fluorinated mannosides inhibit cellular fucosylation

69
doi.org/10.26434/chemrxiv.13082138.v1 Fluorinated Mannosides Inhibit Cellular Fucosylation. Johan Pijnenborg, Emiel Rossing, Marek Noga, Willem Titulaer, Raisa Veizaj, Dirk J. Lefeber, Thomas Boltje Submitted date: 12/10/2020 Posted date: 13/10/2020 Licence: CC BY-NC-ND 4.0 Citation information: Pijnenborg, Johan; Rossing, Emiel; Noga, Marek; Titulaer, Willem; Veizaj, Raisa; Lefeber, Dirk J.; et al. (2020): Fluorinated Mannosides Inhibit Cellular Fucosylation.. ChemRxiv. Preprint. https://doi.org/10.26434/chemrxiv.13082138.v1 Fucose sugars are expressed on mammalian cell membranes as part of glycoconjugates and mediates essential physiological processes. The aberrant expression of fucosylated glycans has been linked to pathologies such as cancer, inflammation, infection, and genetic disorders. Tools to modulate fucose expression on living cells are needed to elucidate the biological role of fucose sugars and the development of potential therapeutics. Herein, we report a novel class of fucosylation inhibitors directly targeting de novo GDP-fucose biosynthesis. We demonstrate that cell permeable fluorinated mannoside 1-phosphate derivatives (Fucotrim I & II) are metabolic prodrugs that are metabolized to their respective GDP-mannose derivatives and efficiently inhibit cellular fucosylation. File list (2) download file view on ChemRxiv Fucotrim_Manuscript.pdf (0.97 MiB) download file view on ChemRxiv Fucotrim_Supplementary information.pdf (3.70 MiB)

Upload: others

Post on 22-Feb-2022

6 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Fluorinated Mannosides Inhibit Cellular Fucosylation

doi.org/10.26434/chemrxiv.13082138.v1

Fluorinated Mannosides Inhibit Cellular Fucosylation.Johan Pijnenborg, Emiel Rossing, Marek Noga, Willem Titulaer, Raisa Veizaj, Dirk J. Lefeber, Thomas Boltje

Submitted date: 12/10/2020 • Posted date: 13/10/2020Licence: CC BY-NC-ND 4.0Citation information: Pijnenborg, Johan; Rossing, Emiel; Noga, Marek; Titulaer, Willem; Veizaj, Raisa;Lefeber, Dirk J.; et al. (2020): Fluorinated Mannosides Inhibit Cellular Fucosylation.. ChemRxiv. Preprint.https://doi.org/10.26434/chemrxiv.13082138.v1

Fucose sugars are expressed on mammalian cell membranes as part of glycoconjugates and mediatesessential physiological processes. The aberrant expression of fucosylated glycans has been linked topathologies such as cancer, inflammation, infection, and genetic disorders. Tools to modulate fucoseexpression on living cells are needed to elucidate the biological role of fucose sugars and the development ofpotential therapeutics. Herein, we report a novel class of fucosylation inhibitors directly targeting de novoGDP-fucose biosynthesis. We demonstrate that cell permeable fluorinated mannoside 1-phosphatederivatives (Fucotrim I & II) are metabolic prodrugs that are metabolized to their respective GDP-mannosederivatives and efficiently inhibit cellular fucosylation.

File list (2)

download fileview on ChemRxivFucotrim_Manuscript.pdf (0.97 MiB)

download fileview on ChemRxivFucotrim_Supplementary information.pdf (3.70 MiB)

Page 2: Fluorinated Mannosides Inhibit Cellular Fucosylation

1

Fluorinated mannosides inhibit cellular fucosylation.

Johan F.A. Pijnenborg[a],†, Emiel Rossing[a],†, Marek Noga[b], Willem Titulaer[a], Raisa Veizaj[c], Dirk J.

Lefeber[b,c] and Thomas J. Boltje*[a]

[a] J.F.A. Pijnenborg, E. Rossing, W. Titulaer, Dr. T.J. Boltje

Department of Synthetic Organic Chemistry

Institute for Molecules and Materials, Radboud University

Heyendaalseweg 135, 6525AJ, Nijmegen, The Netherlands

[email protected]

[b] Dr. M. Noga, Prof. D.J. Lefeber

Department of Laboratory Medicine, Translational Metabolic Laboratory

Radboud Institute for Molecular Life Sciences, Radboud University Medical Center

Geert Grooteplein Zuid 10, 6525GA, Nijmegen, The Netherlands

[c] R. Veizaj, Prof. D.J. Lefeber

Department of Neurology

Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center

Geert Grooteplein Zuid 10, 6525GA, Nijmegen, The Netherlands

[†] These authors contributed equally to this work.

Supporting information for this article is given via a link at the end of the document.

Abstract: Fucose sugars are expressed on mammalian cell

membranes as part of glycoconjugates and mediates essential

physiological processes. The aberrant expression of fucosylated

glycans has been linked to pathologies such as cancer, inflammation,

infection, and genetic disorders. Tools to modulate fucose expression

on living cells are needed to elucidate the biological role of fucose

sugars and the development of potential therapeutics. Herein, we

report a novel class of fucosylation inhibitors directly targeting de novo

GDP-fucose biosynthesis. We demonstrate that cell permeable

fluorinated mannoside 1-phosphate derivatives (Fucotrim I & II) are

metabolic prodrugs that are metabolized to their respective GDP-

mannose derivatives and efficiently inhibit cellular fucosylation.

L-Fucose (Fuc) is a 6-deoxyhexose expressed at the termini of glycan

chains that decorate cell surface proteins and lipids.1 The fucose

residues on glycoconjugates are essential mediators of physiological

processes. For example, the fucose moiety in the tetrasaccharide

sialyl Lewisx (sLex) expressed on leukocytes is recognized by selectin

receptors that regulate leukocyte recruitment and extravasation. The

aberrant expression of sLex and increased fucosylation has been

linked to pathologies, most notably cancer, and has been shown to

promote tumor progression.2,3 Reducing fucosylation of glycans in

cancer using inhibitors of the fucose biosynthesis has therefore been

recognized as a promising therapeutic option. Glycans or proteins are

fucosylated by the action of thirteen fucosyltransferases (FucTs) that

differ in acceptor glycan preference but all utilize GDP-fucose as the

donor substrate.4

Scheme 1. (a) Working model of metabolic fucosylation inhibitors. (b) Previous work showing mechanism of action of different 4,6-dehydratase inhibitor acting on

CDP-D-glucose 4,6-dehydratase. Abbreviations: HK, hexokinase; PMM, phosphomannomutase; GDP, mannose-1-phosphate guanylyltransferase; GMDS, GDP-

mannose 4,6 dehydratase; FX, GDP-4-keto-6-deoxy-D-mannose-3,5-epimerase-4-reductase (GMER); FUK, fucokinase; FPGT, fucose-1-phosphate

guanylyltransferase.

Page 3: Fluorinated Mannosides Inhibit Cellular Fucosylation

2

Cells generate GDP-fucose either via the recycling of

fucose released during glycan turnover in the lysosome (salvage

pathway) or via de novo biosynthesis from mannose 1-phosphate

(Scheme 1). To decrease cellular fucosylation,

fucosyltransferase (FucT) inhibitors have attracted considerable

attention.2 A common strategy has been mimicking the natural

substrate guanosine diphosphate fucose (GDP-Fuc). However,

the essential GDP moiety in these compounds is associated with

a high polarity and low stability thereby limiting their use in vivo by

poor penetration of the cell membrane. This hurdle was recently

overcome with the development of less polar, cell permeable

fucose derivatives which are metabolized to the corresponding

active GDP-fucose analogs through the salvage pathway.5,6,7,8

These compounds target FucTs by competitive inhibition and the

de novo enzymes GMDS and FX by feedback inhibition. This

strategy led to fucosylation inhibitors that are active in vitro and in

vivo, showing promising anticancer effects in liver, breast and

blood cancer models.9,10,11,12,13 Moreover, combining a

fucosylation inhibitor with immunotherapy vaccination in

immunocompetent mice completely protected against tumor

growth due to enhanced antibody-dependent cellular cytotoxicity

(ADCC) with LS174T colorectal carcinoma and A20 lymphoma

cells.11 These inhibitors enter via the fucose salvage pathway yet

it is estimated that ~90% of the GDP-Fuc pool is biosynthesized

via de novo biosynthesis from GDP-mannose.14 Thus, direct

inhibition of de novo GDP-fucose biosynthesis could result in

more potent inhibitors yet this approach remains unexplored so

far to the best of our knowledge. GDP-fucose is biosynthesized

from GDP-mannose by oxidation of the 4-carbon to a ketone and

dehydration of the 6-position by GDP-mannose-4,6-dehydratase

(GMDS, Figure 3a). Subsequent epimerization of the 3- and 5-

position followed by the reduction of the 4-ketone by FX leads to

GDP-Fucose. Hence, GMDS and FX are prime targets to develop

de novo pathway inhibitors.

Scheme 2. Synthesis of 1–10. i) Ac2O, Pyr; ii) H2NNH2·HOAc, DMF; iii) bis(S-

acetyl-2-thioethyl)N,N-diethylphosphoramidite (23), 1H-tetrazole, ACN, then

mCPBA; iv) DMP, NaHCO3, DCM; v) DAST, DCM; vi) Ac2O, H2SO4, AcOH; vii)

TMSCF3, TBAF, THF, then AcOH; viii) NBS, H2O/acetone.

Herein we report the development of highly potent inhibitors

of the de novo pathway based on fluorinated mannose 1-

phosphate derivatives (Scheme 1a).15 We were inspired by a

mechanism-based inhibitor for CDP-D-glucose 4,6-dehydratase

isolated from Yersinia pseudotuberculosis. Use of a 6-deoxy-

difluoride modified CDP-glucose formed a reactive Michael

acceptor inside the active site leading to entrapment (Scheme

1b).16 We hypothesized that GDP-D-6-deoxy-difluoro-mannose

(GDP-D-Rha6F2) could inhibit GMDS, also a 4,6-dehydratase,

through a similar mechanism. To allow passive diffusion over the

cell membrane, lipophilic metabolic precursors were designed.

Two entry points in the de novo biosynthesis were considered with

precursors based on mannose (1–5) and mannose-1-phosphate

(6–10). The lipophilic (thio)ester protecting groups are expected

to be cleaved by esterases inside the cell, allowing the derivatives

to enter the endogenous GDP-mannose biosynthetic pathway. To

investigate the degree of fluorination on the inhibitory potency we

modified the 6-position with none to three fluorides (2–5 & 7–10).

Compounds 1–10 were synthesized as described in

Scheme 2 and the supplementary information. Compounds 1 and

2 were obtained by acetylation of D-mannose and D-rhamnose,

respectively. Mono- and difluorides 3 and 4 were obtained via

DAST mediated fluorination of the 6-OH or aldehyde, respectively,

followed by acetolysis. The synthesis of protected 1-phosphate

analogs 6–9 was achieved in a two-step sequence from their

respective precursors 1–4. Selective deprotection of the anomeric

acetyl ester was achieved using hydrazinium acetate. The

resulting lactol was reacted with phosphoramidite reagent 23

bearing two S-acetyl-2-thioethyl (SATE) groups and subsequently

oxidized to corresponding phosphate triesters 6–9. Trifluorides 5

and 10 were prepared via a homologation approach by

introducing a CF3-group at the C-5 position of the pentose D-

lyxose. To this end, derivative 17 was oxidized and reacted with

a trifluoromethyl anion affording a separable mixture of the

desired 6-trifluoro D-rhamnose (19) and the byproduct L-gulose

(20) derivative. Hydrolysis of the thioacetal with NBS and water

resulted in ring closure and the resulting lactol was acetylated or

phosphorylated to afford 5 and 10, respectively. This synthetic

approach ensures pyranose formation.

Table 1. EC50 values in micromolar for fucose expression inhibition.[a]

THP-1 Jurkat EL4

Compound AAL AOL AAL AOL AAL AOL

DMSO NI NI NI NI NI NI

P-D-Man-1P (6) NI NI NI NI NI NI

P-D-Rha-1P (7) NI 137 NI 153 NI NI

P-D-Rha6F-1P (8) 267 084 174 345 NI NI

P-D-Rha6F2-1P (9) 002.0 005.3 005.9 004.1 014 038

P-D-Rha6F3-1P (10) 000.61 000.45 028 013 NI NI

P-Fuc2F 045 031 115 121 NI NI

[a] Three cell lines (THP-1, Jurkat, EL4) were cultured for 3 days with 0–512 µM

of compound 6–10, P-Fuc2F or DMSO control. The cells were stained with two

fucose specific lectins (AAL & AOL) and analyzed by flow cytometry, presented

as mean percentage lectin binding normalized to control (n=3). N.I, no inhibition

(EC50>500 µM).

