equiv) in DMF (2 μΙ_). After stirring for 1 -3 h at rt, thioglycerol (0.2 μΙ_, 2.7 μηιοΙ, 3.0 equiv) and Et 3 N (0.4 μΙ_, 2.7 μηιοΙ, 3.0 equiv) were added. The reaction mixture was stirred at rt for another 12-24 h. The product was precipitated by slow addition to a stirring solution of EtOH/Et 2 0 (1 :1 , 1 ml_). The precipitate was filtered off, washed with EtOH and dried. Further purification was achieved by ultrafiltration (Sartorius Stedim Vivaspin tubes, 6 ml_, molecular weight cutoff 10 kDa, 5500 rpm). Freeze-drying gave GT1 a polymer 34 (0.28 mg, 25%) as a white solid. According to 1 H NMR, the product contained approximately 57% of the lysine side-chains substituted by the carbohydrate epitope 33.

Scheme 9: Synthesis of the GM1 a-linker2 polymer 38

Reagents and conditions: a) 35, sodium acetate buffer, 58%; b) DL-dithiothreitol, Y-thiobutyrolactone, Et 3 N, DMF, 74%; c) i. 5, DBU, DMF/H 2 0; ii. thioglycerol, Et 3 N, 41 % /V-[0-Methyl-/V-(2-aminoethyl)hv^^

D-galactopyranosyl-(1→4)-[5-acetyl-a-neuraminic acid-(2→3VI-p-D-galactopyranosyl- (1→4)-p-D-glucopyranoside (36):

To a solution of hemiacetal 1 (10.0 mg, 9.80 μηιοΙ) in NaOAc/AcOH buffer (0.1 M, pH 4.5, 98 μΙ_) was added oxyamine 35 (8.8 mg, 98 μηηοΙ, 10 equiv). The reaction mixture was stirred for 24-48 h at 25-40°C. Purification by dialysis gave compound 36 (6.2 mg, 5.63 μηιοΙ, 58%) as a white fluffy solid.

1 H-NMR (500 MHz, D 2 0): δ 4.78 (d, 1 H), 4.57-4.54 (m, 2H), 4.31 (d, 1 H), 4.18-3.51 (m, 33H), 3.38 (t, 1 H), 3.32 (m, 2H), 3.27 (m, 2H), 2.68 (dd, 1 H), 2.05, 2.03 (2s, 6H), 1.95 (t, 1 H).

HRMS (ESI + ): m/z 1071.4177 (calc. for C 4 oH 71 N 4 029 + [M+H] + : 1071 .4198).

/V-(/\/-[2-(2-lv1ercaptobutanamido)ethyll-0-methyl)-p-D-g alactopyranosyl-(1→'3)-2- acetamido-p-D-galactopyranosyl-d ^VfS-acetyl-a-neuraminic acid-(2→3)l-p-D- galactopyranosyl-d→4)-p-D-glucopyranoside (37):

To a suspension of amine 36 (6.1 mg, 5.6 μηηοΙ) in anhyd DMF (1 12 μΙ_) were successively added DL-dithiothreitol (tip of spatula), γ-thiobutyrolactone (4.9 μΙ_, 56 μηιοΙ, 10 equiv) and Et 3 N (7.8 μΙ_, 56 μηηοΙ, 10 equiv). The reaction mixture was stirred for 12-24 h at 25-40°C. After that time, the reaction mixture was concentrated and the solvents co-evaporated with xylene. Purification by P2 size-exclusion chromatography gave compound 37 (5.0 mg, 4.2 μηηοΙ, 74%) as a white fluffy solid.

1 H-NMR (500 MHz, D 2 0): δ 4.78 (d, 1 H), 4.56 (m, 2H), 4.25 (d, 1 H), 4.18-3.37 (m, 34H), 3.46 (m, 2H, Hb), 3.23 (m, 1 H), 3.07 (m, 1 H), 2.77 (m, 1 H), 2.68 (dd, 1 H), 2.41 (t, 2H), 2.05, 2.03 (2s, 6H), 1 .97-1 .93 (m, 3H). MS (ESI-): m/z 1 171.65 (calc. for C 44 H 75 N 4 0 3 oS " [M-Na] " : 1 171.42). GM1 a-linker2-polymer (38):

To a solution of 5 (1 .7 mg, 8.4 μηιοΙ) in DMF (84 μΙ_) were subsequently added compound 37 (5.0 mg, 4.2 μηηοΙ, 0.5 equiv), water (8.4 μΙ_) and a solution of DBU (1 .9 μΙ_, 12.5 μηηοΙ, 1 .5 equiv) in DMF (17 μΙ_). After stirring for 1 -24 h at rt, thioglycerol (2.2 μΙ_, 25 μηηοΙ, 3.0 equiv) and Et 3 N (3.5 μΙ_, 25 μηιοΙ, 3.0 equiv) were added. The reaction mixture was stirred at rt for another 12-24 h. The product was precipitated by slow addition to a stirring solution of EtOH/Et 2 0 (1 :1 , 1 mL). The precipitate was filtered off, washed with EtOH and dried. Further purification was achieved by ultrafiltration (Sartorius Stedim Vivaspin tubes, 6 mL, molecular weight cutoff 10 kDa, 5500 rpm). Freeze-drying gave GM1 a-linker2 polymer 38 (2.5 mg, 41 %) as a white solid. According to 1 H NMR, the product contained approximately 41 % of the lysine side-chains substituted by the carbohydrate epitope 37.

Scheme 10: Synthesis of the GM1 a-linker3 polymer 42

41 [GM1a-N(Me)(0(CH 2 )2)2NHC(0)(CH 2 )3SH]

42 Reagents and conditions: a) 39, sodium acetate buffer, 51 %; b) DL-dithiothreitol, Y-thiobutyrolactone, Et 3 N, DMF, 64%; c) i. 5, DBU, DMF/H 2 0; ii. thioglycerol, Et 3 N, 32%

/V-(/V-Methyl-0-[2-0-(2-aminoethyl)hvdroxylethyl)lhvdroxy lamino)-p-D-galactopyranosyl- (1→3)-2-acetamido-p-D-galactopyranosyl-(1→4)-[5-acetyl-g -neuraminic acid-(2→3)l-p-D- galactopyranosyl-d→4)-p-D-glucopyranoside (40):

To a solution of hemiacetal 1 (10.0 mg, 9.80 μηιοΙ) in NaOAc/AcOH buffer (0.1 M, pH 4.5, 98 μΙ_) was added oxyamine 39 (13.1 mg, 98 μηηοΙ, 10 equiv). The reaction mixture was stirred for 24-48 h at 25-40°C. Purification by dialysis gave compound 40 (7.48 mg, 6.50 μηιοΙ, 51 %) as a white fluffy solid. 1 H-NMR (500 MHz, D 2 0): δ 4.78 (d, 1 H), 4.56 (d, 2H), 4.19 (d, 1 H), 4.18-4.14 (m, 3H), 4.13-3.48 (m, 32H), 3.38 (dd, 1 H), 3.23 (m, 2H), 2.79 (s, 3H), 2.67 (m, 1 H), 2.05, 2.02 (2s, 6H), 1.94 (m, 1 H).

HRMS (ESI + ): m/z 1 1 15.4447 (calc. for C 4 2H 75 N 4 03o + [M+H] + : 1 1 15.4461 ).

/V-(/\/-Methyl-0-[2-0-[2-(2-mercaptobutanamido)ethyllhvdr oxylethyllhvdroxylamino)-p-D- galactopyranosyl-d ^S^-acetamido-p-D-galactopyranosyl-d ^VfS-acetyl-a-neuraminic acid-(2→3)l-p-D-galactopyranosyl-d→4)-p-D-glucopyranosid e (41 ):

To a suspension of amine 40 (7.48 mg, 6.50 μηηοΙ) in anhyd DMF (130 μΙ_) were successively added DL-dithiothreitol (tip of spatula), γ-thiobutyrolactone (5.6 μΙ_, 65 μηιοΙ, 10 equiv) and Et 3 N (9.1 μΙ_, 65 μηηοΙ, 10 equiv). The reaction mixture was stirred for 12-24 h at 25-40°C. After that time, the reaction mixture was concentrated and the solvents co-evaporated with xylene. Purification by P2 size-exclusion chromatography gave compound 41 (5.21 mg, 4.16 μηηοΙ, 64%) as a white fluffy solid.

1 H-NMR (500 MHz, D 2 0): δ 4.78 (d, 1 H), 4.56 (d, 2H), 4.19 (d, 1 H), 4.18-3.52 (m, 36H), 3.41 (t, 2H), 3.37 (dd, 1 H), 2.79 (s, 3H), 2.67 (m, 1 H), 2.57 (t, 2H), 2.39 (t, 2H), 2.05, 2.02 (2s, 6H), 1 .98-1 .88 (m, 3H).

HRMS (ESI + ) : m/z 1239.4412 (calc. for C 46 H 8 oN 4 0 3 iNaS + [M+H] + 1239.4419).

