Abstract
Fucosylation, the conjugation of glycoproteins and glycolipids with the dietary sugar l-fucose, can have key functional and regulatory roles across a range of normal biological and developmental processes. Although the full repertoire of fucosylated proteins and their direct influence on signalling and cellular behaviour remains incompletely understood, it is not surprising that deregulated fucosylation has been increasingly associated with disease contexts, particularly cancer. Importantly, fucosylation regulates the biology of immune and other stromal cells, and emerging studies have elucidated how pathological aberrations in fucosylation can deregulate signalling that governs cellular interactions in the tumour microenvironment, thereby influencing tumour progression and therapeutic responses. Accordingly, fucosylated glycoproteins and glycans have been reported to exhibit potential biomarker utility, associating with cancer type and staging. Notably, fucosylation appears to be therapeutically actionable, as simply administering l-fucose orally can suffice to suppress tumour growth and stimulate antitumour immune responses in preclinical models. However, given that the blockade of fucosylation machinery can elicit similar antitumour effects reflects the diversity of cell-intrinsic and cell-extrinsic roles that fucosylation can divergently have across the tumour microenvironment. Here, we review recent glycobiology discoveries that shed light on the complexity of fucosylation, its mechanistic roles in immune and tumour biology, and how it might be strategically leveraged for the treatment of cancer.
This is a preview of subscription content, access via your institution
Access options
Access Nature and 54 other Nature Portfolio journals
Get Nature+, our best-value online-access subscription
$32.99 / 30 days
cancel any time
Subscribe to this journal
Receive 12 print issues and online access
$259.00 per year
only $21.58 per issue
Buy this article
- Purchase on SpringerLink
- Instant access to the full article PDF.
USD 39.95
Prices may be subject to local taxes which are calculated during checkout



Similar content being viewed by others
References
Peixoto, A., Relvas-Santos, M., Azevedo, R., Santos, L. L. & Ferreira, J. A. Protein glycosylation and tumor microenvironment alterations driving cancer hallmarks. Front. Oncol. 9, 380 (2019).
Varki, A. et al. (eds) Essentials of Glycobiology 4th edn (Cold Spring Harbor Laboratory, 2022). This textbook provides core concepts, pathways and methods of glycobiology, including fucose metabolism and fucosylated glycoconjugates, that underpin interpretation of much of the literature discussed in this Progress.
Adhikari, E. et al. L-Fucose, a sugary regulator of antitumor immunity and immunotherapies. Mol. Carcinog. 61, 439–453 (2022). This review focuses specifically on LF as a regulator of antitumour immunity and immunotherapy responses, motivating the concept of dietary or pharmacological LF manipulation in cancer.
Ng, B. G., Sosicka, P., Xia, Z. & Freeze, H. H. GLUT1 is a highly efficient L-fucose transporter. J. Biol. Chem. 299, 102738 (2023).
Kierans, S. J. & Taylor, C. T. Glycolysis: a multifaceted metabolic pathway and signaling hub. J. Biol. Chem. 300, 107906 (2024).
Sosicka, P. et al. Origin of cytoplasmic GDP-fucose determines its contribution to glycosylation reactions. J. Cell Biol. 221, e202205038 (2022). This study uncovers how the salvage versus de novo pathways give rise to distinct and dynamic intracellular pools of GDP-fucose that are ultimately differentially incorporated into glycans and glycoproteins.
Becker, D. J. & Lowe, J. B. Fucose: biosynthesis and biological function in mammals. Glycobiology 13, 41R–53R (2003). This comprehensive review lays out the biosynthetic pathways and systemic functions of fucose in mammals, forming a conceptual and biochemical foundation for subsequent work on fucosylation in cancer.
Skurska, E. & Olczak, M. Interplay between de novo and salvage pathways of GDP-fucose synthesis. PLoS ONE 19, e0309450 (2024). This study dissects how de novo and salvage GDP-fucose pathways interact, clarifying how exogenous LF is channelled into cellular fucosylation and, thereby, influences tumour and immune cell glycosylation.
Schneider, M., Al-Shareffi, E. & Haltiwanger, R. S. Biological functions of fucose in mammals. Glycobiology 27, 601–618 (2017). This comprehensive review summarizes the diverse biological roles of fucose in mammals, providing essential context for how perturbations in fucosylation can contribute to cancer and other diseases.
Hao, H. et al. FUT10 and FUT11 are protein O-fucosyltransferases that modify protein EMI domains. Nat. Chem. Biol. 21, 598–610 (2025). This study identifies FUT10 and FUT11 as protein O-fucosyltransferases for EMI domain-containing proteins, expanding the enzymatic repertoire and substrate scope of fucosylation with implications for cancer signalling networks.
Cheng, J. Y. et al. Tumor-associated glycan exploits adenosine receptor 2A signaling to facilitate immune evasion. Adv. Sci. 12, e2416501 (2025).
Groux-Degroote, S. & Delannoy, P. Cancer-associated glycosphingolipids as tumor markers and targets for cancer immunotherapy. Int. J. Mol. Sci. 22, 6145 (2021).
Prochazkova, J. et al. Single-cell profiling of surface glycosphingolipids opens a new dimension for deconvolution of breast cancer intratumoral heterogeneity and phenotypic plasticity. J. Lipid Res. 65, 100609 (2024).
Sigal, D. S., Hermel, D. J., Hsu, P. & Pearce, T. The role of Globo H and SSEA-4 in the development and progression of cancer, and their potential as therapeutic targets. Future Oncol. 18, 117–134 (2022).
Blanas, A., Sahasrabudhe, N. M., Rodriguez, E., van Kooyk, Y. & van Vliet, S. J. Fucosylated antigens in cancer: an alliance toward tumor progression, metastasis, and resistance to chemotherapy. Front. Oncol. 8, 39 (2018).