Page 4: Fluorinated Mannosides Inhibit Cellular Fucosylation

3

Figure 1. (a) Effect of 10 or 100 µM Fucotrim I & II and P-Fuc2F on cell surface glycosylation using different lectins, presented as percentage lectin binding compared

to DMSO control. (b) Effect of C6-modifications on potency of inhibition. (c) Potency of Fucotrim I and II compared to known salvage pathway dependent fucose

inhibitors P-Fuc2F, Fucostatin-I and Fucostatin-II using AOL lectin (n=3). (d) Onset and recovery of defucosylation. THP1 cells were incubated with 10 or 100 µM

(Figure S2) compound or DMSO control and fucosylation levels were determined with AAL lectins (Figure S2) for six days. (e) The effect on viability and cytotoxicity

on THP1 cells after incubation with 2, 10, 100 or 250 µM 6–10 or 1–5 (Figure S4) for three days was determined with an XTT and LDH respectively and presented

as percentage cell viability or cytotoxicity compared to DMSO control.

The inhibitory potency of 1–10 was assessed at different

concentrations in human THP-1 cells with fucose specific AAL

and AOL lectins (Figure S1a–d). The EC50 values were

determined, defined as the concentration where a 50% decrease

in lectin binding compared to control was observed (Table 1). The

non-phosphorylated compounds 1–5 did not show any inhibition

of fucosylation. In contrast, from the phosphorylated set 6–10, the

mono-, di-, and trifluorinated derivatives 7–10 displayed inhibition

of fucosylation. A trend was observed where increasing the

number of C-6 fluorides increased its potency (Figure 1b).

Inhibitors P-D-Rha6F2-1P (Fucotrim I, 9) and P-D-Rha6F3-1P

(Fucotrim II, 10) afforded (sub)micromolar inhibition of

fucosylation. Both Fucotrim I (9) & II (10) were more potent than

fucosylation inhibitor P-Fuc2F (1,3,4-tri-O-acetyl-2-deoxy-2-

fluoro-L-fucose), a known salvage pathway dependent inhibitor.

Also in human Jurkat cells potent inhibition for Fucotrim I and II

was observed (Table 1; Figure S1e–f). In mouse cell line EL4

however, only Fucotrim I (9) inhibited fucosylation while both

Fucotrim II (10) and P-Fuc2F did not inhibit fucosylation (Table 1;

Figure S1g–h).

Since Fucotrim I and II (9&10) are mannose-1-phosphate

derivatives, it is possible that other glycosylation pathways such

as mannosylation would be affected. Having established the

ability to inhibit cellular fucosylation with AAL and AOL lectins, the

effect on total cell surface glycosylation was evaluated using

additional lectins after 3 days incubation (Figure 1a; Figure S2).

N-glycans consist of a biantennary mannose containing backbone,

recognized by WGA, and can be branched, recognized by L-Pha.

These branches can be terminated with galactose, recognized by

PNA, or α2,6- and α2,3-linked sialic acids recognized by SNA and

MAL-II, respectively.17,18 Binding of LCA and PSA was decreased

for all fucosylation inhibitors as they are core fucose dependent.

The binding of WGA was unaffected as it recognizes core GlcNAc

residues independent of fucosylation.17 Fucotrim I and P-Fuc2F

showed a highly similar profile, not affecting the binding of L-Pha,

PNA, and MAL-II. Only the binding of SNA increased using these

inhibitors. Fucotrim II (10) on the other hand showed a less

specific profile, decreasing the binding of L-Pha, SNA and MAL-II

and increasing the binding of PNA. This would not be expected

for a selective fucosylation inhibitor as sialylation is also clearly

Page 5: Fluorinated Mannosides Inhibit Cellular Fucosylation

4

affected. From these results we conclude that Fucotrim I has a

similar specificity as known fucosylation inhibitor P-Fuc2F but is

more potent.

To further evaluate the potency of Fucotrim I and II, we

made a direct comparison with known fucosylation inhibitors P-

Fuc2F, P-L-Fuc6F3 (Fucostatin I) and P-L-Fuc-1CH2P (Fucostatin

II) (Figure 1c).5,6,7 The EC50 values for known Fucostatin I and II

were determined at 1.3 µM and 0.41 µM respectively, which is in

the same range as Fucotrim I and II under these conditions (Table

1). P-Fuc2F was significantly less potent with an EC50 of 31 µM.

The onset of inhibition and duration for recovery was

evaluated on THP-1 cells with 10 and 100 µM compound using

the AAL lectin (Figure 1d–e; Figure S3). In line with the potency

data, no inhibition was observed for known inhibitor P-Fuc2F at

10 µM concentration. At the same concentration, Fucotrim I and

II decreased lectin binding by over 50% after 3 days with full

recovery to normal fucosylation levels after 4–5 days. At 100 µM

concentration Fucotrim I and II inhibit fucosylation after 1 day and

to over 50% after 3 days. Fucosylation was fully recovered to

normal levels after 6 days. Similar results were obtained using the

AOL lectin (Figure S3).

Finally, a toxicity profile of 1–10 was established by

monitoring the metabolic activity and cell death of THP-1 cells

after three days of treatment (Figure 1e; Figure S4). Importantly,

none of the compounds were cytotoxic at concentrations up to

250 μM. However, the cell viability was decreased which might be

attributed to the known relation between fucosylation and

proliferation.9,10,19

Figure 2. Nucleotide sugar analysis. THP-1 cells were incubated for indicated

time points with 10 µM Fucotrim I or II or with DMSO control. After sample

preparation, the GDP-Rha6F2 (a), GDP-Rha6F3 (b), GDP-Fuc (c) and GDP-Man

(d) levels were analyzed using reverse-phase ion pairing chromatography

coupled to a triple quadrupole mass spectrometer operating in negative ion

mode and presented as their abundance in the nucleotide sugar pool (Figure

S5) (n=3).

It has been suggested that the reduced proliferation after

incubation with known fucosylation inhibitors is caused by acting

on the de novo biosynthesis of GDP-fucose rather than acting on

fucosyltransferases which correlates with our findings.20

Figure 3. (a) Endogenous mechanism of action for human GMDS.21 (b) Natural substrate GDP-mannose (green carbons) was docked at the 4-fluoro-GDP-mannose

(G4F) binding site in human GMDS (6GPJ21, white carbons) in which also NADP+ (cyan carbons) was cocrystalized. (c–i) Unnatural substrate GDP-D-Rha6F2 (c,

green carbons) and the proposed metabolic intermediates (d–i, green carbons) were docked in the same active site. For each structure, the active site residues

Glu157 and Tyr179 were first protonated or deprotonated and either NADPH or NADP+ was present accordingly with the known mechanism of action on GDP-

mannose. Black dotted lines depict expected interactions between atoms leading to a new product, based on the natural mechanism.21

Page 6: Fluorinated Mannosides Inhibit Cellular Fucosylation

5

To test our hypothesized prodrug strategy, we studied the

metabolism of Fucotrim I and II to their corresponding GDP-

analogs inside cells. To this end, intracellular nucleotide sugar

levels were analyzed. THP-1 cells were treated for different time

points with Fucotrim I and II and extracted metabolites analyzed

using reverse-phase ion pairing chromatography coupled to a

triple quadrupole mass spectrometer operating in negative ion

mode (Figure 2; Figure S5). For analysis of GDP-D-Rha6F2 and

GDP-D-Rha6F3 analogs, theoretical mass transitions were

programmed. After incubations for only 1 hour, both Fucotrim

GDP-analogs were observed with a maximum abundance after

4–8 hours (Figure 2a). Subsequently, GDP-fucose levels

decreased after 1–2 days for both inhibitors, indicating inhibition

of GDP-fucose biosynthesis by Fucotrim I and II (Figure 2b).

GDP-mannose levels were also affected by both inhibitors. For

Fucotrim I, the GDP-mannose levels recovered after 3 days and

for Fucotrim II, GDP-mannose levels didn’t recover within the 3

days of the experiment. This correlates well with the lectin panel

(Figure 1a) where after 3 days incubation Fucotrim I only shows

an effect on fucosylation and Fucotrim II shows an effect on

multiple forms of glycosylation. Importantly, for both Fucotrim I

and II no changes were observed for other nucleotide sugar levels

such as UDP-glucose and UDP-galactose (Figure S5).

Having established the metabolism towards GDP-D-Rha6F2

and GDP-D-Rha6F3 inside the cell we investigated if a mechanism

of inhibition as previously described for a bacterial CDP-glucose

4,6-dehydratase (Figure 1b) would be plausible for Fucotrim I

using in silico experiments.16 The docking experiments were

based on a recent study describing the mechanism of action for

human GMDS in detail (Figure 3a).21 The natural substrate GDP-

mannose was docked at the 4-fluoro-GDP-mannose (G4F)

binding site in a human GMDS crystal structure (6GPJ) in which

cofactor NADP+ was co-crystallized (Figure 3b).21 Docking of

GDP-D-Rha6F2 in the same binding site resulted in similar poses

as for GDP-mannose, suggesting it would still be a substrate for

GMDS. The proposed metabolic intermediates (Figure 3d–i) are

based on the endogenous mechanism of action for GMDS and a

known 4,6-dehydratase inhibitor.16,21 The catalytic residues were

positioned such that oxidation (I, Figure 3d), enolization (II,

Figure 3e), elimination (III, Figure 3f–g) and reduction (IV,

Figure 3h) do not seem to be affected sterically. In the

endogenous mechanism enolization would yield the final product.

For the difluoride derivative however, a second elimination might

occur, affording an electrophilic intermediate (Figure 3i) that

cannot be reduced due to the lack of reductant NADPH in the

active site. It is hypothesized that this intermediate either

competitively binds GMDS, or analogous to previous findings16,

reacts with a nucleophilic amino acid (e.g. Ser156) in GMDS

resulting in covalent inhibition, or with water affording a natural

intermediate for GMDS. The in silico experiments indicate that

GDP-D-Rha6F2 likely acts on GMDS but further studies are

needed to investigate the exact mechanism of action.

In conclusion, we have developed in vitro validated de novo

pathway dependent fucosylation inhibitors. Fucotrim I (P-D-

Rha6F2-1P, 9) and Fucotrim II (P-D-Rha6F3-1P, 10) are new tools

to study fucosylation function and are good candidates for further

therapeutic development. Additionally, this approach could be

considered for targeting other 4,6-dehydratases affording cell

active inhibitors.

Acknowledgements

This work was supported by an ERC-Stg awarded to T.J.B., the

Netherlands Organization for Scientific Research (VIDI Grant

91713359 to D.J.L), and the Radboud Consortium for

Glycoscience. We thank prof. Gosse Adema PhD and the

Molecular Immunology group at the Radiotherapy &

OncoImmunology lab at the Radboud University Medical Centre

for kindly supplying us with the THP-1, Jurkat and EL-4 cell lines.

We would to acknowledge John Allen from Amgen Inc. for

providing Fucostatin I and II.

Keywords: Fucosylation • Inhibition • De novo pathway • Small

molecule • GMDS • mannoside

References:

1. Becker, D. J.; Lowe, J. B., Fucose: biosynthesis and biological function in mammals. Glycobiology 2003, 13 (7), 41R-53R. 2. Tu, Z.; Lin, Y.-N.; Lin, C.-H., Development of fucosyltransferase and fucosidase inhibitors. Chemical Society Reviews 2013, 42 (10), 4459-4475. 3. Varki, A., Essentials of Glycobiology. 3 ed.; 2017; pp 597-609. 4. Narimatsu, Y.; Joshi, H. J.; Nason, R.; Van Coillie, J.; Karlsson, R.; Sun, L.; Ye, Z.; Chen, Y.-H.; Schjoldager, K. T.; Steentoft, C.; Furukawa, S.; Bensing, B. A.; Sullam, P. M.; Thompson, A. J.; Paulson, J. C.; Büll, C.; Adema, G. J.; Mandel, U.; Hansen, L.; Bennett, E. P.; Varki, A.; Vakhrushev, S. Y.; Yang, Z.; Clausen, H., An Atlas of Human Glycosylation Pathways Enables Display of the Human Glycome by Gene Engineered Cells. Molecular Cell 2019, 75 (2), 394-407.e5. 5. Rillahan, C. D.; Antonopoulos, A.; Lefort, C. T.; Sonon, R.; Azadi, P.; Ley, K.; Dell, A.; Haslam, S. M.; Paulson, J. C., Global metabolic inhibitors of sialyl- and fucosyltransferases remodel the glycome. Nat Chem Biol 2012, 8 (7), 661-668. 6. Kizuka, Y.; Nakano, M.; Yamaguchi, Y.; Nakajima, K.; Oka, R.; Sato, K.; Ren, C.-T.; Hsu, T.-L.; Wong, C.-H.; Taniguchi, N., An Alkynyl-Fucose Halts Hepatoma Cell Migration and Invasion by Inhibiting GDP-Fucose-Synthesizing Enzyme FX, TSTA3. Cell Chemical Biology 2017, 24 (12), 1467-1478.e5. 7. Allen, J. G.; Mujacic, M.; Frohn, M. J.; Pickrell, A. J.; Kodama, P.; Bagal, D.; San Miguel, T.; Sickmier, E. A.; Osgood, S.; Swietlow, A.; Li, V.; Jordan, J. B.; Kim, K.-W.; Rousseau, A.-M. C.; Kim, Y.-J.; Caille, S.; Achmatowicz, M.; Thiel, O.; Fotsch, C. H.; Reddy, P.; McCarter, J. D., Facile Modulation of Antibody Fucosylation with Small Molecule Fucostatin Inhibitors and Cocrystal Structure with GDP-Mannose 4,6-Dehydratase. ACS Chemical Biology 2016, 11 (10), 2734-2743. 8. Zandberg, W. F.; Kumarasamy, J.; Pinto, B. M.; Vocadlo, D. J., Metabolic inhibition of sialyl-Lewis X biosynthesis by 5-thiofucose remodels the cell surface and impairs selectin-mediated cell adhesion. J Biol Chem 2012, 287 (47), 40021-40030. 9. Zhou, Y.; Fukuda, T.; Hang, Q.; Hou, S.; Isaji, T.; Kameyama, A.; Gu, J., Inhibition of fucosylation by 2-fluorofucose suppresses human liver cancer HepG2 cell proliferation and migration as well as tumor formation. Sci Rep 2017, 7 (1), 11563-11563. 10. Carrascal, M. A.; Silva, M.; Ramalho, J. S.; Pen, C.; Martins, M.; Pascoal, C.; Amaral, C.; Serrano, I.; Oliveira, M. J.; Sackstein, R.; Videira, P. A., Inhibition of fucosylation in human invasive ductal carcinoma reduces E-selectin ligand expression, cell proliferation, and ERK1/2 and p38 MAPK activation. Mol Oncol 2018, 12 (5), 579-593. 11. Okeley, N. M.; Alley, S. C.; Anderson, M. E.; Boursalian, T. E.; Burke, P. J.; Emmerton, K. M.; Jeffrey, S. C.; Klussman, K.; Law, C.-L.; Sussman, D.; Toki, B. E.; Westendorf, L.; Zeng, W.; Zhang, X.; Benjamin, D. R.; Senter, P. D., Development of orally active inhibitors of protein and cellular fucosylation. Proc Natl Acad Sci U S A 2013, 110 (14), 5404-5409.