GM1 a-linker3-polymer (42): To a solution of 5 (1.72 mg, 8.41 μηηοΙ) in DMF (84 μΙ_) were subsequently added compound 41 (5.21 mg, 4.21 μηιοΙ, 0.5 equiv), water (8.4 μΙ_) and a solution of DBU (1 .9 μΙ_, 13 μηηοΙ, 1 .5 equiv) in DMF (17 μΙ_). After stirring for 1 -24 h at rt, thioglycerol (2.2 μΙ_, 25 μηιοΙ, 3.0 equiv) and Et 3 N (3.5 μΙ_, 25 μηιοΙ, 3.0 equiv) were added. The reaction mixture was stirred at rt for another 12-24 h. The product was precipitated by slow addition to a stirring solution of EtOH/Et 2 0 (1 :1 , 1 ml_). The precipitate was filtered off, washed with EtOH and dried. Further purification was achieved by ultrafiltration (Sartorius Stedim Vivaspin tubes, 6 ml_, molecular weight cutoff 10 kDa, 5500 rpm). Freeze-drying gave GM1 a-linker3-polymer 42 (2.64 mg, 32%) as a white solid. According to 1 H NMR, the product contained approximately 61 % of the lysine side-chains substituted by the carbohydrate epitope 41.

Scheme 11: Synthesis of the GM1a-linker4-polymer 45

OMe

I

HN _ . ,SH

S

43

44 [GM1a-N(OMe CH2) 2 S(CH 2 )2SH]

45

Reagents and conditions: a) 43, sodium acetate buffer, 48%; b) i.5, DBU, DMF/H 2 0; ii. thioglycerol, Et 3 N, 90% /V-(0-Methyl-/V-r2-(2-ethylthio)e^^

acetamido-p-D-galactopyranosyl-f l ^VfS-acetyl-a-neuraminic acid-(2→3VI-p-D- galactopyranosyl-d→4)-p-D-glucopyranoside (44):

To a solution of hemiacetal 1 (10.0 mg, 9.80 μηιοΙ) in NaOAc/AcOH buffer (0.1 M, pH 4.5, 98 μΙ_) was added oxyamine 43 (16 mg, 98 μηηοΙ, 10 equiv). The reaction mixture was stirred for 24-48 h at 25-40°C. Purification by reverse phase chromatography (0→100% MeOH in H 2 0) gave compound 44 (5.5 mg, 4.7 μηηοΙ, 48%) as a white fluffy solid.

1 H-NMR (500 MHz, D 2 0): δ 4.78 (d, 1 H), 4.56, 4.55 (2d, 2H), 4.26 (d, 1 H), 4.21 -3.48 (m, 36H), 3.38 (t, 1 H), 3.35-3.30 (m, 1 H), 3.16 (m, 1 H), 3.04-3.01 (m, 2H), 3.01 -2.99 (m, 2H), 2.89 (t, 2H), 2.68 (dd, 1 H), 2.05 2.02 (2s, 6H), 1 .95 (t, 1 H).

MS (ESP): m/z 1 146.59 (calc. for C 4 2H 72 N 3 029S2 " [M-Na] " 1 146.37). GM1 a-linker4-polymer (45):

To a solution of 5 (1.17 mg, 5.13 μηηοΙ) in DMF (57 μΙ_) were subsequently added compound 44 (3.35 mg, 2.86 μηιοΙ, 0.5 equiv), water (5.8 μΙ_) and a solution of DBU (1 .3 μΙ_, 8.6 μηηοΙ, 1 .5 equiv) in DMF (12 μΙ_). After stirring for 1 -3 h at rt, thioglycerol (1 .5 μΙ_, 17 μηιοΙ, 3.0 equiv) and Et 3 N (2.4 μΙ_, 17 μηιοΙ, 3.0 equiv) were added. The reaction mixture was stirred at rt for another 12-24 h. The product was precipitated by slow addition to a stirring solution of EtOH/Et 2 0 (1 :1 , 1 ml_). The precipitate was filtered off, washed with EtOH and dried. Further purification was achieved by ultrafiltration (Sartorius Stedim Vivaspin tubes, 6 ml_, molecular weight cutoff 10 kDa, 5500 rpm). Freeze-drying gave GM1 a-linker4-polymer 45 (3.08 mg, 90%) as a white solid. According to 1 H NMR, the product contained approximately 30% of the lysine side-chains substituted by the carbohydrate epitope 44.

Scheme 12: Synthesis of Iinker2 35

61 62 35

Reagents and conditions: a) i. MeONH 2 HCI, AcONa, EtOH; ii. NaBH 3 CN, AcCI, 39%; b) TFA, DCM, quant te/f-Butyl (2-(methoxyamino)ethyl)carbamate (62):

To a solution of aldehyde 61 (340 mg, 2.14 mmol) in EtOH (3.5 mL) was added methoxyamine hydrochloride (214 mg, 3.77 mmol, 1.2 equiv) and AcONa (350 mg, 4.27 mmol, 2.0 equiv). The reaction mixture was stirred overnight at rt. After that time, NaBH 3 CN (201 mg, 3.20 mmol, 1 .5 equiv) was added followed by dropwise addition of a freshly prepared solution of 1 M ethanolic HCI (7.0 mL, freshly prepared from AcCI and EtOH). After stirring for 1 h at rt, the reaction was neutralized by addition of satd aq NaHC0 3 . The reaction mixture was diluted with H 2 0 and extracted with DCM (3 x). The organic phases were pooled, washed with brine and dried over anhyd Na 2 S0 4 . The suspension was filtrated and concentrated under reduced pressure. Purification by flash chromatography eluting with PE/Acetone (85:15) yielded the aminoalcohol 62 (158 mg, 0.832 mmol, 39%) as a colourless oil.

1 H-NMR (500 MHz, CDCI 3 ) δ 5.69 (s, 1 H), 4.91 (s, 1 H), 3.54 (s, 3H), 3.30 (m, 2H), 3.00 (t, 2H), 1.46 (s, 9H). 2-(Methoxyamino)ethan-1 -amine (35):

Aminoalcohol 62 (160 mg, 0.84 mmol) was dissolved in DCM (1 .1 mL). The solution was cooled to 0°C and trifluoroacetic acid (TFA, 320 μΐ, 4.2 mmol, 5.0 equiv) was added dropwise to the reaction mixture. After stirring for 1 h at 0°C followed by 3 h at rt, the reaction mixture was diluted with MeOH and neutralized with free base Amberlite resin. The suspension was filtered over cotton and the filtrate was concentrated in vacuo. Purification by flash chromatography eluting with DCM/MeOH (9:1 → 7:3) gave amine 62 (100 mg) as a TFA salt partially.

1 H-NMR (500 MHz, D 2 0) δ 3.47 (s, 3H), 3.13 (m, 2H), 3.10 (m, 2H).

Scheme 13: Synthesis of Iinker3 39

, 0. a) , 0 , b)

HO " NH, HO ' " NHBoc Br ' 'NHBoc

63 64 65

BocN .

"OH

66 d)

BocN . .0, HN . .0,

* 0 "NHBoc " NH,

c) 67 39

Reagents and conditions: a) Boc 2 0, Et 3 N, DCM, 84%; b) i. MsCI, Et 3 N, DCM; ii. LiBr, Acetone, 98%; c) 66, NaH, DMF, 90%; d) TFA, DCM, 96% te/f-Butyl (2-(2-hvdroxyethoxy)ethyl)carbamate (64):

Amine 63 (1.0 mL, 10 mmol) was dissolved in DCM (50 mL). The solution was cooled to 0°C and di-tert-butyl dicarbonate (Boc 2 0, 1.74 g, 8.9 mmol, 0.8 equiv) was added to the solution followed by Et 3 N (1 .4 mL, 10 mmol, 1 .0 equiv). After stirring for 1 h at 0°C followed by 2 h at rt, the reaction mixture was diluted with DCM and washed with H 2 0 and brine. The organic phase was dried over anhyd Na 2 S0 4 . The suspension was filtered over cotton and the filtrate concentrated in vacuo. Purification by flash chromatography eluting with DCM/MeOH (1 :0→ 9:1 ) gave alcohol 64 (1 .37 g, 6.67 mmol, 84%) as a colourless oil.

1 H-NMR (500 MHz, CDCI 3 ) δ 4.88 (s, 1 H), 3.74 (m, 2H), 3.58 (t, 2H), 3.56 (t, 2H), 3.34 (m, 2H), 2.05 (s, 1 H), 1 .45 (s, 9H). te/f-Butyl (2-(2-bromoethoxy)ethyl)carbamate (65):

Alcohol 64 (1 .25 g, 6.09 mmol) was dissolved in DCM (34 mL). The solution was cooled to 0°C and methanesulfonyl chloride (MsCI, 0.80 mL, 10.3 mmol, 1 .7 equiv) was added to the solution followed by Et 3 N (1.9 mL, 13.4 mmol, 2.2 equiv). After stirring for 3 h at rt, the reaction mixture was diluted with acetone (33 mL) and LiBr (8.9 g, 103 mmol, 17 equiv) was added. The reaction mixture was stirred overnight at rt. After that time, the solvents were evaporated under reduced pressure. The crude residue was diluted with EtOAc and washed with H 2 0 and brine. The organic phase was dried over anhyd Na 2 S0 4 . The suspension was filtered over cotton and the filtrate concentrated in vacuo. Purification by flash chromatography eluting with PE/Acetone (85:15→ 8:2) gave bromide 65 (1 .60 g, 5.95 mmol, 98%) as a colourless oil.