Mathieu, S. et al. Transgene expression of α(1,2)-fucosyltransferase-I (FUT1) in tumor cells selectively inhibits sialyl-Lewis x expression and binding to E-selectin without affecting synthesis of sialyl-Lewis a or binding to P-selectin. Am. J. Pathol. 164, 371–383 (2004).
Osman, N. et al. Combined transgenic expression of α-galactosidase and α1,2-fucosyltransferase leads to optimal reduction in the major xenoepitope Galα(1,3)Gal. Proc. Natl Acad. Sci. USA 94, 14677–14682 (1997).
Prieto, P. A. et al. Expression of human H-type α1,2-fucosyltransferase encoding for blood group H(O) antigen in Chinese hamster ovary cells: evidence for preferential fucosylation and truncation of polylactosamine sequences. J. Biol. Chem. 272, 2089–2097 (1997).
Zerfaoui, M., Fukuda, M., Sbarra, V., Lombardo, D. & El-Battari, A. α(1,2)-Fucosylation prevents sialyl Lewis x expression and E-selectin-mediated adhesion of fucosyltransferase VII-transfected cells. Eur. J. Biochem. 267, 53–61 (2000).
Nakahashi, H. et al. Aberrant glycosylation in pancreatic ductal adenocarcinoma 3D organoids is mediated by KRAS mutations. J. Oncol. 2024, 1529449 (2024).
Lau, E. et al. The transcription factor ATF2 promotes melanoma metastasis by suppressing protein fucosylation. Sci. Signal. 8, ra124 (2015).
Agrawal, P. et al. A systems biology approach identifies FUT8 as a driver of melanoma metastasis. Cancer Cell 31, 804–819.e7 (2017). This integrated systems biology study identifies FUT8 as a key driver of melanoma metastasis in vivo, establishing core fucosylation as a major regulator and potential therapeutic target in tumour progression.
Liu, Q. et al. Androgen drives melanoma invasiveness and metastatic spread by inducing tumorigenic fucosylation. Nat. Commun. 15, 1148 (2024). This study reveals that androgen signalling induces a tumorigenic fucosylation programme mediated by FUT4 that drives melanoma invasion and metastasis, linking hormonal cues to pro-metastatic fucosylation changes.
Kyunai, Y. M., Sakamoto, M., Koreishi, M., Tsujino, Y. & Satoh, A. Fucosyltransferase 8 (FUT8) and core fucose expression in oxidative stress response. PLoS ONE 18, e0281516 (2023).
Li, J., Hsu, H. C., Mountz, J. D. & Allen, J. G. Unmasking fucosylation: from cell adhesion to immune system regulation and diseases. Cell Chem. Biol. 25, 499–512 (2018). This review gives a broad overview of the roles of fucosylation in cell adhesion, immune regulation and disease, positioning cancer-related fucosylation within the wider physiological landscape of fucose-regulated biology.
Li, F. et al. α1,6-Fucosyltransferase (FUT8) regulates the cancer-promoting capacity of cancer-associated fibroblasts (CAFs) by modifying EGFR core fucosylation (CF) in non-small cell lung cancer (NSCLC). Am. J. Cancer Res. 10, 816–837 (2020).
Cheng, L. et al. FUT family mediates the multidrug resistance of human hepatocellular carcinoma via the PI3K/Akt signaling pathway. Cell Death Dis. 4, e923 (2013).
Lopez-Cortes, R., Correa Pardo, I., Muinelo-Romay, L., Fernandez-Briera, A. & Gil-Martin, E. Core fucosylation mediated by the FucT-8 enzyme affects TRAIL-induced apoptosis and sensitivity to chemotherapy in human SW480 and SW620 colorectal cancer cells. Int. J. Mol. Sci. 24, 11879 (2023).
Shimoyama, H. et al. Partial silencing of fucosyltransferase 8 gene expression inhibits proliferation of Ishikawa cells, a cell line of endometrial cancer. Biochem. Biophys. Rep. 22, 100740 (2020).
Bastian, K. et al. FUT8 is a critical driver of prostate tumour growth and can be targeted using fucosylation inhibitors. Cancer Med. 14, e70959 (2025).
Tu, C. F., Wu, M. Y., Lin, Y. C., Kannagi, R. & Yang, R. B. FUT8 promotes breast cancer cell invasiveness by remodeling TGF-β receptor core fucosylation. Breast Cancer Res. 19, 111 (2017).
Tada, K. et al. Fucosyltransferase 8 plays a crucial role in the invasion and metastasis of pancreatic ductal adenocarcinoma. Surg. Today 50, 767–777 (2020).
Bastian, K., Scott, E., Elliott, D. J. & Munkley, J. FUT8 alpha-(1,6)-fucosyltransferase in cancer. Int. J. Mol. Sci. 22, 455 (2021).
Loong, J. H. et al. Glucose deprivation-induced aberrant FUT1-mediated fucosylation drives cancer stemness in hepatocellular carcinoma. J. Clin. Invest. 131, e143377 (2021).
Cao, W. et al. Knockdown of FUT11 inhibits the progression of gastric cancer via the PI3K/AKT pathway. Heliyon 9, e17600 (2023).
He, L., Guo, Z., Wang, W., Tian, S. & Lin, R. FUT2 inhibits the EMT and metastasis of colorectal cancer by increasing LRP1 fucosylation. Cell Commun. Signal. 21, 63 (2023).
Guo, Z., He, L., Wang, W., Tian, S. & Lin, R. FUT2-dependent fucosylation of LAMP1 promotes the apoptosis of colorectal cancer cells by regulating the autophagy-lysosomal pathway. Cancer Lett. 619, 217643 (2025).
Dong, C. et al. FUT2 promotes colorectal cancer metastasis by reprogramming fatty acid metabolism via YAP/TAZ signaling and SREBP-1. Commun. Biol. 7, 1297 (2024).
Cheng, T. C. et al. Down-regulation of α-L-fucosidase 1 expression confers inferior survival for triple-negative breast cancer patients by modulating the glycosylation status of the tumor cell surface. Oncotarget 6, 21283–21300 (2015).