Page 7: Fluorinated Mannosides Inhibit Cellular Fucosylation

6

12. Okeley, N. M.; Heiser, R. A.; Zeng, W.; Hengel, S. M.; Wall, J.; Haughney, P. C.; Yap, T. A.; Robert, F.; Sanborn, R. E.; Burris, H.; Chow, L. Q.; Do, K. T.; Gutierrez, M.; Reckamp, K.; Weise, A.; Camidge, D. R.; Strickler, J.; Steuer, C.; Wang, Z.; O'Meara, M. M.; Alley, S. C.; Gardai, S. J., Abstract 5551: SGN-2FF: A small-molecule inhibitor of fucosylation modulates immune cell activity in preclinical models and demonstrates pharmacodynamic activity in early phase 1 analysis. Cancer Research 2018, 78 (13 Supplement), 5551-5551. 13. Disis, M. L.; Corulli, L. R.; Gad, E. A.; Koehnlein, M. R.; Cecil, D. L.; Senter, P. D.; Gardai, S. J.; Okeley, N. M., Therapeutic and prophylactic anti-tumor activity of an oral inhibitor of fucosylation in spontaneous mammary cancers. Molecular Cancer Therapeutics 2020, molcanther.0500.2019. 14. Yurchenco, P. D.; Atkinson, P. H., Fucosyl-glycoprotein and precursor pools in HeLa cells. Biochemistry 1975, 14 (14), 3107-3114. 15. Pijnenborg, J.F.A.; Rossing, E.; Boltje, T.J., New fluorinated hexoses. EP20195308.0, 2020. 16. Chang, C.-W. T.; Chen, X. H.; Liu, H.-w., CDP-6-deoxy-6,6-difluoro-d-glucose:  A Mechanism-Based Inhibitor for CDP-d-glucose 4,6-Dehydratase. Journal of the American Chemical Society 1998, 120 (37), 9698-9699. 17. Gao, C.; Hanes, M. S.; Byrd-Leotis, L. A.; Wei, M.; Jia, N.; Kardish, R. J.; McKitrick, T. R.; Steinhauer, D. A.; Cummings, R. D., Unique Binding Specificities of Proteins toward Isomeric Asparagine-Linked Glycans. Cell Chemical Biology 2019, 26 (4), 535-547.e4. 18. Iskratsch, T.; Braun, A.; Paschinger, K.; Wilson, I. B. H., Specificity analysis of lectins and antibodies using remodeled glycoproteins. Analytical Biochemistry 2009, 386 (2), 133-146. 19. Jia, L.; Zhang, J.; Ma, T.; Guo, Y.; Yu, Y.; Cui, J., The Function of Fucosylation in Progression of Lung Cancer. Front Oncol 2018, 8, 565-565. 20. Dai, Y.; Hartke, R.; Li, C.; Yang, Q.; Liu, J. O.; Wang, L.-X., Synthetic Fluorinated L-Fucose Analogs Inhibit Proliferation of Cancer Cells and Primary Endothelial Cells. ACS Chemical Biology 2020. 21. Pfeiffer, M.; Johansson, C.; Krojer, T.; Kavanagh, K. L.; Oppermann, U.; Nidetzky, B., A Parsimonious Mechanism of Sugar Dehydration by Human GDP-Mannose-4,6-dehydratase. ACS Catalysis 2019, 9 (4), 2962-2968.

Page 8: Fluorinated Mannosides Inhibit Cellular Fucosylation

7

Entry for the Table of Contents

Fucose sugars on mammalian cell membranes play important roles in physiological and pathological processes. Inhibition of

fucosylation has been identified as a therapeutic avenue. We have developed potent cell-active fucosylation inhibitors, directly inhibiting

the de novo biosynthesis of GDP-fucose.

Institute and/or researcher Twitter usernames: @JohanPijnenborg

Page 10: Fluorinated Mannosides Inhibit Cellular Fucosylation

1

Supplementary information for:

Fluorinated mannosides inhibit cellular fucosylation.

Johan F.A. Pijnenborg[a],†, Emiel Rossing[a],†, Marek Noga[b], Willem Titulaer[a], Raisa Veizaj[c],

Dirk J. Lefeber[b,c] and Thomas J. Boltje*[a]

[a] J.F.A. Pijnenborg, E. Rossing, W. Titulaer, Dr. T.J. Boltje

Department of Synthetic Organic Chemistry

Institute for Molecules and Materials, Radboud University

Heyendaalseweg 135, 6525AJ, Nijmegen, The Netherlands

[email protected]

[b] Dr. M. Noga,Prof. D.J. Lefeber

Department of Laboratory Medicine, Translational Metabolic Laboratory

Radboud Institute for Molecular Life Sciences, Radboud University Medical Center

Geert Grooteplein Zuid 10, 6525GA, Nijmegen, The Netherlands

[c] R. Veizaj, Prof. D.J. Lefeber

Department of Neurology

Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center

Geert Grooteplein Zuid 10, 6525GA, Nijmegen, The Netherlands

[†] These authors contributed equally to this work.

Supporting information for this article is given via a link at the end of the document.

Content

Supporting figures and tables ................................................................................................................ 2

Supplementary methods ........................................................................................................................ 7

Reagents ............................................................................................................................................ 7

Procedure for cell culture .................................................................................................................... 7

Procedure for lectin staining of cells ................................................................................................... 7

Procedure for lectin specificity assay ................................................................................................. 8

Procedure for determining the onset and recovery of inhibition ........................................................ 8

Viability/toxicity assays ....................................................................................................................... 9

Nucleotide sugar analysis ................................................................................................................... 9

In silico modeling ................................................................................................................................ 9

Synthetic procedures ............................................................................................................................ 11

General synthetic procedures .......................................................................................................... 11

Synthetic procedures ........................................................................................................................ 11

References ........................................................................................................................................... 23

Page 11: Fluorinated Mannosides Inhibit Cellular Fucosylation

2

Supporting figures and tables

Figure S1: Cell lines THP-1 (a-d), Jurkat (e-f) and EL4 (g-h) were cultured for 3 days with 0–512 µM

compound or DMSO control. The cells were stained with two fucose specific lectins, AAL (a, c, e, g)

& AOL (b, d, f, h) and analysed by flow cytometry, presented as mean percentage lectin binding

normalized to control (n=3).

Page 12: Fluorinated Mannosides Inhibit Cellular Fucosylation

3

Figure S2: Effect of 1, 2, 4, 6–10, P-Fuc2F and DMSO control on total cell surface glycosylation. THP-

1 cells treated for three days with 10 or 100 µM compound or DMSO control were stained with a panel

of biotinylated lectins and streptavidin-PE. Lectin binding was determined in three independent

experiments by flow cytometry and is presented as mean fluorescent intensity relative to DMSO control

± SEM. Bar diagrams show binding of AAL(a), AOL (b), LCA (c), PSA (d), WGA (e), L-Pha (f), PNA

(g), SNA (h) and MAL-II (i).

Page 13: Fluorinated Mannosides Inhibit Cellular Fucosylation

4

Figure S3: Onset and recovery of defucosylation. THP1 cells were incubated with 10 µM (b, Figure

1d) or 100 µM (a,c) compound or DMSO control and fucosylation levels were determined with AAL (a)

and AOL (b,c) lectins for six days (n=3).

Page 14: Fluorinated Mannosides Inhibit Cellular Fucosylation

5

Figure S4: The effect on viability and cytotoxicity on THP1 cells after incubation with 2, 10, 100 or

250 µM 1–5 was determined with an XTT and LDH respectively and presented as percentage cell

viability or cytotoxicity compared to DMSO control.

Page 15: Fluorinated Mannosides Inhibit Cellular Fucosylation

6

Figure S5: Full nucleotide sugar analysis. THP-1 cells were incubated for indicated time points with

10 µM Fucotrim I or II or with DMSO control. After sample preparation, the different nucleotide levels

were analyzed using reverse-phase ion pairing chromatography coupled to a triple quadrupole mass

spectrometer operating in negative ion mode and presented as their abundance in the nucleotide sugar

pool (n=3).

Page 16: Fluorinated Mannosides Inhibit Cellular Fucosylation

7

Supplementary methods

Reagents

Biotinylated AAL, SNA, MAL-II, WGA, LCA, PSA, PNA, PHA-L and GSL-1 lectins and 10x Carbo-free

Blocking Buffer were purchased from Vector laboratories Inc.. Biotinylated AOL lectin was purchased

from TCI Europe. 0.2 mg/mL Streptavidin-phycoerythrin conjugate was purchased from Fischer

Scientific (Invitrogen, eBioscience). Unnatural sugar derivatives were synthesized as described under

synthetic procedures and stored at -20°C at a concentration of 100 mM/DMSO.

Procedure for cell culture

THP-1 cells (TIB-202, ATCC) and Jurkat cells (TIB-152, ATCC) were cultured in RPMI-1640 medium

containing 2mM Glutamine and 25mM HEPES (GibcoTM, Life Technologies), supplemented with 10%

v/v heat-inactivated fetal bovine serum (FBS) (GibcoTM, Life Technologies) and 1X antibiotic-

antimycotic solution (100 units/mL of penicillin, 100 μg/mL of streptomycin, and 0.25 μg/mL Fungizone)

(GibcoTM, Life Technologies) and passaged every 3-4 day by seeding a fraction of 0.5-2 mln cells of

the culture per 10 mL medium.

EL-4 cells (TIB-39, ATCC) were cultured in DMEM medium (GibcoTM, Life Technologies),

supplemented with 10% v/v heat-inactivated fetal bovine serum (FBS) (GibcoTM, Life Technologies),

1X antibiotic-antimycotic solution (100 units/mL of penicillin, 100 μg/mL of streptomycin, and 0.25

μg/mL Fungizone) (GibcoTM, Life Technologies) and 2mM Glutamine (GibcoTM, Life Technologies) and

passaged every 2-4 days by seeding a fraction of 0.5-1 mln cells of the culture per 10 mL medium.

All cells were cultured at 37°C, 5% CO2 in a humidified incubator.

Procedure for lectin staining of cells

Protocol below was repeated until n>=3 for each compounds using cells at different passage numbers.

Cells were cultured in medium containing different concentrations of unnatural sugar derivatives on

96-wells plates (suspension cell lines; 13,000–20,000 cells per well) (Thermo Scientific). DMSO, at

same dilution as the unnatural sugar derivative stock solutions, was used as a positive control for lectin

staining; P-Fuc2F was used for all concentrations as negative control. Cells were incubated for three

days at 37°C and 5% CO2 in a humidified incubator.

Cells were harvested and washed with 100 μl 1X CF-blocking buffer (Vector Laboratories Inc.)

containing 1 mM CaCl2 and 1 mM MgCl2. The cells were then resuspended in 50 μl 0.5 μg/mL of 0.5

ng/mL biotinylated lectin in 1X carbo-free blocking buffer and incubated at 4-8°C for 45-60 min. Cells

were washed with 3x100 μl PBA (PBS containing 1% v/v FBS and 0.1% w/w sodium azide), incubated

with 40 μl 1 μg/mL Streptavidin-phycoerythrin conjugate (Invitrogen, eBioscience) in PBA for 10-15

min at 4-8°C. Cells were then washed again with 3x100 μl PBA, resuspended in PBA and fluorescence

was measured with a flow cytometer (Beckman & Dickinson FACS-Calibur). Each replicate for each

condition with >10,000 gated events. Data was processed using FlowJo (FlowJo LLC). Percentage of

lectin binding was obtained by normalizing the MFI values to the MFI values of the respective DMSO

control.