1 H-NMR (500 MHz, CDCI 3 ): δ 4.91 (s, 1 H), 3.78 (t, 2H), 3.56 (t, 2H), 3.47 (t, 2H), 3.33 (d, 2H), 1 .45 (s, 9H). te f-Butyl (2-(2-((te f-butoxycarbonyl)amino)ethoxy)ethoxy)(methyl)carbamate (67):

NaH (60% in mineral oil, 82 mg, 2.04 mmol, 0.96 equiv) was added at 0°C to a solution of aminoalcohol 66 (313 mg, 2.12 mmol, 1 .0 equiv) in anhyd DMF (1 .4 mL). After stirring for 30 min at that temperature, a solution of bromide 65 (456 mg, 1.70 mmol, 0.8 equiv) was added to the reaction mixture. The reaction mixture was stirred 1 h at 0°C followed by 2 h at rt. After that time, the reaction was quenched by addition of MeOH, concentrated in vacuo and the solvent coevaporated with xylene. Purification by flash chromatography eluting with PE/Acetone (8:2→ 75:25) gave aminoalcohol 67 (510 mg, 1 .53 mmol, 90%) as a colourless oil.

1 H-NMR (500 MHz, CDCI 3 ): δ 5.04 (s, 1 H), 4.04-3.93 (m, 2H), 3.69-3.62 (m, 2H), 3.55 (t, 2H), 3.33 (m, 2H), 3.1 1 (s, 3H), 1 .49 (s, 9H), 1 .44 (s, 9H).

2-(2-((methylamino)oxy)ethoxy)ethan-1 -amine (39)

Aminoalcohol 67 (421 mg, 1.26 mmol) was dissolved in DCM (1 .6 mL). The solution was cooled to 0°C and TFA (480 μΐ, 6.29 mmol, 5.0 equiv) was added dropwise to the reaction mixture. After stirring for 1 h at 0°C followed by 5 h at rt, the reaction mixture was diluted with MeOH and neutralized with free base Amberlite resin. The suspension was filtered over cotton and the filtrate was concentrated in vacuo. Purification by flash chromatography eluting with DCM/MeOH (95:5 → 7:3) gave amine 39 (162 mg, 1 .21 mmol, 96%) as a colourless oil.

1 H-NMR (500 MHz, D 2 0): δ 3.94 (m, 2H), 3.78 (t, 2H), 3.74 (m, 2H), 3.24 (t, 2H), 2.69 (s, 3H).

Scheme 14: Synthesis of Iinker4 43

Reagents and conditions: a) DIBAL-H, DCM, 61 %; b) i. MeONH 2 HCI, AcONa, EtOH; ii. NaBH 3 CN, AcCI, EtOH, 17%

1 ,4-dithian-2-ol (69)

Ester 68 (100 mg, 0.60 mmol) was dissolved in anhyd DCM (1 .2 mL). The solution was cooled to -78°C and DIBAL-H (1 M in Toluene, 0.60 mL, 0.60 mmol, 1 equiv) was added dropwise to the reaction mixture. After stirring for 2 h at -78°C, DIBAL-H (0.3 mL, 0.3 mmol, 0.5 equiv) was added dropwise to the reaction mixture. After stirring for another 30 min at -78°C, potassium sodium tartrate tetrahydrate (1 .7 g) and H 2 0 (2.0 mL) were added to the reaction mixture. After stirring vigourously for 1 h at rt, the aqueous phase was extracted (3 x) with DCM. The organic phases were pooled, washed with brine and dried over Na 2 S0 4 . The suspension was filtered over cotton and the filtrate concentrated in vacuo. Flash chromatography eluting with PE/Acetone (85:15→ 8:2) yielded derivative 69 (50 mg, 0.37 mmol, 61 %) as a white solid.

1 H-NMR (500 MHz, CDCI 3 ): δ 4.93 (dd, 1 H), 3.65 (d, 1 H), 3.46 (dd, 1 H), 3.34 (m, 1 H), 3.1 1 -2.99 (m, 1 H), 2.87 (dd 1 H), 2.71 (m, 1 H), 2.61 (m, 1 H).

2-((2-(methoxyamino)ethyl)thio)ethane-1 -thiol (43)

To a solution of compound 69 (232 mg, 1 .7 mmol) in EtOH (2.8 mL) was added methoxyamine hydrochloride (171 mg, 2.0 mmol, 1 .2 equiv) and AcONa (279 mg, 3.4 mmol, 2.0 equiv). The reaction mixture was stirred overnight at rt. After that time, NaBH 3 CN (160 mg, 2.6 mmol, 1 .5 equiv) was added followed by dropwise addition of a freshly prepared solution of 1 M ethanolic HCI (5.6 mL, freshly prepared from AcCI and EtOH). After stirring for 1 h at rt, the reaction was neutralized by addition of satd aq NaHC0 3 . The reaction mixture was diluted with H 2 0 and extracted with DCM (3 x). The organic phases were pooled, washed with brine and dried over anhyd Na 2 S0 4 . The suspension was filtrated and concentrated under reduced pressure. Purification by flash chromatography eluting with Tol/Acetone (85:15) yielded the aminoalcohol 43 (49 mg, 0.29 mmol, 17%) as a colourless oil.

1 H-NMR (500 MHz, CDCI 3 ): δ 5.90 (s, 1 H), 3.54 (s, 3H), 3.08 (t, 2H), 2.78-2.68 (m, 6H), 1 .72 (t, 1 H). Scheme 15: Synthesis of Iinker5 72

Reagents and conditions: a) i. 71 ; ii. MeONH 2 HCI, AcONa, EtOH; iii. NaBH 3 CN, AcCI, EtOH, 29%

3-(3-(Methoxyamino)propylthio)propane-1 -thiol (72): Acrolein 70 (0.20 ml_, 3.0 mmol) was added dropwise to 1 ,2-ethanedithiol 71 (1 .3 ml_, 15.0 mmol, 5.0 equiv) and the reaction mixture was stirred for 3 h at rt. After that time, the reaction mixture was diluted with EtOH (5.0 mL) and methoxyamine hydrochloride (300 mg, 3.6 mmol) and NaOAc (492 mg, 6.0 mmol) were added and the reaction mixture was stirred overnight at rt. After that time, NaBH 3 CN (282 mg, 4.5 mmol, 1 .5 equiv) was added to the reaction mixture, followed by dropwise addition of 1 M ethanolic HCI (10 mL, freshly prepared from AcCI and EtOH). After stirring for 1 h at rt, the reaction was neutralized by addition of satd aq NaHC0 3 . The reaction mixture was diluted with H 2 0 and extracted with DCM (3 x). The organic phases were pooled, washed with brine and dried over anhyd Na 2 S0 4 . The suspension was filtrated and concentrated under reduced pressure. Purification by flash chromatography eluting with Tol/Acetone (8:2) yielded the aminoalcohol 72 (159 mg, 0.88 mmol, 29%) as a colourless oil.

1 H-NMR (500 MHz, CDCI 3 ): δ 5.60 (s, 1 H), 3.53 (s, 3H), 3.01 (t, 2H), 2.76 (m, 2H), 2.73 (m, 2H), 2.62 (t, 2H), 1 .82 (m, 2H), 1.72 (dd, 1 H).

Scheme 16: Synthesis of the GM4 mimetic conjugate 78

77 [G 4m-N(OMe)-(CH 2 ) 3 -S-(CH 2 ) 2 -SH]

78

Reagents and conditions: a) i. Bu 2 SnO, MeOH; ii. 74, CsF, DME, 23%; b) Pd(OH) 2 , H 2 , THF/H 2 0, 40%; c) 72, AcOH/AcOH buffer, EtOH, 40°C, 64%; d) i. 5, DBU, DMF/H 2 0; ii. thioglycerol, Et 3 N, 67%

Benzyl 2,6-di-0-benzyl-3-0-((1 S)-1 -benzyloxycarbonyl-2-cvclohexyl-ethyl)-p-D- galactopyranoside (75):

Diol 73 (100 mg, 0.22 mmol) was dissolved in anhyd MeOH (5 ml_). Bu 2 SnO (58 mg, 0.23 mmol, 1 .05 equiv) was added and the reaction mixture was refluxed at 80°C for 4 h. After that time, the solvent was evaporated under reduced pressure and the crude residue was dried under high vacuum for 5 h. The crude residue was disolved under Ar in anhyd 1 ,2-dimethoxyethane (DME, 2.5 ml_). Anhyd CsF (67 mg, 0.44 mmol, 2.0 equiv) and triflate 74 (175 mg, 0.44 mmol, 2.0 equiv) were added. After overnight stirring at rt under Ar, the solvent was evaporated under reduced pressure. Flash chromatography (Tol/EtOAc 8:2) yielded alcohol 75 (35 mg, 50 μηιοΙ, 23%). 1 H NMR (500 MHz, CDCI 3 ) δ 7.36-7.23 (m, 20H), 5.19 (d, 1 H), 5.10 (d, 1 H), 4.95 (d, 1 H), 4.94 (d, 1 H), 4.67 (d, 1 H), 4.64 (d, 1 H), 4.63 (d, 1 H), 4.58 (1 d, 1 H), 4.44 (d, 1 H), 4.16 (dd, 1 H), 3.79 (m, 1 H), 3.77 (d, 2H), 3.73 (dd, 1 H), 3.53 (m, 1 H), 3.39 (t, 1 H), 3.30 (dd, 1 H), 1 .70-1 .45 (m, 8H), 1 .06-0.97 (m, 3H), 0.86-0.76 (m, 2H).

