Dynamic Tuning of Galectins and Their Binding Sites During

Integrins (1), cadherins (2–7), caveolins (8), metalloproteinases (9), hormone receptors (10) ... of Clinical Sciences of Companion Animals, Faculty...
0 downloads 0 Views 319KB Size
Downloaded by NORTH CAROLINA STATE UNIV on December 19, 2012 | http://pubs.acs.org Publication Date (Web): December 18, 2012 | doi: 10.1021/bk-2012-1115.ch011

Chapter 11

Dynamic Tuning of Galectins and Their Binding Sites During Mammary Tumor Progression and Metastasis Joana T. de Oliveira and Fátima Gärtner* Institute of Molecular Pathology and Immunology (IPATIMUP), and Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), University of Porto, Porto, Portugal *E-mail: [email protected]

Galectins are essentially bridging molecules and as such their biological effect will be dependent on their levels of expression, binding sites availability on their ligands, presence of other competitive galectins or even members of other families of endogenous lectins. Classically, glycosylation patterns were believed to be fairly constant in cells of the same tissue origin and the process was not thought to be a quickly modulated one able to transduce external signals. Nevertheless, a growing amount of data now suggests glycosylation to be a much more dynamic microenvironment-related mechanism. The consequent formation of different glycans alters the array of lectins which are able to recognize them. Therefore, rather discrete and transient changes in interacting glycans, glycan carriers and glycan receptors may profoundly influence the fate of an invasive tumor cell and ultimately its ability to metastasize to distant sites. This mini-review will focus on the dynamic interplay between galectins and their binding sites in a spontaneous model of mammary tumor progression and invasion, canine mammary tumors (CMT).

© 2012 American Chemical Society In Galectins and Disease Implications for Targeted Therapeutics; Klyosov, A., et al.; ACS Symposium Series; American Chemical Society: Washington, DC, 2012.

Introduction

Downloaded by NORTH CAROLINA STATE UNIV on December 19, 2012 | http://pubs.acs.org Publication Date (Web): December 18, 2012 | doi: 10.1021/bk-2012-1115.ch011

Integrins (1), cadherins (2–7), caveolins (8), metalloproteinases (9), hormone receptors (10), growth factors (11), growth factor receptors (12) and mucins (13, 14) putatively modulate the interactions between mammary tumour cells and their surrounding microenvironment, at least in part, by their well-known interplay with galectin-3 via its carbohydrate recognition domain [reviewed in (15)]. However, the interplay between these ligands and galectin-3 has been shown to be modulated not only by the levels of expression of the lectin but also by differential glycosylation of the ligands in several settings (16–20).

Galectins’ Levels of Expression: Adapting to Presenting Microenvironments in CMT Progression Galectin-3 is down-regulated during the process of malignant transformation of canine mammary glands. Its expression being significantly decreased in malignant when compared with benign canine mammary tumors (CMT) (21). Galectin-3 is a marker of progression in other types of tumors presenting increased expression in non-aggressive tumors and being reduced in aggressive ones, the opposite having also been observed (22–25). Galectin-1 is on the other hand overexpressed in malignant CMT, and found in the nucleus and cytoplasm of tumor cells (21). Several examples of dynamic and differential expression of galectins-1 and -3 in physiological and pathological contexts suggesting they play opposing roles can be found in the literature (26–28). Among other examples, in lymphoma cells, galectin-1 induces apoptosis while galectin-3 induces cell-cell aggregation (29) and blocks the execution of the cell surface apoptotic signal (30). In accordance to its pro-adhesive effects, it comes as no surprise that galectin-3 is highly expressed by vessel-invading tumor cell subpopulations in both primary and metastatic CMT while in well-established metastatic lesions, there is galectin-3 staining almost only in tumor cells surrounding necrotic areas, a pattern resembling that observed in the primary malignant CMT (21). On the opposite galectin-1 is down-regulated in tumour emboli and up-regulated in well-established primary and metastatic lesions (unpublished results by the authors of this paper). Accordingly, galectin-1 serum levels are not elevated in cancer patients (31) while those of galectin-3 have been found to be increased (32). Cytoplasmic staining of galectin-3 is associated with increased aggressiveness in CMT whereas significant down-regulation of nuclear galectin-3 expression is observed in malignant when compared with benign tumors (21). The cytoplasmic subcellular distribution of galectin-3 is an important feature related to malignancy and is suggested to be responsible for increasing apoptosis resistance of tumor cells that migrate and/or are shed into the circulation (33–35). Indeed, xenografts from a CMT cell line with an in vitro homogeneous cytoplasmic-only expression of galectin-3, quickly metastasize. However, a heterogeneous pattern with specific galectin-3 positive areas is observed in well-established xenografts suggesting a role for microenvironment in the regulation of galectin-3 expression. In primary tumor xenografts, cells staining for galectin-3 were specifically located in necrosis182 In Galectins and Disease Implications for Targeted Therapeutics; Klyosov, A., et al.; ACS Symposium Series; American Chemical Society: Washington, DC, 2012.

Downloaded by NORTH CAROLINA STATE UNIV on December 19, 2012 | http://pubs.acs.org Publication Date (Web): December 18, 2012 | doi: 10.1021/bk-2012-1115.ch011