Page 17: Fluorinated Mannosides Inhibit Cellular Fucosylation

8

Procedure for lectin specificity assay

Protocol below was repeated until n>=3 for each compounds using cells at different passage numbers.

Culture medium was prepared in in 96-wells plates (Thermo Scientific). For every unnatural sugar

derivative, 11 wells containing 100 μM of unnatural sugar derivatives and 11 wells containing 10 μM

of the unnatural sugar derivatives were prepared. DMSO, at same dilution as the tested probes, was

used as a positive control for lectin staining. THP-1 Cells were cultured in the plates (20,000 cells per

well) and the cells were incubated for three days at 37°C and 5% CO2.

Cells were harvested and washed with 100 μl 1X CF-blocking buffer (Vector Laboratories Inc.)

containing 1 mM CaCl2 and 1 mM MgCl2. The cells were then resuspended in 50 μl 0.5 μg/mL of either

0.5 ng/mL biotinylated AAL, AOL, SNA, MAL-II, WGA, LCA, PSA, PNA, PHA-L or GSL-1 lectin in 1X

carbo-free blocking buffer or with 50 μl of non-supplemented 1X carbo-free blocking buffer and

incubated at 4-8°C for 45-60 min. Cells were washed with 3x100 μl PBA (PBS containing 1% v/v FBS

and 0.1% w/w sodium azide), incubated with 40 μl 1 μg/mL Streptavidin-phycoerythrin conjugate

(Invitrogen, eBioscience) in PBA for 10-15 min at 4-8°C. Cells were then washed again with 3x100 μl

PBA, resuspended in PBA and fluorescence was measured with a flow cytometer (Beckman &

Dickinson FACS-Calibur). Each replicate for each condition with >10,000 gated events. Data was

processed using FlowJo (FlowJo LLC). Percentage of lectin binding was obtained by normalizing the

MFI values to the MFI values of the respective DMSO control.

Procedure for determining the onset and recovery of inhibition

THP-1 cells were cultured in medium containing different concentrations of unnatural sugar derivatives

on 96-wells plates (suspension cell lines; 13,000-20,000 cells per well) (Thermo Scientific). DMSO, at

same dilution as the unnatural sugar derivative stock solutions, was used as a positive control for lectin

staining; P-Fuc2F was used for all concentrations as negative control. Cells were incubated for eight

days at 37°C and 5% CO2 in a humidified incubator. Medium was changed after three and six days of

incubation with cell culture medium without unnatural sugar derivatives. Cells were passaged 1:2 after

4, 6 and 8 days of incubation. Every day cells were harvested and washed with 100 μl 1X CF-blocking

buffer (Vector Laboratories Inc.) containing 1 mM CaCl2 and 1 mM MgCl2. The cells were then

resuspended in 50 μl 0.5 μg/mL of either 0.5 ng/mL biotinylated AAL, AOL, SNA, MAL-II, WGA, LCA,

PSA, PNA, PHA-L or GSL-1 lectin in 1X carbo-free blocking buffer or with 50 μl of non-supplemented

1X carbo-free blocking buffer and incubated at 4-8°C for 45-60 min. Cells were washed with 3x100 μl

PBA (PBS containing 1% v/v FBS and 0.1% w/w sodium azide), incubated with 40 μl 1 μg/mL

Streptavidin-phycoerythrin conjugate (Invitrogen, eBioscience) in PBA for 10-15 min at 4-8°C. Cells

were then washed again with 3x100 μl PBA, resuspended in PBA and fluorescence was measured

with a flow cytometer (Beckman & Dickinson FACS-Calibur). Each replicate for each condition with

>10,000 gated events. Data was processed using FlowJo (FlowJo LLC). Percentage of lectin binding

was obtained by normalizing the MFI values to the MFI values of the respective DMSO control.

Page 18: Fluorinated Mannosides Inhibit Cellular Fucosylation

9

Viability/toxicity assays

Assays were performed using an XTT Cell Viability Assay kit (Invitrogen, CyQUANT) or a LDH-toxicity

assay kit (Thermo Scientific) respectively. Assays were performed according to enclosed manuals.

THP-1 cells were treated for 3 days with unnatural sugar analogs at 37°C, 5% CO2 in a humidified

incubator before adding the XTT/LDH reagents. For the control, cells were treated with DMSO.

%Viability and %Cytotoxicity were calculated relative to the DMSO control for XTT and lysed untreated

cells for the LDH assay.

Nucleotide sugar analysis

THP-1 cells were incubated for 0, 1, 2, 4, 8, 24, 48 or 72 hrs with 10 μM Fucotrim I, Fucotrim II or

DMSO control in triplo. For the 0, 1, 2, 4, 8 hr timepoints, 3 mln cells were plated in 6-well plates, for

the 24 hour timepoint, 1.5 mln cells were plated per well, for the 48 hour timepoint, 1 million cells were

plated and for the 72 hour timepoint, 750,000 cells were plated per well to ensure a total of 3 to 3.5

million cells at time of harvesting. At each timepoint, cells were harvested, medium was removed and

the cells were washed twice with 2 mL 75 mM ammonium carbonate pH 7.4 buffer in MilliQ water. After

removing the washing buffer, cells were snap-frozen in liquid nitrogen. Samples then were stored at -

80°C until workup.

Samples were thawed at -20°C and incubated for 5 minutes in 1 mL of extraction buffer (1:2:2 MilliQ

water: acetonitrile: methanol). Samples were then spun down at 13.000 rpm in a tabletop centrifuge

and supernatant was transferred into 1.5 mL eppendorf tubes and solvent was removed with a

speedvac (Savant SC100, RVT 100 with an external oil pump). Upon redissolving in 100 μL of MilliQ

water 10 μL was injected in to a Agilent 1290 LC-MS system using a modified version of previously

published method for analysis of nucleotide sugars.1 Agilent 6490A QQQ mass spectrometer operated

in dynamic multiple-reaction monitoring MRM modes using transitions generated in silico by the use of

a script written in Python (v 3.6.7), an RDkit library (2019.09.2), and chemical structures of the target

compound as the input. All LC-MS data files were processed in Skyline version 20.1 using a target list

chosen on the basis of an in silico-generated transition list.2 Resulting peak areas were normalized to

the sum of all nucleotide sugars detected, excluding metabolites in pathways affected by treatment (all

GDP-sugars) to obtain relative abundances. Data visualization was performed in Python (v 3.8.5) using

Pandas (v 1.1.2) and Seaborn (v 0.11) libraries.

In silico modeling

Protein Preparation for Docking. The crystal structure of GMDS in complex with 4-fluoro-α-D-

mannopyrranosyl guanoside was downloaded from the PDB-redo database (https://pdb-redo.eu/ , PDB

code 6GPJ)3 and loaded into MOE. The structure was preprocessed by deleting all non- NADP+ and

G4F ligands. Next, the potential was set up to AMBER14:EHT as force field with R-field solvation

model. Addition of hydrogen atoms, removal of water molecules farther than 4.5 Å away from ligand or

receptor, correction of library errors and tethered energy minimization up to a gradient of 0.1 kcal mol-

1 Å2 was performed using MOE’s QuickPrep module. Afterwards, only the C-chain residues, ligands

and solvent molecules were kept. To mimic each intermediate step in the reaction pathway of GMDS,

changes were made to both the receptor and ligands, based on earlier finding by Pfeiffer et al. using

the MOE Builder module.3 Regarding the protein this concerned the (de)protonation of Tyr179 and

Glu157 and regarding the ligands the presence of either NADP+ or NADPH. After making the

Page 19: Fluorinated Mannosides Inhibit Cellular Fucosylation

10

adjustments, MOE’s QuickPrep module was ran again without protonation, changing the final RMS

gradient for the energy minimization to 1x10-5 kcal mol-1 Å2. Ligand preparation. Ligand structures

were drawn in ChemDraw 18.1, imported into a MOE database in sdf-file format, and energy minimized

up to a gradient of 1x10-5 using the MMFF94x force field. The saved mdb-file was used for the docking

studies. Docking. The ligands were docked into the appropriate protein model, using cocrystalized 4-

fluoro-GDP-mannose as template for placement. After 30 placements the placements were refined to

5 poses using the Induced Fit method with GBVI/WSA rescoring. From the 5 poses, the best pose was

chosen manually for each ligand by a combination of their displacement compared to the template

ligand and the calculated score.

Page 20: Fluorinated Mannosides Inhibit Cellular Fucosylation

11

Synthetic procedures

General synthetic procedures and abbreviations 1H and 13C NMR spectra were recorded on a Varian Iinova 400 MHz or Bruker Avance III 500 MHz spectrometer. Chemical shifts are reported in parts per million (ppm) relative to tetramethylsilane (TMS) as the internal standard. NMR data is presented as follows: Chemical shift, multiplicity (s = singlet, bs = broad singlet, d = doublet, t = triplet, dd = doublet of doublet, dt = doublet of triplet, m = multiplet and/or multiple resonances), integration, coupling constant in Hertz (Hz). All NMR signals were assigned on the basis of 1H, 13C, 19F NMR, COSY and HSQC experiments. Mass spectra were recorded on a JEOL JMS-T100CS AccuTOF mass spectrometer. Automatic column chromatography was performed on Biotage Isolera Spektra One, using SNAP cartridges 10-50g filled with normal silica (Biotage, 30-100 μm, 60 Å) or water resistant iatro beads. Microwave reactions were perfOCH3d on a Biotage Initiator 4.1.3. TLC analysis was conducted on TLC Silicagel, 60, F254, Merck, with detection by UV absorption (254 nm) where applicable, and by spraying with 20% sulfuric acid in methanol followed by charring at ~150 °C or by spraying with a solution of (NH-4)6Mo7O24

.H2O (25 g l-1) in 10% sulfuric acid in methanol followed by charring at ~300°C. DCM, ACN and Tol were freshly distilled. All reactions were carried out under an argon atmosphere. All synthesized samples that were used for cell tests were dissolved in water or a mixture of dioxane/water and subsequently lyophilized. Abbreviations: Ac, acetyl; ACN, Acetonitrile; AcOH Acetic acid; aq., aqueous; DCM dichloromethane; DMAP, dimethylaminopyridine; DMF, N,N-Dimethylformamide; DMP, Dess–Martin periodinane; EtOAc, ethyl acetate; eq., equivalent; Hept, heptane; hrs, hours; mCPBA, meta-chloroperoxybenzoic acid; min, minutes; NBS, N-bromosuccinimide; Pyr, pyridine; r.t., room temperature; sat., saturated; TBDMS, tert-Butyldiphenylsilyl; TEA, triethylamine; THF, tetrahydrofuran; TIPS, triisopropylsilyl; TMS, trimethylsilyl; Tol, toluene

Synthetic procedures

Scheme 1: Synthetic route towards trifluoro derivatives 5 and 10.

Page 21: Fluorinated Mannosides Inhibit Cellular Fucosylation

12

Scheme 2: Synthetic route towards phosphoramidite reagent 23.

General procedure A: Selective 1-deacetylation

Compounds 2–4 (1 eq.) were dissolved DMF (0.3 M) and hydrazine acetate (1.2 eq.) was added at

0 ̊C. After stirring for 5 minutes, the mixture was stirred till completion at r.t. which was typically after 3

hrs. The mixture was diluted with DCM, washed with sat. aq. NaHCO3 and brine. The organic layer

was dried over anhydrous Na2SO4, filtered concentrated in vacuo. The residue was purified by silicagel

flash column chromatography (EtOAc in Hept) to afford 11–13.

General procedure B: 1-Phosphorylation

Similar as described previously,4 1-hydroxy-derivatives of 6–10 (1 eq.) and di-(S-acetyl-2-thioethyl)-

N,N-diethylphosphoroamidite 23 (1.1 eq) were dissolved in in ACN (0.35 M) at 0 °C and 1H-tetrazole

(1.4 eq, 0.45 M in acetonitrile) was slowly added. After full conversion, typically after 1 hr, mCPBA (2.4

eq, 77% wt.) was added at 0 °C and stirred for 30 min. The mixture was diluted with EtOAc and 10%

aq. Na2SO3 and the organic layer was washed with sat. aq. NaHCO3 and brine. The aqueous phases

were extracted two more times with fresh EtOAc and the combined organic layers were dried over

anhydrous Na2SO4, filtered concentrated in vacuo. The residue was purified by silicagel flash column

chromatography (EtOAc in Hept) to afford 6–10.