MS (ESI + ): m/z 717.60 (calc for C 4 3H 5 o0 8 Na + [M+Na] + : m/z 717.34). 3-0-((1 S)-1 -carboxy-2-cvclohexyl-ethyl)-a,p-D-galactopyranose (76):

To a degazed solution of benzyl 75 (25 mg, 40 μηηοΙ) in THF/H 2 0 (4:1 , 1 .0 mL) was added under Ar Pd(OH) 2 /C (10 mg). The reaction mixture was stirred overnight under an H 2 atmosphere. After that time, the reaction mixture was filtered over a PTFE Acrodisc 0.45 μπΊ membrane and concentrated under reduced pressure. Reverse phase chromatography eluting with MeOH in H 2 0 (0→ 100%) gave the corresponding uronate 76 (8.5 mg, 25 μηιοΙ, 63%) as a white fluffy solid.

The a-anomer had: 1 H-NMR (500 MHz, D 2 0) δ 5.17 (d, 1 H), 4.20 (dd, 1 H), 4.01 -3.97 (m, 2H), 3.83 (dd, 1 H), 3.69-3.62 (m, 2H), 3.61 -3.58 (m, 1 H), 1.74-1 .51 (m, 8H), 1.19-1 .07 (m, 3H), 0.93-0.81 (m, 2H). The β-anomer had: 1 H-NMR (500 MHz, D 2 0) δ 4.50 (d, 1 H), 4.18 (dd, 1 H), 3.94 (d, 1 H), 3.69-3.62 (m, 2H), 3.61 -3.58 (m, 1 H), 3.51 (dd, 1 H), 3.40 (dd, 1 H), 1 .74-1.51 (m, 8H), 1 .19-1 .07 (m, 3H), 0.93-0.81 (m, 2H).

MS (ESI + ): m/z 357.32 (calc for Ci 5 H 26 0 8 Na + [M+Na] + : m/z 357.16).

/V-(0-Methyl-/V-r2-(2-ethylthio)propylthiolhvdroxylamino) -3-0-((1 S)-1 -carboxy-2- cvclohexyl-ethvD-p-D-galactopyranoside (77):

To a solution of hemiacetal 76 (8.8 mg, 26 μιτιοΙ) in NaOAc/AcOH buffer (2 M, pH 4.5, 260 μΙ_) was added oxyamine 72 (25 mg, 138 μηιοΙ, 5.2 equiv) and EtOH (520 μΙ_). The reaction mixture was stirred for 48-72 h at 25-40°C. Purification by P2 size-exclusion chromatography followed by reverse phase chromatography (0%→ 100% MeOH in H 2 0) gave compound 77 (8.3 mg, 16.7 μηιοΙ, 64%) as a white fluffy solid. 1 H-NMR (500 MHz, D 2 0) δ 4.17 (d, 1 H), 4.00 (dd, 1 H), 3.94 (dd, 1 H), 3.88 (t, 1 H), 3.80 (dd, 1 H), 3.74 (dd, 1 H), 3.66-3.63 (m, 4H), 3.46 (dd, 1 H), 3.15 (m, 1 H), 3.05-2.96 (m, 2H), 2.82-2.69 (m, 5H), 1.95-1 .88 (m, 2H), 1 .81 (m, 1 H), 1.73-1 .54 (m, 7H), 1 .21 (m, 3H), 1.02- 0.90 (m, 2H).

MS (ESI " ): m/z 496.37 (calc for CziHssOsNSz " [M-H]-: m/z 496.20). GM4 mimetic polymer (78):

To a solution of 5 (2.38 mg, 1 1 .6 μηιοΙ) in DMF (1 16 μΙ_) were subsequently added compound 77 (1 .60 mg, 3.22 μηιοΙ, 0.28 equiv), water (10 μΙ_) and a solution of DBU (2.6 μΙ_, 17.4 μηιοΙ, 1 .5 equiv) in DMF (24 μΙ_). After stirring for 1 -3 h at rt, thioglycerol (3.0 μΙ_, 34.8 μηηοΙ, 3.0 equiv) and Et 3 N (4.9 μΙ_, 34.8 μηηοΙ, 3.0 equiv) were added. The reaction mixture was stirred at rt for another 12-24 h. The product was precipitated by slow addition to a stirring solution of EtOH/Et 2 0 (1 :1 , 1 ml_). The precipitate was filtered off, washed with EtOH and dried. Further purification was achieved by ultrafiltration (Sartorius Stedim Vivaspin tubes, 6 ml_, molecular weight cutoff 10 kDa, 5500 rpm). Freeze-drying gave the GM4 mimetic polymer 78 (3.84 mg, 67%) as a white solid. According to 1 H NMR, the product contained approximately 56% of the lysine side-chains substituted by the carbohydrate epitope 77.

Scheme 17: Synthesis of linker-equipped HNK-1 disaccharide 58

85

58 [(HNK-1 )-N(OMe)-(CH2)3-S-(CH 2 )2-SH]

Reagents and conditions: a) BzCN, DMAP, 4 A MS, DCM, -78°C, 74%; b) 80, NIS, TfOH, DCM, -20°C, 57%; c) NaOAc, MeOH, 64%; d) S0 3 Pyr, DMF, 82%; e) LiOH, THF/H 2 0, 83%; f) Pd(OH) 2 /C, H 2 , H 2 0/MeOH, quant; g) 72, AcOH/AcOH buffer, EtOH, 40°C, 33%;

Benzyl 4-0-benzoyl-2,6-di-0-benzyl-p-D-galactopyranosyl (79):

A solution of diol 73 (285 mg, 0.634 mmol) in anhyd DCM (26 mL) was stirred over freshly activated 4 A MS for 30 min at rt under an Ar atmosphere. The mixture was cooled to -78°C and BzCN (87 mg, 0.665 mmol, 1.05 equiv) and DMAP (7.7 mg, 63 μηιοΙ, 0.1 equiv) were added. The reaction mixture was stirred for 4 h at -78°C under an Ar atmosphere. The reaction was quenched with MeOH and the resulting suspension was filtrated. The filtrate was washed with 10% aq NaHC0 3 and brine and dried over anhyd Na 2 S0 4 . The solution was filtrated and concentrated under reduced pressure. Purification by flash chromatography eluting with toluene/EtOAc (95:5→ 9:1 ) yielded the alcohol 79 (260 mg, 0.469 mmol, 74%) as a white foam.

1 H-NMR (500 MHz, CDCI 3 ) δ 8.12-7.10 (m, 20H), 5.65 (d, 1 H), 5.02 (d, 1 H), 5.00 (d, 1 H), 4.71 (d, 1 H), 4.69 (d, 1 H), 4.57 (d, 1 H), 4.53 (d, 1 H), 4.46 (d, 1 H), 3.90-3.86 (m, 1 H), 3.87- 3.84 (m, 1 H), 3.69-3.65 (dd, 1 H), 3.66-3.61 (m, 2H), 2.42 (d, 1 H). MS (ESI + ): m/z 577.23 (calc for C 34 H 34 0 7 Na + [M+Na] + : m/z 577.22).

Benzyl 2,4-di-Q-acetyl-1 -thio-p-D-glucopyranosidurono-3,6-lactone-(1→3)-4-Q-benzoy l- 2,6-di-Q-benzyl-p-D-galactopyranoside (81 ):

To a solution of acceptor 79 (100 mg, 0.180 mmol) and donor 80 (127 mg, 0.360 mmol, 2.0 equiv) in anhyd DCM (1 .0 mL) was added NIS (164 mg, 0.728 mmol, 2.4 equiv). The reaction mixture was cooled to -20°C and TfOH (1 .6 μΙ_, 0.018 mmol, 0.1 equiv) was added. The reaction mixture was stirred for 1 h at -20°C. The reaction mixture was neutralized with Et 3 N, diluted with DCM, washed with 10% aq Na 2 S 2 0 3 and brine and dried over anhyd Na 2 S0 4 . The suspension was filtrated and concentrated under reduced pressure. Purification by flash chromatography eluting with toluene/EtOAc (85:15→ 8:2) yielded the disaccharide 81 (82 mg, 0.103 mmol, 57%) as a white foam.

1 H-NMR (500 MHz, CDCI 3 ) δ 8.09-8.06, 7.56, 7.46, 7.37-7.22 (m, 20H), 5.54 (d, 1 H), 5.37 (s, 1 H), 5.07 (d, 1 H), 5.03 (d, 1 H), 4.99 (d, 1 H), 4.92 (t, 1 H), 4.77 (t, 1 H), 4.70 (d, 1 H), 4.68 (d, 1 H), 4.51 (d, 1 H), 4.54 (d, 1 H), 4.56 (d, 1 H), 4.19 (d, 1 H), 4.03 (dd, 1 H), 3.92 (dd, 1 H), 3.84 (dd, 1 H), 3.75 (dd, 1 H), 3.58 (dd, 1 H), 2.1 1 , 1 .82 (2s, 6H) MS (ESI + ): m/z 819.39 (calc for C 44 H 44 0i 4 Na + [M+Na] + : m/z 819.26).