surrounding regions and tumor emboli, as observed in spontaneously occurring malignant CMT. Likewise, metastatic lesions displayed low galectin-3 expression which was mainly present in necrosis-surrounding areas and inside tumor vessels (21). The reasons why intravascular tumor cells consistently overexpress galectin-3 when compared to sedentary tumor cells still need clarification. However a few hypothesis are to be considered. Galectin-3 is a hypoxia regulated protein (36). Hypoxia also leads to activation of epidermal growth factor receptor (EGFR) kinase function and hence may enhance tumor cell migration (37, 38). Interestingly, EGFR expression is up-regulated in CMT intravascular tumor cells and viable cells adjacent to necrotic areas paralleling that of galectin-3. This indicates a possible joint role of EGFR and galectin-3 in the survival and invasion process of tumor cells under stress conditions in malignant CMT (39). Adding to that, high level of activity in necrotic areas was found when analyzing EGFR by autoradiography (Berns and Rutteman, unpublished data, Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, the Netherlands) and its levels were significantly associated to the presence of necrosis in CMT (39). Since vessel-invading tumor cells express both galectin-3 and EGFR, the hypothesis that tumor cells which intravasate, are mainly those exposed to stress conditions seems very plausible. Furthermore, as galectin-3, EGFR has also been associated to angiogenesis which could in addition aid in providing an escape pathway for these stress-exposed cells (15, 40). Another line of thinking arises from the fact that galectin-3 expression has been associated to cell differentiation in several contexts (41). Epithelial to mesenchymal transition (EMT) has been suggested to be crucial for cells to acquire invasive characteristics, migrate throughout the ECM and ultimately intravasate (42, 43). In the absence of galectin-3 there is decreased TGFβ-induced EMT, myofibroblast activation and collagen production with reduced β-catenin phosphorylation and translocation to the nucleus (44). Accordingly, in low galectin-3-expressing malignant CMT, there is little nuclear expression of β-catenin (45, 46). This further points to galectin-3 re-expression being implicated in acquisition of aggressive characteristics in stress-exposed cells, such as those under hypoxic conditions. Mixed type malignant CMT (carcinosarcomas) present a common histogenesis between epithelial and mesenchymal cells (47). These mixed malignant mammary tumours are considered a naturally occurring model of EMT [reviewed in (48)]. Galectin-1 was consistently co-expressed with mesenchymal while galectin-3, when present, was co-expressed with epithelial markers in this natural model of EMT (unpublished data). However, the differentiation status of galectin-3-positive intravascular tumor cells could not always be associated with loss or gain of membrane expression of E-cadherin since both E-cadherin-negative and E-cadherin-positive tumor emboli were found in malignant CMT. Interestingly however, β-catenin expression was often found at the cell membrane, in contact sites between some intravascular tumor cells indicating a correct assembly of the adherens junctions complex at these sites (21). Since the hallmark of EMT is loss of E-cadherin expression (49), current findings 183 In Galectins and Disease Implications for Targeted Therapeutics; Klyosov, A., et al.; ACS Symposium Series; American Chemical Society: Washington, DC, 2012.

Downloaded by NORTH CAROLINA STATE UNIV on December 19, 2012 | http://pubs.acs.org Publication Date (Web): December 18, 2012 | doi: 10.1021/bk-2012-1115.ch011

do not support the assumption that all intravascular tumor cells found in CMT have undergone complete EMT and/or the inverse process mesenchymal to epithelial transition (MET) but rather point to the presence of several cells in hybrid states inside vessels (21). The isolated expression either of anti-adhesion, lymph node metastasis-associated, glycan Sialyl Lewis X (50) or the adherens junction E-cadherin at the cell surface of CMT emboli cells, seems to corroborate this assumption (51). Despite this it is the authors’ opinion that MET is crucial for cell-cell aggregation inside vessels insuring anoikis survival for the 2% of the circulating tumor cells that in fact successfully arrive at distant sites and metastasize (52). Thus overexpression of galectin-3 seems to be a characteristic of potentially metastatic tumor cells in malignant CMT but does not appear to be exclusively associated to a differentiation status.

Glycosylation of Galectin-Ligands: Modulating Galectin-Mediated CMT Cell-Cell Adhesion Mucins have been associated to malignancy (13, 14, 53) and are important carriers of tumour-associated glycan antigens (54, 55). Variable glycoforms of MUC1 have been found to be involved in carcinogenesis (56, 57). MUC1 is involved in cell-cell aggregation, (its sialylation being particularly important for the mucin’s anti-aggregation effect) (58, 59); and cell-ECM adhesion (58, 60, 61) in which MUC1 specific O-glycosylation plays a relevant role (62). MUC1 is overexpressed and significantly associated to vascular invasion and distant metastases in malignant CMT (21, 39). In primary tumour cells, the mucin is present in a cytoplasmic vesicular pattern and all around the cell membrane. This high level of MUC1 expression is significantly associated with the above mentioned galectin-3 generalized down-regulation. Furthermore, the two molecules are not co-expressed in primary tumor sedentary cells (39). Despite the described overexpression of its well-known ligand, galectin-3-binding sites are expressed at the tumor cell surface only in moderately differentiated tumor areas, their expression being low in the majority of sedentary primary tumor cells. However, in malignant CMT, intravascular tumor cells and tumor vasculature strongly express galectin-3-binding sites pointing to the existence of galectin-3-mediated cell–cell interactions, which could thus facilitate anoikis survival and metastatic spread (21, 63). Interestingly, in these vessel-invading cell tumor subpopulations MUC1 expression is focally localized at the cell membrane where it co-expresses with galectin-3 (39). Different glycosylation patterns of MUC1 mucin in normal mucosa and colon cancer tissues correlate well with galectin-3-binding sites expression (64). Sialylation thus acts as an important on/off switch mechanism modifying galectin-3 binding to its ligands during tumor progression in malignant CMT. The differential presence of α2, 6- linked sialic acid in sedentary tumor cells and certain tumor subpopulations, such as invasive fronts, may account for differences observed in galectin-3-binding site expression and modulate galectin-3-mediated adhesion between tumor cells at these locations (39). Up- or down-regulating 184 In Galectins and Disease Implications for Targeted Therapeutics; Klyosov, A., et al.; ACS Symposium Series; American Chemical Society: Washington, DC, 2012.

Downloaded by NORTH CAROLINA STATE UNIV on December 19, 2012 | http://pubs.acs.org Publication Date (Web): December 18, 2012 | doi: 10.1021/bk-2012-1115.ch011