Synthetic procedures:

1,2,3,4,6-penta-O-acetyl-D-mannopyranoside (P-D-Man, 1). Synthesis was

performed as described previously and the data is identical to the data previously reported.5. TLC:

(60:40, EtOAc:Hept v/v) Rf = 0.53. 1H NMR (500 MHz, CDCl3, major anomer) δ 6.09 (d, J = 1.9 Hz,

1H, H1), 5.36 – 5.34 (m, 2H, H3 & H4), 5.27 – 5.26 (m, 1H, H2), 4.29 (dd, J = 12.4, 4.8 Hz, 1H, H6a),

4.11 (dd, J = 12.4, 2.5 Hz, 1H, H6b), 4.06 (dtd, J = 6.2, 4.6, 2.5 Hz, 1H, H5), 2.18 (s, 3H, CH3 Ac), 2.17

(s, 3H, CH3 Ac), 2.10 (s, 3H, CH3 Ac), 2.06 (s, 3H, CH3 Ac), 2.01 (s, 3H, CH3 Ac); 13C NMR (126 MHz,

Page 22: Fluorinated Mannosides Inhibit Cellular Fucosylation

13

CDCl3, major anomer) δ 170.73 (CO Ac), 170.08 (CO Ac), 169.83 (CO Ac), 169.63 (CO Ac), 168.15

(CO Ac), 90.71 (C1), 70.72 (C5), 68.85 (C3), 68.45 (C2), 65.66 (C4), 62.21 (C6), 20.96 (CH3 Ac), 20.87

(CH3 Ac), 20.81 (CH3 Ac), 20.76 (CH3 Ac), 20.74 (CH3 Ac); HRMS (m/z): [M+Na]+ calcd for

C16H22NaO11, 413.10598; found, 413.10835.

1,2,3,4-tetra-O-acetyl-D-rhamnopyranoside (P-D-Rha, 2). D-Rhamnose (230

mg; 1.52 mmol) was dissolved in pyridine (2.2 mL; 18 eq.) and acetic anhydride (1.7 mL; 12 eq.) was

slowly added at 0 ̊C. The mixture was stirred for 16 hrs while slowly warming to r.t.. The mixture was

concentrated in vacuo and the residue was dissolved in DCM, washed with aq. HCl (1 M), sat. aq.

NaHCO3 and brine. The organic layer was dried over anhydrous MgSO4, filtered and concentrated in

vacuo. The residue was purified by silicagel flash column chromatography (0% → 20 EtOAc in Hept)

to afford 2 (466 mg; 1.40 mmol; qu.). TLC: (50:50, EtOAc:Hept v/v) Rf = 0.44. 1H NMR (500 MHz,

CDCl3, major anomer) δ 6.02 (d, J = 1.9 Hz, 1H, H1), 5.32 – 5.29 (m, 1H, H3), 5.25 (dd, J = 3.5, 2.0

Hz, 1H, H2), 5.12 (t, J = 10.0 Hz, 1H, H4), 3.94 (dq, J = 9.8, 6.2 Hz, 1H, H5), 2.17 (s, 3H, CH3 Ac),

2.16 (s, 3H, CH3 Ac), 2.07 (s, 3H, CH3 Ac), 2.01 (s, 3H, CH3 Ac), 1.24 (d, J = 6.2 Hz, 3H, 3x H6); 13C

NMR (126 MHz, CDCl3, major anomer) δ 170.17 (CO Ac), 169.92 (CO Ac), 169.90 (CO Ac), 168.47

(CO Ac), 90.76 (C1), 70.59 (C4), 68.89 (C3), 68.83 (C5), 68.76 (C2), 21.02 (CH3 Ac), 20.89 (CH3 Ac),

20.87 (CH3 Ac), 20.79 (CH3 Ac), 17.56 (C6). HRMS (m/z): [M+Na]+ calcd for C14H20NaO9, 355.10050;

found, 355.10295.

1,2,3,4-tetra-O-acetyl-6-fluoro-D-rhamnopyranoside (P-D-Rha6F, 3).

Synthesis was performed as described previously and the data is identical to the data previously

reported.6 TLC: (60:40, EtOAc:Hept v/v) Rf = 0.53. 1H NMR (500 MHz, CDCl3, major anomer) δ 6.10

(d, J = 1.9 Hz, 1H, H1), 5.39 – 5.36 (m, 2H, H3 & H4), 5.27 (td, J = 2.2, 0.6 Hz, 1H, H2), 4.54 – 4.42

(m, 2H, H6a & H6b), 4.08 – 3.99 (m, 1H, H5), 2.18 (s, 3H, CH3 Ac), 2.17 (s, 3H, CH3 Ac), 2.08 (s, 3H,

CH3 Ac), 2.02 (s, 3H, CH3 Ac); 13C NMR (126 MHz, CDCl3, major anomer) δ 170.11 (CO Ac), 169.87

(CO Ac), 169.57 (CO Ac), 168.14 (CO Ac), 90.62 (C1), 81.29 (d, J = 175.8 Hz, C6), 71.42 (d, J = 19.5

Hz, C5), 68.78 (C3), 68.36 (C2), 65.24 (d, J = 6.6 Hz, C4), 20.94 (CH3 Ac), 20.83 (CH3 Ac), 20.74 (2x

CH3 Ac); 19F NMR (470 MHz, CDCl3, major anomer) δ -232.46 (td, J = 47.1, 22.9 Hz, 6F). HRMS (m/z):

[M+Na]+ calcd for C14H19FNaO9, 373.09108; found, 373.09255.

1,2,3,4-tetra-O-acetyl-6,6-difluoro-D-rhamnopyranoside (P-D-Rha6F2, 4).

Synthesis was performed as described previously.7 TLC: (50:50, EtOAc:Hept v/v) Rf = 0.55. HRMS

(m/z): [M+Na]+ calcd for C14H18F2NaO9, 391.08166; found, 391.08289. 1H NMR (500 MHz, CDCl3) δ

6.12 (d, J = 2.0 Hz, 1H, H1), 5.83 (td, J = 54.2, 3.1 Hz, 1H, H6), 5.53 (t, J = 10.0 Hz, 1H, H4), 5.37 (dd,

J = 10.0, 3.5 Hz, 1H, H3), 5.26 (dd, J = 3.5, 2.0 Hz, 1H, H2), 4.04 (qd, J = 9.9, 3.1 Hz, 1H, H5), 2.19

Page 23: Fluorinated Mannosides Inhibit Cellular Fucosylation

14

(s, 3H, CH3 Ac), 2.18 (s, 3H, CH3 Ac), 2.06 (s, 3H, CH3 Ac), 2.02 (s, 3H, CH3 Ac). 13C NMR (126 MHz,

CDCl3) δ 170.04 (CO Ac), 169.80 (CO Ac), 169.42 (CO Ac), 167.93 (CO Ac), 113.62 (t, J = 245.6 Hz,

C6), 90.27 (C1), 70.79 (t, J = 24.3 Hz, C5), 68.28 (C3), 68.02 (C2), 63.97 (dd, J = 3.7, 2.0 Hz, C4),

20.91 (CH3 Ac), 20.82 (CH3 Ac), 20.72 (CH3 Ac), 20.67 (CH3 Ac); 19F NMR (471 MHz, CDCl3) δ -126.43

(ddd, J = 295.5, 53.8, 9.4 Hz, 6Fa), -130.59 (ddd, J = 295.6, 54.5, 11.2 Hz, 6Fb); HRMS (m/z): [M+Na]+

calcd for C14H18F2NaO9, 391.08166; found, 391.08289.

1,2,3,4-tetra-O-acetyl-6,6,6-trifluoro-D-rhamnopyranoside (P-D-Rha6F3, 5).

Compound 21 (40 mg; 0.12 mmol) was dissolved in pyridine (190 µL; 20 eq.) and acetic anhydride

(110 µL; 10 eq.) was slowly added at 0 ̊C. The mixture was then stirred for 4 hrs at r.t.. The mixture

was concentrated in vacuo and the residue was dissolved in DCM, washed with aq. HCl (1 M) and sat.

aq. NaHCO3. The organic layer was dried over anhydrous MgSO4, filtered and concentrated in vacuo.

The residue was purified by silicagel flash column chromatography (0% → 30% EtOAc in Hept) to

afford 5 (39.4 mg; 102 µmol; 88%). TLC: (50:50, EtOAc:Hept v/v) Rf = 0.46. 1H-NMR (500 MHz, CDCl3)

δ 6.17 (d, J = 2.0 Hz, 1H, H1), 5.59 (t, J = 10.0 Hz, 1H, H4), 5.37 (dd, J = 10.0, 3.4 Hz, 1H, H3), 5.26

(dd, J = 3.4, 2.1 Hz, 1H, H2), 4.22 (dt, J = 10.1, 5.6 Hz, 1H, H5), 2.20 (s, 3H, CH3 Ac), 2.19 (s, 3H, CH3

Ac), 2.06 (s, 3H, CH3 Ac), 2.03 (s, 3H, CH3 Ac). 13C-NMR (126 MHz, CDCl3) δ 169.97 (CO Ac), 169.79

(CO Ac), 169.02 (CO Ac), 167.64 (CO Ac), 122.84 (q, J = 280.8 Hz, C6), 90.16 (C1), 70.22 (q, J = 31.5

Hz, C5), 68.09 (C3), 67.80 (C2), 63.71 (C4), 20.87 (CH3 Ac), 20.81 (CH3 Ac), 20.69 (CH3 Ac), 20.58

(CH3 Ac); 19F NMR (471 MHz, CDCl3) δ -75.56 (d, J = 5.8 Hz, CF3); HRMS (m/z): [M+Na]+ calcd for

C14H17F3NaO9, 409.07224; found, 409.07328.

Di-(S-acetyl-2-thioethyl)-(2,3,4,6-tetra-O-acetyl-α-D-

mannopyranoside) phosphate (P-D-Man-1P, 6). 2,3,4,6-tetra-O-acetyl-D-mannopyranoside8 was

reacted as described in general procedure B and purified by silicagel flash column chromatography

(0% → 50% EtOAc in Hept) affording 6 (61.6 mg; 97.4 µmol; 42%). TLC: before oxidation (80:20,

EtOAc:Hept v/v) Rf = 0.76 & after oxidation (80:20, EtOAc:Hept v/v) Rf = 0.51. 1H NMR (500 MHz,

CDCl3) δ 5.65 (dd, J = 6.5, 1.7 Hz, 1H, H1), 5.38 – 5.32 (m, 3H, H2 & H3 & H4), 4.31 (dd, J = 12.4, 4.7

Hz, 1H, H6a), 4.24 – 4.16 (m, 5H, H5 & 2x CH2-O), 4.14 (dd, J = 12.4, 2.4 Hz, 1H, H6b), 3.20 (td, J =

6.4, 4.7 Hz, 4H, 2x CH2-S), 2.37 (s, 6H, 2x CH3 SAc), 2.18 (s, 3H, CH3 OAc), 2.11 (s, 3H, CH3 OAc),

2.06 (s, 3H, CH3 OAc), 2.00 (s, 3H, CH3 OAc); 13C NMR (126 MHz, CDCl3) δ 194.72 (CO SAc), 194.70

(CO SAc), 170.64 (CO OAc), 169.87 (CO OAc), 169.68 (CO OAc), 169.66 (CO OAc), 95.44 (d, J = 5.4

Hz, C1), 70.59 (C5), 68.85 (d, J = 10.9 Hz, C2), 68.34 (C3), 66.83 – 66.59 (m, 2x CH2-O), 65.41 (C4),

62.13 (C6), 30.66 (CH3 SAc), 30.64 (CH3 SAc), 29.22 (d, J = 7.3 Hz, 2x CH2-S), 20.86 (CH3 OAc),

20.83 (CH3 OAc), 20.78 (CH3 OAc), 20.72 (CH3 OAc). 31P NMR (202 MHz, CDCl3) δ -4.14 (1-OP);

HRMS (m/z): [M+Na]+ calcd for C22H33NaO15PS2, 655.08962; found, 655.08718.

Page 24: Fluorinated Mannosides Inhibit Cellular Fucosylation

15

Di-(S-acetyl-2-thioethyl)-(2,3,4-tri-O-acetyl-α-D-

rhamnopyranoside) phosphate (P-D-Rha-1P, 7). Compound 11 was reacted as described in general

procedure B and purified by silicagel flash column chromatography (0% → 50% EtOAc in Hept)

affording 7 (234 mg; 407 µmol; 43%). TLC: before oxidation (60:40, EtOAc:Hept v/v) Rf = 0.61 & after

oxidation (60:40, EtOAc:Hept v/v) Rf = 0.25. 1H NMR (500 MHz, CDCl3) δ 5.58 (dd, J = 6.3, 1.9 Hz,

1H, H1), 5.34 – 5.29 (m, 2H, H2 & H3), 5.11 (t, J = 10.0 Hz, 1H, H4), 4.23 – 4.05 (m, 5H, 2x CH2-O &

H5), 3.20 (td, J = 6.4, 4.6 Hz, 4H, 2x CH2-S), 2.37 (s, 6H, 2x CH3 SAc), 2.16 (s, 3H, CH3 OAc), 2.07

(s, 3H, CH3 OAc), 1.99 (s, 3H, CH3 OAc), 1.26 (d, J = 6.2 Hz, 3H, 3x H6); 13C NMR (126 MHz, CDCl3)

δ 194.79 (CO SAc), 194.76 (CO SAc), 169.97 (2x CO OAc), 169.78 (CO OAc), 95.56 (d, J = 5.3 Hz,

C1), 70.35 (C4), 69.14 (d, J = 11.0 Hz, C2), 68.73 (C5), 68.38 (C3), 66.72 – 66.48 (m, 2x CH2-O),

30.66 (2x CH3 SAc), 29.25 (d, J = 7.4 Hz, 2x CH2-S), 20.88 (2x CH3 OAc), 20.77 (CH3 OAc), 17.48

(C6); 31P NMR (202 MHz, CDCl3) δ -3.97 (1-OP); HRMS (m/z): [M+Na]+ calcd for C20H31NaO13PS2,

597.08414; found, 597.08291.

