Tumour-Associated Carbohydrates as Cancer Targets: Comparison
Please note this is a comparison between Version 2 by Catherine Yang and Version 1 by Mariano Rolando Gabri.

Glycans are essential players involved in the interaction between cells and the microenvironment, making them ideal targets for the development of new therapies against infections and cancer. Since they modulate membrane receptor affinities, immune recognition, protein-protein interactions, and cell signaling, among others, glycans have been shown to play a relevant role in normal and malignant cell behaviour. Therefore, the alteration of the glycophenotype derived from malignant transformation is one of the adaptive mechanisms that provides tumour cells with growth advantages over normal cells, selected in order to circumvent the control mechanisms of the tissue microenvironment and ensure tumour survival.

  • active immunotherapy
  • TACA
  • aberrant glycosylation
  • mimicry

1. Introduction

Glycans expressed in malignant cells that participate in tumour behaviour are known as tumour-associated carbohydrate antigens (TACAs). TACAs represent an opportunity for the identification of new antigens that could potentially be suitable as immunotherapy targets. Given that they are not presented by major histocompatibility complex (MHC) molecules and that they induce immune tolerance, the development of immunotherapeutic strategies against TACAs is a real challenge. However, antitumor strategies based on mimetics of TACAs have been developed and show promising results. Active immunotherapies based on TACA mimicry can currently be grouped into strategies based on the use of mimetic peptides and anti-idiotype (Id) antibodies. All these alternatives seek to stimulate the immune system against a particular TACA to further direct a specific immunological response able to attack and eliminate tumour cells.

2. TACAs as Cancer Targets

According to their structural similarities, TACAs can be classified into five groups: (1) the globo-series glycosphingolipids, including Globo-H, Stage-specific embryonic antigen-3 (SSEA-3) and Stage-specific embryonic antigen-4 (SSEA-4); (2) the gangliosides, including GD2, GD3, GM2, NeuGcGM3, and Fuc-GM1; (3) the blood groups or Lewis antigens, including Lewis X (Lex), Lewis Y (Ley), sialyl Lewis X (SLex), and sialyl Lewis A (SLea); (4) the truncated O-glycans, including Thomsennouveau (Tn), sialyl-Tn (STn), and Thomsen−Friendreich (TF); and (5) the polysialic acid.
Glycosphingolipids share the characteristic of being composed of complex glycans covalently linked to a ceramide backbone. Globo-H ceramide is thought to be the most prevalent cancer-associated glycosphingolipid since its overexpression was detected by immunohistochemistry in a wide variety of epithelial cancers such as breast, uterus, ovary, prostate, lung, liver, and colon cancers [1][2]. Considering its expression is restricted to undifferentiated embryonic stem cells and the luminal surface of glandular tissues, some publications propose Globo-H as a potential target for cancer immunotherapy [3]. SSEA-3 and SSEA-4 were originally identified as human embryonic stem cell-specific markers, but subsequent research demonstrated their association with malignant behaviour in tumour cells [4]. The SSEA-4 expression is positively related to breast [5], glioblastoma [6], and prostate [7] cancer aggressiveness. Since its expression is related to the loss of cell-to-cell interaction and the cell migratory phenotype, SSEA-4 has been associated with epithelial-mesenchymal transition [8]. In breast cancer samples, a high percentage of positivity was reported for SSEA-3 (77.5%), which may correlate with tumorigenicity and multidrug resistance [9][10]. Similarly, in non-small cell lung cancer (NSCLC), SSEA-3 expression increases in samples resistant to multiple anticancer drugs [11]. Moreover, SSEA-3 was postulated as a novel cell surface marker in human colorectal cancer since the SSEA-3-expressing cell subpopulation has a greater proliferative capacity in vitro and a higher tumorigenicity in vivo than SSEA-3-negative cells [12].