Waidely, E., Al-Youbi, A. O., Bashammakh, A. S., El-Shahawi, M. S. & Leblanc, R. M. Alpha-L-fucosidase immunoassay for early detection of hepatocellular carcinoma. Anal. Chem. 89, 9459–9466 (2017).
Bonin, S. et al. Reduced expression of alpha-L-fucosidase-1 (FUCA-1) predicts recurrence and shorter cancer specific survival in luminal B LN + breast cancer patients. Oncotarget 9, 15228–15238 (2018).
Vecchio, G. et al. Human α-L-fucosidase-1 attenuates the invasive properties of thyroid cancer. Oncotarget 8, 27075–27092 (2017).
Ezawa, I. et al. Novel p53 target gene FUCA1 encodes a fucosidase and regulates growth and survival of cancer cells. Cancer Sci. 107, 734–745 (2016).
Tsuchida, N., Ikeda, M. A., Iotashino, U., Grieco, M. & Vecchio, G. FUCA1 is induced by wild-type p53 and expressed at different levels in thyroid cancers depending on p53 status. Int. J. Oncol. 50, 2043–2048 (2017).
Baudot, A. D. et al. p53 directly regulates the glycosidase FUCA1 to promote chemotherapy-induced cell death. Cell Cycle 15, 2299–2308 (2016).
Xu, L. et al. Downregulation of α-L-fucosidase 1 suppresses glioma progression by enhancing autophagy and inhibiting macrophage infiltration. Cancer Sci. 111, 2284–2296 (2020).
Jia, L. et al. The function of fucosylation in progression of lung cancer. Front. Oncol. 8, 565 (2018).
Liang, W. et al. Ablation of core fucosylation attenuates the signal transduction via T cell receptor to suppress the T cell development. Mol. Immunol. 112, 312–321 (2019).
Liang, W. et al. Core fucosylation of the T cell receptor is required for T cell activation. Front. Immunol. 9, 78 (2018).
Li, W. et al. Core fucosylation of IgG B cell receptor is required for antigen recognition and antibody production. J. Immunol. 194, 2596–2606 (2015).
Li, W. et al. Core fucosylation of μ heavy chains regulates assembly and intracellular signaling of precursor B cell receptors. J. Biol. Chem. 287, 2500–2508 (2012).
Moriwaki, K. & Miyoshi, E. Fucosylation and gastrointestinal cancer. World J. Hepatol. 2, 151–161 (2010).
Lester, D. K. et al. Fucosylation of HLA-DRB1 regulates CD4+ T cell-mediated anti-melanoma immunity and enhances immunotherapy efficacy. Nat. Cancer 4, 222–239 (2023). This study reveals that fucosylation of melanoma-expressed HLA-DRB1 elicits CD4+ T cell-mediated antitumour immunity, whereas LF enhances intrinsic CD4+ T cell signalling to promote a central memory phenotype, collectively demonstrating that modulating fucosylation can promote antitumour immunity and improved immunotherapy efficacy.
Ilca, F. T. & Boyle, L. H. The glycosylation status of MHC class I molecules impacts their interactions with TAPBPR. Mol. Immunol. 139, 168–176 (2021).
Ryan, S. O. & Cobb, B. A. Roles for major histocompatibility complex glycosylation in immune function. Semin. Immunopathol. 34, 425–441 (2012).
Burton, C. et al. The functional role of L-fucose on dendritic cell function and polarization. Front. Immunol. 15, 1353570 (2024).
Iijima, J. et al. Core fucose is critical for CD14-dependent Toll-like receptor 4 signaling. Glycobiology 27, 1006–1015 (2017).
Tong, X. et al. Fucosylation promotes cytolytic function and accumulation of NK cells in B cell lymphoma. Front. Immunol. 13, 904693 (2022).
Huang, Y. et al. FUT8-mediated aberrant N-glycosylation of B7H3 suppresses the immune response in triple-negative breast cancer. Nat. Commun. 12, 2672 (2021). This work shows that FUT8-mediated fucosylation of the immune checkpoint protein B7H3 drives immune suppression in TNBC, highlighting fucosylation of immunoregulatory molecules as a therapeutic target.
Geijtenbeek, T. B. et al. Identification of DC-SIGN, a novel dendritic cell-specific ICAM-3 receptor that supports primary immune responses. Cell 100, 575–585 (2000).
Amin, R. et al. DC-SIGN-expressing macrophages trigger activation of mannosylated IgM B-cell receptor in follicular lymphoma. Blood 126, 1911–1920 (2015).
Pinioti, S. et al. A metabolic gene survey pinpoints fucosylation as a key pathway underlying the suppressive function of regulatory T cells in cancer. Cancer Immunol. Res. 11, 1611–1629 (2023). This study identifies fucosylation as a central metabolic pathway that sustains the suppressive function of regulatory T cells in tumours, directly linking fucose metabolism to immune evasion.
Nie, H. et al. Targeting branched N-glycans and fucosylation sensitizes ovarian tumors to immune checkpoint blockade. Nat. Commun. 15, 2853 (2024). This study shows that pharmacological targeting of branched N-glycans and fucosylation sensitizes ovarian tumours to immune checkpoint blockade, providing preclinical support for glyco-immunotherapy combinations.
Chu, C. W. et al. Variable PD-1 glycosylation modulates the activity of immune checkpoint inhibitors. Life Sci. Alliance 7, e202302368 (2024).
Zhang, N. et al. Loss of core fucosylation enhances the anticancer activity of cytotoxic T lymphocytes by increasing PD-1 degradation. Eur. J. Immunol. 50, 1820–1833 (2020).
Miyoshi, E. et al. Fucosylation is a promising target for cancer diagnosis and therapy. Biomolecules 2, 34–45 (2012).