Di-(S-acetyl-2-thioethyl)-(2,3,4-tri-O-acetyl-6-fluoro-α-D-

rhamnopyranoside) phosphate (P-D-Rha6F-1P, 8). Compound 12 was reacted as described in

general procedure B and purified by silicagel flash column chromatography (0% → 60% EtOAc in Hept)

affording 8 (10.7 mg; 18.0 µmol; 19%). TLC: before oxidation (60:40, EtOAc:Hept v/v) Rf = 0.61 & after

oxidation (60:40, EtOAc:Hept v/v) Rf = 0.34. 1H NMR (500 MHz, CDCl3) δ 5.66 (dd, J = 6.5, 1.8 Hz,

1H, H1), 5.40 – 5.32 (m, 3H, H2 & H3 & H4), 4.58 – 4.42 (m, 2H, H6a & H6b), 4.24 – 4.15 (m, 5H, H5

& 2x CH2-O), 3.23 – 3.17 (m, 4H, 2x CH2-S), 2.37 (s, 3H, CH3 SAc), 2.36 (s, 2H, CH3 SAc), 2.17 (s,

3H, CH3 OAc), 2.08 (s, 3H, CH3 OAc), 2.01 (s, 3H, CH3 OAc); 13C NMR (126 MHz, CDCl3) δ 194.79

(CO SAc), 194.78 (CO SAc), 169.92 (CO OAc), 169.75 (CO OAc), 169.65 (CO OAc), 95.39 (d, J = 5.4

Hz, C1), 81.21 (d, J = 176.0 Hz, C6), 71.31 (d, J = 19.2 Hz, C5), 68.79 (d, J = 11.2 Hz, C3), 68.31 (C2),

66.85 – 66.62 (m, 2x CH2-O), 64.97 (d, J = 6.7 Hz, C4), 30.66 (2x CH3 SAc), 29.27 – 29.15 (m, 2x CH2-

S), 20.86 (CH3 Ac), 20.79 (CH3 Ac), 20.74 (CH3 Ac); 31P NMR (202 MHz, CDCl3) δ -4.05 (1-OP); 19F

NMR (470 MHz, CDCl3) δ -232.24 (td, J = 47.2, 23.0 Hz, 6F); HRMS (m/z): [M+Na]+ calcd for

C20H30FNaO13PS2, 615.07472; found, 615.07346.

Page 25: Fluorinated Mannosides Inhibit Cellular Fucosylation

16

Di-(S-acetyl-2-thioethyl)-(2,3,4-tri-O-acetyl-6,6-difluoro-α-D-

rhamnopyranoside) phosphate (P-D-Rha6F2-1P, 9). Compound 13 was reacted as described in

general procedure B and purified by silicagel flash column chromatography (0% → 50% EtOAc in Hept)

affording 9 (50.0 mg; 81.9 µmol; 32%). TLC: before oxidation (60:40, EtOAc:Hept v/v) Rf = 0.65 & after

oxidation (60:40, EtOAc:Hept v/v) Rf = 0.42; 1H NMR (500 MHz, CDCl3) δ 5.86 (td, J = 54.0, 3.1 Hz,

1H, H6), 5.68 (dd, J = 6.5, 2.0 Hz, 1H, H1), 5.50 (t, J = 10.0 Hz, 1H, H4), 5.38 (dd, J = 9.9, 3.4 Hz, 1H,

H3), 5.34 (dd, J = 3.5, 2.0 Hz, 1H, H2), 4.20 (dddd, J = 14.4, 9.6, 5.5, 2.5 Hz, 5H, H5 & 2x CH2-O),

3.20 (q, J = 6.3 Hz, 4H, 2x CH2-S), 2.37 (s, 3H, CH3 SAc), 2.37 (s, 3H, CH3 SAc), 2.17 (s, 3H, CH3

OAc), 2.07 (s, 3H, CH3 OAc), 2.01 (s, 3H, CH3 OAc); 13C NMR (126 MHz, CDCl3) δ 194.70 (CO SAc),

194.69 (CO SAc), 169.78 (CO OAc), 169.60 (CO OAc), 169.44 (CO OAc), 113.40 (t, J = 245.7 Hz,

C6), 94.95 (d, J = 5.3 Hz, C1), 70.42 (t, J = 23.9 Hz, C5), 68.39 (d, J = 11.0 Hz, C2), 67.74 (C3), 66.90

– 66.69 (m, 2x CH2-O), 63.87 (t, J = 2.8 Hz, C4), 30.60 (2x CH3 SAc), 29.18 – 29.08 (m, 2x CH2-S),

20.76 (CH3 OAc), 20.65 (2x CH3 OAc); 19F NMR (471 MHz, CDCl3) δ -127.31 (ddd, J = 295.3, 53.9,

10.3 Hz, 6Fa), -130.57 (ddd, J = 295.2, 54.3, 10.3 Hz, 6Fb); 31P NMR (202 MHz, CDCl3) δ -4.05 (1OP);

HRMS (m/z): [M+Na]+ calcd for C20H29F2NaO13PS2, 633.06529; found, 633.06322.

Di-(S-acetyl-2-thioethyl)-(2,3,4-tri-O-acetyl-6,6,6-trifluoro-α-D-

mannopyranoside) phosphate (P-D-Rha6F3-1P, 10). Compound 21 was reacted as described in

general procedure B and purified by silicagel flash column chromatography (0% → 40% EtOAc in Hept)

affording 10 (95.0 mg; 151 µmol; 35%). TLC: before oxidation (60:40, EtOAc:Hept v/v) Rf = 0.65 &

after oxidation (60:40, EtOAc:Hept v/v) Rf = 0.39; 1H-NMR (500 MHz, CDCl3) δ 5.71 (dd, J = 6.6, 1.9

Hz, 1H, H1), 5.58 (t, J = 9.9 Hz, 1H, H4), 5.38 (dd, J = 9.9, 3.4 Hz, 1H, H3), 5.35 (dd, J = 3.4, 2.0 Hz,

1H, H2), 4.41 (dq, J = 11.1, 5.6 Hz, 1H, H5), 4.27 – 4.14 (m, 4H, 2x CH2-O), 3.22 – 3.17 (m, 4H, 2x

CH2-S), 2.37 (s, 3H, CH3 SAc), 2.37 (s, 3H, CH3 SAc), 2.19 (s, 3H, CH3 OAc), 2.07 (s, 3H, CH3 OAc),

2.02 (s, 3H, CH3 OAc); 13C-NMR (126 MHz, CDCl3) δ 194.68 (CO SAc), 194.65 (CO SAc), 169.71 (CO

Ac), 169.60 (CO Ac), 169.03 (CO Ac), 122.84 (q, J = 280.8 Hz, C6), 94.79 (d, J = 5.3 Hz, C1), 69.81

(q, J = 31.6 Hz, C5), 68.16 (d, J = 11.0 Hz, C2), 67.54 (C3), 67.03 – 66.80 (m, 2xCH2-O), 63.48 (C4),

30.61 (CH3 SAc), 30.60 (CH3 SAc), 29.19 – 29.05 (m, 2x CH2-S), 20.75 (CH3 OAc), 20.63 (CH3 OAc),

20.55 (CH3 OAc); 19F NMR (471 MHz, CDCl3) δ -75.48 (d, J = 5.7 Hz, CF3); 31P NMR (202 MHz, CDCl3)

δ -4.05 (1-OP); HRMS (m/z): [M+Na]+ calcd for C20H28F3NaO13PS2, 651.05587; found, 651.05355.

Page 26: Fluorinated Mannosides Inhibit Cellular Fucosylation

17

2,3,4-tri-O-acetyl-D-rhamnopyranoside (11). Compound 2 was reacted as

described in general procedure A and purified by silicagel flash column chromatography (0% → 50%

EtOAc in Hept) affording 11 (323 mg; 1.11 mmol; 83%). TLC: (50:50, EtOAc:Hept v/v) Rf = 0.33. 1H

NMR (500 MHz, CDCl3, major anomer) δ 5.37 (dd, J = 10.1, 3.4 Hz, 1H, H3), 5.27 (dd, J = 3.5, 1.8 Hz,

1H, H2), 5.17 – 5.15 (m, 1H, H1), 5.08 (t, J = 10.0 Hz, 1H, H4), 4.17 – 4.10 (m, 1H, H5), 3.44 (d, J =

3.7 Hz, 1H, 1OH), 2.16 (s, 3H, CH3 Ac), 2.06 (s, 3H, CH3 Ac), 2.00 (s, 3H, CH3 Ac), 1.22 (d, J = 6.3

Hz, 3H, 3x H6). 13C NMR (126 MHz, CDCl3, major anomer) δ 170.45 (CO Ac), 170.30 (CO Ac), 170.26

(CO Ac), 92.23 (C1), 71.27 (C4), 70.43 (C2), 68.97 (C3), 66.50 (C5), 21.05 (CH3 Ac), 20.94 (CH3 Ac),

20.86 (CH3 Ac), 17.59 (C6). HRMS (m/z): [M+Na]+ calcd for C12H18NaO8, 313.08994; found,

313.09287.

2,3,4-tri-O-acetyl-6-fluoro-D-rhamnopyranoside (12). Compound 3 was reacted

as described in general procedure A and purified by silicagel flash column chromatography (0% →

35% EtOAc in Hept) affording 12 (29.0 mg; 94.1 µmol; 66%). TLC: (60:40, EtOAc:Hept v/v) Rf = 0.41. 1H NMR (500 MHz, CDCl3, major anomer) δ 5.44 (dd, J = 10.0, 3.3 Hz, 1H, H3), 5.31 – 5.27 (m, 2H,

H4 & H2), 5.25 (d, J = 1.8 Hz, 1H, H1), 4.54 – 4.42 (m, 2H, H6a & H6b), 4.24 (dddd, J = 22.9, 10.3,

4.4, 2.8 Hz, 1H, H5), 3.96 (s, 1H, 1-OH), 2.16 (s, 3H, CH3 Ac), 2.07 (s, 3H, CH3 Ac), 2.01 (s, 3H, CH3

Ac); 13C NMR (126 MHz, CDCl3, major anomer) δ 170.47 (CO Ac), 170.30 (CO Ac), 170.03 (CO Ac),

92.24 (C1), 81.85 (d, J = 174.2 Hz, C6), 70.11 (C2), 69.29 (d, J = 19.0 Hz, C5), 68.90 (C3), 65.85 (d,

J = 7.0 Hz, C4), 21.00 (CH3 Ac), 20.81 (2x CH3 Ac); 19F NMR (470 MHz, CDCl3, major anomer) δ -

231.87 (td, J = 47.2, 22.9 Hz, 6F); HRMS (m/z): [M+Na]+ calcd for C12H17FNaO8, 331.08051; found,

331.08349.

2,3,4-tri-O-acetyl-6,6-difluoro-D-rhamnopyranoside (13). Compound 4 was

reacted as described in general procedure A and purified by silicagel flash column chromatography

(0% → 40% EtOAc in Hept) affording 13 (84.0 mg; 257 µmol; 83%). TLC: (60:40, EtOAc:Hept v/v) Rf

= 0.51. 1H-NMR (500 MHz, CDCl3, major anomer) δ 5.83 (td, J = 54.3, 3.2 Hz, 1H, H6), 5.47 – 5.40

(m, 2H, H4 & H3), 5.28 – 5.24 (m, 2H, H1 & H2), 4.65 (s, 1H, 1-OH), 4.24 (ddq, J = 13.3, 6.3, 3.3 Hz,

1H, H5), 2.16 (s, 3H, CH3 Ac), 2.06 (s, 3H, CH3 Ac), 2.02 (s, 3H, CH3 Ac); 13C-NMR (126 MHz, CDCl3,

major anomer) δ 170.55 (CO Ac), 170.43 (CO Ac), 170.04 (CO Ac), 114.05 (t, J = 244.8 Hz, C6), 92.13

(C1), 69.85 (C2), 68.92 – 68.47 (m, C5 & C3), 64.84 (t, J = 3.1 Hz, C4), 20.91 (CH3 Ac), 20.74 (CH3

Ac), 20.69 (CH3 Ac); 19F NMR (471 MHz, CDCl3, major anomer) δ -127.38 (ddd, J = 293.8, 54.4, 11.4

Hz, 6Fa), -130.25 (ddd, J = 293.7, 54.3, 10.1 Hz, 6Fb). HRMS (m/z): [M+Na]+ calcd for C12H16F2NaO8,

349.07109; found, 349.07331.

Page 27: Fluorinated Mannosides Inhibit Cellular Fucosylation

18

D-lyxose diethyl dithioacetal (14). D-Lyxose (6.00 g; 40.0 mmol) was dissolved in conc.