of sialyltransferases is a well-known mechanism of motility tuning, turning galectin-3-binding positive cells into negative ones and vice-versa, allowing cells to migrate in a physiological context. Indeed there is an increase of both galectin-1 and -3 binding following alpha 2,6 neuraminidase treatment of glycans at the cell surface in vitro (65). The Thomsen-Friedenreich antigen (T antigen) is one of the most common glycoforms of MUC1 found in breast cancer patients. MUC1 interaction with galectin-3 in vitro via T antigen, causes clustering of the mucin at the cell surface with consequent exposure of smaller adhesion molecules. This has been proven to increase E-cadherin mediated cell-cell adhesion and ICAM-1 mediated cellendothelial adhesion (17, 66). A similar effect has been observed for galectins-2, 4 and -8 by interaction with MUC1 via T antigen (31). Most cells of primary malignant CMT show high levels of sialylated T antigen when compared to the un-substituted form of the antigen which is expressed in low levels and is often negative. However, surprisingly, intravascular tumor cells express mainly the unsubstituted form of the T antigen. This allows a proven physical interaction in vivo between galectin-3 and MUC1-carried T antigen, therefore supporting an important role in metastasis (39). Alterations in the glycosylation patterns of its ligands, namely increased disialylated core 1 O-glycan structures, have been implied to alter galectin-3 affinity at the cell surface with consequent differences in cell biological behaviour (67). Core 1 biosynthesis is dependent on the activity of core 1 beta1,3 Gal-transferase (68) which has been found to be impaired by mutations in the molecular chaperone Cosmc leading to increased expression of Tn and sialyl Tn antigens (69). In order to have the unsubstituted core 1 (T antigen), there must not be action of ST6GalNacI and II (70). The action of these enzymes leads to the biosynthesis of Sialyl Tn and/or Sialyl-6T antigen. ST6GalNAc-I is the key-enzyme leading to sialyl-Tn biosynthesis in MUC1-Tn glycoform (71) while ST6GalNAc-II sialylates better the T antigen (70). MUC1 is also a natural carrier of Sialyl Tn (72). It is of note that ST6GalNacI and II enzymes perform very fast (70). Their subcellular localization has been suggested to influence their activity in breast cancer cells (73) as has the tumour microenvironment which was found to importantly modulate Sialyl-Tn expression (74). Galectin-3 has been implicated in the apical sorting of several proteins (75) but in the absence of galectin-3 there is still apical targeting of MUC1. However, this can be blocked by overexpression of an alpha 2,6 sialyltransferase (76) further corroborating the importance of sialylation in the interplay between MUC1 and the galectins family. In a normal context MUC1 is localized at the apical cell membrane while EGFR is localized basolaterally. MUC1-EGFR interaction in a non-tumour context is a sign of temporary polarization disruption and mediates cell survival programs. These are thought to be persistently present in several types of carcinoma where there is loss of polarization with permanent interaction of transmembrane mucins and growth factor receptors (77). In breast cancer cells this interaction is at least in part mediated by galectin-3. In vitro and in vivo results show the lectin’s co-expression with both of its ligands in intravascular tumour cells, further supporting its importance for cell survival in this context (39, 78). 185 In Galectins and Disease Implications for Targeted Therapeutics; Klyosov, A., et al.; ACS Symposium Series; American Chemical Society: Washington, DC, 2012.

Downloaded by NORTH CAROLINA STATE UNIV on December 19, 2012 | http://pubs.acs.org Publication Date (Web): December 18, 2012 | doi: 10.1021/bk-2012-1115.ch011

Microenvironment seems therefore to play a vital role in the availability of ligands for galectin-3-binding in CMT. This is corroborated in the CMT metastatic cell line which shows a homogeneous expression of galectin-3-binding sites in vitro (21) which turns to a heterogeneous one in tumor xenografts (39). Also, galectin-3-binding site expression is low in the vast majority of sedentary primary tumor cells to the opposite of invasive fronts where it is high akin to spontaneously occurring CMT. In addition, binding of galectin-3 is again increased, following neuraminidase treatment in galectin-3-binding sites negative areas, resembling the observations in the spontaneous model (39). Other than downregulation of sialyltransferases which can at least in part mask galectin-3-ligands in these cells, there may be higher activity in invading tumor cells of specific glycosyltranferases (79) some of which responsible for assembling ligands for galectin-3, such as the metastasis-related GnTV and concomitant lower activity of competitive GnTIII, which leads to the production of N-glycans not recognized by the lectin (80, 81) Indeed, recently GnT-III-mediated glycosylation, was found to be altered upon EMT/MET-inducing microenvironment changes (82).

Glycosylation of Galectin-Ligands: Modulating Galectin-Mediated CMT Cell-ECM Adhesion Downregulation of galectin-3-binding sites in the ECM parallels the malignant transformation of canine mammary glands. In fact, galectin-3-binding sites are significantly decreased in the ECM of malignant tissue when compared with that of normal-adjacent glands. In most benign lesions the ECM also presents strong expression of galectin-3-binding sites. A coordinated decrease of galectin-3-binding sites in the ECM may further account for the loss of galectin-3-mediated cell–ECM adhesion in the tumor microenvironment (21). The decreased galectin-3 binding to the ECM is attributable, at least in part, to binding sites occupancy by other galectins and altered stromal glycosylation. Although there are substantial differences in the type of glycans recognized by galectins-1 and 3 they often compete for the same ligands (83). Galectin-3 affinity for the T antigen, for instance, is two times higher than that of galectin-1 (84). These differences might account at least in part for their divergent biological functions. Galectin-1 is scattered throughout the tumor stroma and could thus be leading to galectin-3-binding site occupancy (21). The decrease in galectin-3-binding sites is also most likely due to differential ECM glycosylation in malignant CMT. Sialylation is a prevalent type of glycosylation in tumour cells but not in the ECM of malignant CMT (85). However, in normal adjacent mammary tissue, both ECA and PNA bind to matrix glycoproteins in tissue stroma and gland mucus secretion in addition to the apical border of luminal cells while in the ECM of malignant CMT, there is a striking decrease in ECA- and PNA-binding sites pointing to an overall decrease in galactosylation (21). Collagen glycosylation is known to affect tumor cell adhesion to and spreading on collagen IV (86). In normal mammary glands, the collagen-galactosylating enzyme, GLT25D1 (87), is expressed with slightly 186 In Galectins and Disease Implications for Targeted Therapeutics; Klyosov, A., et al.; ACS Symposium Series; American Chemical Society: Washington, DC, 2012.

higher levels than in benign CMT. However, the enzyme expression levels decrease considerably in malignant CMT (21). Galectin-3 knockdown causes a decrease in GLT25D1 mRNA levels in vitro. This suggests that downregulation of galectin- 3 may be at least partially responsible for the decreased GLT25D1 mRNA expression levels found in spontaneously occurring malignant CMT (21).

Downloaded by NORTH CAROLINA STATE UNIV on December 19, 2012 | http://pubs.acs.org Publication Date (Web): December 18, 2012 | doi: 10.1021/bk-2012-1115.ch011

Concluding Remarks Circulating tumour cells (88), micrometastases (89) and well-established metastases (90) may coexist in the patient. In these, changes in the expression of galectins, glycans and the glycoproteins which carry them are at least in part microenvironment related. Galectin-3 has been shown to play a crucial role in cancer drug resistance [reviewed in (91)]. Inhibition of galectin-3/T antigen interaction was shown to reduce experimental metastatic disease (92). However, other galectins seem to be able to step up and perform its functions. Novel potential specific galectin inhibitors and galectin-targeted therapy are increasingly being studied (93–95) but there is an increasing need to broaden their galectin-specter of action and take into account the relevance of the glycosylation status of their ligands in other to achieve therapeutic efficacy.

References 1.

2.

3.

4.

5.