Benzyl (methyl 2,4-di-0-acetyl-p-D-glucopyranuronate)-(1→3)-4-0-benzoyl-2 ,6-di-0- benzyl-p-D-galactopyranoside (82):

Lactone 81 (109 mg, 138 μηιοΙ) was dissolved at 0°C in anhyd DCM/MeOH (1 :4, 2.5 mL). Anhyd NaOAc (10 mg, 124 μηηοΙ, 0.9 equiv) was added and the reaction mixture was stirred overnight at 4°C. After that time, the mixture was neutralised by addition of Amberlyst H + resin. The suspension was filtered and the filtrate concentrated under reduced pressure. Flash chromatography using PE/Acet (75:25→ 7:3) gave the desired alcohol 82 (73 g, 88.2 μηιοΙ, 64%) as a white foam.

1 H-NMR (500 MHz, CDCI 3 ) δ 8.07-8.03, 7.60-7.55, 7.45, 7.40-7.22 (m, 20H), 5.67 (d, 1 H), 5.13 (dd, 1 H), 5.00 (d, 1 H), 4.98 (d, 1 H), 4.97 (d, 1 H), 4.74 (dd, 1 H), 4.71 (d, 1 H), 4.64 (d, 1 H), 4.56 (d, 1 H), 4.52 (d, 1 H), 4.48 (d, 1 H), 4.05 (dd, 1 H), 3.85 (m, 1 H), 3.84 (d, 1 H), 3.82 (dd, 1 H), 3.69 (s, 3H), 3.65-3.62 (m, 2H), 3.59 (m, 1 H), 2.70 (d, 1 H), 2.06 (s, 3H), 1 .79 (s, 3H).

MS (ESI + ): m/z 851 .47 (calc for C 45 H 48 0i 5 Na + [M+Na] + : m/z 851 .29).

Benzyl (methyl 2,4-di-0-acetyl-3-0-sulfo-p-D-glucopyranuronate)-(1 ^3)-4-0-benzoyl -2,6- di-O-benzyl-p-D-galactopyranoside (83):

To a solution of alcohol 82 (72 mg, 87 μηηοΙ) in anhyd DMF (0.4 mL) was added S0 3 -Py (41 mg, 26 mmol, 3.0 equiv) at 0°C under Ar. After stirring for 2 h at rt, the reaction mixture was quenched by addition of NaHC0 3 (146 mg) and the reaction mixture was stirred for 30 min at rt. The suspension was filtered, the filtrate concentrated and the solvents were co-evaporated with xylene. Flash chromatography using DCM/MeOH (95:5 → 9:1 ) gave the sulfate 83 (67 mg, 72 μηιοΙ, 82%).

1 H-NMR (500 MHz, CDCI 3 ) δ 7.98, 7.52-7.45, 7.40-7.20 (m, 20H), 5.61 (d, 1 H), 5.19 (t, 1 H), 4.99 (d, 1 H), 4.98 (d, 1 H), 4.92 (d, 1 H), 4.83 (t, 1 H), 4.69 (d, 1 H), 4.63 (d, 1 H), 4.55 (d, 1 H), 4.50 (d, 1 H), 4.46 (m, 1 H), 4.45 (d, 1 H), 3.98 (dd, 1 H), 3.88 (d, 1 H), 3.83 (t, 1 H), 3.79 (dd, 1 H), 3.65 (s, 3H), 3.63-3.52 (m, 2H), 1 .95 (s, 3H), 1 .75 (s, 3H).

MS (ESI-): /z 907.45 (calc for C 45 H 47 0i 8 S " [M-Na] " : m/z 907.25).

Benzyl (sodium 3-0-sulfo-p-D-glucopyranuronate)-(1→3)-2,6-di-Q-benzyl-p-D - galactopyranoside (84):

Acetate 83 (70 mg, 75 μηιοΙ) was dissolved in a solution of THF/H 2 0 (10:1 , 1 .8 mL). The reaction mixture was cooled to 0°C and a 2.0 M aq LiOH solution (0.4 mL, 1 15 mmol, 9.5 equiv) was slowly added. The reaction mixture was stirred overnight and allowed to slowly reach rt. The next morning, the reaction was neutralised by addition of Amberlyst H + resin. The reaction mixture was filtered and concentrated under reduced pressure. Reverse phase chromatography eluting with MeOH in H 2 0 (0% → 50%) gave the corresponding uronate 84 (47 mg, 62 μηηοΙ, 83%) as a white foam. 1 H-NMR (500 MHz, MeOD) δ 7.43-7.20 (m, 15H), 4.92 (d, 1 H), 4.78 (d, 1 H), 4.77 (d, 1 H), 4.67 (d, 1 H), 4.62 (d, 1 H), 4.51 (d, 1 H), 4.59 (d, 1 H), 4.32 (t, 1 H), 4.12 (d, 1 H), 3.83 (dd, 1 H), 3.79-3.71 (m, 4H), 3.68-3.64 (m, 2H), 3.58 (dd, 1 H).

MS (ESI-): /z 705.43 (calc for Css^C^S " [M-2Na+H] " : m/z 705.19). Sodium 3-0-sulfo-p-D-glucopyranuronate-(1→3)-a,p-D-galactopyranos e (85):

To a solution of benzyl 84 (46 mg, 61 μηηοΙ) in H 2 0/MeOH (10:1 , 5.2 mL) was added Pd(OH) 2 /C (20 mg). The reaction mixture was stirred under an H 2 atmosphere for 6 h. After that time, the reaction mixture was filtered over a PTFE Acrodisc 0.45 μηη membrane and concentrated under reduced pressure. Reverse phase chromatography eluting with H 2 0 gave the corresponding uronate 85 (29 mg, 60 μηηοΙ, quant) as a white fluffy solid.

The a-anomer had: 1 H-NMR (500 MHz, D 2 0) δ 5.30 (d, 1 H), 4.78 (d, 1 H), 4.35 (t, 1 H), 4.26 (dd, 1 H), 4.12 (ddd, 1 H), 4.00 (m, 2H), 3.82 (m, 1 H), 3.77-3.70 (m, 3H), 3.62 (dd, 1 H).

The β-anomer had: 1 H NMR (500 MHz, D 2 0) δ 4.78 (d, 1 H), 4.65 (d, 1 H), 4.35 (t, 1 H), 4.20 (d, 1 H), 3.82 (dd, 1 H), 3.77-3.70 (m, 5H), 3.66 (dd, 1 H), 3.62 (dd, 1 H).

MS (ESI " ): m/z 434.96 (calc for Ci 2 H 19 0i 5 S " [M-2Na+H] " : m/z 435.05).

/V-(0-Methyl-/V-r2-r(2-ethylthio)propylthiolhvdroxylamine )-(sodium 3-Q-sulfo-p-D- glucopyranuronate)-(1→3)-p-D-galactopyranoside (58)

To a solution of hemiacetal 85 (1 1.3 mg, 23.5 μηιοΙ) in NaOAc/AcOH buffer (2 M, pH 4.5, 235 μΙ_) was added oxyamine 72 (21 mg, 1 17 μηιοΙ, 10 equiv) and EtOH (450 μΙ_). The reaction mixture was stirred for 24-48 h at 25-40°C. Purification by P2 size-exclusion chromatography followed by reverse phase chromatography (100% H 2 0) gave compound 58 (5.07 mg, 7.88 μηιοΙ, 33%) as a white fluffy solid.

1 H-NMR (500 MHz, D 2 0) δ 4.80 (d, 1 H), 4.36 (t, 1 H), 4.24-4.17 (m, 2H), 3.93 (dd, 1 H), 3.84 (dd, 1 H), 3.82 (d, 1 H), 3.80-3.74 (m, 2H), 3.73 (dd, 1 H), 3.67 (m, 1 H), 3.65 (s, 3H), 3.64 (dd, 1 H), 3.18 (m, 1 H), 3.03-2.96 (m, 5H), 2.74 (t, 2H), 1.94 (t, 2H).