HCl (27 mL, 1.5 M) and ethanethiol (6.3 mL; 81.9 mmol; 2.05 eq.; 97% wt.) was added dropwise at

0 ̊C. After stirring for 27 hrs at 0 ̊C the mixture was extracted with EtOAc (4x) and the combined organic

phases were neutralized by portion wise addition of solid NaHCO3, dried over Na2SO4, filtered and

concentrated in vacuo to afford 14 as white crystals (9.90 g; 38.6 mmol; 97%). TLC: (20:80,

MeOH:DCM v/v) Rf = 0.60. 1H NMR (500 MHz, (CD3)2SO)) δ 4.98 (d, J = 6.1 Hz, 1H, 2-OH), 4.46 (t, J

= 5.6 Hz, 1H, 5-OH), 4.23 (d, J = 8.2 Hz, 1H, 3-OH), 4.21 (d, J = 1.5 Hz, 1H, H1), 4.17 (d, J = 6.7 Hz,

1H, 4-OH), 3.84 (ddd, J = 9.3, 6.1, 1.6 Hz, 1H, H2), 3.71 (d, J = 6.8 Hz, 1H, H4), 3.54 (t, J = 8.5 Hz,

1H, H3), 3.44 – 3.35 (m, 2H, 2x H5), 2.68 – 2.59 (m, 4H, 2x CH2 SEt), 1.19 (dt, J = 8.7, 7.4 Hz, 6H, 2x

CH3 SEt); 13C NMR (126 MHz, (CD3)2SO)) δ 73.85 (C2), 70.34 (C3), 69.56 (C4), 62.94 (C5), 54.98

(C1), 24.84 (CH2 SEt), 24.67 (CH2 SEt), 14.74 (CH3 SEt), 14.71 (CH3 SEt); HRMS (m/z): [M+Na]+ calcd

for C9H20NaO4S2, 279.07007; found, 279.07299.

5-O-triisopropylsilyl-D-lyxose diethyl dithioacetal (15). Compound 14 (9.90 g; 38.6

mmol) and 4-dimethylaminopyridine (472 mg; 3.86 mmol; 0.1 eq.) were dissolved in pyridine (110 mL;

0.35 M) and subsequently triisopropylsilylchloride (9.3 mL; 42.5 mmol; 1.1 eq.; 97% wt.) was added

dropwise at 0 ̊C. After 48 hrs most of the starting material had converted and the mixture was

concentrated in vacuo. The residue was dissolved in EtOAc, extracted with water, dried over Na2SO4,

filtered and concentrated in vacuo. The residue was used for the next reaction without further

purification. TLC: (50:50, EtOAc:Hept v/v) Rf = 0.69; 1H NMR (500 MHz, CDCl3) δ 4.23 (d, J = 2.9 Hz,

1H, H1), 4.04 (q, J = 5.3 Hz, 1H, H4), 3.97 – 3.88 (m, 4H, H2 & H3 & H5a & H5b), 3.12 (d, J = 5.3 Hz,

1H, 3-OH), 2.90 (d, J = 5.8 Hz, 1H, 4-OH), 2.87 (d, J = 4.0 Hz, 1H, 2-OH), 2.74 (q, J = 7.4 Hz, 2H, CH2

SEt), 2.67 (q, J = 7.4 Hz, 2H, CH2 SEt), 1.29 (td, J = 7.4, 3.8 Hz, 6H, 2x CH3 SEt), 1.18 – 1.10 (m, 3H,

3x CH TIPS), 1.08 (d, J = 6.5 Hz, 18H, 3x CH3 TIPS); 13C NMR (126 MHz, CDCl3) δ 73.30 (C2), 72.20

(C3), 69.67 (C4), 66.75 (C5), 54.84 (C1), 25.94 (CH2 SEt), 25.82 (CH2 SEt), 18.06 (3x CH3 TIPS),

18.05 (3x CH3 TIPS), 14.85 (CH3 SEt), 14.69 (CH3 SEt), 11.93 (3x CH TIPS). HRMS (m/z): [M+Na]+

calcd for C18H40NaO4S2Si, 435.20350; found, 435.20401.

Page 28: Fluorinated Mannosides Inhibit Cellular Fucosylation

19

2,3,4-tri-O-acetyl-5-O-triisopropylsilyl-D-lyxose diethyl dithioacetal (16). Crude

15 was dissolved in pyridine (190 mL; 60 eq.) and acetic anhydride (109 mL; 30 eq.) was slowly added

at 0 ̊C. The mixture was stirred for 28 hrs while slowly warming to r.t.. The mixture was concentrated

in vacuo and the residue was dissolved in DCM, washed with aq. HCl (1 M), sat. aq. NaHCO3 and

brine. The organic layer was dried over anhydrous MgSO4, filtered and concentrated in vacuo. The

residue was purified by silicagel flash column chromatography (0% → 10% EtOAc in Hept) to afford

16 (12.09 g; 22.444 mmol; 58% over 2 steps). TLC: (10:90, EtOAc:Hept v/v) Rf = 0.24; 1H NMR (500

MHz, CDCl3) δ 5.76 (dd, J = 6.9, 1.8 Hz, 1H, H3), 5.39 – 5.33 (m, 2H, H2 & H4), 3.96 (d, J = 5.4 Hz,

1H, H1), 3.74 – 3.64 (m, 2H, 2x H5), 2.76 – 2.59 (m, 4H, 2x CH2 Et), 2.10 (s, 3H, CH3 Ac), 2.08 (s, 3H,

CH3 Ac), 2.07 (s, 3H, CH3 Ac), 1.25 (dt, J = 8.4, 7.4 Hz, 6H, 2x CH3 Et), 1.10 – 1.02 (m, 21H, 6x CH3

TIPS & 3x CH TIPS); 13C NMR (126 MHz, CDCl3) δ 170.36 (CO Ac), 169.87 (CO Ac), 169.74 (CO Ac),

71.65 (C2), 70.74 (C4), 70.26 (C3), 61.85 (C5), 51.76 (C1), 25.58 (CH2 Et), 25.16 (CH2 Et), 21.17 (CH3

Ac), 20.92 (2x CH3 Ac), 17.99 (6x CH3 TIPS), 14.40 (CH3 Et), 14.16 (CH3 Et), 11.99 (3x CH TIPS);

HRMS (m/z): [M+Na]+ calcd for C24H46NaO7S2Si, 561.23519; found, 561.23388.

2,3,4-tri-O-acetyl-D-lyxose diethyl dithioacetal (17). Compound 16 (12.00 g; 22.27

mmol) was dissolved in THF (150 mL; 0.15 M) in a plastic bottle and hydrogen fluoride pyridine complex

(15 mL; ~70%/30%) was added in portions over 3.5 hrs at 0 ̊C. After stirring for an additional 14 hrs at

r.t. all starting material was consumed and the mixture was diluted with EtOAc (100 mL) and carefully

quenched with sat. aq. NaHCO3. The organic layer was washed with 10% aq. CuSO4 (2x) and the

combined organic layers were dried over anhydrous MgSO4, filtered and concentrated in vacuo. The

residue was purified by silicagel flash column chromatography (0% → 40% EtOAc in Hept) to afford

17 (4.85 g; 12.7 mmol; 57%). TLC: (40:60, EtOAc:Hept v/v) Rf = 0.18. 1H NMR (400 MHz, CDCl3) δ

5.59 (dd, J = 8.8, 1.7 Hz, 1H, H3), 5.39 (dd, J = 8.8, 3.5 Hz, 1H, H2), 5.06 (ddd, J = 7.8, 6.2, 1.7 Hz,

1H, H4), 3.86 (d, J = 3.5 Hz, 1H, H1), 3.53 (dt, J = 12.0, 5.9 Hz, 1H, H5a), 3.38 (dd, J = 11.7, 7.8 Hz,

1H, H5b), 2.80 (d, J = 7.1 Hz, 1H, 5-OH), 2.71 – 2.52 (m, 4H, 2x CH2 SEt), 2.11 (s, 3H, CH3 Ac), 2.02

(s, 3H, CH3 Ac), 2.01 (s, 3H, CH3 Ac), 1.18 (td, J = 7.4, 5.9 Hz, 6H, 2x CH3 SEt); 13C-NMR (101 MHz,

CDCl3) δ 171.15 (CO Ac), 170.54 (CO Ac), 169.64 (CO Ac), 70.86 (C2), 70.62 (C4), 70.45 (C3), 60.05

(C5), 51.56 (C1), 25.81 (CH2 SEt), 25.36 (CH2 SEt), 20.93 (CH3 Ac), 20.90 (CH3 Ac), 20.70 (CH3 Ac),

14.36 (CH3 SEt), 14.15 (CH3 SEt); HRMS (m/z): [M+Na]+ calcd for C15H26NaO7S2, 405.10176; found,

405.10253.

Page 29: Fluorinated Mannosides Inhibit Cellular Fucosylation

20

2,3,4-tri-O-acetyl-5-al-D-lyxose diethyl dithioacetal (18). Compound 17 (4.85 g; 12.7

mmol) was dissolved in dry DCM (106 mL; 0.12 M) and subsequently NaHCO3 (10.7 g; 127 mmol; 10

eq.) and Dess-Martin periodinane (8.07 g; 19.0 mmol; 1.5 eq.) were added at 0 ̊C. After 10 minutes the

ice bath was removed and the mixture was stirred for an additional 3 hrs at r.t.. The mixture was diluted

with DCM (100 mL) and an aqueous solution (150 mL) of Na2SO3 (15 g) and NaHCO3 (5 g) was added

at 0 ̊C and stirred for 30 min. The organic phase was separated, dried over anhydrous MgSO4, filtered

and concentrated in vacuo. The residue was purified by silicagel flash column chromatography (0% →

60% EtOAc in Hept) to afford 18 (3.65 g; 9.59 mmol; 76%) which was immediately used for the next

step. TLC: (50:50, EtOAc:Hept v/v) Rf = 0.32. HRMS (m/z): [M+MeOH+Na]+ calcd for C16H28NaO8S2,

435.11233; found, 435.11239

2,3,4-tri-O-acetyl-6,6,6-trifluoro-D-rhamnose diethyl dithioacetal (19)

& 2,3,4-tri-O-acetyl-6,6,6-trifluoro-L-gulose diethyl dithioacetal (20). Synthetic procedure was

adapted from a previously described procedure.9 Compound 18 and trifluoromethyl(trimethyl)silane

(1.45 mL; 9.56 mmol; 1.6 eq; 97% wt.) were dissolved in dry THF (20 mL; 0.3 M) and a

tetrabutylammonium fluoride solution (598 µL; 598 µmol; 0.1 eq.; 1.0 M in THF) was added dropwise

at 0 ̊C. After stirring at 0 ̊C for 1.5 hrs the mixture was concentrated in vacuo. The residue was dissolved

in CHCl3, washed with water, dried over anhydrous Na2SO4, filtered and concentrated in vacuo. To

remove the 5-OTMS groups, the residue was dissolved in 80% AcOH (0.25 M; 24 mL) and kept for 3

hrs at 50 ̊C. The mixture was concentrated in vacuo and the residue was purified by silicagel flash

column chromatography (0% → 20% → 30% EtOAc in Hept) to afford first 19 (813 mg; 1.80 mmol;

30%) and then 20 (816; 1.81 mmol; 30%). Compound 19: TLC: (30:70, EtOAc:Hept v/v) Rf = 0.23. 1H-

NMR (500 MHz, CDCl3) δ 5.82 (dd, J = 8.4, 1.3 Hz, 1H, H3), 5.48 (dd, J = 9.5, 1.3 Hz, 1H, H4), 5.33

(dd, J = 8.4, 4.2 Hz, 1H, H2), 3.97 (d, J = 4.2 Hz, 1H, H1), 3.81 (dq, J = 9.4, 6.3 Hz, 1H, H5), 2.77 –

2.60 (m, 5H, OH & 2x CH2 SEt), 2.22 (s, 3H, CH3 Ac), 2.10 (s, 3H, CH3 Ac), 2.07 (s, 3H, CH3 Ac), 1.26

(q, J = 7.3 Hz, 7H, 2x CH3 SEt); 13C-NMR (126 MHz, CDCl3) δ 172.28 (CO), 169.85 (CO), 169.47 (CO),

124.34 (q, J = 282.2 Hz, C6), 71.14 (C3), 71.06 (C2), 68.05 – 67.27 (m, C5&C4), 51.67 (C1), 26.06

(CH2 SEt), 25.68 (CH2 SEt), 21.22 (CH3 Ac), 20.95 (CH3 Ac), 20.68 (CH3 Ac), 14.40 (CH3 SEt), 14.26

(CH3 SEt). 19F NMR (471 MHz, CDCl3) δ -75.75 (d, J = 6.4 Hz, CF3); HRMS (m/z): [M+Na]+ calcd for

C16H25F3NaO7S2, 473.08915; found, 473.08916. Compound 20: TLC: (30:70, EtOAc:Hept v/v) Rf =

0.18. 1H-NMR (500 MHz, CDCl3) δ 5.79 (dd, J = 8.0, 2.5 Hz, 1H, H3), 5.60 (dd, J = 2.5, 1.4 Hz, 1H,

Page 30: Fluorinated Mannosides Inhibit Cellular Fucosylation

21

H4), 5.33 (dd, J = 8.0, 4.2 Hz, 1H, H2), 4.29 – 4.21 (m, 1H, H5), 3.93 (d, J = 4.2 Hz, 1H, H1), 2.79 (d,

J = 9.8 Hz, 1H, OH), 2.74 – 2.61 (m, 4H, 2x CH2 SEt), 2.13 (s, 3H, CH3 Ac), 2.13 (s, 3H, CH3 Ac), 2.10

(s, 3H, CH3 Ac), 1.25 (q, J = 7.5 Hz, 7H, 2x CH3 SEt); 13C-NMR (126 MHz, CDCl3) δ 170.13 (CO),

169.94 (CO), 169.14 (CO), 123.68 (q, J = 283.1 Hz, C6), 72.06 (C3), 71.20 (C2), 70.38 (q, J = 31.4

Hz, C5), 65.43 (d, J = 1.9 Hz, C4), 51.40 (C1), 25.86 (CH2 SEt), 25.68 (CH2 SEt), 21.07 (CH3 Ac),

20.93 (CH3 Ac), 20.68 (CH3 Ac), 14.41 (CH3 SEt), 14.21 (CH3 SEt); 19F NMR (471 MHz, CDCl3) δ -

78.18 (d, J = 6.8 Hz, CF3); HRMS (m/z): [M+Na]+ calcd for C16H25F3NaO7S2, 473.08915; found,

473.08860.