Sobierajska, K., Ciszewski, W. M., Sacewicz-Hofman, I. & Niewiarowska, J. Endothelial cells in the tumor microenvironment. Adv. Exp. Med. Biol. 1234, 71–86 (2020).
Moehler, T. M. et al. Involvement of α 1-2-fucosyltransferase I (FUT1) and surface-expressed Lewisy (CD174) in first endothelial cell-cell contacts during angiogenesis. J. Cell Physiol. 215, 27–36 (2008).
Munro, J. M. et al. Expression of sialyl-Lewis X, an E-selectin ligand, in inflammation, immune processes, and lymphoid tissues. Am. J. Pathol. 141, 1397–1408 (1992).
Farrar, C. A. et al. Collectin-11 detects stress-induced L-fucose pattern to trigger renal epithelial injury. J. Clin. Invest. 126, 1911–1925 (2016).
Vasta, G. R. et al. F-Type lectins: a highly diversified family of fucose-binding proteins with a unique sequence motif and structural fold, involved in self/non-self-recognition. Front. Immunol. 8, 1648 (2017).
Amin, M. A. et al. A key role for Fut1-regulated angiogenesis and ICAM-1 expression in K/BxN arthritis. Ann. Rheum. Dis. 74, 1459–1466 (2015).
Atiya, H., Frisbie, L., Pressimone, C. & Coffman, L. Mesenchymal stem cells in the tumor microenvironment. Adv. Exp. Med. Biol. 1234, 31–42 (2020).
Liu, Q. et al. Cancer-associated adipocytes release FUCA2 to promote aggressiveness in TNBC. Endocr. Relat. Cancer 29, 139–149 (2022).
Yang, J. & Cui, Y. TM9SF3 is a mammalian Golgiphagy receptor that safeguards Golgi integrity and glycosylation fidelity. Autophagy 21, 2526–2527 (2025).
Yang, J. et al. TM9SF3 is a Golgi-resident ATG8-binding protein essential for Golgi-selective autophagy. Dev. Cell 60, 2862–2879.e8 (2025).
Yang, S. et al. Multidimensional proteomic landscape reveals distinct activated pathways between human brain tumors. Adv. Sci. 12, e2410142 (2025).
Adhikari, E. et al. Brain metastasis-associated fibroblasts secrete fucosylated PVR/CD155 that induces breast cancer invasion. Cell Rep. 42, 113463 (2023). This study reveals that hypoxia induces expression of FUT11 in breast cancer brain metastasis-associated fibroblasts, which fucosylates PVR, triggering its secretion to promote tumour cell invasion deeper into the brain parenchyma, illustrating how stromal fucosylation shapes metastatic niches.
Sun, W. et al. Mechanisms of pulmonary fibrosis induced by core fucosylation in pericytes. Int. J. Biochem. Cell Biol. 88, 44–54 (2017).
Vestweber, D., Luhn, K., Marquardt, T. & Wild, M. in Leucocyte Trafficking. Ernst Schering Research Foundation Workshop Vol. 44 (eds Hamann, A. et al.) 53–74 (Springer, 2004).
US National Library of Medicine. ClinicalTrials.gov https://www.clinicaltrials.gov/study/NCT05462587 (2024).
US National Library of Medicine. ClinicalTrials.gov https://www.clinicaltrials.gov/study/NCT05754450 (2024).
US National Library of Medicine. ClinicalTrials.gov https://www.clinicaltrials.gov/study/NCT03354533 (2019).
Marquardt, T. et al. Leukocyte adhesion deficiency II syndrome, a generalized defect in fucose metabolism. J. Pediatr. 134, 681–688 (1999).
Yao, Y. et al. L-Fucose increases the fucosylation of colorectal cancer cells via promoting the accumulation of serine. Food Funct. 14, 4314–4326 (2023).
Zhu, B. et al. L-Fucose inhibits the progression of cholangiocarcinoma by causing microRNA-200b overexpression. Chin. Med. J. 135, 2956–2967 (2022).
Tomsik, P. S. et al. L-Rhamnose and L-fucose suppress cancer growth in mice. Cent. Eur. J. Biol. 6, 1–9 (2011).
Keeley, T., Lin, S., Lester, D. K., Lau, E. K. & Yang, S. The fucose salvage pathway inhibits invadopodia formation and extracellular matrix degradation in melanoma cells. PLoS ONE 13, e0199128 (2018).
McCallum, N. & Najlah, M. The anticancer activity of monosaccharides: perspectives and outlooks. Cancers 16, 2775 (2024).
Shi, C. et al. Fucoidan MF4 from Fucus vesiculosus inhibits Lewis lung cancer via STING-TBK1-IRF3 pathway. Int. J. Biol. Macromol. 267, 131336 (2024).
Gao, H. et al. A highly branched glucomannan from the fruiting body of Schizophyllum commune: structural characteristics and antitumor properties analysis. Int. J. Biol. Macromol. 282, 137460 (2024).
He, C. et al. The DDX39B/FUT3/TGFβR-I axis promotes tumor metastasis and EMT in colorectal cancer. Cell Death Dis. 12, 74 (2021).
Hirakawa, M. et al. Fucosylated TGF-β receptors transduces a signal for epithelial-mesenchymal transition in colorectal cancer cells. Br. J. Cancer 110, 156–163 (2014).
Blanas, A. et al. FUT9-driven programming of colon cancer cells towards a stem cell-like state. Cancers 12, 2580 (2020).
Auslander, N. et al. An integrated computational and experimental study uncovers FUT9 as a metabolic driver of colorectal cancer. Mol. Syst. Biol. 13, 956 (2017).
Yang, X., Liu, S. & Yan, Q. Role of fucosyltransferase IV in epithelial-mesenchymal transition in breast cancer cells. Cell Death Dis. 4, e735 (2013).
Antonarelli, G. et al. Targeting post-translational modifications to improve combinatorial therapies in breast cancer: the role of fucosylation. Cells 12, 840 (2023).