MS (ESI " ): m/z 598.19 (calc for Ci 8 H 32 0i 5 S 3 " [M-2Na+H] " : m/z 598.04). Scheme 18: Synthesis of HNK-1 -linear polylysine glycoconjugate 86

Reagents and conditions: a) i. 5, DBU, DMF/H 2 0; ii. thioglycerol, Et 3 N, 87% HNK-1 polymer (86): To a solution of 5 (3.59 mg, 17.5 μηιοΙ) in DMF (175 μΙ_) were subsequently added compound 58 (5.07 mg, 7.88 μηιοΙ, 0.45 equiv), water (28 μΙ_) and a solution of DBU (3.9 μΙ_, 26 μηιοΙ, 1 .5 equiv) in DMF (36 μΙ_). After stirring for 1 -3 h at rt, thioglycerol (4.5 μΙ_, 53 μηιοΙ, 3.0 equiv) and Et 3 N (7.3 μΙ_, 53 μηιοΙ, 3.0 equiv) were added. The reaction mixture was stirred at rt for another 12-24 h. The product was precipitated by slow addition to a stirring solution of EtOH/Et 2 0 (1 :1 , 1 ml_). The precipitate was filtered off, washed with EtOH and dried. Further purification was achieved by ultrafiltration (Sartorius Stedim Vivaspin tubes, 6 ml_, molecular weight cutoff 10 kDa, 5500 rpm). Freeze-drying gave HNK-1 polymer 86 (7.4 mg, 87%) as a white solid. According to 1 H NMR, the product contained approximately 40% of the lysine side-chains substituted by the carbohydrate epitope 58. Scheme 19: Synthesis of HNK-1 -polylysine dendrimer glycocon jugate 86

87 88

89

Reagents and conditions: a) (CIAc) 2 0, DMF/2,6-lutidine, 89%; b) i. 58, DBU, DMF/H 2 0; thioglycerol, Et 3 N, 58% Chloroacetylated dendrimer (88):

Poly-L-lysine dendrimer (generation 6, 64 outer amine groups, TFA salt, 10 mg, 41.8 μηιοΙ) was dissolved under Ar in anhyd DMF/2,6-lutidine (4:1 , 130 μΙ_) The solution was cooled to 0°C and a solution of (CIAc) 2 0 (7.0 mg, 52 μηηοΙ, 1.25 equiv) in anhyd DMF (17 μΙ_) was added dropwise. The reaction mixture was stirred overnight at 4°C. After that time, the dendrimer was precipitated by slow addition of the reaction mixture to a stirring solution of Et 2 0/EtOH (1 :1 , 2 ml_). The precipitate was filtered off, washed with Et 2 0/EtOH (1 :1 ) and dried to obtain chloroacetylated dendrimer 88 (7.2 mg, 89%) as an off-white solid.

1 H NMR (500 MHz, DMSO) δ 8.27 (s, 1 H), 8.17 (s, 1 H), 7.99 (s, 1 H), 7.81 (s, 1 H), 4.26 (s, 1 H), 4.19 (s, 1 H), 4.10 (s, 2H), 4.02 (s, 2H), 3.08-2.96 (s, 4H), 1 .66-1.17 (m, 12H).

HNK-1/dentrimeric polylysine conjugate (89):

To a solution of 88 (2.12 mg, 10.9 μηηοΙ) in DMF (109 μΙ_) were subsequently added compound 58 (3.5 mg, 5.44 μηιοΙ, 0.5 equiv), water (20 μΙ_) and a solution of DBU (2.4 μΙ_, 16 μηιοΙ, 1 .5 equiv) in DMF (22 μΙ_). After stirring for 1 -3 h at rt, thioglycerol (2.8 μΙ_, 33 μηιοΙ, 3.0 equiv) and Et 3 N (4.6 μΙ_, 33 μηιοΙ, 3.0 equiv) were added. The reaction mixture was stirred at rt for another 12-24 h. The product was precipitated by slow addition to a stirring solution of EtOH/Et 2 0 (1 :1 , 2 ml_). The precipitate was filtered off, washed with EtOH and dried. Further purification was achieved by ultrafiltration (Sartorius Stedim Vivaspin tubes, 6 ml_, molecular weight cutoff 10 kDa, 5500 rpm). Freeze-drying gave HNK-1/dentrimeric polylysine conjugate 89 (3.54 mg, 58%) as a white solid. According to 1 H NMR, the product contained approximately 48% of the lysine side-chains substituted by the carbohydrate epitope 58.

Scheme 20: Synthesis of HNK-1 -ornithine conjugate 93

93

Reagents and conditions: a) i. Resin OH " , H 2 0; ii. 10% aq PTSA, 91 %; b) (CIAc) 2 0, DMF/2,6-lutidine, 63%; c) 58, DBU, then thioglycerol, Et 3 N, DMF/H 2 0, 61 %

Tosylate salt of poly-L-ornithine (91 ):

Poly-L-ornithine hydrobromide (25 mg dissolved in 0.25 ml water) was passed through an anion exchange column, (Ambersep 900 hydroxide form, 5 x 0.5 cm). The effluent solution was neutralized with 10% aq p-toluenesulfonic acid (PTSA). Lyophilisation gave the tosylate salt of poly-L-ornithine (33.5 mg, 91 %) as a white fluffy solid. 1 H NMR (500 MHz, D 2 0) δ 7.86 (s, 1 H), 7.48 (t, 2H), 7.1 1 (t, 2H), 4.15 (s, 1 H), 2.76 (s, 2H), 1 .75-1 .46 (m, 4H).

Chloroacetylated poly-L-ornithine (92):

The tosylate salt of poly-L-ornithine (33 mg, 1 16 μηηοΙ) was dissolved under Ar in anhyd DMF/2,6-lutidine (4:1 , 360 μΙ_) The solution was cooled to 0°C and a solution of (CIAc) 2 0 (25 mg, 145 μηηοΙ, 1.25 equiv) in DMF (48 μΙ_) was added dropwise. The reaction mixture was stirred overnight at 4°C. After that time, the polymer was precipitated by slow addition of the reaction mixture to a stirring solution of Et 2 0/EtOH (1 :1 , 4 ml_). The precipitate was filtered off, washed with Et 2 0/EtOH (1 :1 ) and dried to obtain chloroacetylated poly-L- ornitine 92 (14 mg, 73 μηιοΙ, 63%) as an off-white solid.

1 H NMR (500 MHz, DMSO) δ 8.24 (s, 1 H), 4.04 (s, 2H), 3.88 (m, 1 H), 3.13 (s, 2H), 2-04- 1 .38 (m, 6H).

HNK-1 polyornithine conjugate (93):

To a solution of 92 (2.6 mg, 13.7 μηηοΙ) in DMF (137 μΙ_) were subsequently added compound 58 (4.0 mg, 6.17 μηιοΙ, 0.45 equiv), water (24 μΙ_) and a solution of DBU (3.1 μΙ_, 21 μηιοΙ, 1 .5 equiv) in DMF (28 μΙ_). After stirring for 1 -3 h at rt, thioglycerol (3.6 μΙ_, 41 μηιοΙ, 3.0 equiv) and Et 3 N (5.7 μΙ_, 41 μηιοΙ, 3.0 equiv) were added. The reaction mixture was stirred at rt for another 12-24 h. The product was precipitated by slow addition to a stirring solution of EtOH/Et 2 0 (1 :1 , 2 ml_). The precipitate was filtered off, washed with EtOH and dried. Further purification was achieved by ultrafiltration (Sartorius Stedim Vivaspin tubes, 6 ml_, molecular weight cutoff 10 kDa, 5500 rpm). Freeze-drying gave HNK-1/polyornithine conjugate 93 (4.9 mg, 61 %) as a white solid. According to 1 H NMR, the product contained approximately 60% of the ornithine side- chains substituted by the carbohydrate epitope 58. Scheme 21 : Synthesis of lactose-linker5 conjugate 56 and conjugate 97

Reagents and conditions: a) 72, AcOH/AcOH buffer, EtOH, 40°C, 78%; b) 95, Cs 2 C0 3 , DMF, 75%; c) 20% piperidine in DMF, 64% /V-(0-Methyl /V-[2-[(2-ethylthiolpropylthiolhvdroxylamine)-p-D-galactopyr anosyl-(1→'4)-p-D- glucopyranoside (56)

To a solution of hemiacetal 94 (107 mg, 0.297 mmol) in NaOAc/AcOH buffer (2 M, pH 4.5, 1 .5 mL) was added oxyamine 72 (270 mg, 1.50 mmol, 5.0 equiv) and EtOH (3.0 ml_). The reaction mixture was stirred for 24-48 h at 25-40°C. After that time, the solvents were evaporated under reduced pressure. The crude residue was suspended under Ar in H 2 0 (5.0 mL). DL-Dithiothreitol (460 mg, 2.98 mmol, 10 equiv) was added to the reaction mixture followed by 1 M aq NaOH (until pH consistently 9). After stirring for 2 h at rt, the reaction mixture was directly loaded onto the C18 column. Purification by reverse phase chromatography (0→ 100% MeOH in H 2 0) gave compound 56 (1 17 mg, 0.231 mmol, 78%) as a white fluffy solid. 1 H-NMR (500 MHz, D 2 0) δ 4.47 (d, 1H), 4.23 (d, 1H), 3.98 (dd, 1H), 3.95 (d, 1H), 3.86- 3.77 (m, 3H), 3.75 (m, 1H), 3.71-3.60 (m, 4H), 3.65 (s, 3H), 3.56 (dd, 1H), 3.55 (m, 1H), 3.19 (m, 1H), 3.01 (m, 1H), 2.83 (m, 2H), 2.79 (m, 2H), 2.72 (m, 2H), 1.92 (m, 2H).

MS (ESI + ): m/z 528.29 (calcfor Ci 8 H 35 0iiNS 2 Na + [M+Na] + : m/z 528.15). /V-(0-Methyl Λ/-Γ2-Γ(2-Γ2- fluorenylmethyloxycarbamate)ethyl1e^

galactopyranosyl-d→4)-p-D-glucopyranoside (96)

Thiol 56 (28 mg, 49 μηηοΙ) was dissolved in anhyd DMF (1.0 mL). The solution was degazed then flushed with Ar. Bromide 95 (56 mg, 0.163 mmol, 3.3 equiv) and Cs 2 C0 3 (32 mg, 99 μηηοΙ, 2.0 equiv) were added to the reaction mixture. After stirring for 2 h at rt under Ar, the reaction mixture was directly loaded onto the C18 column. Reverse phase chromatography eluting with MeCN in H 2 0 (0%→ 95%) gave the corresponding Fmoc- protected amine 96 (28 mg, 37 μηηοΙ, 75%) as a white foam.