Gangliosides are the second TACA group with a glycosphingolipid nature that are composed of at least one sialic acid in the carbohydrate structure [13][14]. The disialylganglioside GD2 is only expressed in a few normal tissues, like the brain, peripheral sensory nerve fibres, and in particular melanocytes from the skin. Nonetheless, this ganglioside is overexpressed in neuroblastomas, melanomas, retinoblastomas, and breast cancer [15][16][17][18]. It was established that GD2 is the most important complex ganglioside present in neuroblastoma, whose expression correlates with tumour progression and poor patient outcomes [19][20]. Another ganglioside widely described in association with malignant transformation is GD3, which is expressed normally in embryonic cells and in some tissues under disease conditions, such as the nervous system in neurodegenerative disorders and tumours of neuroectodermal origin [21]. Interestingly, this TACA is detected in 60% of primary melanoma and 75% of metastatic melanoma, even though it is not expressed in normal melanocytes [22]. In this indication, the upregulation of GD3 on the cell surface is associated with tumour progression, brain metastasis, and poor clinical outcome [23]. GM3 is a monosialoganglioside that can contain a N-acetylneuraminic acid (NeuAc), which is abundant in normal tissues, or a N-glycolylneuraminic acid (NeuGc), which is considered a ‘non-human’ sialic acid due to a mutation in the gene that encodes the enzyme capable of catalysing the conversion of NeuAc to NeuGc [24]. Whereas NeuGcGM3 is rarely detected in normal tissue, it is found in multiple types of tumours such as melanoma [25], neuroblastoma [26], retinoblastoma [27], colon [28], bladder [29] and lung cancer [30], among others. Most literature ascribes the presence of this sialic acid in malignant cells to the metabolic assimilation of NeuGc, obtained principally from red meat, under hypoxic conditions [31][32][33]. On the other side, Fuc-GM1 is a fucose-containing glycolipid that is naturally expressed in a subset of peripheral sensory neurons and dorsal root ganglia [34][35]. However, multiple reports have demonstrated enhanced levels of this antigen in small cell lung cancer (SCLC), even though only 20% and 10% of squamous epithelial and large cell lung cancer specimens were positive, respectively. In addition, no Fuc-GM1 was detected in lung adenocarcinoma, bronchial carcinoma, or normal lung or bronchus, so it could be considered a hallmark of SCLC and useful for sample stratification as well as for targeted immunotherapies [36][37].
Lewis antigens correspond to a family of terminal glycan structures characterised by the presence of one or two fucoses linked to a disaccharide of N-acetylglucosamine and galactose that can also be sialylated. Lex antigen, also known as Stage-Specific Embryonic Antigen-1 (SSEA-1) or CD15, is mainly found in epithelial tissues like the stomach, colon, salivary glands, kidneys, bladder, uterus, cervix, and medulla [38]. Nevertheless, the overexpression of Lex can be found on the surface of various types of cancer cells, which usually correlates with tumour progression and patient survival [39]. In bladder, hepatic, and breast cancer, this molecule is strongly associated with systemic dissemination, possibly related to its role in the adhesion of cancer cells to blood vessel walls [40][41][42]. Particularly in glioblastoma multiforme, this epitope has been proposed as a cancer stem cell marker, and it could be a potential marker of malignant glioma progression [43][44]. Its sialylated version, the terminal glycan SLex, is usually detected on the surface of lymphocytes, neutrophils, and a subset of T lymphocytes [45]. However, expression of this well-known sialofucosylated motif was also reported in high-grade gliomas [46] and gastrointestinal cancers [47]. SLex binds to selectins, mainly E-selectin, expressed on the surface of endothelial cells, facilitating leukocyte extravasation and inflammatory processes [48]. In accordance, in vivo studies have shown that Slex-positive tumour cells exhibit increased metastatic potential by arresting tumour cells in the vasculature of target organs through adhesive interactions [49][50][51]. Another member of this family is SLea, also called cancer antigen 19-9 (CA19-9), which is the most widely used and best validated marker for pancreatic cancers. It is also present in other malignancies such as gastric, endometrial, and colorectal cancers. Initially found in glycolipids, SLea is now known to also be present on glycoproteins and proteoglycans [52]. Although to date it could be considered an indicator for predicting patient prognosis, further studies are needed to fully understand its involvement in cancer biology [53][54][55]. The last TACA of this family, Ley antigen, is a prognostic factor associated with cell dedifferentiation and proliferation in NSCLC and hepatocellular carcinoma [56][57]. It has been demonstrated that the in vitro blockade of Ley significantly reduced the invasion and metastasis of ovarian cancer cells [58]. In addition, numerous publications have shown that overexpression of Ley on these tumour cells is associated with chemotherapy resistance [59][60][61].
Altered O-glycan expression is observed in several tumour indications [62][63][64]. Truncated glycans refer to a set of molecules composed of a small number of carbohydrates present as a consequence of related O-glycan core expression. In this regard, Tn and STn antigens show little or no expression in normal adult cells and tissues. Many studies have shown that Tn antigen is detected in various human carcinomas, such as those of the cervix, ovary, breast, bladder, prostate, gastric, and colon [65][66][67]. Indeed, in many cases, its expression has been associated with tumour progression, dissemination, and patient prognosis [68][69][70]. Tn and STn are frequently co-expressed in tumour cells, possibly because they share synthetic pathways. STn detection is observed in more than 80% of human carcinomas, including gastric, endometrial, and bladder tumours, and in all cases, STn expression is associated with an adverse outcome and decreased overall survival for the patients [71][72][73][74]. Although the mechanism by which STn participates in the interaction between tumour cells and the microenvironment is not fully understood, it has been reported to promote the separation of individual cells from the primary tumour mass by disrupting the interaction of galectins with terminal galactose residues. In this scenario, STn antigen could promote the escape of cancer cells participating in the migration and invasion of the surrounding tissue [75]. TF is a truncated O-glycan highly expressed in approximately 70–90% of carcinomas like ovarian, prostate, breast, colon, stomach, and bladder malignant tissues, although the percentage of positive cases varies among the carcinoma types [76]. Aberrant expression of TF antigen is usually related to the dissipation of the pH gradient in the Golgi apparatus of tumour cells, which can relocate certain glycosyltransferases, leading to an abnormal synthesis of mucin-type O-glycans [77][78]. This TACA plays a crucial role in the interaction between cancer cells and galectin-3, a carbohydrate-binding protein of the endothelium, promoting tumour aggressiveness and metastasis [79][80][81].