Lu, H. H. et al. Fucosyltransferase 4 shapes oncogenic glycoproteome to drive metastasis of lung adenocarcinoma. eBioMedicine 57, 102846 (2020).
Zhan, L., Chen, L. & Chen, Z. Knockdown of FUT3 disrupts the proliferation, migration, tumorigenesis and TGF-β induced EMT in pancreatic cancer cells. Oncol. Lett. 16, 924–930 (2018).
Gao, N. et al. c-Jun transcriptionally regulates alpha 1, 2-fucosyltransferase 1 (FUT1) in ovarian cancer. Biochimie 107, 286–292 (2014).
Yin, X., Rana, K., Ponmudi, V. & King, M. R. Knockdown of fucosyltransferase III disrupts the adhesion of circulating cancer cells to E-selectin without affecting hematopoietic cell adhesion. Carbohydr. Res. 345, 2334–2342 (2010).
Shan, X. et al. Role of fucosyltransferase IV in the migration and invasion of human melanoma cells. IUBMB Life 72, 942–956 (2020).
Zhou, Y. et al. Inhibition of fucosylation by 2-fluorofucose suppresses human liver cancer HepG2 cell proliferation and migration as well as tumor formation. Sci. Rep. 7, 11563 (2017).
Do, K. T. et al. First-in-human, first-in-class, phase I trial of the fucosylation inhibitor SGN-2FF in patients with advanced solid tumors. Oncologist 26, 925–e1918 (2021).
Dai, Y. et al. Synthetic fluorinated L-fucose analogs inhibit proliferation of cancer cells and primary endothelial cells. ACS Chem. Biol. 15, 2662–2672 (2020).
Allen, J. G. et al. Facile modulation of antibody fucosylation with small molecule fucostatin inhibitors and cocrystal structure with GDP-mannose 4,6-dehydratase. ACS Chem. Biol. 11, 2734–2743 (2016).
Orozco-Moreno, M. et al. Targeting aberrant sialylation and fucosylation in prostate cancer cells using potent metabolic inhibitors. Glycobiology 33, 1155–1171 (2023).
Pijnenborg, J. F. A. et al. Cellular fucosylation inhibitors based on fluorinated fucose-1-phosphates. Chemistry 27, 4022–4027 (2021).
Zimmermann, M., Nguyen, M., Schultheiss, C. M., Kolmar, H. & Zimmer, A. Use of 5-thio-L-fucose to modulate binding affinity of therapeutic proteins. Biotechnol. Bioeng. 118, 1818–1831 (2021).
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. 287, 40021–40030 (2012).
Kizuka, Y. et al. An alkynyl-fucose halts hepatoma cell migration and invasion by inhibiting GDP-fucose-synthesizing enzyme FX, TSTA3. Cell Chem. Biol. 24, 1467–1478.e5 (2017). This study shows that an alkynyl-fucose analogue inhibits FX, reduces GDP-fucose synthesis and halts hepatoma cell migration and invasion, highlighting de novo fucose biosynthesis as a druggable target in cellular fucosylation.
Skurska, E. et al. Incorporation of fucose into glycans independent of the GDP-fucose transporter SLC35C1 preferentially utilizes salvaged over de novo GDP-fucose. J. Biol. Chem. 298, 102206 (2022).
Zimmermann, M., Ehret, J., Kolmar, H. & Zimmer, A. Impact of acetylated and non-acetylated fucose analogues on IgG glycosylation. Antibodies 8, 9 (2019).
Okeley, N. M. et al. Development of orally active inhibitors of protein and cellular fucosylation. Proc. Natl Acad. Sci. USA 110, 5404–5409 (2013).
Ma, C. et al. Differential labeling of glycoproteins with alkynyl fucose analogs. Int. J. Mol. Sci. 21, 6007 (2020).
Pijnenborg, J. F. A. et al. Fluorinated rhamnosides inhibit cellular fucosylation. Nat. Commun. 12, 7024 (2021).
Yuan, K. et al. Alterations in human breast cancer adhesion-motility in response to changes in cell surface glycoproteins displaying alpha-L-fucose moieties. Int. J. Oncol. 32, 797–807 (2008).
Benesova, I. et al. N-Glycan profiling of tissue samples to aid breast cancer subtyping. Sci. Rep. 14, 320 (2024).
Brito, A. E., Kletter, D., Singhal, M. & Bern, M. Benchmark study of automatic annotation of MALDI-TOF N-glycan profiles. J. Proteom. 129, 71–77 (2015).
Chen, H. et al. Mass spectrometric profiling reveals association of N-glycan patterns with epithelial ovarian cancer progression. Tumour Biol. 39, 1010428317716249 (2017).
Ma, C. et al. Improvement of core-fucosylated glycoproteome coverage via alternating HCD and ETD fragmentation. J. Proteom. 146, 90–98 (2016).
Song, E. & Mechref, Y. Defining glycoprotein cancer biomarkers by MS in conjunction with glycoprotein enrichment. Biomark. Med. 9, 835–844 (2015).
Wang, D., Hincapie, M., Rejtar, T. & Karger, B. L. Ultrasensitive characterization of site-specific glycosylation of affinity-purified haptoglobin from lung cancer patient plasma using 10 μm i.d. porous layer open tubular liquid chromatography-linear ion trap collision-induced dissociation/electron transfer dissociation mass spectrometry. Anal. Chem. 83, 2029–2037 (2011).
Zhang, Y., Jiao, J., Yang, P. & Lu, H. Mass spectrometry-based N-glycoproteomics for cancer biomarker discovery. Clin. Proteom. 11, 18 (2014).
Alatrash, G. et al. Fucosylation enhances the efficacy of adoptively transferred antigen-specific cytotoxic T lymphocytes. Clin. Cancer Res. 25, 2610–2620 (2019). This study demonstrates that enhancing the fucosylation of antigen-specific cytotoxic T lymphocytes improves their trafficking and antitumour efficacy in vivo, illustrating how fucosylation can be harnessed to optimize adoptive cell therapies.