1 H NMR (500 MHz, MeOD) δ 7.81-7.77 (m, 2H), 7.73-7.65 (m, 2H), 7.41-7.36 (m, 2H), 7.33-7.29 (m, 2H), 4.36 (d, 1H), 4.05 (d, 1H), 3.86-3.83 (m, 2H), 3.81 (d, 1H), 3.78 (dd, 1H), 3.70 (dd, 1H), 3.63 (d, 1H), 3.61 (s, 3H), 3.58 (m, 1H), 3.57-3.50 (m, 5H), 3.48 (dd, 1H), 3.33 (m, 1H), 3.12 (m, 1H), 3.00-2.99 (m, 2H), 2.94 (dt, 1H), 2.75-2.62 (m, 6H), 1.89- 1.80 (m, 1H), 1.72-1.63 (m, 1H).

MS (ESI + ): m/z 793.41 (calc for C 35 H 5 oOi 3 N 2 S 2 Na + [M+Na] + : m/z 793.26). /V-(0-Methyl /V-r2-r2-r2-aminoethyllethylthiolpropylthiolhvdroxylamine)-B -D- galactopyranosyl-d→4)-p-D-glucopyranoside (97)

Derivative 96 (28 mg, 37 μηηοΙ) was dissolved under Ar in anhyd DMF (1.0 mL). Piperidine (0.2 mL) was added to the solution under Ar. After stirring for 4 h at rt under Ar, the solvents were coevaporated with toluene (3x). Reverse phase chromatography eluting with MeOH in 0.1% aq TFA (0%→60%) gave the corresponding amine 97 (13 mg, 23.7 μηηοΙ, 64%) as a white foam.

1 H NMR (500 MHz, D 2 0) δ 4.47 (d, 1H), 4.23 (d, 1H), 3.99 (dd, 1H), 3.95 (d, 1H), 3.85- 3.76 (m, 3H), 3.75 (m, 1H), 3.71-3.59 (m, 4H), 3.64 (s, 3H), 3.56 (dd, 1H), 3.55 (m, 1H), 3.26 (t, 2H), 3.18 (m, 1H), 3.01 (m, 1H), 2.92 (t, 2H), 2.88-2.86 (m, 5H), 2.73 (t, 1H), 1.95- 1.89 (m,2H). MS (ESI + ): m/z 549.31 (calc for C 2 oH4iOii N 2 S2 + [M+H] + : m/z 549.21 ).

Scheme 22: Synthesis of lactose-chitosan conjugate 100

100

Reagents and conditions: a) 56, DBU, DMF/H 2 0, 58%; b) 0.1 M aq NaOH, 40°C, 59% Chitosan derivative (99)

To a solution of chloracetylated chitosan 98 (5.0 mg, 12.2 μηηοΙ) in DMF (0.4 mL) were subsequently added compound 56 (24.0 mg, 39.1 μηιοΙ, 3.2 equiv) and DBU (7.0 μΙ_, 48.8 μηηοΙ, 4.0 equiv). After stirring for 2 h at rt, the reaction mixture was heated at 50°C for 1 h. After that time, H 2 0 (20 μΙ_) was added and the reaction mixture was stirred at 50°C for another 1 h. The product was precipitated by slow addition to a stirring solution of EtOH/Et 2 0 (1 :1 , 4 mL). The precipitate was filtered off, washed with EtOH and dried to obtain chitosan conjugate 99 (12.8 mg, 58%) as a white solid.

IR (KBr) v 3400 (vs, b, OH), 2926, 2067, 1734 (CO es ter), 1651 (CO amide ), 1419, 1382, 1274, 1207, 1 1 19, 1076, 1034, 894, 784, 702, 622, 600 Lactose-chitosan conjugate (100)

Chitosan derivative 99 (16 mg, 8.9 μηηοΙ) was suspended in 0.1 M aq NaOH (0.32 mL). The suspension was stirred at 40°C for 90 min. The solid was filtered off, washed with H 2 0, EtOH and Et 2 0 and dried to obtain lactose-chitosan conjugate 100 (3.7 mg, 59%) as a white solid. IR (KBr) v 3436 (vs, b, OH), 2921 , 1648 (CO amide ), 1553, 1377, 1075, 1034, 894

Scheme 23: Synthesis of lactose-polyglutamic acid conjugate 102

Reagents and conditions: a) Sulfo-NHS, EDC ' HCI, NaHC0 3 , phosphate buffer, 45% Lactose-polyglutamic acid conjugate (102)

To a solution of poly-L-glutamic acid sodium salt (from Alamanda Polymers, n = 250, 2.50 mg, 16.5 μηιοΙ) in phosphate buffer (100 mM, pH 5.0, 81 μΙ_) was added a solution of /V-hydroxysulfosuccinimide sodium salt (sulfo-NHS, 60 mg, 0.26 mmol, 15.6 equiv) and N- (3-dimethylaminopropyl)-/V'-ethylcarbodiimide hydrochloride (EDC ' HCI, 37 mg, 0.19 mmol, 1 1.5 equiv) in phosphate buffer (100 mM, pH 5.0, 417 μΙ_) was added After stirring for 15 min at rt, amine 97 (13 mg, 23.7 μηηοΙ, 1 .4 equiv) was added followed by addition of satd aq NaHC0 3 until the pH was consistenly 7. After stirring for 2 h at rt, ethanolamine was added to the reaction mixture to reach a final concentration of 10 mM. After stirring for 10 min at rt, the reaction mixture was transferred into an ultrafiltration tube (Sartorius Stedim Vivaspin tubes, 6 mL, molecular weight cutoff 10 kDa). Purification by ultrafiltration (5500 rpm) and freeze-drying gave lactose-polyglutamic acid conjugate 102 (4.9 mg, 45%) as a white fluffy solid. According to 1 H NMR, the product contained approximately 100% of the glutamic acid side-chains substituted by the carbohydrate epitope 56. Patient Sera

Sera from seven neuropathy patients were investigated. They all were tested positive for anti-ganglioside antibodies in the clinic. Serum anti-ganglioside antibody titers were determined by an ELISA assays from Bijhlmann Laboratories (Schonenbuch, Switzerland). Sera were either obtained from Bijhlmann Laboratories (Schonenbuch, Switzerland) or the clinical laboratory of the University Hospital Basel (Basel, Switzerland). Sera from individuals undergoing neuro-immunological evaluation with negative anti- ganglioside reactivity served as control. Use of sera for our study was approved by the ethics committee of northwestern and central Switzerland (EKNZ UBE-15/46). Competitive Binding Assay

The synthesized carbohydrate polymers 6 (GM1 a epitope), 26 (GD1 b epitope), and 34 (GT1 a epitope) were tested in the GanglioCombi(-Light) ELISA and/or, in case of compound 6, the anti-GM1 ELISA (all kits from Bijhlmann Laboratories, Schonenbuch, Switzerland). The 96 well microtiter plates coated with purified gangliosides from bovine cauda equina were washed two times with washing buffer (300 μΙ/well) before adding the carbohydrate polymers in eight different concentrations, 25 μΙ/well. The patient sera containing anti-ganglioside IgG or IgM antibodies were added in the appropriate dilutions, 25 μΙ/well (2x concentrated), to obtain a total of 50 μΙ volume per well. The plate was covered with a plate sealer and incubated for 2 h at 4-8°C. The wells were washed three times with wash buffer (300 μΙ/well) before either the anti-human IgM antibody- horseradish peroxidase conjugate or the anti-human IgG antibody-horseradish peroxidase conjugate was added (100 μΙ/well). The plate was incubated for 2 h at 4-8°C. After washing the wells (3 x 300 μΙ/well), a substrate solution of tetramethylbenzidin (TMB in citrate buffer with hydrogen peroxide) was added (100 μΙ/well) and the plate incubated for further 30 minutes at 600 rpm and room temperature, protected from light. Finally, a stop solution (0.25 M sulfuric acid) was added (100 μΙ/well) and the degree of colorimetric reaction was determined by absorption measurement at 450 nm with a microplate reader (Spectramax 190, Molecular Devices, California, USA).

The synthesized carbohydrate polymer 86 (HNK-1 epitope mimetic 58) was tested in the anti-MAG ELISA (kit from Bijhlmann Laboratories, Schonenbuch, Switzerland). The assay protocol was performed according to the one described above for the GanglioCombi(- Light) ELISA. To determine the in vitro IC 50 of polymer 86, the assay was performed with co-incubation of polymer (25 μΙ/well) and a mouse monoclonal anti-HNK-1 (anti-MAG) IgM antibody (25 μΙ/well) at a final dilution of 1 :1000. To determine the in vivo efficacy of polymer 86, the assay was performed by incubation of mouse plasma diluted 1 :100 (50 μΙ/well). Both, the mouse monoclonal anti-HNK-1 (anti-MAG) IgM and anti-HNK-1 (anti- MAG) IgM in plasma of immunized BALB/c mice (pre- and post-treatment) were detected with goat anti-mouse IgM HRP conjugate (Sigma Aldrich, A8786) diluted 1 :10Ό00.