6.

7.

Restucci, B.; De Vico, G.; Maiolino, P. Expression of beta 1 integrin in normal, dysplastic and neoplastic canine mammary gland. J. Comp .Pathol. 1995, 113 (2), 165–173. Gama, A.; Paredes, J.; Milanezi, M. F.; Reis-Filho, J. S.; Gärtner, F.; Schmitt, F. C. P-cadherin expression in canine lactating mammary gland. J. Cell, Biochem. 2002, 86 (3), 420–421. Reis, A. L.; Carvalheira, J.; Schmitt, F. C.; Gärtner, F. Immunohistochemical study of the expression of E-cadherin in canine mammary tumours. Vet. Rec. 2003, 152 (20), 621–624. Gama, A.; Paredes, J.; Albergaria, A.; Gärtner, F.; Schmitt, F. P-cadherin expression in canine mammary tissues. J. Comp. Pathol. 2004, 130 (1), 13–20. Sarli, G.; Preziosi, R.; De Tolla, L.; Brunetti, B.; Benazzi, C. E-cadherin immunoreactivity in canine mammary tumors. J. Vet. Diagnostic Invest. 2004, 16, 542–547. Matos, A. J.; Lopes, C.; Carvalheira, J.; Santos, M.; Rutteman, G. R.; Gärtner, F. E-cadherin expression in canine malignant mammary tumours: Relationship to other clinico-pathological variables. J. Comp. Pathol. 2006, 134 (2−3), 182–189. Nowak, M.; Madej, J. A.; Podhorska-Okolow, M.; Dziegiel, P. Expression of extracellular matrix metalloproteinase (MMP-9), E-cadherin and proliferation-associated antigen Ki-67 and their reciprocal correlation in canine mammary adenocarcinomas. In vivo 2008, 22, 463–470. 187 In Galectins and Disease Implications for Targeted Therapeutics; Klyosov, A., et al.; ACS Symposium Series; American Chemical Society: Washington, DC, 2012.

8.

9.

Downloaded by NORTH CAROLINA STATE UNIV on December 19, 2012 | http://pubs.acs.org Publication Date (Web): December 18, 2012 | doi: 10.1021/bk-2012-1115.ch011

10.

11.

12.

13.

14.

15. 16.

17.

18.

19.

20.

Pereira, P. D.; Lopes, C. C.; Matos, A. J.; Cortez, P. P.; Gärtner, F.; Medeiros, R.; Lopes, C. Caveolin-1 in diagnosis and prognosis of canine mammary tumours: Comparison of evaluation systems. J. Comp. Pathol. 2010, 143 (2−3), 87–93. Santos, A.; Lopes, C.; Frias, C.; Amorim, I.; Vicente, C.; Gärtner, F.; Matos, A. Immunohistochemical evaluation of MMP-2 and TIMP-2 in canine mammary tumours: A survival study. Vet. J. 2011, 190 (3), 396–402. Geraldes, M.; Gärtner, F.; Schmitt, F. Immunohistochemical study of hormonal receptors and cell proliferation in normal canine mammary glands and spontaneous mammary tumours. Vet. Rec. 2000, 146 (14), 403–406. Santos, A. A.; Oliveira, J. T.; Lopes, C. C.; Amorim, I. F.; Vicente, C. M.; Gärtner, F. R.; Matos, A. J. Immunohistochemical expression of vascular endothelial growth factor in canine mammary tumours. J. Comp. Pathol. 2010, 143 (4), 268–275. Gama, A.; Gärtner, F.; Alves, A.; Schmitt, F. Immunohistochemical expression of Epidermal Growth Factor Receptor (EGFR) in canine mammary tissues. Res. Vet. Sci. 2009, 87 (3), 432–437. de Oliveira, J. T.; Pinho, S. S.; de Matos, A. J.; Hespanhol, V.; Reis, C. A.; Gärtner, F. MUC1 expression in canine malignant mammary tumours and relationship to clinicopathological features. Vet. J. 2009, 182 (3), 491–493. Tavares, C.; de Oliveira, J. T.; Lopes, C.; Carvalheira, J.; de Matos, A. J.; Rutteman, G. R.; Reis, CA; Gärtner, F. Mucin 6 and Tn antigen expression in canine mammary tumours: Correlation with pathological features. J. Comp. Pathol. 2012, Epub ahead of print; DOI: 10.1016/j.jcpa.2012.03.007. Nangia-Makker, P.; Balan, V.; Raz, A. Regulation of tumor progression by extracellular galectin-3. Cancer Microenviron. 2008, 1, 43–51. Fry, S. A.; Van den Steen, P. E.; Royle, L.; Wormald, M. R.; Leathem, A. J.; Opdenakker, G.; McDonnell, J. M.; Dwek, R. A.; Rudd, P. M. Cancerassociated glycoforms of gelatinase B exhibit a decreased level of binding to galectin-3. Biochemistry 2006, 45 (51), 15249–15258. Yu, L. G.; Andrews, N.; Zhao, Q.; McKean, D.; Williams, J. F.; Connor, L. J.; Gerasimenko, O. V.; Hilkens, J.; Hirabayashi, J.; Kasai, K.; Rhodes, J. M. Galectin-3 interaction with Thomsen-Friedenreich disaccharide on cancerassociated MUC1 causes increased cancer cell endothelial adhesion. J. Biol. Chem. 2007, 282 (1), 773–781. Goetz, J. G.; Joshi, B.; Lajoie, P.; Strugnell, S. S.; Scudamore, T.; Kojic, L. D.; Nabi, I. R. Concerted regulation of focal adhesion dynamics by galectin-3 and tyrosine-phosphorylated caveolin-1. J. Cell Biol. 2008, 180 (6), 1261–1275. Zhuo, Y.; Chammas, R.; Bellis, S. L. Sialylation of beta1 integrins blocks cell adhesion to galectin-3 and protects cells against galectin-3-induced apoptosis. J. Biol. Chem. 2008, 283, 22177–22185. Markowska, A. I.; Jefferies, K. C.; Panjwani, N. Galectin-3 protein modulates cell surface expression and activation of vascular endothelial growth factor receptor 2 in human endothelial cells. J. Biol. Chem. 2011, 286 (34), 29913–29921. 188 In Galectins and Disease Implications for Targeted Therapeutics; Klyosov, A., et al.; ACS Symposium Series; American Chemical Society: Washington, DC, 2012.