Myers, J. et al. Fucose-deficient hematopoietic stem cells have decreased self-renewal and aberrant marrow niche occupancy. Transfusion 50, 2660–2669 (2010).
Zhou, J. et al. N-Alkyl-1,5-dideoxy-1,5-imino-L-fucitols as fucosidase inhibitors: synthesis, molecular modelling and activity against cancer cell lines. Bioorg. Chem. 84, 418–433 (2019).
Hottin, A. et al. A second-generation ferrocene-iminosugar hybrid with improved fucosidase binding properties. Bioorg. Med. Chem. Lett. 26, 1546–1549 (2016).
Zhang, Z. et al. Suppression of FUT1/FUT4 expression by siRNA inhibits tumor growth. Biochim. Biophys. Acta 1783, 287–296 (2008).
Su, K. J. et al. Combinations of FUT2 gene polymorphisms and environmental factors are associated with oral cancer risk. Tumour Biol. 37, 6647–6652 (2016).
Sadeghzadeh, Z., Khosravi, A., Jazi, M. S. & Asadi, J. Upregulation of fucosyltransferase 3, 8 and protein O-fucosyltransferase 1, 2 genes in esophageal cancer stem-like cells (CSLCs). Glycoconj. J. 37, 319–327 (2020).
Rostami Abookheili, A., Asadi, J., Khosravi, A. & Gorji, A. Fucosyltransferase 3 and 8 promote the metastatic capacity of cancer stem-like cells via CD15s and E-cadherin in esophageal cancer. Iran. J. Basic Med. Sci. 27, 985–995 (2024).
Zhao, L. et al. miR-493-5p attenuates the invasiveness and tumorigenicity in human breast cancer by targeting FUT4. Oncol. Rep. 36, 1007–1015 (2016).
Zheng, Q. et al. miR-200b inhibits proliferation and metastasis of breast cancer by targeting fucosyltransferase IV and α1,3-fucosylated glycans. Oncogenesis 6, e358 (2017).
Feng, X. et al. Increased fucosylation has a pivotal role in multidrug resistance of breast cancer cells through miR-224-3p targeting FUT4. Gene 578, 232–241 (2016).
Liu, E., Qian, X., He, Y. & Chen, K. FUT4 promotes the progression of cholangiocarcinoma by modulating epithelial-mesenchymal transition. Cell Cycle 23, 218–231 (2024).
Jassam, S. A., Maherally, Z., Ashkan, K., Pilkington, G. J. & Fillmore, H. L. Fucosyltransferase 4 and 7 mediates adhesion of non-small cell lung cancer cells to brain-derived endothelial cells and results in modification of the blood-brain-barrier: in vitro investigation of CD15 and CD15s in lung-to-brain metastasis. J. Neurooncol. 143, 405–415 (2019).
Higai, K., Miyazaki, N., Azuma, Y. & Matsumoto, K. Interleukin-1β induces sialyl Lewis X on hepatocellular carcinoma HuH-7 cells via enhanced expression of ST3Gal IV and FUT VI gene. FEBS Lett. 580, 6069–6075 (2006).
Guo, Q. et al. Functional analysis of α1,3/4-fucosyltransferase VI in human hepatocellular carcinoma cells. Biochem. Biophys. Res. Commun. 417, 311–317 (2012).
Liu, M., Zheng, Q., Chen, S., Liu, J. & Li, S. FUT7 promotes the epithelial-mesenchymal transition and immune infiltration in bladder urothelial carcinoma. J. Inflamm. Res. 14, 1069–1084 (2021).
Wang, S., Zhang, X., Yang, C. & Xu, S. MicroRNA-198-5p inhibits the migration and invasion of non-small lung cancer cells by targeting fucosyltransferase 8. Clin. Exp. Pharmacol. Physiol. 46, 955–967 (2019).
Cao, W. et al. Hypoxia-related gene FUT11 promotes pancreatic cancer progression by maintaining the stability of PDK1. Front. Oncol. 11, 675991 (2021).
Sun, M. A. et al. Gene expression analysis suggests immunosuppressive roles of endolysosomes in glioblastoma. PLoS ONE 19, e0299820 (2024).
Yu, X. et al. Alpha-L-fucosidase: a novel serum biomarker to predict prognosis in early stage esophageal squamous cell carcinoma. J. Thorac. Dis. 11, 3980–3990 (2019).
Yu, X. Y. et al. Association and prognostic significance of alpha-L-fucosidase-1 and matrix metalloproteinase 9 expression in esophageal squamous cell carcinoma. World J. Gastrointest. Oncol. 14, 498–510 (2022).
Zhong, A., Chen, T., Xing, Y., Pan, X. & Shi, M. FUCA2 is a prognostic biomarker and correlated with an immunosuppressive microenvironment in pan-cancer. Front. Immunol. 12, 758648 (2021).
do Nascimento, J. C. et al. Fut3 role in breast invasive ductal carcinoma: investigating its gene promoter and protein expression. Exp. Mol. Pathol. 99, 409–415 (2015).
Zhang, B., van Roosmalen, I. A. M., Reis, C. R., Setroikromo, R. & Quax, W. J. Death receptor 5 is activated by fucosylation in colon cancer cells. FEBS J. 286, 555–571 (2019).
Li, N. et al. MicroRNA-106b targets FUT6 to promote cell migration, invasion, and proliferation in human breast cancer. IUBMB Life 68, 764–775 (2016).
Wang, Q. et al. FUT6 inhibits the proliferation, migration, invasion, and EGF-induced EMT of head and neck squamous cell carcinoma (HNSCC) by regulating EGFR/ERK/STAT signaling pathway. Cancer Gene Ther. 30, 182–191 (2023).
Yao, Y. et al. LncRNA PART1 promotes malignant biological behaviours associated with head and neck cancer cells via synergistic action with FUT6. Cancer Cell Int. 24, 185 (2024).