Tumours are not only characterised by having a high expression of monosialylated structures such as SSEA-4 or Slex, among others, but also of polysialylated glycans. These TACAs present a limited expression in normal tissue and are associated with particular proteins and cell types, especially those linked to neuronal cells. However, in clinical samples from patients with NSCLC, breast cancer, and glioblastoma, it was shown that the expression of polysialic acids and the glycosyltransferase responsible for its synthesis correlate with cell migration, invasion, and poor prognosis [12][82].
TACAs are promising targets for the development of novel therapeutic strategies due to their active participation in tumour biology (Table 1). Successful development of active immunotherapeutic approaches requires an understanding of how glycans interact with the immune system and the identification of strategies to trigger appropriate immune responses against them.
Table 1.
Main tumour-associated carbohydrate antigens (TACAs).
Name Structure Classification of Glycan Type of Cancer
Globo H Fucα1-2Galβ1-3GalNAcβ1-3Galα1-4Galβ1-4Glcβ Globo Series Breast, Uterus, Ovary, Prostate, Lung, Liver, Colon
SSEA-3 Galβ1-3GalNAcβ1-3Galα1-4Galβ1 Globo Series Breast, NSCLC, Colon
SSEA-4 Neu5Acα1-3Galβ1-3GalNAcβ1-3Galα1-4Galβ1-4Glcβ Globo Series Breast, Glioblastoma, Prostate
GD2 GalNAcβ1,4(Neu5Acα2,

8Neu5Acα2,3)Galβ1, 4Glcβ1Cer
Gangliosides Neuroblastoma, Melanoma, Retinoblastoma, Breast
GD3 Neu5Acα2,8Neu5Acα2,3Galβ1, 4Glcβ1Cer Gangliosides Neuroectodermal, Melanoma
NeuGcGM3 Neu5Gcα2-3Galβ1-4GlcβCer Gangliosides Melanoma, Neuroblastoma, Retinoblastoma, Colon, Bladder, NSCLC
Fuc-GM1 Fucα1-2Galβ1-3GalNAcβ1-4(Neu5Acα2-3)Galβ1-4GlcβCer Gangliosides SCLC
Lex Galβ1-4(Fucα1-3)GlcNAc- Lewis antigens Bladder, Hepatic, Breast, Glioblastoma
SLex Neu5Acα2-3Galβ1-4(Fucα1-3)GlcNAc Lewis antigens Glioma, Gastrointestinal
SLea Neu5Acα2-3Galβ1-3(Fucα1-4)GlcNAc Lewis antigens Pancreatic, Gastric, Endometrial, Colon
Ley Fucα1-2Galβ1-4(Fucα1-3)GlcNAc- Lewis antigens NSCLC, Hepatocellular, Ovarian
Tn GalNAcαSer/Thr O-glycans Cervix, Ovarian, Breast, Prostate, Colon
STn Neu5Acα2-6GalNAcαSer/Thr O-glycans Gastric, Endometrial, Bladder
TF Galβ1-3GalNAcαSer/Thr O-glycans Ovarian, Prostate, Breast, Colon, Stomach and Bladder
Polysialic acid α2,8-/α2,9 NeuAc Polysialylated NSCLC, Breast, Glioblastoma

References

  1. Astronomo, R.D.; Burton, D.R. Carbohydrate vaccines: Developing sweet solutions to sticky situations? Nat. Rev. Drug Discov. 2010, 9, 308–324.
  2. Zhang, S.; Cordon-Cardo, C.; Zhang, H.S.; Reuter, V.E.; Adluri, S.; Hamilton, W.B.; Lloyd, K.O.; Livingston, P.O. Selection of Tumor Antigens as Targets for Immune Attack Using immunohistochemistry: I. focus on Gangliosides. J. Cancer 1997, 73, 42–49.
  3. Yu, A.L.; Hung, J.T.; Ho, M.Y.; Yu, J. Alterations of Glycosphingolipids in Embryonic Stem Cell Differentiation and Development of Glycan-Targeting Cancer Immunotherapy. Stem Cells Dev. 2016, 25, 1532–1548.