Downloaded by NORTH CAROLINA STATE UNIV on December 19, 2012 | http://pubs.acs.org Publication Date (Web): December 18, 2012 | doi: 10.1021/bk-2012-1115.ch011

21. de Oliveira, J. T.; de Matos, A. J.; Gomes, J.; Vilanova, M.; Hespanhol, V.; Manninen, A.; Rutteman, G.; Chammas, R.; Gärtner, F.; Bernardes, E. S. Coordinated expression of galectin-3 and galectin-3-binding sites in malignant mammary tumors: Implications for tumor metastasis. Glycobiology 2010, 20 (11), 1341–1352. 22. Brown, E. R.; Doig, T.; Anderson, N.; Brenn, T.; Doherty, V.; Xu, Y.; Bartlett, J. M.; Smyth, J. F.; Melton, D. W. Association of galectin-3 expression with melanoma progression and prognosis. Eur. J. Cancer 2011, 48 (6), 865–874. 23. Buljan, M.; Šitum, M.; Tomas, D.; Milošević, M.; Krušlin, B. Prognostic value of galectin-3 in primary cutaneous melanoma. J. Eur. Acad. Dermatol. Venereol. 2010, 25 (10), 1174–1181. 24. Canesin, G.; Gonzalez-Peramato, P.; Palou, J.; Urrutia, M.; CordónCardo, C.; Sánchez-Carbayo, M. Galectin-3 expression is associated with bladder cancer progression and clinical outcome. Tumour Biol. 2010, 31 (4), 277–285. 25. Ege, C. B.; Akbulut, M.; Zekioğlu, O.; Ozdemir, N. Investigation of galectin3 and heparanase in endometrioid and serous carcinomas of the endometrium and correlation with known predictors of survival. Arch. Gynecol. Obstet. 2011, 284 (5), 1231–1239. 26. Froehlich, R.; Hambruch, N.; Haeger, J. D.; Dilly, M.; Kaltner, H.; Gabius, H. J.; Pfarrer, C. Galectin fingerprinting detects differences in expression profiles between bovine endometrium and placentomes as well as early and late gestational stages. Placenta 2012, 33 (3), 195–201. 27. Gál, P.; Vasilenko, T.; Kostelníková, M.; Jakubco, J.; Kovác, I.; Sabol, F.; André, S.; Kaltner, H.; Gabius, H. J.; Smetana, K., Jr Open wound healing in vivo: Monitoring binding and presence of adhesion/growth-regulatory galectins in rat skin during the course of complete re-epithelialization. Acta Histochem. Cytochem. 2011, 44 (5), 191–199. 28. Kopitz, J.; Ballikaya, S.; André, S.; Gabius, H. J. Ganglioside GM1/ galectin-dependent growth regulation in human neuroblastoma cells: Special properties of bivalent galectin-4 and significance of linker length for ligand selection. Neurochem. Res. 2012, 37 (6), 1267–1276. 29. Suzuki, O.; Hirsch, B.; Abe, M.; Dürkop, H.; Stein, H. Galectin-1-mediated cell death is increased by CD30-induced signaling in anaplastic large cell lymphoma cells but not in Hodgkin lymphoma cells. Lab Invest. 2012, 92 (2), 191–199, DOI:10.1038/labinvest.2011.151. 30. Mazurek, N.; Byrd, J. C.; Sun, Y.; Hafley, M.; Ramirez, K.; Burks, J.; Bresalier, R. S. Cell-surface galectin-3 confers resistance to TRAIL by impeding trafficking of death receptors in metastatic colon adenocarcinoma cells. Cell Death Differ. 2012, 19 (3), 523–33, DOI:10.1038/cdd.2011.123. 31. Barrow, H.; Guo, X.; Wandall, H. H.; Pedersen, J. W.; Fu, B.; Zhao, Q.; Chen, C.; Rhodes, J. M.; Yu, L. G. Serum galectin-2, -4, and -8 are greatly increased in colon and breast cancer patients and promote cancer cell adhesion to blood vascular endothelium. Clin. Cancer Res. 2011, 17 (22), 7035–7046. 189 In Galectins and Disease Implications for Targeted Therapeutics; Klyosov, A., et al.; ACS Symposium Series; American Chemical Society: Washington, DC, 2012.

Downloaded by NORTH CAROLINA STATE UNIV on December 19, 2012 | http://pubs.acs.org Publication Date (Web): December 18, 2012 | doi: 10.1021/bk-2012-1115.ch011

32. Iurisci, I.; Tinari, N.; Natoli, C.; Angelucci, D.; Cianchetti, E.; Iacobelli, S. Concentrations of galectin-3 in the sera of normal controls and cancer patients. Clin. Cancer Res. 2000, 6 (4), 1389–1393. 33. Califice, S.; Castronovo, V.; Bracke, M.; van den Brûle, F. Dual activities of galectin-3 in human prostate cancer: Tumor suppression of nuclear galectin-3 vs tumor promotion of cytoplasmic galectin-3. Oncogene 2004, 23, 7527–7536. 34. Glinsky, V. V.; Raz, A. Modified citrus pectin anti-metastatic properties: One bullet, multiple targets. Carbohydr. Res. 2009, 344 (14), 1788–1791. 35. Cheong, T. C.; Shin, J. Y.; Chun, K. H. Silencing of galectin-3 changes the gene expression and augments the sensitivity of gastric cancer cells to chemotherapeutic agents. Cancer Sci. 2010, 101 (1), 94–102. 36. Greijer, A. E.; van der Groep, P.; Kemming, D.; Shvarts, A.; Semenza, G. L.; Meijer, G. A.; van de Wiel, MA; Belien, J. A.; van Diest, P. J.; van der Wall, E. Up-regulation of gene expression by hypoxia is mediated predominantly by hypoxia-inducible factor 1 (HIF-1). J. Pathol. 2005, 206 (3), 291–304. 37. Bockhorn, M.; Jain, R. K.; Munn, L. L. Active versus passive mechanisms in metastasis: Do cancer cells crawl into vessels, or are they pushed? Lancet Oncol. 2007, 8, 444–448. 38. Wang, X.; Schneider, A. HIF-2alpha-mediated activation of the epidermal growth factor receptor potentiates head and neck cancer cell migration in response to hypoxia. Carcinogenesis 2010, 31, 1202–1210. 39. de Oliveira, J. T.; de Matos, A. J.; Santos, A. L.; Pinto, R.; Gomes, J.; Hespanhol, V.; Chammas, R.; Manninen, A.; Bernardes, E. S.; Albuquerque Reis, C.; Rutteman, G.; Gärtner, F. Sialylation regulates galectin-3/ligand interplay during mammary tumour progression: A case of targeted uncloaking. Int. J. Dev. Biol. 2011, 55 (7−9), 823–834. 40. Soares, R.; Guo, S.; Gärtner, F.; Schmitt, F. C.; Russo, J. 17 beta-estradiolmediated vessel assembly and stabilization in tumor angiogenesis requires TGF beta and EGFR crosstalk. Angiogenesis 2003, 6 (4), 271–281. 41. Betka, J.; Plzák, J.; Smetana, K., Jr; Gabius, H. J. Galectin-3, an endogenous lectin, as a tool for monitoring cell differentiation in head and neck carcinomas with implications for lectin-glycan functionality. Acta Otolaryngol. 2003, 123 (2), 261–263. 42. Sethi, S.; Macoska, J.; Chen, W.; Sarkar, F. H. Molecular signature of epithelial-mesenchymal transition (EMT) in human prostate cancer bone metastasis. Am. J. Transl. Res. 2010, 3 (1), 90–99. 43. Ding, W.; You, H.; Dang, H.; LeBlanc, F.; Galicia, V.; Lu, S. C.; Stiles, B.; Rountree, C. B. Epithelial-to-mesenchymal transition of murine liver tumor cells promotes invasion. Hepatology 2010, 52 (3), 945–953. 44. Mackinnon, A. C.; Gibbons, M. A.; Farnworth, S. L.; Leffler, H.; Nilsson, U. J.; Delaine, T.; Simpson, A. J.; Forbes, S. J.; Hirani, N.; Gauldie, J; Sethi, T. Regulation of transforming growth factor-β1-driven lung fibrosis by galectin3. Am. J. Respir. Crit. Care Med. 2012, 185 (5), 537–546. 45. de Matos, A. J.; Lopes, C. C.; Faustino, A. M.; Carvalheira, J. G.; Rutteman, G. R.; Gärtner, F. E-cadherin, beta-catenin, invasion and lymph 190 In Galectins and Disease Implications for Targeted Therapeutics; Klyosov, A., et al.; ACS Symposium Series; American Chemical Society: Washington, DC, 2012.