Immunological mouse model for anti-MAG neuropathy

Six gender matched BALB/c wild type mice at the age of 6 weeks were injected subcutaneously at multiple sites on the lower back with a total of 100 μg of the glycosphingolipids SGPG and SGLPG purified from bovine cauda equina (both glycolipids contain the HNK-1 carbohydrate epitope). The isolation of glycolipids was performed according to a protocol described by Burger et al. (Journal of Immunological Methods 1991 , 140, 31 -36). These glycosphingolipids were taken up in PBS, mixed with KLH (1 .4 mg/ml final concentration) and emulsified with an equal volume of TiterMax® Gold. Two booster injections were performed after 2 and 4 weeks with 20 μg of purified SGPG/SGLPG mixed with KLH and TiterMax® Gold. Blood samples were taken by puncture of the tail vein and transferred to tubes containing 1 μΙ of 0.5 M EDTA and centrifuged 15 min at 1 '800 rpm. The supernatant (plasma) was transferred to new tubes and stored at -55 °C. The glycopolymer 86, dissolved in PBS, was administered by i.v. injection of the tail vein. Mouse plasma samples were analyzed by the above described anti-MAG ELISA.

As indicated, the synthesized carbohydrate polymers 6 (GM1 a epitope), 26 (GD1 b epitope), and 34 (GT1 a epitope) were tested in the GanglioCombi(-Light) ELISA and/or, in case of compound 6, the anti-GM1 ELISA (all kits from Bijhlmann Laboratories, Schonenbuch, Switzerland). These ELISAs are used to support the clinical diagnosis of immune-mediated neuropathies. The assays allow the determination of the anti- ganglioside IgM/lgG antibodies titer (e.g. gangliosides GM1 , GD1 a, and GQ1 b) in serum samples from patients. We used these ELISAs as competitive binding assays. The synthesized compounds and patient serum samples (containing anti-ganglioside antibodies) were given into 96 well plates, coated with purified gangliosides from bovine cauda equina. Immobilized gangliosides and the synthesized compounds competed for binding to the anti-ganglioside antibodies. After a washing step ganglioside-bound antibodies (IgM/lgG) were detected with horseradish peroxidase labeled anti-human IgM or anti-human IgG antibodies, followed by a colorimetric reaction. Successful competition of the compounds with gangliosides led to a decrease in measured OD 45 o nm (optical density), because they block the binding sites of anti-ganglioside antibodies, preventing their binding to immobilized gangliosides. The principle of the assay is depicted in FIG. 1 . For the evaluation of the compounds, sera from seven patients (anti-GM1 a: PP IgG Pos., P21 , P3, P4; anti-GD1 b: P22; anti-GQ1 b: EK-GCO 1803, P23), tested positive for anti- ganglioside reactivity during clinical laboratory routine analysis, were chosen. IgG and IgM antibody titers were determined for each serum in preliminary experiments. Serum dilutions with measured OD 45 o nm values around 1 .0 (0.7-1 .3) were chosen for the assay, to be able to compare the measured IC 50 values (half maximal inhibitory concentration) which are antibody concentration dependent. Serum dilutions: PP IgG Pos. 1 :1 '200, P21 1 :1 '300, P3 1 :50, P4 1 :400, P22 1 :50, EK-GCO 1803 V. 300, P23 1 :50). The sera that served as negative controls (dilution 1 :50) showed no antibody binding to gangliosides.

IC 5 o values of compound 6 were determined for sera PP IgG Pos. (IgG), P21 (IgG), P3 (IgM) and P4 (IgM). Compound 26 were evaluated with serum P22 (IgG). The IC 50 values of compound 34 were determined for sera EK-GCO 1803 (IgG) and P23 (IgG). The results are shown in the Table below. The inhibition curves are shown in FIG. 2.

Table: IC½ values of glvcopolymers 6, 26, and 34 tested with a total of seven neuropathy patient sera including standard deviations.

Serum Ganglioside Compound 6 Compound 26 Polymer 34

reactivity PL(GM1 a) 28 PL(GD1 b) 20 PL(GT1 a) 58

(antibody IC50 IC50 IC50 isotype)

PP IgG Pos. GM1 a

28.0 ± 13.5 μΜ

(igG)

P21 GM1 a

218.6 ± 77.8 nM

(igG)

P3 GM1 a

374.9 ± 157.0 nM

(IgM)

P4 GM1 a

59.4 ± 62.9 pM

(IgM)

P22 GD1 a

313.1 ± 1 12.3 μΜ

(igG)

EK-GCO GQ1 b/GT1 a

12.5 ± 4.1 μΜ 1803 (igG)

P23 GQ1 b/GT1 a

347.6 ± 92.0 μΜ

(igG) The inventive polymers 6, 26, 34 are glycopolymers that imitate the natural glycoepitopes of the GM1 a-, GD1 b-, and the GT1 a-gangliosides. These and other glycoepitopes are involved in autoimmune neurological diseases; they are targets for antibodies that trigger demyelination and neurodegeneration (H. J. Willison and N. Yuki, Brain, 2002, 125, 2591 - 2625). The prepared glycopolymers are based on a biodegradable poly-L-lysine backbone and are designed for a therapeutic application in patients, where pathogenic anti-glycan antibodies could be selectively neutralized and removed by these polymers.

For the biological evaluation of the prepared glycopolymers, patient sera were used. These sera have been tested positive in the clinic for anti-ganglioside antibodies. The synthetic glycopolymers were tested with sera presenting an antibody response against the ganglioside epitopes displayed by the conjugates (e.g. sera with anti-GM1 a IgG or IgM antibodies for the evaluation of the PL(GM1 a) 2 8 polymer 6). The IC 50 values obtained during the biological characterization in the competitive binding ELISA assay showed the different neutralization effects of the glycopolymers for anti-ganglioside antibodies from different patients with reactivity against the same glycoepitope. This is probably due to interindividual differences of antibody characteristics (isotype, affinity, specificity, serum concentration, monoclonal/polyclonal, etc.) between the different patients. However, the inhibitory effect of the glycopolymers is given for antibody reactivities against different gangliosides. Furthermore, the data on compound 6 shows that glycopolymers mimicking a specific glycoepitope can neutralize antibodies of different isotypes, e.g. antibodies of the IgG and/or the IgM type. It is also interesting to note, that partial glycoepitope structures can be sufficient to retain affinity to anti-ganglioside antibodies. This is the case for the competitive binding assay of GT1 a-glycoconjugate 34 with sera EK-GCO 1803 (IgG) and P23 (IgG), where the antibodies target the GQ1 b epitope (characteristic for e.g. Miller-Fischer syndrome and Bickerstaff brainstem encephalitis). Even though the GT1 a epitope, displayed by conjugate 34, lacks one sialic acid compared to the GQ1 b ganglioside, the patient sera directed against GQ1 b were neutralized by glycopolymer 34.

The IC 50 value of compound 86 was determined for the mouse monoclonal anti-HNK-1 IgM antibody. This antibody shows comparable reactivity with the HNK-1 glycoepitope as monoclonal anti-MAG IgM antibodies of anti-MAG neuropathy patients. The results are shown in the Table 2 below. The inhibition curve is shown in FIG. 2E. Table 2: ICsn value of glycopolymer 86, tested with the mouse monoclonal anti-HNK-1 IgM antibody including standard deviation.

The inventive polymer 86 is a glycopolymer that imitates the natural trisaccharide glycoepitope HNK-1 which is present in the peripheral nervous system as part of the glycosphingolipids SGPG and SGLPG but also the glycoprotein MAG. This HNK-1 glycoepitope is the target of an autoimmune attack in the neurological disorder anti-MAG neuropathy. The prepared glycopolymer is based on a biodegradable poly-L-lysine backbone of an average of 400 lysines, wherein 40% of the lysine side chains are loaded with the HNK-1 mimetic 58. The remaining 60% of side chains are caped with thioglycerole to improve the water solubility of the polymer. The polymer is designed for a therapeutic application in anti-MAG neuropathy patients (or patients with other neurological diseases with the same or similar antibodies), where pathogenic anti-HNK-1 (MAG/SGPG/SGLPG) antibodies could be selectively neutralized and removed by this polymer. Polymer 86 inhibits the binding of the mouse monoclonal anti-HNK-1 IgM to the HNK-1 epitope on MAG at nanomolar concentrations (Table 2). The therapeutic utility of polymer 86 is further supported by in vivo data (Fig. 3). The compound PL(HNK- 1 mimetic(58)) 4 o was administered intravenously to immunized BALB/c mice (n = 6) with induced high levels of anti-HNK-1 (anti-MAG) IgM antibodies. These mouse antibodies are a model for pathogenic human anti-HNK-1 (anti-MAG) IgM antibodies of anti-MAG neuropathy patients. A dose of 10mg/kg or polymer 86 significantly reduced the levels of mouse anti-HNK-1 (anti-MAG) IgM antibodies up to seven days after administration.