Zhang, Y., Cui, K., Qiang, R. & Wang, L. FUT10 is related to the poor prognosis and immune infiltration in clear cell renal cell carcinoma. Transl. Cancer Res. 14, 827–842 (2025).
Keeley, T. S., Yang, S. & Lau, E. The diverse contributions of fucose linkages in cancer. Cancers 11, 1241 (2019).
Kim, J. H. et al. Anti-melanogenic effects of a polysaccharide isolated from Undaria pinnatifida sporophyll extracts. Int. J. Mol. Sci. 25, 10624 (2024).
Reis, M. B. E. et al. A fucose-containing sulfated polysaccharide from Spatoglossum schroederi potentially targets tumor growth rather than cytotoxicity: distinguishing action on human melanoma cell lines. Mar. Biotechnol. 26, 181–198 (2024).
El-Sheekh, M. M., Ward, F., Deyab, M. A., Al-Zahrani, M. & Touliabah, H. E. Chemical composition, antioxidant, and antitumor activity of fucoidan from the brown alga Dictyota dichotoma. Molecules 28, 7175 (2023).
Lu, J. et al. A novel polysaccharide from Tremella fuciformis alleviated high-fat diet-induced obesity by promoting AMPK/PINK1/PRKN-mediated mitophagy in mice. Mol. Nutr. Food Res. 69, e202400699 (2025).
Huang, J. et al. A glucomannan from defatted Ganoderma lucidum spores: structural characterization and immunomodulatory activity via activating TLR4/MyD88/NF-κB signaling pathway. Int. J. Biol. Macromol. 294, 139195 (2025).
Staack, L., Della Pia, E. A., Jorgensen, B., Pettersson, D. & Rangel Pedersen, N. Cassava cell wall characterization and degradation by a multicomponent NSP-targeting enzyme (NSPase). Sci. Rep. 9, 10150 (2019).
Saikumar, A. & Badwaik, L. S. Rheological, functional, thermal and physicochemical properties of mucilage extracted from gelatinous pulp of Dillenia indica. L (elephant apple). Int. J. Biol. Macromol. 307, 141924 (2025).
Jin, W. et al. Isolation, in vitro antioxidant capacity, hypoglycemic activity and immunoactivity evaluation of polysaccharides from Coriandrum sativum L. Antioxidants 14, 149 (2025).
Kimura, M. et al. Convenient preparation of an antigenic oligosaccharide from white kidney bean powder: a useful plant oligosaccharide for synthesis of immunoactive glycopolymer. Int. J. Biol. Macromol. 153, 1016–1023 (2020).
Kobayashi, Y. et al. A novel core fucose-specific lectin from the mushroom Pholiota squarrosa. J. Biol. Chem. 287, 33973–33982 (2012).
Bianchet, M. A., Odom, E. W., Vasta, G. R. & Amzel, L. M. Structure and specificity of a binary tandem domain F-lectin from striped bass (Morone saxatilis). J. Mol. Biol. 401, 239–252 (2010).
Nivetha, R., Meenakumari, M., Peroor Mahi Dev, A. & Janarthanan, S. Fucose-binding lectins: purification, characterization and potential biomedical applications. Mol. Biol. Rep. 50, 10589–10603 (2023).
Watanabe, K. et al. Fucosylation is associated with the malignant transformation of intraductal papillary mucinous neoplasms: a lectin microarray-based study. Surg. Today 46, 1217–1223 (2016).
Moriwaki, K. & Miyoshi, E. Basic procedures for lectin flow cytometry. Methods Mol. Biol. 1200, 147–152 (2014).
Terao, N. et al. Fucosylation is a common glycosylation type in pancreatic cancer stem cell-like phenotypes. World J. Gastroenterol. 21, 3876–3887 (2015).
Wang, M. et al. Spatial omics-based machine learning algorithms for the early detection of hepatocellular carcinoma. Commun. Med. 4, 258 (2024).
Boyaval, F. et al. N-Glycomic signature of stage II colorectal cancer and its association with the tumor microenvironment. Mol. Cell Proteom. 20, 100057 (2021).
Caval, T., Alisson-Silva, F. & Schwarz, F. Roles of glycosylation at the cancer cell surface: opportunities for large scale glycoproteomics. Theranostics 13, 2605–2615 (2023).
Trinchera, M., Aronica, A. & Dall’Olio, F. Selectin ligands sialyl-Lewis a and sialyl-Lewis x in gastrointestinal cancers. Biology 6, 16 (2017).
Huang, C. et al. Glycomic profiling of carcinoembryonic antigen isolated from human tumor tissue. Clin. Proteom. 12, 17 (2015).
Fujita, K. et al. Serum core-type fucosylated prostate-specific antigen index for the detection of high-risk prostate cancer. Int. J. Cancer 148, 3111–3118 (2021).
Araki, E. et al. [Clinical assessment of urinary free L-fucose levels]. Rinsho Byori 40, 868–874 (1992).
Manchil, P. R. et al. Correlation of serum levo-fucose levels as a biomarker with tumor node metastasis staging in oral cancer patients. J. Pharm. Bioallied Sci. 8, S147–S150 (2016).
Sakai, T. et al. Rapid, simple enzymatic assay of free L-fucose in serum and urine, and its use as a marker for cancer, cirrhosis, and gastric ulcers. Clin. Chem. 36, 474–476 (1990).
Hanzawa, K. et al. Increased levels of acidic free-N-glycans, including multi-antennary and fucosylated structures, in the urine of cancer patients. PLoS ONE 17, e0266927 (2022).
Tanaka-Okamoto, M. et al. Combination of urinary free-glycan markers for the diagnosis of various malignant tumors. Sci. Rep. 15, 10109 (2025).