46.

Downloaded by NORTH CAROLINA STATE UNIV on December 19, 2012 | http://pubs.acs.org Publication Date (Web): December 18, 2012 | doi: 10.1021/bk-2012-1115.ch011

47.

48. 49.

50.

51.

52. 53.

54.

55.

56.

57.

58.

node metastases in canine malignant mammary tumours. APMIS 2007, 115 (4), 327–334. Gama, A.; Paredes, J.; Gärtner, F.; Alves, A.; Schmitt, F. Expression of E-cadherin, P-cadherin and beta-catenin in canine malignant mammary tumours in relation to clinicopathological parameters, proliferation and survival. Vet. J. 2008, 177 (1), 45–53. Gärtner, F.; Geraldes, M.; Cassali, G.; Rema, A.; Schmitt, F. DNA measurement and immunohistochemical characterization of epithelial and mesenchymal cells in canine mixed mammary tumours: Putative evidence for a common histogenesis. Vet. J. 1999, 158 (1), 39–47. Cardiff, R. D. The pathology of EMT in mouse mammary tumorigenesis. J. Mammary Gland Biol. Neoplasia 2010, 15 (2), 225–233. Perl, A. K.; Wilgenbus, P.; Dahl, U.; Semb, H.; Christofori, G. A causal role for E-cadherin in the transition from adenoma to carcinoma. Nature 1998, 392, 190–193. Pinho, S. S.; Matos, A. J.; Lopes, C.; Marcos, N. T.; Carvalheira, J.; Reis, C. A.; Gärtner, F. Sialyl Lewis x expression in canine malignant mammary tumours: Correlation with clinicopathological features and E-cadherin expression. BMC Cancer 2007, 7, 124. Pinho, S. S.; Reis, C. A.; Gärtner, F.; Alpaugh, M. L. Molecular plasticity of E-cadherin and sialyl lewis x expression, in two comparative models of mammary tumorigenesis. PLoS One 2009, 4 (8), e6636. Chambers, A. F.; Groom, A. C.; MacDonald, I. C. Dissemination and growth of cancer cells in metastatic sites. Nat. Rev .Cancer 2002, 2, 563–572. Reis, C. A.; David, L.; Carvalho, F.; Mandel, U.; de Bolós, C.; Mirgorodskaya, E.; Clausen, H.; Sobrinho-Simões, M. Immunohistochemical study of the expression of MUC6 mucin and co-expression of other secreted mucins (MUC5AC and MUC2) in human gastric carcinomas. J. Histochem. Cytochem. 2000, 48 (3), 377–388. Carvalho, F.; David, L.; Aubert, J. P.; López-Ferrer, A.; De Bolós, C.; Reis, C. A.; Gärtner, F.; Peixoto, A.; Alves, P.; Sobrinho-Simões, M. Mucins and mucin-associated carbohydrate antigens expression in gastric carcinoma cell lines. Virchows Arch. 1999, 435 (5), 479–485. Pinto, R.; Carvalho, A. S.; Conze, T.; Magalhães, A.; Picco, G.; Burchell, J. M.; Taylor-Papadimitriou, J.; Reis, C. A.; Almeida, R.; Mandel, U.; Clausen, H.; Söderberg, O.; David, L. Identification of new cancer biomarkers based on aberrant mucin glycoforms by in situ proximity ligation. J. Cell. Mol. Med. 2011, DOI:10.1111/j.1582-4934.2011.01436.x. Reis, C. A.; David, L.; Seixas, M.; Burchell, J.; Sobrinho-Simões, M. Expression of fully and under-glycosylated forms of MUC1 mucin in gastric carcinoma. Int. J. Cancer 1998, 79 (4), 402–410. Santos-Silva, F.; Fonseca, A.; Caffrey, T.; Carvalho, F.; Mesquita, P.; Reis, C.; Almeida, R.; David, L.; Hollingsworth, M. A. ThomsenFriedenreich antigen expression in gastric carcinomas is associated with MUC1 mucin VNTR polymorphism. Glycobiology 2005, 15 (5), 511–517. Ligtenberg, M. J.; Buijs, F.; Vos, H. L.; Hilkens, J. Suppression of cellular aggregation by high levels of episialin. Cancer Res 1992, 52 (8), 2318–2324. 191 In Galectins and Disease Implications for Targeted Therapeutics; Klyosov, A., et al.; ACS Symposium Series; American Chemical Society: Washington, DC, 2012.