  4. Ho, M.Y.; Yu, A.L.; Yu, J. Glycosphingolipid dynamics in human embryonic stem cell and cancer: Their characterization and biomedical implications. Glycoconj. J. 2017, 34, 765–777.
  5. Aloia, A.; Petrova, E.; Tomiuk, S.; Bissels, U.; Déas, O.; Saini, M.; Zickgraf, F.M.; Wagner, S.; Spaich, S.; Sütterlin, M.; et al. The sialyl-glycolipid stage-specific embryonic antigen 4 marks a subpopulation of chemotherapy-resistant breast cancer cells with mesenchymal features. Breast Cancer Res. 2015, 17, 1–17.
  6. Lou, Y.W.; Wang, P.Y.; Yeh, S.C.; Chuang, P.K.; Li, S.T.; Wu, C.Y.; Khoo, K.H.; Hsiao, M.; Hsu, T.L.; Wong, C.H. Stage-specific embryonic antigen-4 as a potential therapeutic target in glioblastoma multiforme and other cancers. Proc. Natl. Acad. Sci. USA 2014, 111, 2482–2487.
  7. Nakamura, Y.; Miyata, Y.; Matsuo, T.; Shida, Y.; Hakariya, T.; Ohba, K.; Taima, T.; Ito, A.; Suda, T.; itiroh Hakomori, S.; et al. Stage-specific embryonic antigen-4 is a histological marker reflecting the malignant behavior of prostate cancer. Glycoconj. J. 2019, 36, 409–418.
  8. 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. 2022, 18, 117–134.
  9. Chang, W.W.; Chien, H.L.; Lee, P.; Lin, J.; Hsu, C.W.; Hung, J.T.; Lin, J.J.; Yu, J.C.; Shao, L.E.; Yu, J.; et al. Expression of Globo H and SSEA3 in breast cancer stem cells and the involvement of fucosyl transferases 1 and 2 in Globo H synthesis. Proc. Natl. Acad. Sci. USA 2008, 105, 11667–11672.
  10. Chuang, P.K.; Hsiao, M.; Hsu, T.L.; Chang, C.F.; Wu, C.Y.; Chen, B.R.; Huang, H.W.; Liao, K.S.; Chen, C.C.; Chen, C.L.; et al. Signaling pathway of globo-series glycosphingolipids and β1,3-galactosyltransferase V (β3GalT5) in breast cancer. Proc. Natl. Acad. Sci. USA 2019, 116, 3518–3523.
  11. Levina, V.; Marrangoni, A.M.; DeMarco, R.; Gorelik, E.; Lokshin, A.E. Drug-Selected Human Lung Cancer Stem Cells: Cytokine Network, Tumorigenic and Metastatic Properties. PLoS ONE 2008, 3, e3077.
  12. Suzuki, Y.; Haraguchi, N.; Takahashi, H.; Uemura, M.; Nishimura, J.; Hata, T.; Takemasa, I.; Mizushima, T.; Ishii, H.; Doki, Y.; et al. SSEA-3 as a novel amplifying cancer cell surface marker in colorectal cancers. Int. J. Oncol. 2013, 42, 161–167.
  13. Krengel, U.; Bousquet, P.A. Molecular recognition of gangliosides and their potential for cancer immunotherapies. Front. Immunol. 2014, 5, 325.
  14. Yu, R.K.; Tsai, Y.T.; Ariga, T.; Yanagisawa, M. Structures, biosynthesis, and functions of gangliosides–an overview. J. Oleo Sci. 2011, 60, 537–544.
  15. Ohmi, Y.; Kambe, M.; Ohkawa, Y.; Hamamura, K.; Tajima, O.; Takeuchi, R.; Furukawa, K.; Furukawa, K. Differential roles of gangliosides in malignant properties of melanomas. PLoS ONE 2018, 13, e0206881.
  16. Portoukalian, J.; David, M.-J.; Richard, M.; Gain, P. Shedding of GD2 ganglioside in patients with retinoblastoma. Int. J. Cancer 1993, 53, 948–951.
  17. Schnaar, R.L. Gangliosides of the vertebrate nervous system. J. Mol. Biol. 2016, 428, 3325.
  18. Shao, C.; Anand, V.; Andreeff, M.; Battula, V.L. Ganglioside GD2: A novel therapeutic target in triple-negative breast cancer. Ann. N. Y. Acad. Sci. 2022, 1508, 35–53.
  19. Sariola, H.; Terava, H.; Rapola, J.; Saarinen, U.M. Cell-surface ganglioside GD2 in the immunohistochemical detection and differential diagnosis of neuroblastoma. Am. J. Clin. Pathol. 1991, 96, 248–252.