Acknowledgements
The authors thank H. Kashfi (Lau laboratory) for the critical readings and feedback on this manuscript. This work was supported in part by a Judith Ann Mogan Foundation Graduate Fellowship to A.B. and NCI grants to E.K.L. (R01CA241559 and R21CA286226) and to the Oregon Health & Science University Knight Cancer Institute (P30CA069533).
Author information
Authors and Affiliations
Contributions
The authors contributed equally to all aspects of the article.
Corresponding author
Ethics declarations
Competing interests
The authors declare no competing interests.
Peer review
Peer review information
Nature Reviews Cancer thanks the anonymous reviewers for their contribution to the peer review of this work.
Additional information
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Glossary
- Adipocytes
-
Adipose tissue-specialized cells that control the storage and release of free fatty acids throughout the body and are regulated by endocrine regulatory signals.
- Adoptive cell transfer
-
The process of isolating immune cells from a patient (autologous) or donor (allogeneic), which are modified ex vivo, and then re-infused into a patient to treat cancer.
- Androgen receptor
-
(AR). An intracellular receptor that, upon binding to male sex hormones (androgens) including testosterone, can translocate into the nucleus to serve as a transcription factor, thereby regulating cellular responses to androgen.
- Click chemistry
-
A class of modular, high-yield reactions used to attach molecular probes to biomolecules such as proteins, DNA and carbohydrates; often involving azides and alkynes to form carbon-heteroatom bonds designed to be highly selective and to allow for their visualization, tracking and functionalization.
- Elastin microfibril interface (EMI) domains
-
Cysteine-enriched domains located at the N terminus of proteins of the elastin microfibril interface-located protein 1 (EMILIN1) protein family, which are involved in the interaction between elastin and microfibrils in the extracellular matrix.
- Epidermal growth factor (EGF)-like repeats
-
Conserved protein domains, consisting of six conserved cysteine residues, which form three disulfide bonds that stabilize the protein structure.
- Fibroblasts
-
Connective tissue-specialized cells, the predominant functions of which are related to building and remodelling the extracellular matrix.
- Galactosylation
-
The enzymatic process by which a galactose residue is transferred from a sugar nucleotide donor (for example, UDP-galactose) to an acceptor substrate, forming a glycosidic linkage and modifying glycoproteins or glycolipids.
- Glycans
-
Complex carbohydrate molecules composed of n number of monosaccharides connected by glycosidic bonds.
- Hydrazide chemistry
-
A biochemical method using hydrazide (–C(=O)NHNH2) functional groups to react with oxidized glycans (aldehydes) for covalent capture or enrichment of glycoproteins and peptides.
- Hydrophilic interaction liquid chromatography
-
(HILIC). A liquid chromatography mode that uses a polar stationary phase (for example, silica) and a mobile phase with high organic solvent (for example, acetonitrile) to separate hydrophilic and polar analytes.
- Lectin
-
A naturally occurring carbohydrate-binding protein derived mainly from plant sources and can be highly specific for sugar groups.
- Lewis antigens
-
A family of fucosylated oligosaccharide determinants expressed on glycoproteins or glycolipids (typically on type 1 or type 2 lactosamine chains) that are part of the histo-blood group systems and mediate cell surface recognition events including blood-group phenotypes, leukocyte adhesion and pathogen binding.
- Macropinocytosis
-
An endocytic process initiated by invagination of the cell membrane to engulf large soluble extracellular contents into primary endocytic vesicles.
- Mannosylation
-
The enzymatic addition of a mannose residue from a sugar nucleotide donor (for example, guanosine diphosphate (GDP)-mannose) onto an acceptor molecule to form a glycosidic linkage.
- Matrix-assisted laser desorption/ionization mass spectrometry imaging
-
(MALDI-MSI). A technique that uses a mass spectrometer to generate images of the spatial distribution of molecules in a sample, typically a thin tissue section.
- Monosaccharide
-
The simplest sugar structure found in nature (for example, glucose, galactose and fructose); complex structures can be made by glycosidic linkages between monosaccharides to form polysaccharides.
- Mucin
-
A high molecular weight glycoprotein synthesized by epithelial mucosal cells, heavily O-glycosylated with oligosaccharides on serine/threonine residues, that contributes to mucus formation and barrier and adhesive functions.
- N-Acetylglucosamine
-
(GlcNAc). An amide derivative of the monosaccharide glucose and secondary derivative of glucosamine and acetic acid.
- N-Glycans
-
Glycan branches conjugated onto asparagine residues in a peptide or protein, by a N-glycosidic bond.
- O-Glycans
-
Glycans conjugated onto the oxygen of a serine or threonine residue.
- Oxidative stress
-
A physiological or pathological state in which the production of reactive oxygen species exceeds the capacity of antioxidant and repair systems, leading to biomolecular damage.
- Sialylation
-
A post-translational modification in which sialic acid, a type of alpha-keto sugar with a nine-carbon backbone, is added to the terminal ends of glycoprotein and glycolipid chains and has roles in various cellular processes, including cell recognition, immune responses and development.
- Sialyl Lewis X
-
(sLeX). A fucosylated, sialylated tetrasaccharide glycan (Neu5Acα2-3Galβ1-4[Fuc〈1-3]GlcNAc) that serves as a key ligand for the selectin family of cell adhesion molecules (E-selectin, P-selectin and L-selectin), thereby mediating leukocyte rolling and extravasation in inflammation.
- Thrombospondin type I repeats
-
(TSRs). Conserved domains of approximately 60 amino acids found in matrix proteins such as the thrombospondin family of proteins; exhibit a unique 3-stranded fold with a positive charge groove, which may mediate ligand–receptor interactions.
Rights and permissions
Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.
About this article
Cite this article
Bitaraf, A., Jimenez, M.C., Kakirde, C. et al. l-Fucose: a dietary sugar with multifaceted potential in the biology and therapy of cancer. Nat Rev Cancer (2026). https://doi.org/10.1038/s41568-025-00901-z
Accepted:
Published:
Version of record:
DOI: https://doi.org/10.1038/s41568-025-00901-z