Downloaded by NORTH CAROLINA STATE UNIV on December 19, 2012 | http://pubs.acs.org Publication Date (Web): December 18, 2012 | doi: 10.1021/bk-2012-1115.ch011

59. Wesseling, J.; van der Valk, S. W.; Hilkens, J A mechanism for inhibition of E-cadherin-mediated cell-cell adhesion by the membrane-associated mucin episialin/MUC1. Mol. Biol. Cell 1996, 7 (4), 565–577. 60. Wesseling, J.; van der Valk, S. W.; Vos, H. L.; Sonnenberg, A.; Hilkens, J. Episialin (MUC1) overexpression inhibits integrin-mediated cell adhesion to extracellular matrix components. J .Cell. Biol. 1995, 129 (1), 255–265. 61. Kondo, K.; Kohno, N.; Yokoyama, A.; Hiwada, K. Decreased MUC1 expression induces E-cadherin-mediated cell adhesion of breast cancer cell lines. Cancer Res. 1998, 58 (9), 2014–2019. 62. Satoh, S.; Hinoda, Y.; Hayashi, T.; Burdick, M. D.; Imai, K.; Hollingsworth, M. A. Enhancement of metastatic properties of pancreatic cancer cells by MUC1 gene encoding an anti-adhesion molecule. Int. J. Cancer 2000, 88 (4), 507–518. 63. Inohara, H.; Raz, A. Functional evidence that cell surface galectin-3 mediates homotypic cell adhesion. Cancer Res. 1995, 55 (15), 3267–3271. 64. Saeland, E.; Belo, A. I.; Mongera, S.; van Die, I.; Meijer, G. A.; van Kooyk, Y. Differential glycosylation of MUC1 and CEACAM5 between normal mucosa and tumour tissue of colon cancer patients. Int. J .Cancer 2011, 131 (1), 117–128, DOI:10.1002/ijc.26354. 65. Stowell, S. R.; Arthur, C. M.; Mehta, P.; Slanina, K. A.; Blixt, O.; Leffler, H.; Smith, D. F.; Cummings, R. D. Galectin-1, -2, and -3 exhibit differential recognition of sialylated glycans and blood group antigens. J. Biol. Chem. 2008, 283 (15), 10109–10123. 66. Zhao, Q.; Guo, X.; Nash, G. B.; Stone, P. C.; Hilkens, J.; Rhodes, J. M.; Yu, L. G. Circulating galectin-3 promotes metastasis by modifying MUC1 localization on cancer cell surface. Cancer Res. 2009, 69, 6799–6806. 67. Antonopoulos, A.; Demotte, N.; Stroobant, V.; Haslam, S. M.; van der Bruggen, P.; Dell, A. Loss of effector function of human cytolytic T lymphocytes is accompanied by major alterations in N- and O-glycosylation. J. Biol. Chem. 2012, 287 (14), 11240–11251. 68. Brockhausen, I.; Yang, J.; Dickinson, N.; Ogata, S.; Itzkowitz, S. H. Enzymatic basis for sialyl-Tn expression in human colon cancer cells. Glycoconjugate J. 1998, 15 (6), 595–603. 69. Ju, T.; Lanneau, G. S.; Gautam, T.; Wang, Y.; Xia, B.; Stowell, S. R.; Willard, M. T.; Wang, W.; Xia, J. Y.; Zuna, R. E.; Laszik, Z.; Benbrook, D. M.; Hanigan, M. H.; Cummings, R. D. Human tumor antigens Tn and sialyl Tn arise from mutations in Cosmc. Cancer Res. 2008, 68 (6), 1636–1646. 70. Marcos, N. T.; Pinho, S.; Grandela, C.; Cruz, A.; Samyn-Petit, B.; Harduin-Lepers, A.; Almeida, R.; Silva, F.; Morais, V.; Costa, J; Kihlberg, J.; Clausen, H.; Reis, C. A. Role of the human ST6GalNAc-I and ST6GalNAc-II in the synthesis of the cancer-associated sialyl-Tn antigen. Cancer Res. 2004, 64 (19), 7050–7057. 71. Marcos, N. T.; Bennett, E. P.; Gomes, J.; Magalhaes, A.; Gomes, C.; David, L.; Dar, I.; Jeanneau, C.; DeFrees, S.; Krustrup, D.; Vogel, L. K.; Kure, E. H.; Burchell, J.; Taylor-Papadimitriou, J.; Clausen, H.; Mandel, U.; Reis, C. A. ST6GalNAc-I controls expression of sialyl-Tn antigen in gastrointestinal tissues. Front. Biosci. (Elite Ed) 2011, 3, 1443–1455. 192 In Galectins and Disease Implications for Targeted Therapeutics; Klyosov, A., et al.; ACS Symposium Series; American Chemical Society: Washington, DC, 2012.

Downloaded by NORTH CAROLINA STATE UNIV on December 19, 2012 | http://pubs.acs.org Publication Date (Web): December 18, 2012 | doi: 10.1021/bk-2012-1115.ch011