  20. Wu, Z.-L.; Ladisch, S.; Feig, S.; Ulsh, L.; Schwartz, E.; Floutsis, G.; Wiley, F.; Lenarsky, C.; Seeger, R. Shedding of GD2 ganglioside by human neuroblastoma. Int. J. Cancer 1987, 39, 73–76.
  21. Cavdarli, S.; Groux-Degroote, S.; Delannoy, P. Gangliosides: The Double-Edge Sword of Neuro-Ectodermal Derived Tumors. Biomolecules 2019, 9, 311.
  22. Hersey, P.; Jamal, O. Expression of the gangliosides GD3 and GD2 on lymphocytes in tissue sections of melanoma. Pathology 1989, 21, 51–58.
  23. Ramos, R.I.; Bustos, M.A.; Wu, J.; Jones, P.; Chang, S.C.; Kiyohara, E.; Tran, K.; Zhang, X.; Stern, S.L.; Izraely, S.; et al. Upregulation of cell surface GD3 ganglioside phenotype is associated with human melanoma brain metastasis. Mol. Oncol. 2020, 14, 1760–1778.
  24. Varki, A. N-glycolylneuraminic acid deficiency in humans. Biochimie 2001, 83, 615–622.
  25. Carr, A.; Mullet, A.; Mazorra, Z.; Vázquez, A.M.; Alfonso, M.; Mesa, C.; Rengifo, E.; Pérez, R.; Fernández, L.E. A Mouse IgG1 Monoclonal Antibody Specific for N-Glycolyl GM3 Ganglioside Recognized Breast and Melanoma Tumors. Hybridoma 2004, 19, 241–247.
  26. Scursoni, A.M.; Galluzzo, L.; Camarero, S.; Lopez, J.; Lubieniecki, F.; Sampor, C.; Segatori, V.I.; Gabri, M.R.; Alonso, D.F.; Chantada, G.; et al. Detection of N-glycolyl GM3 ganglioside in neuroectodermal tumors by immunohistochemistry: An attractive vaccine target for aggressive pediatric cancer. Clin. Dev. Immunol. 2011, 2011, 245181.
  27. Torbidoni, A.V.; Scursoni, A.; Camarero, S.; Segatori, V.; Gabri, M.; Alonso, D.; Chantada, G.; Dávila, M.T.G.D. Immunoreactivity of the 14F7 Mab raised against N-Glycolyl GM3 Ganglioside in retinoblastoma tumours. Acta Ophthalmol. 2015, 93, e294–e300.
  28. Lahera, T.; Calvo, A.; Torres, G.; Rengifo, C.E.; Quintero, S.; Arango, M.d.C.; Danta, D.; Vazquez, J.; Escobar, X.; Carr, A. Prognostic Role of 14F7 Mab Immunoreactivity against N-Glycolyl GM3 Ganglioside in Colon Cancer. J. Oncol. 2014, 2014, 482301.
  29. Albertó, M.; Cuello, H.A.; Gulino, C.A.; Pifano, M.; Belgorosky, D.; Gabri, M.R.; Eiján, A.N.A.M.; Segatori, V.I. Expression of bladder cancer—Associated glycans in murine tumor cell lines. Oncol. Lett. 2019, 17, 3141–3150.
  30. Blanco, R.; Domínguez, E.; Morales, O.; Blanco, D.; Martínez, D.; Rengifo, C.E.; Viada, C.; Cedeño, M.; Rengifo, E.; Carr, A. Prognostic Significance of N-Glycolyl GM3 Ganglioside Expression in Non-Small Cell Lung Carcinoma Patients: New Evidences. Patholog. Res. Int. 2015, 2015, 132326.
  31. Bardor, M.; Nguyen, D.H.; Diaz, S.; Varki, A. Mechanism of uptake and incorporation of the non-human sialic acid N-glycolylneuraminic acid into human cells. J. Biol. Chem. 2005, 280, 4228–4237.
  32. Wang, J.; Shewell, L.K.; Day, C.J.; Jennings, M.P. N-glycolylneuraminic acid as a carbohydrate cancer biomarker. Transl. Oncol. 2023, 31, 101643.
  33. Yin, J.; Hashimoto, A.; Izawa, M.; Miyazaki, K.; Chen, G.; Takematsu, H.; Kozutsumi, Y.; Suzuki, A.; Furuhata, K.; Cheng, F.; et al. Hypoxic Culture Induces Expression of Sialin, a Sialic Acid Transporter, and Cancer-Associated Gangliosides Containing Non-Human Sialic Acid on Human Cancer Cells. Cancer Res. 2006, 66, 2937–2946.
  34. Kusunoki, S.; Inoue, K.; Iwamori, M.; Nagai, Y.; Mannen, T. Fucosylated glycoconjugates in human dorsal root ganglion cells with unmyelinated axons. Neurosci. Lett. 1991, 126, 159–162.