2,3,4-tri-O-acetyl-6,6,6-trifluoro-D-rhamnopyranoside (21). Compound 19 (763

mg; 1.69 mmol) was dissolved in a 5:1 acetone/water mixture (34 mL; 0.05 M) and NBS (1.51 g; 8.47

mmol; 5 eq.) was added at 0 ̊C. After stirring for 45 min at 0 ̊C, subsequently sat. aq. NaHCO3 (15 mL)

and sat. aq. Na2S2O3 (15 mL) were added and the mixture was stirred for 15 min. The organic solvent

was evaporated in vacuo and the remaining mixture was extracted with Et2O (2x), dried over anhydrous

MgSO4, filtered and concentrated in vacuo. The residue was purified by silicagel flash column

chromatography (0% → 30% EtOAc in Hept) to afford 21 (225 mg; 654 µmol; 39%). TLC: (50:50,

EtOAc:Hept v/v) Rf = 0.40; 1H-NMR (500 MHz, CDCl3) δ 5.54 (t, J = 9.9 Hz, 1H, H4), 5.44 (dd, J = 10.0,

3.3 Hz, 1H, H3), 5.32 (s, 1H, H1), 5.26 (dd, J = 3.3, 1.9 Hz, 1H, H2), 4.42 (dq, J = 9.9, 5.9 Hz, 1H, H5),

4.20 (s, 1H, OH), 2.17 (s, 3H, CH3 Ac), 2.06 (s, 3H, CH3 Ac), 2.02 (s, 3H, CH3 Ac); 13C-NMR (126 MHz,

CDCl3) δ 170.50 (CO Ac), 170.31 (CO Ac), 169.54 (CO Ac), 123.47 (q, J = 280.4 Hz, C6), 92.26 (C1),

69.56 (C2), 68.71 – 67.90 (m, C5 & C3), 64.32 (C4), 20.93 (CH3 Ac), 20.74 (CH3 Ac), 20.62 (CH3 Ac); 19F NMR (471 MHz, CDCl3) δ -75.44 (d, J = 6.0 Hz, 3x 6F); HRMS (m/z): [M+Na]+ calcd for

C12H15F3NaO8, 367.06167; found, 367.06424.

S-acetyl-2-thioethyl alcohol (22). Synthesis was performed as described previously

and the data was identical.10 TLC: (5:95, EtOAc:DCM v/v) Rf = 0.15. 1H NMR (500 MHz, CDCl3) δ 3.67

(t, J = 6.3 Hz, 2H, CH2-O), 3.01 (t, J = 6.3 Hz, 2H, CH2-S), 2.30 (s, 3H, CH3 Ac); 13C NMR (126 MHz,

CDCl3) δ 196.42 (CO), 61.48 (CH2OH), 31.89 (CH2-S), 30.60 (CH3 Ac)

Di-(S-acetyl-2-thioethyl)-N,N-diethylphosphoroamidite (23). Compound 22

(5.88 g; 48.9 mmol; 2 eq.) was dissolved in anhydrous THF (31 mL; 1.6 M) and subsequently molecular

sieves (4Å) and anhydrous TEA (6.1 mL; 44.0 mmol; 1.8 eq.) were added. The mixture was cooled to

0° C, diethylphosphoramidous dichloride (3.56 mL; 24.5 mmol; 1 eq.). After stirring for 2 hrs the mixture

was diluted with heptane, filtered and the filtrate was concentrated and directly purified by silicagel

flash column chromatography (3% TEA in Hept) to afford 23 (3.90 g; 11.4 mmol; 47%). The dry

phosphoroamidite reagent was stored at -80 ̊C and after 11 months no changes in 1H NMR were

Page 31: Fluorinated Mannosides Inhibit Cellular Fucosylation

22

observed. TLC: (30:67:3, EtOAc:Hept:TEA v/v) Rf = 0.53. 1H NMR (500 MHz, CDCl3) δ 3.73 (dddt, J

= 32.3, 10.7, 8.4, 6.5 Hz, 4H), 3.11 (t, J = 6.6 Hz, 4H), 3.06 (dq, J = 9.5, 7.1 Hz, 4H), 2.34 (s, 6H), 1.06

(t, J = 7.1 Hz, 6H); 13C NMR (126 MHz, CDCl3) δ 195.08, 61.76 (d, J = 16.0 Hz), 37.31 (d, J = 20.7

Hz), 30.49, 30.37 (d, J = 6.5 Hz), 14.98 (d, J = 3.2 Hz); 31P NMR (202 MHz, CDCl3) δ 147.22

(PNEt2(OCH2)2); HRMS (m/z): [M+Na]+ calcd for C12H24NNaO5PS2, 380.07312; found, 380.07340.

Page 32: Fluorinated Mannosides Inhibit Cellular Fucosylation

23

References

1. van Scherpenzeel, M.; Conte, F.; Büll, C.; Ashikov, A.; Hermans, E.; Willems, A.; van Tol, W.; Kragt, E.; Moret, E. E.; Heise, T.; Langereis, J. D.; Rossing, E.; Zimmermann, M.; Rubio-Gozalbo, M. E.; de Jonge, M. I.; Adema, G. J.; Zamboni, N.; Boltje, T.; Lefeber, D. J., Dynamic analysis of sugar metabolism reveals the mechanisms of action of synthetic sugar analogs. bioRxiv 2020, 2020.09.15.288712. 2. MacLean, B.; Tomazela, D. M.; Shulman, N.; Chambers, M.; Finney, G. L.; Frewen, B.; Kern, R.; Tabb, D. L.; Liebler, D. C.; MacCoss, M. J., Skyline: an open source document editor for creating and analyzing targeted proteomics experiments. Bioinformatics 2010, 26 (7), 966-968. 3. Pfeiffer, M.; Johansson, C.; Krojer, T.; Kavanagh, K. L.; Oppermann, U.; Nidetzky, B., A Parsimonious Mechanism of Sugar Dehydration by Human GDP-Mannose-4,6-dehydratase. ACS Catalysis 2019, 9 (4), 2962-2968. 4. Yu, S.-H.; Boyce, M.; Wands, A. M.; Bond, M. R.; Bertozzi, C. R.; Kohler, J. J., Metabolic labeling enables selective photocrosslinking of O-GlcNAc-modified proteins to their binding partners. Proceedings of the National Academy of Sciences 2012, 109 (13), 4834-4839. 5. Timmer, B. J. J.; Flos, M. A.; Jørgensen, L. M.; Proverbio, D.; Altun, S.; Ramström, O.; Aastrup, T.; Vincent, S. P., Spatially well-defined carbohydrate nanoplatforms: synthesis, characterization and lectin interaction study. Chemical Communications 2016, 52 (83), 12326-12329. 6. Khan, S. H.; Jain, R. K.; Abbas, S. A.; Matta, K. L., Synthesis of some monodeoxyfluorinated methyl and 4-nitrophenyl α-d-mannobiosides and a related 4-nitrophenyl α-d-mannotrioside. Carbohydrate Research 1990, 198 (2), 259-273. 7. Crich, D.; Vinogradova, O., Synthesis and Glycosylation of a Series of 6-Mono-, Di-, and Trifluoro S-Phenyl 2,3,4-Tri-O-benzyl-thiorhamnopyranosides. Effect of the Fluorine Substituents on Glycosylation Stereoselectivity. Journal of the American Chemical Society 2007, 129 (38), 11756-11765. 8. Tosin, M.; Murphy, P. V., Synthesis of Structurally Defined Scaffolds for Bivalent Ligand Display Based on Glucuronic Acid Anilides. The Degree of Tertiary Amide Isomerism and Folding Depends on the Configuration of a Glycosyl Azide. The Journal of Organic Chemistry 2005, 70 (10), 4107-4117. 9. Nakai, K.; Takagi, Y.; Tsuchiya, T., Synthesis and antitumor activity of 7-O-[2,6-dideoxy-2-fluoro-5-C-(trifluoromethyl)-α-l-talopyranosyl]-daunomycinone and -adriamycinone. Carbohydrate Research 1999, 316 (1), 47-57. 10. Liras, M.; García, O.; Guarrotxena, N.; Palacios-Cuesta, M.; Quijada-Garrido, I., Versatile thiolated thermosensitive polymers synthesized by ATRP of MEO2MA and AcSEMA, a new methacrylic monomer with a protected thiol group. Polymer Chemistry 2013, 4 (24), 5751-5759.

Page 33: Fluorinated Mannosides Inhibit Cellular Fucosylation

24

Page 34: Fluorinated Mannosides Inhibit Cellular Fucosylation

25

Page 35: Fluorinated Mannosides Inhibit Cellular Fucosylation

26

Page 36: Fluorinated Mannosides Inhibit Cellular Fucosylation

27

[EMPTY]

Page 37: Fluorinated Mannosides Inhibit Cellular Fucosylation

28

Page 38: Fluorinated Mannosides Inhibit Cellular Fucosylation

29

[EMPTY]

Page 39: Fluorinated Mannosides Inhibit Cellular Fucosylation

30

Page 40: Fluorinated Mannosides Inhibit Cellular Fucosylation

31

[EMPTY]

Page 41: Fluorinated Mannosides Inhibit Cellular Fucosylation

32

Page 42: Fluorinated Mannosides Inhibit Cellular Fucosylation

33

[EMPTY]

Page 43: Fluorinated Mannosides Inhibit Cellular Fucosylation

34

Page 44: Fluorinated Mannosides Inhibit Cellular Fucosylation

35

[EMPTY]

Page 45: Fluorinated Mannosides Inhibit Cellular Fucosylation

36

Page 46: Fluorinated Mannosides Inhibit Cellular Fucosylation

37

Page 47: Fluorinated Mannosides Inhibit Cellular Fucosylation

38

Page 48: Fluorinated Mannosides Inhibit Cellular Fucosylation

39

Page 49: Fluorinated Mannosides Inhibit Cellular Fucosylation

40

Page 50: Fluorinated Mannosides Inhibit Cellular Fucosylation

41

Page 51: Fluorinated Mannosides Inhibit Cellular Fucosylation

42

Page 52: Fluorinated Mannosides Inhibit Cellular Fucosylation

43

Page 53: Fluorinated Mannosides Inhibit Cellular Fucosylation

44

[EMPTY]

Page 54: Fluorinated Mannosides Inhibit Cellular Fucosylation

45

Page 55: Fluorinated Mannosides Inhibit Cellular Fucosylation

46

[EMPTY]

Page 56: Fluorinated Mannosides Inhibit Cellular Fucosylation

47

Page 57: Fluorinated Mannosides Inhibit Cellular Fucosylation

48

Page 58: Fluorinated Mannosides Inhibit Cellular Fucosylation

49

Page 59: Fluorinated Mannosides Inhibit Cellular Fucosylation

50

Page 60: Fluorinated Mannosides Inhibit Cellular Fucosylation

51

Page 61: Fluorinated Mannosides Inhibit Cellular Fucosylation

52

[EMPTY]

Page 62: Fluorinated Mannosides Inhibit Cellular Fucosylation

53

Page 63: Fluorinated Mannosides Inhibit Cellular Fucosylation

54

[EMPTY]

Page 64: Fluorinated Mannosides Inhibit Cellular Fucosylation

55

Page 65: Fluorinated Mannosides Inhibit Cellular Fucosylation

56

[EMPTY]

Page 66: Fluorinated Mannosides Inhibit Cellular Fucosylation

57

Page 67: Fluorinated Mannosides Inhibit Cellular Fucosylation

58

Page 68: Fluorinated Mannosides Inhibit Cellular Fucosylation

59

[EMPTY]