72. Burdick, M. D.; Harris, A.; Reid, C. J.; Iwamura, T.; Hollingsworth, M. A. Oligosaccharides expressed on MUC1 produced by pancreatic and colon tumor cell lines. J. Biol. Chem. 1997, 272 (39), 24198–24202. 73. Sewell, R.; Bäckström, M.; Dalziel, M.; Gschmeissner, S.; Karlsson, H.; Noll, T.; Gätgens, J.; Clausen, H.; Hansson, G. C.; Burchell, J.; Taylor-Papadimitriou, J. The ST6GalNAc-I sialyltransferase localizes throughout the Golgi and is responsible for the synthesis of the tumor-associated sialyl-Tn O-glycan in human breast cancer. J. Biol. Chem. 2006, 281 (6), 3586–3594. 74. Brockhausen, I.; Yang, J.; Lehotay, M.; Ogata, S.; Itzkowitz, S. Pathways of mucin O-glycosylation in normal and malignant rat colonic epithelial cells reveal a mechanism for cancer-associated Sialyl-Tn antigen expression. Biol. Chem. 2001, 382 (2), 219–232. 75. Mattila, P. E.; Youker, R. T.; Mo, D.; Bruns, J. R.; Cresawn, K. O.; Hughey, R. P.; Ihrke, G.; Weisz, O. A. Multiple biosynthetic trafficking routes for apically secreted proteins in MDCK cells. Traffic 2012, 13 (3), 433–442, DOI:10.1111/j.1600-0854.2011.01315.x. 76. Kinlough, C. L.; Poland, P. A.; Gendler, S. J.; Mattila, P. E.; Mo, D.; Weisz, O. A.; Hughey, R. P. Core-glycosylated mucin-like repeats from MUC1 are an apical targeting signal. J. Biol. Chem. 2011, 286 (45), 39072–39081. 77. Kufe, D. W. Mucins in cancer: Function, prognosis and therapy. Nat. Rev. Cancer 2009, 9, 874–885. 78. Ramasamy, S.; Duraisamy, S.; Barbashov, S.; Kawano, T.; Kharbanda, S.; Kufe, D. The MUC1 and galectin-3 oncoproteins function in a microRNAdependent regulatory loop. Mol. Cell. 2007, 27, 992–1004. 79. Gomes, J.; Marcos, N. T.; Berois, N.; Osinaga, E.; Magalhães, A.; Pinto-deSousa, J.; Almeida, R.; Gärtner, F.; Reis, C. A. Expression of UDP-N-acetylD-galactosamine: Polypeptide N-acetylgalactosaminyltransferase-6 in gastric mucosa, intestinal metaplasia, and gastric carcinoma. J. Histochem. Cytochem. 2009, 57 (1), 79–86. 80. Nangia-Makker, P.; Honjo, Y.; Sarvis, R.; Akahani, S.; Hogan, V.; Pienta, K. J.; Raz, A. Galectin-3 induces endothelial cell morphogenesis and angiogenesis. Am. J. Pathol. 2000, 156 (3), 899–909. 81. Pinho, S. S.; Reis, C. A.; Paredes, J.; Magalhães, A. M.; Ferreira, A. C.; Figueiredo, J.; Xiaogang, W.; Carneiro, F.; Gärtner, F.; Seruca, R. The role of N-acetylglucosaminyltransferase III and V in the post-transcriptional modifications of E-cadherin. Hum. Mol. Genet. 2009, 18 (14), 2599–2608. 82. Pinho, S. S.; Oliveira, P.; Cabral, J.; Carvalho, S.; Huntsman, D.; Gärtner, F.; Seruca, R.; Reis, C. A.; Oliveira, C. Loss and recovery of Mgat3 and GnT-III mediated E-cadherin N-glycosylation is a mechanism involved in epithelialmesenchymal-epithelial transitions. PLoS One 2012, 7 (3), e33191. 83. Song, X.; Xia, B.; Stowell, S. R.; Lasanajak, Y.; Smith, D. F.; Cummings, R. D. Novel fluorescent glycan microarray strategy reveals ligands for galectins. Chem. Biol. 2009, 16 (1), 36–47. 84. Bian, C. F.; Zhang, Y.; Sun, H.; Li, D. F.; Wang, D. C. Structural basis for distinct binding properties of the human galectins to Thomsen-Friedenreich antigen. PLoS One 2011, 6 (9), e25007. 193 In Galectins and Disease Implications for Targeted Therapeutics; Klyosov, A., et al.; ACS Symposium Series; American Chemical Society: Washington, DC, 2012.

Downloaded by NORTH CAROLINA STATE UNIV on December 19, 2012 | http://pubs.acs.org Publication Date (Web): December 18, 2012 | doi: 10.1021/bk-2012-1115.ch011

85. Pinho, S. S.; Osório, H.; Nita-Lazar, M.; Gomes, J.; Lopes, C.; Gärtner, F.; Reis, C. A. Role of E-cadherin N-glycosylation profile in a mammary tumour model. Biochem. Biophys. Res. Commun. 2009, 379, 1091–1096. 86. Lauer-Fields, J. L.; Malkar, N. B.; Richet, G.; Drauz, K.; Fields, G. B. Melanoma cell CD44 interaction with the alpha 1(IV)1263-1277 region from basement membrane collagen is modulated by ligand glycosylation. J. Biol. Chem. 2003, 278 (16), 14321–14330. 87. Schegg, B.; Hülsmeier, A. J.; Rutschmann, C.; Maag, C.; Hennet, T. Core glycosylation of collagen is initiated by two beta (1-O) galactosyltransferases. Mol. Cell. Biol. 2009, 29, 943–952. 88. da Costa, A.; Oliveira, J. T.; Gärtner, F.; Kohn, B.; Gruber, A. D.; Klopfleisch, R. Potential markers for detection of circulating canine mammary tumor cells in the peripheral blood. Vet. J. 2011, 190 (1), 165–168. 89. Matos, A. J.; Faustino, A. M.; Lopes, C.; Rutteman, G. R.; Gärtner, F. Detection of lymph node micrometastases in malignant mammary tumours in dogs by cytokeratin immunostaining. Vet. Rec. 2006, 158 (18), 626–30. 90. Misdorp, W. Tumours of the Mammary Gland. In Tumours in Domestic Animals, 4th ed.; Meuten, D. J., Ed.; Iowa State Press: Ames, IA, 2002; pp 575−606. 91. Turner, J. G.; Dawson, J; Sullivan, D. M. Nuclear export of proteins and drug resistance in cancer. Biochem. Pharmacol. 2012, 83 (8), 1021–1032. 92. Glinskii, O. V.; Sud, S.; Mossine, V. V.; Mawhinney, T. P.; Anthony, D. C.; Glinsky, G. V.; Pienta, K. J.; Glinsky, V. V. Inhibition of Prostate cancer bone metastasis by synthetic TF antigen mimic/galectin-3 inhibitor lactulosel-leucine. Neoplasia 2012, 14 (1), 65–73. 93. Collins, P. M.; Oberg, C. T.; Leffler, H.; Nilsson, U. J.; Blanchard, H. Taloside inhibitors of galectin-1 and galectin-3. Chem. Biol. Drug Des. 2012, 79 (3), 339–346, DOI:10.1111/j.1747-0285.2011.01283.x. 94. Yang, Y.; Zhou, Z.; He, S.; Fan, T.; Jin, Y.; Zhu, X.; Chen, C.; Zhang, Z. R.; Huang, Y. Treatment of prostate carcinoma with (galectin-3)-targeted HPMA copolymer-(G3-C12)-5-Fluorouracil conjugates. Biomaterials 2012, 33 (7), 2260–2271. 95. Kopansky, E.; Shamay, Y.; David, A. Peptide-directed HPMA copolymerdoxorubicin conjugates as targeted therapeutics for colorectal cancer. J. Drug Target 2011, 19 (10), 933–943.

194 In Galectins and Disease Implications for Targeted Therapeutics; Klyosov, A., et al.; ACS Symposium Series; American Chemical Society: Washington, DC, 2012.