  35. Kusunoki, S.; Inoue, K.; Iwamori, M.; Nagai, Y.; Mannen, T.; Kanazawa, I. Developmental changes of fucosylated glycoconjugates in rabbit dorsal root ganglia. Neurosci. Res. 1992, 15, 74–80.
  36. Brezicka, F.; Olling, S.; Nilsson, O.; Bergh, J.; Holmgren, J.; Sörenson, S.; Yngvason, F.; Lindholm, L. Immunohistological detection of fucosyl-GM1 ganglioside in human lung cancer and normal tissues with monoclonal antibodies. Cancer Res. 1989, 49, 1300–1305.
  37. Livingston, P.O.; Hood, C.; Krug, L.M.; Warren, N.; Kris, M.G.; Brezicka, T.; Ragupathi, G. Selection of GM2, fucosyl GM1, globo H and polysialic acid as targets on small cell lung cancers for antibody mediated immunotherapy. Cancer Immunol. Immunother. 2005, 54, 1018–1025.
  38. Blanas, A.; Sahasrabudhe, N.M.; Rodríguez, E.; van Kooyk, Y.; van Vliet, S.J. Fucosylated Antigens in Cancer: An Alliance toward Tumor Progression, Metastasis, and Resistance to Chemotherapy. Front. Oncol. 2018, 8, 39.
  39. Kadota, A.; Masutani, M.; Takei, M.; Horie, T. Evaluation of expression of CD15 and sCD15 in non-small cell lung cancer. Int. J. Oncol. 1999, 15, 1081–1089.
  40. Koh, Y.W.; Lee, H.J.; Ahn, J.H.; Lee, J.W.; Gong, G. Expression of Lewis X Is Associated With Poor Prognosis in Triple-Negative Breast Cancer. Am. J. Clin. Pathol. 2013, 139, 746–753.
  41. Konety, B.R.; Ballou, B.; Jaffe, R.; Singh, J.; Reiland, J.; Hakala, T.R. Expression of SSEA-1 (Lewisx) on Transitional Cell Carcinoma of the Bladder. Urol. Int. 1997, 58, 69–74.
  42. Torii, A.; Nakayama, A.; Harada, A.; Nakao, A.; Nonami, T.; Sakamoto, J.; Watanabe, T.; Lto, M.; Takagi, H. Expression of the C D I 5 Antigen in Hepatocellular Carcinoma. Br. J. Cancer 2015, 112, 1911–1920.
  43. Son, M.J.; Woolard, K.; Nam, D.H.; Lee, J.; Fine, H.A. SSEA-1 is an enrichment marker for tumor-initiating cells in human glioblastoma. Cell Stem Cell 2009, 4, 440–452.
  44. Wang, P.; Gong, S.; Liao, B.; Pan, J.; Wang, J.; Zou, D.; Zhao, L.; Xiong, S.; Deng, Y.; Yan, Q.; et al. HIF1α/HIF2α induces glioma cell dedifferentiation into cancer stem cells through Sox2 under hypoxic conditions. J. Cancer 2022, 13, 1–14.
  45. Jin, F.; Wang, F. The physiological and pathological roles and applications of sialyl Lewis x, a common carbohydrate ligand of the three selectins. Glycoconj. J. 2020, 37, 277–291.
  46. Cuello, H.A.; Ferreira, G.M.; Gulino, C.A.; Toledo, A.G.; Segatori, V.I.; Gabri, M.R. Terminally sialylated and fucosylated complex N-glycans are involved in the malignant behavior of high-grade glioma. Oncotarget 2021, 11, 4822–4835.
  47. Trinchera, M.; Aronica, A.; Dall’Olio, F. Selectin Ligands Sialyl-Lewis a and Sialyl-Lewis x in Gastrointestinal Cancers. Biology 2017, 6, 16.
  48. Munro, J.M.; Lo, S.K.; Corless, C.; Robertson, M.J.; Lee, N.C.; Barnhill, R.L.; Weinberg, D.S.; Bevilacqua, M.P. Expression of sialyl-Lewis X, an E-selectin ligand, in inflammation, immune processes, and lymphoid tissues. Am. J. Pathol. 1992, 141, 1397.
  49. Baldus, S.E.; Zirbes, T.K.; Mönig, S.P.; Engel, S.; Monaca, E.; Rafiqpoor, K.; Hanisch, F.G.; Hanski, C.; Thiele, J.; Pichlmaier, H.; et al. Histopathological subtypes and prognosis of gastric cancer are correlated with the expression of mucin-associated sialylated antigens: Sialosyl-Lewis(a), Sialosyl-Lewis(x) and sialosyl-Tn. Tumour Biol. 1998, 19, 445–453.
  50. Esposito, M.; Mondal, N.; Greco, T.M.; Wei, Y.; Spadazzi, C.; Lin, S.C.; Zheng, H.; Cheung, C.; Magnani, J.L.; Lin, S.H.; et al. Bone vascular niche E-selectin induces mesenchymal-epithelial transition and Wnt activation in cancer cells to promote bone metastasis. Nat. Cell Biol. 2019, 21, 627–639.
  51. Nakamori, T.I.S.; Kameyama, M.; Imaoka, S.; Furukawa, H.; Ishikawa, O.; Sasaki, Y.; Kabuto, T.; Iwanaga, T.; Matsushita, Y. Increased expression of sialyl Lewisx antigen correlates with poor survival in patients with colorectal carcinoma: Clinicopathological and immunohistochemical study. Cancer Res. 1993, 53, 3632–3637. Available online: https://pubmed.ncbi.nlm.nih.gov/8101764/ (accessed on 1 August 1993).
  52. Goonetilleke, K.S.; Siriwardena, A.K. Systematic review of carbohydrate antigen (CA 19-9) as a biochemical marker in the diagnosis of pancreatic cancer. Eur. J. Surg. Oncol. 2007, 33, 266–270.
  53. Kolben, T.; Müller, L.; Meister, S.; Keilmann, L.; Buschmann, C.; Trillsch, F.; Burges, A.; Czogalla, B.; Mitter, S.; Schmoeckel, E.; et al. Blood group antigens SLeX, SLeA, and LeY as prognostic markers in endometrial cancer. J. Cancer Res. Clin. Oncol. 2022, 148, 3323–3335.
  54. Moriyama, J.; Oshima, Y.; Nanami, T.; Suzuki, T.; Yajima, S.; Shiratori, F.; Funahashi, K.; Shimada, H. Prognostic impact of CEA/CA19-9 at the time of recurrence in patients with gastric cancer. Surg. Today 2021, 51, 1638–1648.
  55. Rao, H.; Wu, H.; Huang, Q.; Yu, Z.; Zhong, Z. Clinical Value of Serum CEA, CA24-2 and CA19-9 in Patients with Colorectal Cancer. Clin. Lab. 2021, 67, 1079–1089.
  56. Tanaka, F.; Miyahara, R.; Ohtake, Y.; Yanagihara, K.; Fukuse, T.; Hitomi, S.; Wada, H. Lewis Y antigen expression and postoperative survival in non-small cell lung cancer. Ann. Thorac. Surg. 1998, 66, 1745–1750.
  57. Wakabayashi, M.; Shiro, T.; Seki, T.; Nakagawa, T.; Lmamura, M.; Shiozaki, Y.; Lnoue, K.; Okamura, A. Lewis Y Antigen Expression in Hepatocellular Carcinoma An Immunohistochemical Study. Cancer 1995, 75, 2827–2835.
  58. Zhuang, H.; Hu, Z.; Tan, M.; Zhu, L.; Liu, J.; Liu, D.; Yan, L.; Lin, B. Overexpression of Lewis y antigen promotes human epididymis protein 4-mediated invasion and metastasis of ovarian cancer cells. Biochimie 2014, 105, 91–98.
  59. Gao, J.; Zhu, L.; Zhuang, H.; Lin, B. Human Epididymis Protein 4 and Lewis y Enhance Chemotherapeutic Resistance in Epithelial Ovarian Cancer Through the p38 MAPK Pathway. Adv. Ther. 2022, 39, 360–378.
  60. Liu, J.; Zheng, M.; Qi, Y.; Wang, H.; Liu, M.; Liu, Q.; Lin, B. Lewis(y) antigen-mediated positive feedback loop induces and promotes chemotherapeutic resistance in ovarian cancer. Int. J. Oncol. 2018, 53, 1774–1786.
  61. Zhang, D.; Gao, J.; Zhu, L.; Hu, Z.; Hou, R.; Liu, S.; Tan, M.; Liu, J.; Lin, B. Chemoresistance is associated with MUC1 and Lewis y antigen expression in ovarian epithelial cancers. Int. J. Mol. Sci. 2013, 14, 11024–11033.
  62. Dalziel, M.; Whitehouse, C.; McFarlane, I.; Brockhausen, I.; Gschmeissner, S.; Schwientek, T.; Clausen, H.; Burchell, J.M.; Taylor-Papadimitriou, J. The Relative Activities of the C2GnT1 and ST3Gal-I Glycosyltransferases Determine O -Glycan Structure and Expression of a Tumor-associated Epitope on MUC1. J. Biol. Chem. 2001, 276, 11007–11015.
  63. Ju, T.; Aryal, R.P.; Kudelka, M.R.; Wang, Y.; Cummings, R.D. The Cosmc connection to the Tn antigen in cancer. Cancer Biomark. 2014, 14, 63–81.
  64. Cuello, H.A.; Segatori, V.I.; Albertó, M.; Gulino, C.A.; Aschero, R.; Camarero, S.; Mutti, L.G.; Madauss, K.; Alonso, D.F.; Lubieniecki, F.; et al. Aberrant O-glycosylation modulates aggressiveness in neuroblastoma. Oncotarget 2018, 9, 34176–34188.
  65. Desai, P.R. Immunoreactive T and Tn antigens in malignancy: Role in carcinoma diagnosis, prognosis, and immunotherapy. Transfus. Med. Rev. 2000, 14, 312–325.
  66. Springer, G.F. T and Tn, general carcinoma autoantigens. Science 1984, 224, 1198–1206.
  67. Rømer, T.B.; Aasted, M.K.M.; Dabelsteen, S.; Groen, A.; Schnabel, J.; Tan, E.; Pedersen, J.W.; Haue, A.D.; Wandall, H.H. Mapping of truncated O-glycans in cancers of epithelial and non-epithelial origin. Br. J. Cancer 2021, 125, 1239.
  68. Festari, M.F.; Da Costa, V.; Rodríguez-Zraquia, S.A.; Costa, M.; Landeira, M.; Lores, P.; Solari-Saquieres, P.; Kramer, M.G.; Freire, T. The tumor-associated Tn antigen fosters lung metastasis and recruitment of regulatory T cells in triple negative breast cancer. Glycobiology 2022, 32, 366–379.
  69. Hamada, S.-i.; Furumoto, H.; Kamada, M.; Hirao, T.; Aono, T. High expresion rate of Tn antigen in metastatic lesions of uterine cervical cancers. Cancer Lett. 1993, 74, 167–173.
  70. Konno, A.; Hoshino, Y.; Terashima, S.; Motoki, R.; Kawaguchi, T. Carbohydrate expression profile of colorectal cancer cells is relevant to metastatic pattern and prognosis. Clin. Exp. Metastasis 2002, 19, 61–70.
  71. Ferreira, J.A.; Videira, P.A.; Lima, L.; Pereira, S.; Silva, M.; Carrascal, M.; Severino, P.F.; Fernandes, E.; Almeida, A.; Costa, C.; et al. Overexpression of tumour-ssociated carbohydrate antigen sialyl-Tn in advanced bladder tumours. Mol. Oncol. 2013, 7, 719–731.
  72. Hashiguchi, Y.; Kasai, M.; Fukuda, T.; Ichimura, T.; Yasui, T.; Sumi, T. Serum Sialyl-Tn (STN) as a Tumor Marker in Patients with Endometrial Cancer. Pathol. Oncol. Res. 2016, 22, 501–504.
  73. Itzkowitz, S.; Kjeldsen, T.; Friera, A.; Hakomori, S.; Yang, U.S.; Kim, Y.S. Expression of Tn, sialosyl Tn, and T antigens in human pancreas. Gastroenterology 1991, 100, 1691–1700.
  74. Julien, S.; Videira, P.A.; Delannoy, P. Sialyl-tn in cancer: (How) did we miss the target? Biomolecules 2012, 2, 435–466.
  75. Fu, C.; Zhao, H.; Wang, Y.; Cai, H.; Xiao, Y.; Zeng, Y.; Chen, H. Tumor-associated antigens: Tn antigen, sTn antigen, and T antigen. HLA 2016, 88, 275–286.
  76. Karsten, U.; Goletz, S. What controls the expression of the core-1 (Thomsen-Friedenreich) glycotope on tumor cells? Biochemistry 2015, 80, 801–807.
  77. Rivinoja, A.; Kokkonen, N.; Kellokumpu, I.; Kellokumpu, S. Elevated Golgi pH in breast and colorectal cancer cells correlates with the expression of oncofetal carbohydrate T-antigen. J. Cell. Physiol. 2006, 208, 167–174.
  78. Thorens, B.; Vassalli, P. Chloroquine and ammonium chloride prevent terminal glycosylation of immunoglobulins in plasma cells without affecting secretion. Nature 1986, 321, 618–620.
  79. Monzavi-Karbassi, B.; Pashov, A.; Kieber-Emmons, T. Tumor-Associated Glycans and Immune Surveillance. Vaccines 2013, 1, 174–203.
  80. Takenaka, Y.; Fukumori, T.; Raz, A. Galectin-3 and metastasis. Glycoconj. J. 2002, 19, 543–549.
  81. Yu, L.G. The oncofetal Thomsen-Friedenreich carbohydrate antigen in cancer progression. Glycoconj. J. 2007, 24, 411–420.
  82. Soukhtehzari, S.; Berish, R.B.; Fazli, L.; Watson, P.H.; Williams, K.C. The different prognostic significance of polysialic acid and CD56 expression in tumor cells and lymphocytes identified in breast cancer. NPJ Breast Cancer 2022, 8, 78.
More
ScholarVision Creations